[go: up one dir, main page]

WO2025185733A1 - Combination therapy comprising a pd-1/pd-l1 and vegf/vegfr binding agent and a chemotherapy for cancer treatment - Google Patents

Combination therapy comprising a pd-1/pd-l1 and vegf/vegfr binding agent and a chemotherapy for cancer treatment

Info

Publication number
WO2025185733A1
WO2025185733A1 PCT/CN2025/081278 CN2025081278W WO2025185733A1 WO 2025185733 A1 WO2025185733 A1 WO 2025185733A1 CN 2025081278 W CN2025081278 W CN 2025081278W WO 2025185733 A1 WO2025185733 A1 WO 2025185733A1
Authority
WO
WIPO (PCT)
Prior art keywords
agent
use according
cancer
antibody
chemotherapy agent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2025/081278
Other languages
French (fr)
Other versions
WO2025185733A8 (en
Inventor
Guoqiang Hu
Jing Liu
Juan Zhang
Yankui OU
Jie Dong
Ugur Sahin
Na Li
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biotheus Inc
Biontech SE
Original Assignee
Biotheus Inc
Biontech SE
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/CN2024/080506 external-priority patent/WO2025184858A1/en
Priority claimed from ARP240102655A external-priority patent/AR134001A1/en
Priority claimed from PCT/CN2024/124994 external-priority patent/WO2025185161A1/en
Application filed by Biotheus Inc, Biontech SE filed Critical Biotheus Inc
Publication of WO2025185733A1 publication Critical patent/WO2025185733A1/en
Publication of WO2025185733A8 publication Critical patent/WO2025185733A8/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against B7 molecules, e.g. CD80, CD86
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/31Immunoglobulins specific features characterized by aspects of specificity or valency multispecific
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/569Single domain, e.g. dAb, sdAb, VHH, VNAR or nanobody®

Definitions

  • the invention inter alia relates to methods for treating cancer in a subject using a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction (which can, e.g., be a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) ) in combination with a chemotherapy agent, optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ⁇ 1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
  • a binding agent comprising (i) a first binding region which
  • the invention further concerns a method for determining whether a cancer in a subject is susceptible to treatment with the above-mentioned binding agent and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject the PD-L1 expression before the treatment by CPS or another integrating scoring algorithm, wherein a CPS of ⁇ 1 indicates that the subject is susceptible to the treatment, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
  • ICIs immune checkpoint inhibitors
  • PD-1 programmed cell death protein 1
  • PD-L1 programmed death-ligand 1
  • Predictive biomarkers such as scoring PD-L1 expression in cancer tissue, are therefore sometimes used to evaluate the likelihood to respond to ICI treatment alone or in combination with chemotherapy, thereby defining treatment-eligible patient groups (Ulas, Ezgi B et al. “Predictive Value of Combined Positive Score and Tumour Proportion Score for Immunotherapy Response in Advanced NSCLC. ” JTO clinical and research reports vol. 4, 9 100532.25 May. 2023, doi: 10.1016/j. jtocrr. 2023.100532) .
  • Cancer types are scored for PD-L1 expression for example by using the combined positive score (CPS) , which covers the PD-L1 expression on both the tumour cells and the immune cells in the tumour microenvironment.
  • CPS combined positive score
  • Studies have explored the relationship between the expression of programmed death ligand 1 (PD-L1) and prognosis in cancer, and high PD-L1 expression determined by immunohistochemistry is suggested to be a predictive biomarker of response to immunotherapy in several cancers (Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N Engl J Med. 2022; 387 (3) : 217-226.
  • PD-L1 pathway alone or in combination with chemotherapy leads to unsatisfactory responses especially in subjects having a low PD-L1 expression on cancer cells.
  • the approved anti-PD-L1 antibody pembrolizumab plus chemotherapy regime is currently considered the standard of care (SOC) in the first-line treatment of patients with metastatic TNBC whose tumours express PD-L1 with a CPS score of ⁇ 10; and clinically significant improvements have been observed in progression-free survival (PFS) (9.7 months for pembrolizumab plus chemotherapy versus 5.6 months for chemotherapy alone) and overall survival (OS) (23.0 months for pembrolizumab plus chemotherapy versus 16.1 months for chemotherapy alone) .
  • PFS progression-free survival
  • OS overall survival
  • these benefits have not been observed in patients with a CPS of ⁇ 10.
  • the SOC remains chemotherapy alone, with a median PFS of only about 6 months and a median OS of about 16 months (Cortes et al. 2022) .
  • the invention provides a method of treating cancer in a subject, the method comprising administering to the subject: a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent.
  • the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
  • the invention further provides a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating cancer, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent.
  • the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
  • the invention further provides a chemotherapy agent for use in a method of treating cancer, the method comprising administering to the subject a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
  • the invention further provides a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating cancer in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent.
  • the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
  • the invention provides a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy, preferably a chemotherapy agent; optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • PD-L1 programmed death-ligand 1
  • VEGF Vascular Endothelial Growth Factor
  • the present application provides in an embodiment a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy, preferably a chemotherapy agent.
  • PD-L1 programmed death-ligand 1
  • VEGF Vascular Endothelial Growth Factor
  • the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • CPS combined positive score
  • the invention concerns in an embodiment a method of treating cancer in a subject, the method comprising administering to the subject a bispecific antibody that specifically binds to PD-L1 and VEGF in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • a bispecific antibody that specifically binds to PD-L1 and VEGF in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • CPS combined positive score
  • the present application provides in an embodiment a method of treating cancer in a subject, the method comprising administering to the subject a bispecific antibody that specifically binds to PD-L1 and VEGF in combination with chemotherapy. It can be preferred that the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • CPS combined positive score
  • the invention also concerns in an embodiment a chemotherapy agent for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. a bispecific antibody that specifically binds to PD-L1 and VEGF; and b. the chemotherapy agent; optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • CPS combined positive score
  • a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent.
  • a bispecific antibody that specifically binds to PD-L1 and VEGF in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy agent.
  • a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent.
  • a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. a bispecific antibody that specifically binds to PD-L1 and VEGF; and b. the chemotherapy agent.
  • the invention is concerned with a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoi
  • the invention is further concerned with a composition
  • a composition comprising a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a chemotherapy agent selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  • a platinum-based chemotherapy agent preferably carboplatin, cisplatin or oxaliplatin
  • an antimetabolite chemotherapy agent preferably 5-fluor
  • the composition is a pharmaceutical composition.
  • kits comprising a. a composition comprising a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a composition comprising a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which
  • composition comprising a chemotherapy agent selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  • a chemotherapy agent selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or o
  • each composition of the kit is a pharmaceutical composition.
  • the composition comprising a chemotherapy agent may be more than one composition, namely if the chemotherapy agent is a combination of chemotherapy agents and each chemotherapy agent of the combination may be provided in a separate composition.
  • the invention also provides in an embodiment a kit of parts comprising the bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy agent.
  • a kit of the invention typically includes instructions.
  • the invention also concerns a method for determining whether a cancer in a subject is susceptible to treatment with a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject the PD-L1 expression before the treatment by determining the combined positive scope (CPS) or another score, preferably the tumour area positivity (TAP) score or the tumour proportion score (TPS) , wherein a CPS of ⁇ 1 indicates a PD-L1 expression that renders the subject susceptible to treatment with the bispecific antibody and the chemotherapy, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined
  • the invention also concerns in an embodiment a method for determining whether a cancer in a subject is susceptible to treatment with a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy, wherein the method comprises detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score, wherein the CPS of ⁇ 1 indicates that the subject is susceptible to treatment with the bispecific antibody and the chemotherapy.
  • CPS combined positive score
  • FIG. 1 depicts boxplots of AUC tau, ss by 20 mg/kg Q2W (A) versus 1200 mg Q2W (B) or 1500 mg Q2W (C) dosing regimens.
  • N is the number of participants; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of AUC tau, ss , the box represents the inter-quartile distance, and the whiskers represent ⁇ 1.5 times the inter-quartile range (75th -25th quartile) .
  • the number at the top of the figure represents the number of participants included in each category.
  • Fig. 2 (A) depicts boxplots of AUC tau, ss for 30 mg/kg Q3W versus 2100 mg Q3W dosing regimens.
  • N is the number of simulated patients in the different body weight ranges as indicated; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of AUC tau, ss , the box represents the inter-quartile distance, and the whiskers represent ⁇ 1.5 times the inter-quartile range (75th -25th quartile) .
  • Fig. 2 (B) depicts boxplots of c max, ss for 30 mg/kg Q3W versus 2100 mg Q3W dosing regimens.
  • N is the number of simulated patients in the different body weight ranges as indicated; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of c max, ss , the box represents the inter-quartile distance, and the whiskers represent ⁇ 1.5 times the inter-quartile range (75th -25th quartile) .
  • Fig. 2 (C) depicts boxplots of c min, ss for 30 mg/kg Q3W versus 2100 mg Q3W dosing regimens.
  • N is the number of simulated patients in the different body weight ranges as indicated; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of c min, ss , the box represents the inter-quartile distance, and the whiskers represent ⁇ 1.5 times the inter-quartile range (75th -25th quartile) .
  • a chemotherapy preferably a chemotherapy agent
  • the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score; or optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ⁇ 1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
  • CPS combined positive score
  • TPS tumour area positivity
  • TPS tumour proportion score
  • the PD-L1 staining cells are tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
  • the PD-L1 staining cells are tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining. 6.
  • the bispecific antibody for use according to any of the preceding embodiments wherein the PD- L1 expression score has been determined using a TAP scoring algorithm in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer sample; and/or; (ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated stroma containing tumour-associated immune cells; and/or (iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or (iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive tumour cells and immune cells; and/or (v) %PD-L1 positive tumour cells and immune cells /tumour area means the
  • the PD- L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer tissue sample; and/or; (ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or (iii) viable tumour cells are determined by staining with a viability dye; and/or (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining. 10.
  • the stain comprises a hematoxylin and eosin (H&E) stain, preferably wherein the viability dye is propidium iodide.
  • H&E hematoxylin and eosin
  • TAP tumour area positivity
  • TPS tumour proportion score
  • each cycle has up to 28 days, preferably 28 or 21 days.
  • 25. The bispecific antibody for use according to any of the preceding embodiments, wherein the subject has not been previously treated for cancer.
  • 26. The bispecific antibody for use according to any of embodiments 1-24, wherein the subject has been previously treated for cancer, preferably wherein the subject had at least one previous chemotherapy treatment.
  • 27. The bispecific antibody for use according to any of the preceding embodiments, wherein the bispecific antibody is administered every 6 weeks, preferably every 4 weeks, more preferably every 3 weeks or every 2 weeks. 28.
  • the bispecific antibody for use according to any of the preceding embodiments wherein the chemotherapy is administered once or more within the first 20 days or the first 21 days of each cycle, wherein the chemotherapy is administered twice or more within the first 20 days or the first 21 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 20 days or the first 21 days of each cycle.
  • the chemotherapy is administered once or more within the first 14 days or the first 15 days of each cycle, wherein the chemotherapy is administered twice or more within the first 14 days or the first 15 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 14 days or the first 15 days of each cycle.
  • the bispecific antibody for use according to any of the preceding embodiments wherein median progression-free survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  • the cancer comprises one or more solid tumours.
  • the bispecific antibody for use according to any of the preceding embodiments wherein the cancer is selected from the group consisting of melanoma, lung, liver, stomach, renal cell, urothelial, cervical, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancers, preferably wherein the cancer is selected from urothelial, breast and esophagus cancer.
  • the cancer is small cell lung cancer (SCLC) , non-small cell lung cancer (NSCLC) or triple-negative breast cancer (TNBC) , preferably advanced triple-negative breast cancer. 38.
  • SCLC small cell lung cancer
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • bispecific antibody for use according to any of the preceding embodiments, wherein the bispecific antibody comprises a Fab, Fab’, F (ab’) 2, Fd, Fv, dAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH. 44.
  • the bispecific antibody for use according to any of the preceding embodiments, wherein the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1 or 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO:2 or 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3 or 38. 46.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity-determining region 3
  • the bispecific antibody for use according to embodiment 45 wherein the amino acid sequence of the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 9. 47.
  • a heavy chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and wherein a light chain variable region of the anti-VEGF antibody comprises: (i) a complementarity- determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
  • the bispecific antibody for use according to embodiment 50 wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 11; or wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 11. 52.
  • the bispecific antibody for use according to any of the preceding embodiments wherein the amino acid sequence of the heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17. 53.
  • the chemotherapy comprises a chemotherapy agent selected from lurbinectedin, topotecan, taxane such as paclitaxel, docetaxel, or nanoparticle albumin-bound paclitaxel (nab-paclitaxel) , pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof. 56.
  • the method of treatment comprises administering the bispecific antibody in combination with paclitaxel to a subject having small cell lung cancer. 58.
  • the bispecific antibody for use according to any of embodiments 1-55, wherein the method of treatment comprises administering the bispecific antibody in combination with irinotecan, 5-fluorouracil, calcium folinate to a subject having unresectable neuroendocrine neoplasm.
  • the method of treatment comprises administering the bispecific antibody in combination with etoposide and platinum to a subject having small cell lung cancer, preferably as first line treatment and/or extensive-stage small cell lung cancer, most preferably as first line treatment of extensive-stage small cell lung cancer or as second-line treatment of limited-stage small cell lung cancer.
  • the bispecific antibody for use according to any of embodiments 1-55 wherein the method of treatment comprises administering the bispecific antibody every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg or 30 mg/kg being particularly preferred) in combination with paclitaxel, lurbinectedin, or topotecan to a subject having small cell lung cancer, preferably as second line treatment, and/or extensive-stage small cell lung cancer, more preferably wherein the method of treatment comprises administering to a patient suffering from advanced small cell lung cancer as second line treatment the bispecific antibody every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with (i) paclitaxel every 3 weeks at a dosage of 175 mg/m 2 , optionally for 5 cycles, or (ii) topotecan at a dosage of 1.5 mg/m 2 administered IV or 2.3 mg/m 2 administered orally once per day on days 1 to 5 of every 3 weeks cycle.
  • the bispecific antibody for use according to any of embodiments 1-55, wherein the method of treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 10 mg/kg to 20 mg/kg in combination with nab-paclitaxel, paclitaxel, or gemcitabine with carboplatin to a subject having triple-negative breast cancer. 66.
  • the bispecific antibody for use according to any of embodiments 1-55 wherein the method of treatment comprises administered the bispecific antibody every 2 weeks at a dosage of 1000 mg to 2000 mg to a subject having triple-negative breast cancer, preferably on the 1 st and 15 th day of a 28-day treatment cycle, preferably wherein the bispecific antibody is administered in combination with a chemotherapy, preferably on the 1 st , 8 th , and 15 th day of a 28-day treatment cycle. 67.
  • a method of treating cancer, preferably triple-negative breast cancer (TNBC) in a subject comprising administering to the subject a bispecific antibody that specifically bind to PD-L1 and VEGF in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1, preferably from 1 to 20, preferably from 1 to ⁇ 10, or other integrating scoring algorithm defining a similar PD-L1 expression score; or optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ⁇ 1, preferably from 1 to 20, preferably from 1 to ⁇ 10, when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
  • CPS combined positive score
  • TAP tumour area positivity
  • the method is a method for extending progression-free survival in said subject compared to the chemotherapy or an anti-PD-L1 antibody or an anti-PD-1 antibody or the bispecific antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab. 69.
  • the method is a method for increased overall survival in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab. 70.
  • a method for determining whether a cancer in a subject is susceptible to treatment with a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy comprises detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score, wherein the CPS of ⁇ 1 indicates that the subject is susceptible to treatment with the bispecific antibody and the chemotherapy; or wherein the method comprises detecting in a sample of the subject a PD-L1 expression before the treatment by determining the combined positive scope (CPS) or another score, preferably the tumour area positivity (TAP) score or the tumour proportion score (TPS) , wherein a CPS of ⁇ 1 indicates a PD-L1 expression that renders the subject susceptible to treatment with the bispecific antibody and the chemotherapy, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm
  • 71 The method according to embodiment 70, wherein the cancer is selected from the group consisting of melanoma, lung, liver, stomach, renal cell, urothelial, cervical, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancers, preferably wherein the cancer is selected from urothelial, breast and esophagus cancer.
  • 72. The method according to embodiment 70 or 71, wherein the sample is a cancer tissue sample.
  • the bispecific antibody comprises an anti-PD-L1 antibody or fragment thereof.
  • the bispecific antibody comprises an anti-VEGF antibody or fragment thereof.
  • the bispecific antibody comprises a Fab, Fab', F (ab') 2, Fd, Fv, sdAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH.
  • the bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably each VHH is fused to the C-terminus of the anti-VEGF antibody.
  • the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region
  • the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1 or 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 2 or 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3 or 38.
  • the amino acid sequence of the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9. 79.
  • the anti-VEGF antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably, the constant region is selected from the constant region of human IgGl, IgG2, IgG3 or IgG4. 80.
  • the anti-VEGF antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO:13 or having at least 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 13.
  • the bispecific antibody specifically binds to VEGF-A.
  • a heavy chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and a light chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
  • the method comprises the step of determining the CPS in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer tissue sample; and/or (ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour infiltrating) lymphocytes and macrophages; and/or (iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody 22C3) ; and/or (iv) viable tumour cells are determined by staining with a viability dye; and/or (v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour
  • PD-L1 staining cells are tumour cells with partial or complete linear membrane staining that is distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining. 87.
  • the method comprises the step of determining the TAP score in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer sample; and/or; (ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated stroma containing tumour-associated immune cells; and/or (iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or (iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive tumour cells and immune cells; and/or (v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 positive tumour
  • the method comprises the step of determining the TPS in a test sample of the subject by determining the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer tissue sample; and/or; (ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or (iii) viable tumour cells are determined by staining with a viability dye; and/or (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining. 89.
  • the cancer tissue sample is a tissue section of a tumour biopsy, preferably wherein the tissue section is a formalin fixed and embedded in paraffin wax (FFPE) tissue section.
  • FFPE paraffin wax
  • IHC immunohistochemistry
  • the tissue section is stained.
  • the stain comprises a hematoxylin and eosin (H&E) stain, preferably wherein the viability dye is propidium iodide.
  • a chemotherapy agent for use in a method of treating a subject with cancer comprising administering to the subject: a. a bispecific antibody that specifically binds to PD-L1 and VEGF; and b. the chemotherapy agent; optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score; or optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ⁇ 1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
  • CPS combined positive score
  • TPS tumour area positivity
  • TPS tumour proportion score
  • the other integrating scoring algorithms are selected from TAP and TPS; or wherein the subject has a PD-L1 expression before the treatment that provides a score of ⁇ 1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , preferably when determined by the tumour area positivity (TAP) score.
  • CPS combined positive score
  • TPS tumour proportion score
  • the PD-L1 staining cells are tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
  • the viable tumour cells and the number of lymphocytes and macrophages are counted in the tumour nests and the adjacent supporting stroma of the tumour tissue sample.
  • the chemotherapy agent for use according to embodiment 103 wherein the tumour area is determined using a hematoxylin and eosin (H&E) stain.
  • H&E hematoxylin and eosin
  • 105 The chemotherapy agent for use according to embodiment 103 or 104, wherein the PD-L1 positive tumour cells and immune cells are PD-L1 staining cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and immune cells (lymphocytes and macrophages) within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
  • 106 hematoxylin and eosin
  • the chemotherapy agent for use according to any of embodiments 98-105 wherein the PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer tissue sample; and/or; (ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or (iii) viable tumour cells are determined by staining with a viability dye; and/or (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining. 107.
  • IHC immunohistochemistry
  • FFPE paraffin wax
  • the chemotherapy agent for use according to embodiment 110, wherein the stain comprises a hematoxylin and eosin (H&E) stain, preferably wherein the viability dye is propidium iodide. 112.
  • TAP tumour area positivity
  • TPS tumour proportion score
  • the chemotherapy agent for use according to any of embodiments 98-116, wherein the bispecific antibody dosage is in a range of from 0.1 mg/kg to 45 mg/kg body weight, preferably 1 mg/kg to 30 mg/kg body weight.
  • the chemotherapy agent for use according to any of embodiments 98-117, wherein the bispecific antibody and/or the chemotherapy is administered intravenously, preferably wherein the bispecific antibody and the chemotherapy are administered intravenously.
  • the chemotherapy agent for use according to any of embodiments 98-118, wherein the bispecific antibody and/or the chemotherapy is administered via an IV injection or IV infusion. 120.
  • the chemotherapy agent for use according to any of embodiments 98-127 wherein the chemotherapy is administered once or more within the first 14 days or the first 15 days of each cycle, wherein the chemotherapy is administered twice or more within the first 14 days or the first 15 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 14 days or the first 15 days of each cycle.
  • the chemotherapy agent for use according to any of embodiments 98-129 wherein overall survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  • the chemotherapy agent for use according to any of embodiments 98-130 wherein median progression-free survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  • the chemotherapy agent for use according to any of embodiments 98-132 wherein the cancer is selected from the group consisting of melanoma, lung, liver, stomach, renal cell, urothelial, cervical, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancers, preferably wherein the cancer is selected from urothelial, breast and esophagus cancer.
  • SCLC small cell lung cancer
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • the bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably wherein each VHH is fused to the C-terminus of the anti-VEGF antibody. 142.
  • the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region
  • the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1 or 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO:2 or 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3 or 38.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity-determining region 3
  • the chemotherapy agent for use according to embodiment 142 wherein the amino acid sequence of the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 9.
  • a heavy chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and a light chain variable region of the anti-VEGF antibody comprises: (i) a complementarity- determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
  • the chemotherapy agent for use according to any of embodiments 98-148 wherein the amino acid sequence of the heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17.
  • the chemotherapy agent is selected from lurbinectedin, topotecan, taxane such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) , or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof. 153.
  • the chemotherapy agent for use according to any of embodiments 98-154, wherein the method of treatment comprises administering the bispecific antibody in combination with nab-paclitaxel to a subject having triple-negative breast cancer.
  • the method of treatment comprises administering the bispecific antibody in combination with oxaliplatin, calcium folinate, and 5-fluorouracil to a subject having hepatocellular carcinoma, preferably wherein the method of treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg being particularly preferred) .
  • the chemotherapy agent for use according to any of embodiments 98-154, wherein the method of treatment comprises administering the bispecific antibody in combination with irinotecan, 5-fluorouracil, calcium folinate to a subject having unresectable neuroendocrine neoplasm.
  • the chemotherapy agent for use according to any of embodiments 98-154, wherein the method of treatment comprises administering the bispecific antibody in combination with etoposide and platinum to a subject having small cell lung cancer, preferably as first line treatment and/or extensive-stage small cell lung cancer, most preferably as first line treatment of extensive-stage small cell lung cancer or as second-line treatment of limited-stage small cell lung cancer.
  • the chemotherapy agent for use according to any of embodiments 98-154 wherein the method of treatment comprises administering the bispecific antibody every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg or 30 mg/kg being particularly preferred) in combination with paclitaxel, lurbinectedin, or topotecan to a subject having small cell lung cancer, preferably as second line treatment and/or extensive-stage small cell lung cancer, more preferably wherein the method of treatment comprises administering to a patient suffering from advanced small cell lung cancer as second line treatment the bispecific antibody every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with (i) paclitaxel every 3 weeks at a dosage of 175 mg/m 2 , optionally for 5 cycles, or (ii) topotecan at a dosage of 1.5 mg/m 2 administered IV or 2.3 mg/m 2 administered orally once per day on days 1 to 5 of every 3 weeks cycle.
  • the chemotherapy agent for use according to any of embodiments 98-154, wherein the method of treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 10 mg/kg to 20 mg/kg in combination with nab-paclitaxel, paclitaxel, or gemcitabine with carboplatin to a subject having triple-negative breast cancer. 163.
  • the chemotherapy agent for use according to any of embodiments 98-154 wherein the method of treatment comprises administered the bispecific antibody every 2 weeks at a dosage of 1000 mg to 2000 mg to a subject having triple-negative breast cancer, preferably on the 1 st and 15 th day of a 28-day treatment cycle and preferably wherein the bispecific antibody is administered in combination with a chemotherapy, preferably on the 1 st , 8 th , and 15 th day of a 28-day treatment cycle.
  • adjuvant relates to a compound which prolongs, enhances or accelerates an immune response.
  • adjuvants comprise a heterogeneous group of compounds such as oil emulsions (e.g., Freund’s adjuvants) , mineral compounds (such as alum) , bacterial products (such as Bordetella pertussis toxin) , or immune-stimulating complexes.
  • adjuvants include, without limitation, LPS, GP96, CpG oligodeoxynucleotides, growth factors, and cytokines, such as monokines, lymphokines, interleukins, chemokines.
  • the chemokines may be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, INFa, INF- ⁇ , GM-CSF, LT-a.
  • Further known adjuvants are aluminum hydroxide, Freund’s adjuvant or oil such as ISA51.
  • Suitable adjuvants for use in the present disclosure include lipopeptides, such as Pam3Cys, as well as lipophilic components, such as saponins, trehalose-6, 6-dibehenate (TDB) , monophosphoryl lipid-A (MPL) , monomycoloyl glycerol (MMG) , or glucopyranosyl lipid adjuvant (GLA) .
  • lipopeptides such as Pam3Cys
  • lipophilic components such as saponins, trehalose-6, 6-dibehenate (TDB) , monophosphoryl lipid-A (MPL) , monomycoloyl glycerol (MMG) , or glucopyranosyl lipid adjuvant (GLA) .
  • Antibodies are glycoproteins belonging to the immunoglobulin superfamily.
  • the term "full-length antibody” may refer to an immunoglobulin molecule that binds to a target molecule and contains four peptide chains: two heavy chains and two light chains which are connected to each other through disulfide bonds.
  • An antibody may recognise an antigen via the fragment antigen-binding (Fab) variable region.
  • the fragment crystallizable region (Fc region) is the tail region of an antibody that may allow antibodies to activate the immune system.
  • the hinge region is a stretch of heavy chains linking the Fab and Fc regions.
  • the heavy chain and light chain may each comprise a variable region and one or more constant domains.
  • a heavy chain comprises a variable region (VH) and three constant domains (CH1, CH2, and CH3) and a light chain comprises a variable region (VL) and one constant domain (CL) .
  • VH variable region
  • CH1, CH2, and CH3 constant domains
  • VL variable region
  • CL constant domain
  • examples antibodies include a human antibody, a humanized antibody, a chimeric antibody, a multispecific antibody, a monoclonal antibody, and a polyclonal antibody.
  • antibody fragment may refer to a fragment of an antibody, or a genetically engineered product of one of more fragments of an antibody, which fragment is involved in binding with the target molecule.
  • antibody fragments include an antigen-binding fragment (Fab) , a Fab', a Fab'-SH, a fragment antibody (F (ab’) 2) , a variable region (Fv) , a single chain antibody (scFv) , a single-domain antibody (sdAb) , and a camelid antibody (VHH) .
  • Fab antigen-binding fragment
  • Fab fragment of an antibody that binds to antigens and is composed of one constant and one variable region of each of the heavy and the light chain.
  • fragment antibody or “F (ab’) 2” refers to a region of an antibody that remains following digestion of the Fc region while leaving intact some of the hinge region.
  • Fab refers to a fragment formed by the reduction of a F (ab') 2 fragment.
  • Fab Fab’-SH refers to a Fab’ fragment with a free sulfhydryl group.
  • binding agent in the context of the present disclosure refers to any agent capable of binding to desired antigens.
  • the binding agent is an antibody, antibody fragment, or construct thereof.
  • the binding agent may also comprise synthetic, modified or non-naturally occurring moieties, in particular non-peptide moieties. Such moieties may, for example, link desired antigen-binding functionalities or regions such as antibodies or antibody fragments.
  • the binding agent is a synthetic construct comprising antigen-binding CDRs or variable regions.
  • binding in the context of the binding of an antibody to a predetermined antigen or epitope typically is a binding with an affinity corresponding to a K D of about 10 -7 M or less, such as about 10 -8 M or less, such as about 10 -9 M or less, about 10 -10 M or less, or about 10 -11 M or even less, when determined, for instance, using Bio-Layer Interferometry (BLI) or using surface plasmon resonance (SPR) technology in a BIAcore 3000 instrument using the antigen as the ligand and the antibody as the analyte.
  • BLI Bio-Layer Interferometry
  • SPR surface plasmon resonance
  • the antibody binds to the predetermined antigen with an affinity corresponding to a K D that is at least ten-fold lower, such as at least 100-fold lower, for instance at least 1,000-fold lower, such as at least 10,000-fold lower, for instance at least 100,000-fold lower than its K D for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen.
  • a non-specific antigen e.g., BSA, casein
  • the amount with which the affinity is higher is dependent on the K D of the antibody, so that when the K D of the antibody is very low (that is, the antibody is highly specific) , then the degree to which the affinity for the antigen is lower than the affinity for a non-specific antigen may be at least 10,000-fold.
  • k d (sec -1 )
  • Said value is also referred to as the k off value.
  • K D (M)
  • M dissociation equilibrium constant of a particular antibody-antigen interaction.
  • a binding region as defined herein means that the binding region is antagonistic towards the PD-1/PD-L1 signaling pathway.
  • such binding region which antagonizes the PD-1/PD-L1 interaction totally or partially inhibits, reduces, interferes with or negatively modulates the PD-1/PD-L1 signaling pathway by binding to PD-1, PD-L1, or both.
  • the binding region which antagonizes the PD-1/PD-L1 interaction mediates its antagonistic activity in a manner that totally inhibits (i.e., blocks) or partially reduces the binding of PD-L1 to PD-1.
  • a binding region as defined herein (such as an antibody or a fragment thereof as defined herein) "antagonizes the VEGF/VEGFR interaction" means that the binding region is antagonistic towards the VEGF/VEGFR signaling pathway.
  • binding region which antagonizes the VEGF/VEGFR interaction totally or partially inhibits, reduces, interferes with or negatively modulates the VEGF/VEGFR signaling pathway by binding to VEGF, VEGFR, or both.
  • the binding region which antagonizes the VEGF/VEGFR interaction mediates its antagonistic activity in a manner that totally inhibits (i.e., blocks) or partially reduces the binding of VEGF to VEGFR.
  • cancer disease refers to or describes the physiological condition in an individual that is typically characterized by unregulated cell growth and typically a disease characterized by aberrantly regulated cellular growth, proliferation, differentiation, adhesion, and/or migration.
  • cancer cell is meant an abnormal cell that grows by a rapid, uncontrolled cellular proliferation and continues to grow after the stimuli that initiated the new growth cease.
  • cancer metastases is meant the spread of cancer cells from its original site to another part of the body.
  • metastasis is a very complex process and depends on detachment of malignant cells from the primary tumor, invasion of the extracellular matrix, penetration of the endothelial basement membranes to enter the body cavity and vessels, and then, after being transported by the blood, infiltration of target organs. Finally, the growth of a new tumor, i.e. a secondary tumor or metastatic tumor, at the target site depends on angiogenesis. Tumor metastasis often occurs even after the removal of the primary tumor because tumor cells or components may remain and develop metastatic potential.
  • the term "metastasis" according to the present disclosure relates to "distant metastasis" which relates to a metastasis which is remote from the primary tumor and the regional lymph node system.
  • carrier refers to a component which may be natural, synthetic, organic, inorganic in which the active component is combined in order to facilitate, enhance or enable administration of the pharmaceutical composition.
  • a carrier as used herein may be one or more compatible solid or liquid fillers, diluents or encapsulating substances, which are suitable for administration to subject. Suitable carriers include, without limitation, sterile water, Ringer, Ringer lactate, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy-propylene copolymers.
  • the pharmaceutical composition of the present disclosure includes isotonic saline.
  • Pharmaceutically acceptable carriers, excipients or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington’s Pharmaceutical Sciences, Mack Publishing Co. (A. R Gennaro edit. 1985) .
  • Pharmaceutical carriers, excipients or diluents can be selected with regard to the intended route of administration and standard pharmaceutical practice.
  • CDR complementarity determining region (s) in an immunoglobulin variable region.
  • the variable regions of the heavy and light chains each contain three CDRs, designated CDR1, CDR2 and CDR3.
  • CDR1, CDR2 and CDR3 The precise boundaries of these CDRs can be defined according to various numbering systems known in the art (see e.g. Dondelinger, M., et al., 2018. Frontiers in immunology, 9, p. 2278) , such as the Kabat numbering system (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed.
  • IMGT/3Dstructure-DB and IMGT/DomainGapAlign a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF [J] . Nucleic acids research, 2009; 38 (suppl_1) : D301-D307) .
  • CDRs defined by each numbering system. Also, the correspondence between different numbering systems is well known to those skilled in the art (for example, see Lefranc et al., Dev. Comparat. Immunol. 27: 55-77, 2003) .
  • chemotherapeutic agent or “chemotherapeutical agent” or “chemotherapy agent” can be used interchangeably herein.
  • a chemotherapeutic agent is a chemical compound useful in the treatment of cancer.
  • Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytotoxic/antitumour antibiotics, topoisomerase inhibitors, photosensitizers, anti-estrogens and selective estrogen receptor modulators (SERMs) , anti-progesterones, estrogen receptor down-regulators (ERDs) , estrogen receptor antagonists, leutinizing hormone-releasing hormone agonists, anti-androgens, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, anti-sense oligonucleotides that that inhibit expression of genes implicated in abnormal cell proliferation or tumour growth.
  • Chemotherapeutic agents useful in the treatment methods of the present invention include
  • Chimeric antibody refers to an antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain (s) is identical with or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
  • a particular species e.g., human
  • another species e.g., mouse
  • the term “comprising” is used in the context of the present disclosure to indicate that further members may optionally be present in addition to the members of the list introduced by “comprising” . It is, however, contemplated as specific embodiments of the present invention that each time the term “comprising” is used, this shall also encompass the possibility of no further members being present, i.e., for the purpose of this embodiment “comprising” can be understood as having the meaning of “consisting of” .
  • Combination therapy refers to the bispecific antibody disclosed herein that specifically binds to PD-L1 and VEGF in combination with the chemotherapy disclosed herein for use in a method of treating a subject with cancer, optionally wherein the subject has been determined to have a combined positive score (CPS) of ⁇ 1 before the treatment.
  • CPS combined positive score
  • Each component of the combination therapy, i.e., the bispecific antibody and the chemotherapy may be administered separately.
  • the “combined positive score” or “CPS, ” refers to a well-known algorithm for determining a PD-L1 expression score from a tumour sample of a subject (see for example Kulangara, Karina & Hanks, Debra & Waldroup, Stephanie & Peltz, Lindsay & Shah, Supriya & Roach, Charlotte & Juco, Jonathan & Emancipator, Kenneth & Stanforth, Dave. (2017) , Development of the combined positive score (CPS) for the evaluation of PD-L1 in solid tumours with the immunohistochemistry assay PD-L1 IHC 22C3 pharmDx. Journal of Clinical Oncology) .
  • the PD-L1 expression score is determined by taken into account the number of PD-L1 staining cells (i.e., tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer tissue sample; and/or (ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour infiltrating) lymphocytes and macrophages; and/or (iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody 22C3) ; and/or (iv) viable tumour cells are determined by staining with a viability dye; and/or (v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 staining lymphocytes and
  • the CPS has been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula: wherein (i) the test sample is a cancer tissue sample; (ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour infiltrating) lymphocytes and macrophages; (iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody 22C3) ; (iv) viable tumour cells are determined by staining with a viability dye; and (v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 staining lymphocytes and PD-L1 staining macrophages.
  • the CPS is approved as companion diagnostic for the treatment of cancer using pembrolizumab.
  • the term “disease control rate” or DCR is the percentage of all subjects in a study or treatment group who have either a complete response, a partial response, or a stable disease to the treatment (CR, PR or SD) .
  • the DCR can be calculated by adding the numbers of subjects having CR, the numbers of subjects having PR, and the numbers of subjects having SD and dividing the resulting sum by the total number of subjects in the treatment group.
  • the DCR eval i.e., the DCR of all evaluable subjects in a study or treatment group, is the percentage of all evaluable subjects in a study or treatment group who have either a complete response, a partial response, or a stable disease to the treatment (CR, PR or SD) .
  • Duration of response only applies to subjects whose confirmed best overall response is CR or PR and is defined as the time from the first documentation of objective tumor response (CR or PR) to the date of first PD or death due to underlying cancer.
  • diluent relates to a diluting and/or thinning agent.
  • the term “diluent” includes any one or more of fluid, liquid or solid suspension and/or mixing media. Examples of suitable diluents include ethanol, glycerol, and water.
  • DNA relates to a nucleic acid molecule which is entirely or at least substantially composed of deoxyribonucleotide residues. In preferred embodiments, the DNA contains all or a majority of deoxyribonucleotide residues.
  • deoxyribonucleotide refers to a nucleotide which lacks a hydroxyl group at the 2’-position of a ⁇ -D-ribofuranosyl group.
  • DNA encompasses without limitation, double stranded DNA, single stranded DNA, isolated DNA such as partially purified DNA, essentially pure DNA, synthetic DNA, recombinantly produced DNA, as well as modified DNA that differs from naturally occurring DNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations may refer to addition of non-nucleotide material to internal DNA nucleotides or to the end (s) of DNA. It is also contemplated herein that nucleotides in DNA may be non-standard nucleotides, such as chemically synthesized nucleotides or ribonucleotides. For the present disclosure, these altered DNAs are considered analogs of naturally-occurring DNA.
  • a molecule contains “a majority of deoxyribonucleotide residues” if the content of deoxyribonucleotide residues in the molecule is more than 50% (such as at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%) , based on the total number of nucleotide residues in the molecule.
  • DNA may be recombinant DNA and may be obtained by cloning of a nucleic acid, in particular cDNA.
  • the cDNA may be obtained by reverse transcription of RNA.
  • the term “effective amount” refers to an amount of a given substance that is sufficient in quantity to produce a desired effect, including an improvement or remediation of the disease, disorder, or symptoms of the disease or condition.
  • the combination therapy described herein is to be administered to a patient in need therefore in an effective amount.
  • RNA molecule can encode an RNA molecule (e.g., by a transcription process that includes a DNA-dependent RNA polymerase enzyme) .
  • RNA molecule can encode a polypeptide (e.g., by a translation process) .
  • a gene, a cDNA, or a single-stranded RNA encodes a polypeptide if transcription and translation of mRNA corresponding to that gene produces the polypeptide in a cell or other biological system.
  • a coding region of a single-stranded RNA encoding a target polypeptide agent refers to a coding strand, the nucleotide sequence of which is identical to the mRNA sequence of such a target polypeptide agent.
  • a coding region of a single-stranded RNA encoding a target polypeptide agent refers to a non-coding strand of such a target polypeptide agent, which may be used as a template for transcription of a gene or cDNA.
  • the phrase “nucleic acid encoding a peptide or protein” means that the polynucleotide, if present in the appropriate environment, for example within a cell and/or in a cell-free translation system, can direct the assembly of amino acids to produce the peptide or protein via a process of translation.
  • epitopes refers to the part of an antigen that as used herein, refers to an agent that elicits an immune response; and/or (ii) an agent that binds to a T cell receptor (e.g., when presented by an MHC molecule) or to an antibody.
  • epitopes are the discrete, three-dimensional sites on an antigen, which are recognized by the immune system.
  • Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
  • excipient refers to a substance which may be present in a pharmaceutical composition of the present disclosure but is not an active ingredient.
  • excipients include without limitation, carriers, binders, diluents, lubricants, thickeners, surface active agents, preservatives, stabilizers, emulsifiers, buffers, flavoring agents, or colorants.
  • a gene refers to a DNA sequence in a chromosome that codes for a protein.
  • a gene includes coding sequence (i.e., sequence that encodes a particular protein) ; in some embodiments, a gene includes non-coding sequence.
  • a gene may include both coding (e.g., exonic) and non-coding (e.g., intronic) sequences.
  • a gene may include one or more regulatory elements that, for example, may control or impact one or more aspects of gene expression (e.g., cell-type-specific expression, inducible expression, etc. ) .
  • Human antibody refers to an antibody that comprises human immunoglobulin protein sequences only.
  • a human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell.
  • mouse antibody or rat antibody refer to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
  • Humanized antibody refers to forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin.
  • the humanized antibody will comprise substantially all of at least one, and typically two, variable regions, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
  • an immune cell means any cell of hematopoietic lineage involved in regulating an immune response against an antigen (e.g., a bacterial or viral infection or an auto-antigen) .
  • an immune cell is a leukocyte, such as a white blood cell.
  • Immune cells include neutrophils, eosinophils, basophils, lymphocytes, and/or monocytes. Lymphocytes include T lymphocytes and B lymphocytes. Immune cells can also be dendritic cells, natural killer (NK) cells, and/or a mast cell.
  • immunoglobulin relates to proteins of the immunoglobulin superfamily, preferably to antigen receptors such as antibodies or the B cell receptor (BCR) .
  • the immunoglobulins are characterized by a structural domain, i.e., the immunoglobulin domain, having a characteristic immunoglobulin (Ig) fold.
  • the term encompasses membrane bound immunoglobulins as well as soluble immunoglobulins.
  • Membrane bound immunoglobulins are also termed surface immunoglobulins or membrane immunoglobulins, which are generally part of the BCR. Soluble immunoglobulins are generally termed antibodies.
  • the structure of immunoglobulins has been well characterized. See, e.g., Fundamental Immunology Ch.
  • immunoglobulins generally comprise several chains, typically two identical heavy chains and two identical light chains which are linked via disulfide bonds. These chains are primarily composed of immunoglobulin domains or regions, such as the V L or VL (variable light chain) domain/region, C L or CL (constant light chain) domain/region, V H or VH (variable heavy chain) domain/region, and the C H or CH (constant heavy chain) domains/regions C H 1 (CH1) , C H 2 (CH2) , C H 3 (CH3) , and C H 4 (CH4) .
  • V L or VL variable light chain domain/region
  • C L or CL constant light chain domain/region
  • V H or VH variable heavy chain domain/region
  • C H or CH constant heavy chain domains/regions C H 1 (CH1) , C H 2 (CH2) , C H 3 (CH3) , and C H 4 (CH4) .
  • the heavy chain constant region typically is comprised of three domains, CH1, CH2, and CH3.
  • the hinge region is the region between the CH1 and CH2 domains of the heavy chain and is highly flexible. Disulfide bonds in the hinge region are part of the interactions between two heavy chains in an IgG molecule.
  • Each light chain typically is comprised of a VL and a CL.
  • the light chain constant region typically is comprised of one domain, CL.
  • the VH and VL regions may be further subdivided into regions of hypervariability (or hypervariable regions which may be hypervariable in sequence and/or form of structurally defined loops) , also termed complementarity determining regions (CDRs) , interspersed with regions that are more conserved, termed framework regions (FRs) .
  • CDRs complementarity determining regions
  • Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 (see also Chothia and Lesk, J. Mol. Biol. 196, 901-917 (1987) ) .
  • an "instructional material” or “instructions” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the compositions and methods described herein.
  • the instructional material of the kit of the disclosure may, for example, be affixed to a container which contains the compositions of the disclosure or be shipped together with a container which contains the compositions. Alternatively, the instructional material may be shipped separately from the container with the intention that the instructional material and the compositions be used cooperatively by the recipient.
  • Kabat as used herein, means an immunoglobulin alignment and numbering system pioneered by Elvin a. Kabat ( (1991) Sequences of Proteins of Immunological Interest, 5 th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. ) or Kabat et al., 1992, Sequences of Proteins of Immunological Interest, DIANE Publishing: 2719.
  • kit as used herein is synonymous with the term “kit of part” or “kit of parts” and refers to a plurality of compositions that may, e.g., be shipped or stored together, in particular because they are relevant for or required for an administration regimen as disclosed herein.
  • MIC Mononuclear inflammatory cells
  • the term "monoclonal antibody” refers to an antibody obtained from a substantially homogeneous population of antibodies. The individual antibodies composing the population may be identical except for possible naturally occurring mutations, which may be present in minor amounts. Monoclonal antibodies are highly specific and target a single antigenic epitope. In contrast, polyclonal antibody preparations typically include a large number of antibodies which are specific for different epitopes.
  • nucleoside relates to compounds which can be thought of as nucleotides without a phosphate group. While a nucleoside is a nucleobase linked to a sugar (e.g., ribose or deoxyribose) , a nucleotide is composed of a nucleoside and one or more phosphate groups. Examples of nucleosides include cytidine, uridine, pseudouridine, adenosine, and guanosine. The five standard nucleosides which usually make up naturally occurring nucleic acids are uridine, adenosine, thymidine, cytidine and guanosine.
  • thymidine is more commonly written as “dT” ( “d” represents “deoxy” ) as it contains a 2’-deoxyribofuranose moiety rather than the ribofuranose ring found in uridine. This is because thymidine is found in deoxyribonucleic acid (DNA) and not ribonucleic acid (RNA) . Conversely, uridine is found in RNA and not DNA. The remaining three nucleosides may be found in both RNA and DNA.
  • RNA In RNA, they would be represented as A, C and G, whereas in DNA they would be represented as dA, dC and dG.
  • a modified purine (A or G) or pyrimidine (C, T, or U) base moiety is, in some embodiments, modified by one or more alkyl groups, e.g., one or more C 1-4 alkyl groups, e.g., one or more methyl groups.
  • modified purine or pyrimidine base moieties include N 7 -alkyl-guanine, N 6 -alkyl-adenine, 5-alkyl-cytosine, 5-alkyl-uracil, and N (1) -alkyl-uracil, such as N 7 -C 1-4 alkyl-guanine, N 6 -C 1-4 alkyl-adenine, 5-C 1-4 alkyl-cytosine, 5-C 1-4 alkyl-uracil, and N (1) -C 1-4 alkyl-uracil, preferably N 7 -methyl-guanine, N 6 -methyl-adenine, 5-methyl-cytosine, 5-methyl-uracil, and N (1) -methyl-uracil.
  • ORR object response rate
  • the term “objective response rate” or ORR is the percentage of all subjects in a study or treatment group who have either a partial or complete response to the treatment.
  • the ORR can be calculated by adding the numbers of subjects having CR and the numbers of subjects having PR and dividing the resulting sum by the total number of subjects in the treatment group.
  • the ORR eval i.e., the ORR of all evaluable subjects in a study or treatment group, is the percentage of all evaluable subjects in a study or treatment group who have either a partial or complete response to the treatment.
  • OS is defined as the number of days from Day 1 in Cycle 1 to death due to any cause. If a subject is not known to have died, then OS will be censored at the latest date the subject was known to be alive (on or before the cut-off date) .
  • a “PD-1 antibody” or “anti-PD-1 antibody” is an antibody as described above, which is directed against the antigen PD-1 and which preferably binds specifically to the antigen PD-1.
  • a “PD-L1 antibody” or “anti-PD-L1 antibody” is an antibody as described above, which is directed against the antigen PD-L1 and which preferably binds specifically to the antigen PD-L1.
  • PD-L1 expression score as used herein can be determined using a PD-L1 scoring algorithm such as CPS, TPS, or TAP. As known to the skilled person, the same PD-L1 expression can lead to different numerical values depending on the PD-L1 scoring algorithm.
  • composition relates to a composition comprising a therapeutically effective agent, preferably together with pharmaceutically acceptable carriers, diluents and/or excipients. Said pharmaceutical composition is useful for treating, preventing, or reducing the severity of a disease by administration of said pharmaceutical composition to a subject.
  • pharmaceutically acceptable carrier or “pharmaceutically acceptable excipient” means solvents, dispersion media, coatings, antibacterial agents and antifungal agents, isotonic agents, and absorption delaying agents, and the like, which are compatible with pharmaceutical administration.
  • pharmaceutically acceptable carrier or excipient is not naturally occurring.
  • platinum-containing chemotherapy also known as platins refers to the use of chemotherapeutic agent (s) used to treat cancer that are coordination complexes of platinum.
  • Platinum-containing chemotherapeutic agents are alkylating agents that crosslink DNA, resulting in ineffective DNA mismatch repair and generally leading to apoptosis.
  • platins include cisplatin, carboplatin, and oxaliplatin.
  • polynucleotide and “nucleic acid” can be used interchangeably herein to refer to polymers of nucleotides.
  • polynucleotide comprises deoxyribonucleic acid (DNA) , ribonucleic acid (RNA) , combinations thereof, and modified forms thereof.
  • the term comprises genomic DNA, cDNA, mRNA, recombinantly produced and chemically synthesized molecules.
  • a polynucleotide is DNA.
  • a polynucleotide is RNA.
  • a polynucleotide is a mixture of DNA and RNA.
  • a polynucleotide may be present as a single-stranded or double-stranded and linear or covalently circularly closed molecule.
  • a polynucleotide can be isolated.
  • isolated polynucleotide means, according to the present disclosure, that the polynucleotide (i) was amplified in vitro, for example via polymerase chain reaction (PCR) for DNA or in vitro transcription (using, e.g., an RNA polymerase) for RNA, (ii) was produced recombinantly by cloning, (iii) was purified, for example, by cleavage and separation by gel electrophoresis, or (iv) was synthesized, for example, by chemical synthesis.
  • PCR polymerase chain reaction
  • RNA polymerase RNA polymerase
  • polypeptide ” “peptide, ” and “protein” are used interchangeably herein to refer to polymers of amino acids.
  • PFS progression-free survival
  • recombinant when used in the context of a polynucleotide means a polynucleotide having nucleotide sequences that are not naturally joined together and can be made by artificially combining two otherwise separated segments of sequence. This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, for example, by genetic engineering techniques.
  • Recombinant polynucleotides include vectors comprising an amplified or assembled polynucleotide, which can be used to transform or transfect a suitable host cell.
  • a host cell that comprises the recombinant polynucleotide is referred to as a “recombinant host cell. ”
  • the polynucleotide is then expressed in the recombinant host cell to produce a “recombinant polypeptide. ”
  • a recombinant polynucleotide can also comprise a non-coding function.
  • sequence identity is obtained by determining the number of identical positions at which the sequences to be compared correspond, dividing this number by the number of positions compared (e.g., the number of positions in the reference sequence) and multiplying this result by 100.
  • a “single domain antibody” is an antibody composed of a single variable region (e.g., heavy chain variable region) composed of antibody fragments.
  • a single domain antibody, domain antibody or nanobody consists of 4 framework regions and 3 complementarity determining regions, the 4 framework regions are respectively FR1-FR4, and the 3 complementarity determining regions are respectively CDR1 -CDR3.
  • the single domain antibody of the present application may have a structure of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4. These antibodies do not require light chain variable regions to bind antigens with high affinity and specificity.
  • framework region refers to those amino acid residues in an antibody variable region other than the CDR residues as defined above.
  • An antibody that “specifically binds to” a specified target protein is an antibody that exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity.
  • An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g., without producing undesired results such as false positives.
  • Antibodies, or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins.
  • an antibody is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g., the amino acid sequence of a mature human PD-L1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
  • a “subject” is a human of either gender (a male or a female) .
  • the subject may be of any age.
  • the subject is female.
  • the subject is male.
  • the subject is a subject having cancer, in particular a female subject having cancer and/or a male subject having cancer.
  • the term “TAP” score refers to the “Tumour Area Positivity” score.
  • the PD-L1 expression score can be determined using a TAP scoring algorithm by determining in a test sample of the subject the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer sample; and/or (ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated stroma containing tumour-associated immune cells; and/or (iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or (iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive tumour cells and immune cells; and/or (v) %PD-L1 positive tumour
  • the PD-L1 expression score has been determined using a TAP scoring algorithm in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula: wherein (i) the test sample is a cancer sample; (ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated stroma containing tumour-associated immune cells; (iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1; immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; (iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive tumour cells and immune cells; and (v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 positive tumour cells and immune cells relative to the tumour area expressed as %.
  • the TAP score might be referred to as total percentage of the tumour area (tumour and any desmoplastic stroma) covered with tumour cells with PD-L1 membranous staining at any intensity and tumour-associated immune cells with any pattern.
  • the term “TPS” refers to the “Tumour Proportion Score” .
  • the PD-L1 expression score can be determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula: optionally wherein (i) the test sample is a cancer tissue sample; and/or (ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or (iii) viable tumour cells are determined by staining with a viability dye; and/or (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
  • the PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula: wherein (i) the test sample is a cancer tissue sample; (ii) the cancer tissue sample comprises at least 100 viable tumour cells; (iii) viable tumour cells are determined by staining with a viability dye; and (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
  • treating when used in the context of a disease or disease condition means ameliorating, improving or remedying a disease, disorder, or symptom of a disease or condition associated with the disease, or can mean completely or partially stopping, on a molecular level, the biochemical basis of the disease, etc. It describes an act that leads to the elimination, reduction, alleviation, reversal, or prevention or delay of onset or recurrence of any symptom of a disease.
  • treatment cycle is herein defined as the time period, within the effects of separate dosages of the therapeutic agent (e.g., the anti-PD-1/PD-L1xVEGF/VEGFR binding agent or combination partner) cease to exist due to its pharmacodynamics, or in other words the time period after the subject's body is essentially cleared from the administrated therapeutic agent.
  • Multiple small doses in a small time window e.g. within 2-24 few hours, such as 2-12 hours or on the same day, might be equal to a larger single dose.
  • TNBC human epidermal growth factor receptor 2
  • ER estrogen receptors
  • PR progesterone receptors
  • Tumor as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumours and secondary neoplasms.
  • a solid tumour is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Different types of solid tumours are named for the type of cells that form them. Examples of solid tumours are sarcomas, carcinomas, and lymphomas. Leukemias (cancers of the blood) generally do not form solid tumours (National Cancer Institute, Dictionary of Cancer Terms) .
  • variable regions as used herein means the segment of an antibody which contains three CDRs, designated CDR1, CDR2 and CDR3.
  • a “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination.
  • the variable region of the heavy chain may be referred to as “VH. ”
  • the variable region of the light chain may be referred to as “VL. ”
  • the variable regions of both the heavy and light chains comprise three hypervariable regions, the CDRs, which are located within relatively conserved framework regions (FR) .
  • the CDRs are usually aligned by the framework regions, enabling binding to a specific epitope.
  • both light and heavy chains variable regions comprise FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it was individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as” ) , provided herein is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.
  • VEGF antibody or "anti-VEGF antibody” is an antibody as described above, which is directed against the antigen VEGF and which preferably binds specifically to the antigen VEGF.
  • a “VEGFR antibody” or “anti-VEGFR antibody” is an antibody as described above, which is directed against the antigen VEGFR and which preferably binds specifically to the antigen VEGFR.
  • the present invention inter alia provides a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction (including, e.g., a bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b.
  • a binding agent including, e.g., the bispecific antibody
  • a chemotherapy preferably a chemotherapy agent optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 10, preferably ⁇ 1, more preferably 1 to 20, most preferably 1 to ⁇ 10 or other integrating scoring algorithm defining a similar PD-L1 expression score. It can be preferred that the subject has the afore-mentioned PD-L1 expression score before the treatment.
  • CPS combined positive score
  • the present invention provides a bispecific antibody that specifically binds to PD-L1 and VEGF for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy, preferably a chemotherapy agent optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 1, preferably 1 to 20, more preferably 1 to ⁇ 10.
  • CPS combined positive score
  • the present invention provides a bispecific antibody that specifically binds to PD-L1 and VEGF for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy, preferably a chemotherapy agent optionally wherein the subject has a PD-L1 expression score before the treatment as determined by an integrating scoring algorithm defining a PD-L1 expression score similar or identical to the CPS of ⁇ 1, preferably 1 to 20, more preferably 1 to ⁇ 10.
  • a PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS means an ⁇ 70%or ⁇ 80%, preferably ⁇ 85%, more preferably ⁇ 90%, most preferably ⁇ 95%average positive agreement (APA) and ⁇ 70%or ⁇ 80%, preferably ⁇ 85%, more preferably ⁇ 90%, most preferably ⁇ 95%average negative agreement (ANA) , and ⁇ 70%or ⁇ 80%, preferably ⁇ 85%, more preferably ⁇ 90%, most preferably ⁇ 95%overall percent agreement (OPA) between and within readers with 95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS.
  • APA ⁇ 95%average positive agreement
  • ANA ⁇ 95%average negative agreement
  • OPA overall percent agreement
  • the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ⁇ 70%average positive agreement (APA) and ⁇ 70%average negative agreement (ANA) , and ⁇ 70%overall percent agreement (OPA) between and within readers with ⁇ 95%confidence intervals (Cis) when comparing the scoring algorithm TAP with the CPS.
  • APA ⁇ 70%average positive agreement
  • ANA ⁇ 70%average negative agreement
  • OPA ⁇ 70%overall percent agreement
  • Ce ⁇ 95%confidence intervals
  • the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ⁇ 85%average positive agreement (APA) and ⁇ 85%average negative agreement (ANA) , and ⁇ 85%overall percent agreement (OPA) between and within readers with ⁇ 95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS.
  • APA ⁇ 85%average positive agreement
  • ANA ⁇ 85%average negative agreement
  • OPA ⁇ 85%overall percent agreement
  • the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ⁇ 90%average positive agreement (APA) and ⁇ 90%average negative agreement (ANA) , and ⁇ 90%overall percent agreement (OPA) between and within readers with ⁇ 95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS.
  • APA ⁇ 90%average positive agreement
  • ANA ⁇ 90%average negative agreement
  • OPA ⁇ 90%overall percent agreement
  • the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ⁇ 95%average positive agreement (APA) and ⁇ 95%average negative agreement (ANA) , and ⁇ 95%overall percent agreement (OPA) between and within readers with ⁇ 95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS.
  • APA ⁇ 95%average positive agreement
  • ANA ⁇ 95%average negative agreement
  • OPA ⁇ 95%overall percent agreement
  • the other integrating scoring algorithms are selected from TAP and TPS. In some preferred embodiments, the other integrating scoring algorithm is TAP.
  • the integrating scoring algorithm defining a similar PD-L1 expression score preferably is a tumour cell integrating scoring algorithm such TPS or a tumour cell and immune cell integrating scoring algorithm such as CPS or TAP.
  • VEGF Vascular endothelial growth factor
  • VPF vascular permeability factor
  • vasculotropin is a highly specific homodimer that promotes the growth of vascular endothelial cells.
  • VEGF family proteins include VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF-F and placental growth factor (PIGF) , among which VEGF-Ais involved in the early formation of blood vessels play an important role.
  • PIGF placental growth factor
  • VEGF vascular endothelial growth factor
  • VEGF is involved in the pathogenesis and progression of many angiogenesis-dependent diseases, including cancer, certain inflammatory diseases, and diabetic retinopathy. Therefore, VEGF is an important target in antitumour drug research.
  • VEGFA can bind to VEGFR1 and VEGFR2, activate endogenous kinase activation, and promote new blood vessels.
  • Blocking the binding of VEGF to the receptor can be applied to the treatment of various cancers, such as breast cancer, colon cancer, lung cancer, colorectal, ovarian cancer, endometrial cancer, mesothelioma (including malignant pleural mesothelioma [MPM] and malignant peritoneal mesothelioma [MPeM] ) , cervical cancer, kidney cancer (Rakesh R. Ramjiawan, Arjan W. Griffioen, and Dan G. Duda, Angiogenesis. 2017 20 (2) : 185–204. ) .
  • various cancers such as breast cancer, colon cancer, lung cancer, colorectal, ovarian cancer, endometrial cancer, mesothelioma (including malignant pleural mesothelioma [MPM] and malignant peritoneal mesothelioma [MPeM] ) , cervical cancer, kidney cancer (Rakesh R. Ramjiawan,
  • VEGF angiogenesis-modulating capabilities
  • VEGF-A is associated with a range of immunosuppressive effects at successive steps in the cancer-immunity cycle, such as diminished antigen presentation, T cell priming, T cell trafficking, and T cell tumour infiltration.
  • the binding agent including, e.g., the bispecific antibody
  • VEGF-A can release immunosuppression.
  • an anti-VEGF antibody treatment alone results in increased gene expression associated with Th1 chemokines involved with T-cell trafficking, tumour MHC-I protein expression and infiltration of tumour-specific T-cell clones demonstrating that an anti-VEGF antibody is capable of inducing anti-tumour immune responses.
  • the binding agent may reverse VEGF-A mediated inhibition of monocyte-to-dendritic cell maturation thereby increasing numbers of dendritic cells (DCs) , and reverse VEGF-A mediated inhibition of dendritic cell maturation thereby increasing tumour-antigen presentation.
  • the binding agent may further reverse or reduce VEGF-A mediated enhanced T cell exhaustion, as VEGF-A binding to VEGFR-2 on the surface of CD8+ T cells has been shown in preclinical studies to result in upregulated expression of the immune-checkpoint molecules (and exhaustion markers) PD-1, Cytotoxic T Lymphocyte antigen 4 (CTLA-4) , and T cell immunoglobulin mucin receptor 3 (TIM3) as well as that of lymphocyte activation gene 3 protein (LAG3) .
  • CTLA-4 Cytotoxic T Lymphocyte antigen 4
  • TIM3 T cell immunoglobulin mucin receptor 3
  • the binding agent may further reverse or reduce VEGF-A mediated proliferation of Treg cells, and reverse VEGF-A mediated downregulation of adhesion molecules (e.g. ICAM-1, or CD34) to allow for more efficient immune cell infiltration of the tumour.
  • VEGF thus has a major role in the generation of an immunosuppressive tumour microenvironment.
  • blocking the VEGF-A/VEGFR-2 interaction can induce physical changes in the tumour vasculature such as vascular normalization, a process whereby hypoxia is transiently alleviated and the tumour vasculature reverts back to resemble that of a nonmalignant tissue (Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58–62 (2005) ) .
  • PD-L1 Programmed death-ligand 1
  • CD274 is a member of the B7 family and is a ligand of PD-1.
  • PD-L1 is a type I transmembrane protein with a total of 290 amino acids, including an IgV-like domain, an IgC-like domain, a transmembrane hydrophobic domain and an intracellular domain consisting of 30 amino acids.
  • PD-L1 negatively regulates immune responses. Studies have found that PD-L1 is mainly expressed in activated T cells, B cells, macrophages and dendritic cells, etc.
  • PD-L1 is also expressed in other tissues such as thymus, heart, placenta, etc. endothelial cells, as well as various non-lymphoid lineages such as melanoma, lung cancer, liver cancer, gastric cancer, renal cell cancer, urothelial cancer, cervical cancer, colorectal cancer, ovarian cancer, colon cancer, breast cancer, esophageal cancer, head and neck cancer, etc. (Akintunde Akinleye&Zoaib Rasool, Journal of Hematology&Oncology volume 12, Article number: 92 (2019) ) .
  • PD-L1 regulates autoreactive T-and B-cells, and immune tolerance, and plays a role in peripheral tissue T-and B-cell responses. Interfering with the PD-1: PD-L1 interaction reinvigorates exhausted T cells, as PD-1 is a major regulator of T-cell exhaustion. Hence blocking the PD-1: PD-L1 pathway restores T-cell function and improves tumour eradication.
  • the combination therapy disclosed herein comprising the binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction (including, e.g., a bispecific antibody that specifically binds to PD-L1 and VEGF) and the chemotherapy shows particularly encouraging anti-tumour activity even when the subject has a low PD-L1 expression score.
  • the binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/
  • the combination of the PD-L1 and VEGF binding agent (including, e.g., the bispecific antibody) of the invention with the chemotherapy is highly effective in cancer treatment, even in subjects having a low PD-L1 expression score as, e.g., determined by the PD-L1 scoring algorithm CPS of ⁇ 10 or ⁇ 1.
  • the afore-mentioned combination shows a good safety profile for cancer subjects.
  • the release in immunosuppression by blocking VEGF and PD-L1 and the consequential influx of immune cells is contemplated to lead to an increased expression in PD-L1 on tumour cells, further enriching for the binding agent (including, e.g., the bispecific antibody) of the invention in the tumour microenvironment.
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent is contemplated to transform PD-L1 low-expressing tumours into more inflamed, immune cell infiltrated “hot tumours” .
  • the combination therapy comprising the binding agent (including, e.g., the bispecific antibody) and chemotherapy can be used in a method of treating cancer in a subject as disclosed herein.
  • the binding agent including, e.g., the bispecific antibody
  • chemotherapy can be used in a method of treating cancer in a subject as disclosed herein.
  • the binding agent (including, e.g., the bispecific antibody) and chemotherapy disclosed herein can provide for substantial improvement in a subject’s overall survival (OS) , progression-free survival (PFS) , objective response rate (ORR) , duration of response, and/or disease control rate.
  • OS overall survival
  • PFS progression-free survival
  • ORR objective response rate
  • duration of response and/or disease control rate.
  • PFS the binding agent (including, e.g, the bispecific antibody) and chemotherapy disclosed herein can provide for a PFS of at least about 9 months, preferably of at least about 13 months in patients suffering from TNBC (see also example 3 herein) .
  • Objective Response Rate refers to the number (%) of subjects with at least one visit response of Complete Response (CR) or Partial Response (PR) per RECIST 1.1.
  • Duration of Response refers to the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (i.e., date of PFS event or censoring –date of first response + 1) .
  • Disease Control Rate refers to the rate of best objective response of CR, PR, or stable disease (SD) according to RECIST 1.1. Determination of the PD-L1 expression
  • the PD-L1 expression score as described herein is determined by histologically analyzing a tumour sample and applying a scoring algorithm, the scoring algorithm according to the invention is the combined positive score (CPS) or any other scoring algorithm defining a similar PD-L1 expression score as the CPS.
  • CPS is a well-established scoring algorithm which is approved for the treatment of cancer using pembrolizumab in multiple indications.
  • the treatment method comprises the step of applying the scoring algorithm to determine the PD-L1 expression score and thereby whether the subject shall be treated with the combination therapy of the invention.
  • the PD-L1 expression score (e.g., CPS or similar score type) is already known and provided before the start of the method of the invention.
  • the PD-L1 expression score can be determined in form of the CPS in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula: wherein preferably the PD-L1 staining cells are tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
  • the lymphocytes and macrophages are preferably directly associated with the response against the cancer.
  • the PD-L1 staining cells are tumour cells with convincing partial or complete linear membrane staining (at any intensity) that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining (at any intensity) .
  • the PD-L1 staining cells are preferably viable PD-L1 staining cells.
  • CPS scoring algorithm is for example described in Kulangara, Karina & Hanks, Debra & Waldroup, Stephanie & Peltz, Lindsay & Shah, Supriya & Roach, Charlotte & Juco, Jonathan & Emancipator, Kenneth & Stanforth, Dave. (2017) , Development of the combined positive score (CPS) for the evaluation of PD-L1 in solid tumours with the immunohistochemistry assay PD-L1 IHC 22C3 pharmDx. Journal of Clinical Oncology, which disclosure is incorporated herein in its entirety.
  • the number of viable tumour cells in the tumour tissue sample are determining by flow cytometry.
  • a tissue sample analyzed by flow cytometry can be contacted with a viability dye prior to analysis, e.g., propidium iodide. Any convenient viability stain may be employed, with many examples known in the art.
  • the tumour proportion score (TPS) (an integrating scoring algorithm) is used instead of the CPS.
  • the TPS can be obtained by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 (PD-L1 staining tumour cells) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:
  • the number of total tumour cells and tumour cells positive for PD-L1 can be assessed using the PD-L1 IHC 22C3 pharmDx assay (Agilent Technologies, Carpinteria, CA, USA) according to the manufacturer’s Instructions for Use.
  • the number of tumour cells can be measured in formalin-fixed tumour samples obtained by core-needle or excisional biopsy of a tumour lesion or from tissue resected at or after the time the cancer was diagnosed.
  • positivity is defined as complete circumferential or partial cell membrane staining of viable tumour cells with 1+ to 3+ intensity.
  • Nonspecific staining can be recorded on a 0 to 3 intensity scale, in 0.25 grade increments.
  • Tumour-associated immune cells are preferably excluded from PD-L1 scoring.
  • Cytoplasmic staining, if present, is preferably excluded from the scoring. Scoring can be recorded as percentage of PD-L1-positive tumour cells over total tumour cells in the denominator (TPS) .
  • the TAP scoring algorithm is used as the integrating scoring algorithm instead of CPS.
  • the TAP score can be obtained by determining in a test sample of the subject the number of PD-L1 positive tumour and immune cells (PD-L1 staining tumour and immune cells) and the tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:
  • the TAP score can be determined as described in Liu, Chunyan et al. “Tumour Area Positivity (TAP) score of programmed death-ligand 1 (PD-L1) : a novel visual estimation method for combined tumour cell and immune cell scoring. ” Diagnostic pathology vol. 18, 1 48.19 Apr. 2023, doi: 10.1186/s13000-023-01318-8, which disclosure is incorporated by reference herein in its entirety.
  • TAP Tuour Area Positivity
  • the tumour area is determined using the hematoxylin and eosin (H&E) stain.
  • the PD-L1 positive tumour cells and immune cells are PD-L1 staining cells with partial or complete linear membrane staining that is distinct from cytoplasmic staining, and immune cells (lymphocytes and macrophages) within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
  • tumour area area occupied by all viable tumour cells (TC) and the tumour-associated stroma containing tumour-associated immune cells (IC) ) .
  • the immune cells measured in the nominator are tumour-associated stroma containing tumour-associated immune cells. If tumour nests are separated by non-neoplastic tissue, they can be included as part of the tumour area as long as the tumour nests are bordered on both sides of a 10x field; the intervening non-neoplastic tissue can also be included in the tumour area (commonly abbreviated as 10x field rule) .
  • Tumour-associated IC are preferably intra-and peri-tumoural, including those present within the tumour proper, between tumour nests, and within any tumour-associated reactive stroma. In lymph nodes with focal or discrete tumour metastases, only IC immediately adjacent to the leading edge of the metastatic tumour nest are preferably defined as tumour-associated IC.
  • the TAP can be determined on the IHC slide by visually aggregating/estimating the area covered by PD-L1 positive TC (PD-L1 staining TC) and tumour-associated IC relative to the total tumour area.
  • PD-L1 staining TC PD-L1 staining TC
  • tumour-associated IC PD-L1 positive staining
  • Intra-luminal macrophage staining is preferably not included in the TAP score unless the macrophages completely fill the luminal space and are in direct contact with the TC.
  • Staining of multi-nucleated giant cells, granulomas, and IC located within blood vessels and lymphatics are preferably not included in the TAP.
  • Off-target staining e.g., fibroblasts, endothelial cells, neuroendocrine cells, smooth muscle, and nerves
  • the cancer tissue sample is a tissue section of a tumour biopsy, preferably a solid tumour biopsy.
  • Biopsies of interest can include tumour and/or non-neoplastic biopsies of skin (melanomas, carcinomas, etc. ) , soft tissue, bone, breast, colon, liver, kidney, adrenal, gastrointestinal, pancreatic, gall bladder, salivary gland, cervical, ovary, uterus, testis, prostate, lung, thymus, thyroid, parathyroid, pituitary (adenomas, etc. ) , brain, spinal cord, ocular, nerve, and skeletal muscle, etc.
  • the subject from which the biopsy is obtained has a malignancy is selected from the list consisting of gastric cancer, esophageal carcinomas, head and neck cancer (e.g., head and neck squamous cell carcinoma, or HNSCC) , renal cell carcinoma, urothelial/bladder carcinoma, colorectal cancer, ovarian carcinoma, myeloma, melanoma, lung cancer, classical Hodgkin lymphoma, and breast cancer (e.g., triple-negative breast cancer, hormone receptor positive (ER and/or PR) and Her2 positive breast cancer) , small cell lung cancer including extensive stage small cell lung cancer and limited stage small cell lung cancer, salivary gland carcinoma, vulvar carcinoma, thyroid carcinoma, anal canal carcinoma, biliary carcinoma, mesothelioma, cervical carcinoma, and neuroendocrine carcinoma.
  • gastric cancer esophageal carcinomas
  • head and neck cancer e.g., head and neck squamous cell carcinoma, or HNSCC
  • the subject from which the biopsy is obtained has a malignancy selected from melanoma, liver, stomach, renal cell, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancer.
  • the biopsy of interest can include a tumour and optionally non-neoplastic biopsy of any of a melanoma, liver, stomach, renal cell, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck solid tumour.
  • the biopsy is obtained from a subject having urothelial, breast, or esophageal cancer, most preferably breast cancer such as TNBC.
  • the CPS is calculated from a stained tumour tissue biopsy section (e.g., on a slide) or serial tumour tissue biopsy sections by immunohistochemistry (IHC) staining, in-situ hybridization (ISH; e.g., fluorescence-in-situ-hybridization, or FISH) , histological stain, and a combination thereof.
  • a tumour tissue biopsy section is analyzed by IHC.
  • the percentage of viable PD-L1 positive and negative tumour cells and PD-L1 positive mononuclear inflammatory cells (MIC) is determined within the tumour nests and the adjacent supporting stroma.
  • cells are positive for PD-L1 staining if they display partial or complete membrane staining relative to all viable tumour cells present in the sample.
  • PD-L1 is detected by immunohistochemistry (IHC) staining.
  • the number of viable tumour cells can be determining by flow cytometry. For example, a tissue sample analyzed by flow cytometry can be contacted with a viability dye prior to analysis, e.g., propidium iodide or any convenient viability stain known in the art.
  • ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇ ⁇
  • the tumour tissue section is formalin fixed and embedded in paraffin wax (FFPE) .
  • FFPE paraffin wax
  • the tissue section has been fixed in a different way, including tissue sections that have been fixed in, e.g., acrolein, glyoxal, osmium tetroxide, carbodiimide, mercuric chloride, zinc salts, picric acid, potassium di chromate, ethanol, methanol, acetone, and/or acetic acid.
  • the PD-L1 scoring algorithm (such as CPS, TPS, or TAP) is calculated from a tumour tissue sample that is not a fixed section on a slide.
  • the CPS is calculated using flow cytometric analysis of a cell suspension from the tumour tissue sample.
  • the tumour tissue cell suspension can be stained with a detectable PD-L1 binding agent (e.g., a fluorescently labeled antibody) and analyzed on a flow cytometer for counting the number of tumour cells and MIC cells (i.e., lymphocytes and macrophages) expressing PD-L1.
  • a detectable PD-L1 binding agent e.g., a fluorescently labeled antibody
  • MIC cells i.e., lymphocytes and macrophages
  • Tumour cells and MIC cells in the sample can be distinguished using any convenient flow cytometric parameter, e.g., forward scatter (FS) , side scatter (SS) , or by the expression of one or more additional markers using corresponding detectable binding agents for the one or more additional markers, e.g., markers specific or MIC or tumour cells.
  • the cells in the tumour tissue sample can be analyzed on a cell-by-cell basis for mRNA expression of PD-L1 and any other desired target, e.g., using single-cell nucleic acid sequencing methods for gene expression profiling (e.g., next generation sequencing methods) .
  • the tissue section is stained.
  • the stain comprises a hematoxylin and eosin (H&E) stain.
  • Hematoxylin a basic dye, stains nuclei blue due to an affinity to nucleic acids in the cell nucleus; eosin, an acidic dye, stains the cytoplasm pink.
  • a hematoxylin and eosin (H&E) stained section can be used for the evaluation of an acceptable tumour tissue sample or of a tumour area.
  • the staining of a target (e.g., PD-L1) in cells from a tumour tissue biopsy is generally done by contacting the cells with one or more detectable target-specific binding agents under suitable conditions to allow for binding of the target-specific binding agent to its desired target (while minimizing nontarget binding) .
  • a target e.g., PD-L1
  • target-specific binding agent means any agent that specifically binds to a target or analyte of interest, e.g., a target of interest that is present in a tissue section as described herein (e.g., a polypeptide or polynucleotide) .
  • the target-specific binding agent is an antibody (or target-binding fragments thereof) , e.g., as used in IHC and flow cytometry.
  • Staining may be performed with primary and secondary antibodies or without using secondary antibodies (e.g., where the primary antibody is detectably labeled) .
  • Non-limiting examples of anti-PD-L1 antibodies include, but are not limited to, clone 22C3 (Merck & Co. ) , clone 28-8 (Bristol-Myers Squibb) , clones SP263 (Roche) , SP142 (Spring Biosciences) , and clone E1L3N (Cell Signaling Technology) .
  • a preferred anti-PD-L1 antibody is clone SP263 (Roche)
  • another preferred anti-PD-L1 antibody is clone E1L3N (Cell Signaling Technology) .
  • Clone 22C3 is a well-known, commercially available anti-PD-L1 antibody described e.g., in US 9,709,568 B2, which disclosure is incorporated by reference in its entirety.
  • Clone 22C3 comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 23, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 24, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 25; and the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 20, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 21, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 22.
  • the heavy and light chain variable regions are set forth in SEQ ID NO: 27 and 26.
  • Clone 28-8 is a well-known commercially available anti-PD-L1 antibody described e.g., in US 9,212,224 B2, which disclosure is incorporated by reference in its entirety.
  • Clone 28-8 comprises a heavy and light chain variable region (i.e., VH and VL) set forth in SEQ ID NO: 28 and 29.
  • SP263 is a well-known commercially available anti-PD-L1 antibody described e.g., in US 2010/0343556 A1.
  • Clone SP263 comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 33, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 34, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 35; and the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 30, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 31, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 32.
  • the heavy and light chain variable region are set forth in SEQ ID NO: 36 and 37.
  • a target specific binding agent is a nucleic acid or nucleic acid binding agent, e.g., as employed in in situ hybridization (ISH) reactions.
  • the target binding reagent can be a DNA, RNA, DNA/RNA hybrid molecule, peptide nucleic acid (PNA) , and the like. No limitation in the metes and bounds of a target-specific binding agent that finds use in the subject disclosure is intended.
  • the target-specific binding agent (or any secondary reagent used to detect the target-specific binding agent) may be attached to any suitable detectable label (or chromogen) or enzyme capable of producing a detectable label.
  • the first or second label is produced by an enzymatic reaction, e.g., by the activity of horseradish peroxidase, alkaline phosphatase, and the like. Any convenient enzymatic label/chromogen deposition system can be employed (e.g., as used in standard IHC methods) , and as such, no limitation in this regard is intended.
  • the detectable label is a fluorescent tag.
  • the staining reagents used may include a target-specific antibody (e.g., a PD-L1 specific antibody) .
  • the staining reagents used may include one or more additional antibodies that each bind to a different antigen.
  • a set of antibodies may include a first antibody that binds to a first antigen (e.g., PD-L1) , a second antibody that binds to a second antigen, optionally a third antibody that binds to a third antigen and, optionally a fourth antibody that binds to a fourth antigen and/or further antibodies that bind to further antigens.
  • the antibody/antibodies used are primary antibodies that are detected by use of a secondary antibody (or other reagent) .
  • the staining steps thus may be done by incubating the cells of the tissue sample, e.g., a tissue section or cell suspension, with the primary antibody/antibodies and then, after the primary antibody has bound to the desired target in/on the cells, incubating the cells with the labeled secondary antibody/antibodies (e.g., as is done in standard IHC protocols) .
  • each of the primary antibodies for each different target is from a different species (e.g., goat, rabbit, mouse, camel, chicken, donkey, etc. ) and the corresponding secondary antibodies specific for each different primary antibody are distinguishably labeled from each other.
  • the viable tumour cells and the number of lymphocytes and macrophages are counted in the tumour nests and the adjacent supporting stroma of the tumour tissue sample.
  • the maximum score is preferably defined as 100 and a minimum of 100 viable tumour cells are preferably required to calculate the score. See Park et al. Cancer Res. Treat., 2020; 52 (3) : 661-670 and Yamashita et al., Gastric Cancer, 2020, 23: 95-104.
  • an in vitro diagnostic device (IVD) bridging study can be performed to determine whether that scoring algorithm defines a similar PD-L1 expression score as the established CPS.
  • IVD in vitro diagnostic device
  • a test for determining PD-L1 expression score other than the CPS companion diagnostic can be used, if it can be demonstrated that the other IVD companion diagnostic has performance characteristics that are very similar to those of the CPS companion diagnostic. This is generally demonstrated through a bridging study between the two tests, using the original clinical trial samples and a pre-specified statistical analysis plan, to show that results with the candidate IVD companion diagnostic are very similar to those of the CPS companion diagnostic.
  • a bridging study can evaluate efficacy of the therapeutic product in subjects whose marker status is determined by the candidate IVD companion diagnostic by assessing both concordance and discordance between the two tests using the same specimens from subjects who were tested for trial eligibility. The analysis needs to consider any potential impact of missing samples not available for the concordance study.
  • the ability of the candidate IVD companion diagnostic to predict the efficacy of the therapeutic product can be supported indirectly by high analytical concordance with the CPS companion diagnostic on a large number of representative samples, including samples from subjects excluded from the trial because they were marker-negative by the CPS companion diagnostic.
  • the assessment of the clinical validity of the candidate IVD companion diagnostic can rely on extrapolating the clinical performance characteristics of the CPS companion diagnostic to the clinical performance characteristics of the candidate IVD companion diagnostic.
  • the ideal bridging study is one in which all samples tested with the trial test are retested with the candidate IVD companion diagnostic and valid test results are obtained and used to assess comparative performance.
  • a bridging study with specimens from an all-comers trial also allows an analysis of efficacy using the results of the candidate IVD companion diagnostic. Note, however, that care should preferably be taken in understanding the analytical performance of the IVD prior to the bridging study because adjustments to the IVD should not be made from results obtained with the clinical trial samples. Whether a clinical trial enrolls subjects irrespective of the test result or enrolls only the subset of subjects identified by the test result, both the test-negative and test-positive clinical trial samples should preferably be included in bridging studies to avoid bias due to prescreening.
  • the characteristics of the subset adequately reflect the characteristics that affect test performance (e.g., tumour size, histology, melanin content, necrotic tissue, resected tissue versus core needle biopsy) and that the characteristics of the subjects that may affect therapeutic product efficacy (e.g., patient demographics, stage of disease, stratification factors) are proportionally preserved in the retest sample set when compared to the samples in the original set.
  • test performance e.g., tumour size, histology, melanin content, necrotic tissue, resected tissue versus core needle biopsy
  • therapeutic product efficacy e.g., patient demographics, stage of disease, stratification factors
  • a re-analysis of the primary outcome data should preferably be made according to the final test results with the retest sample set in order to assure that any reclassification that occurs does not alter conclusions about the safety and efficacy of the therapeutic product in the selected population.
  • a second re-analysis can be conducted in which missing data for the final test are imputed.
  • the nature of the re-analysis will be product-specific and may be discussed with the appropriate IVD review center.
  • additional analytical validation can potentially be requested to support satisfactory concordance across methods where discordance may arise, e.g., precision, limit of detection, and accuracy. In the event there is discordance in a marker-positive-only trial, it is possible that the candidate IVD companion diagnostic will more accurately predict responders, a difference that would represent an advantage for optimal use of the therapeutic product.
  • the TAP has performance characteristics that are very similar to those of the CPS companion diagnostic using the described bridging study.
  • a PD-L1 expression score determined using different anti-PD L1 antibodies such as SP263 or 22C3 or E1L3N.
  • a PD-L1 expression score obtained from using 22C3 and the CPS scoring algorithm has characteristics that are very similar to those of a PD-L1 expression score obtain from using 28-8 and the CPS scoring algorithm using the described bridging study.
  • this bispecific antibody may comprise in a general aspect an anti-VEGF antibody (e.g., an anti-VEGF-A antibody) or fragment thereof, and an antagonist of the PD1/PD-L1 interaction and/or a PD-1 or PD-L1 blocking antibody.
  • the bispecific antibody comprises an anti-VEGF antibody (e.g., an anti-VEGF-A antibody) or fragment thereof, and a PD-1 or PD-L1 blocking antibody.
  • the bispecific antibody comprises an anti-VEGF antibody (e.g., an anti-VEGF-A antibody) or fragment thereof, and an anti-PD-1 or anti-PD-L1 antibody or fragment thereof.
  • the anti-PD-1 antibody can be selected from any anti-PD-1 antibody known in the art such as pembrolizumab or nivolumab.
  • An exemplary anti-PD-L1 antibody includes atezolizumab.
  • the anti-VEGF antibody can be selected from any VEGF antibody known in the art such as bevacizumab, ranibizumab or an VEGF binding region derived from the anti-VEGF fusion protein Aflibercept (see, e.g., US 7070959) .
  • An exemplary anti-VEGF antibody includes bevacizumab.
  • the present disclosure relates to a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction.
  • This binding agent which is alternatively referred to herein as “anti-PD-1/PD-L1xVEGF/VEGFR binding agent” or simply as “binding agent” will be discussed in more detail in the following sections.
  • First binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
  • One component of the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction.
  • this first binding region comprises an antagonist of the PD-1/PD-L1 interaction, such as an anti-PD-1 antibody, a PD-1 binding fragment thereof, an anti-PD-L1 antibody, or a PD-L1 binding fragment thereof.
  • Anti-PD-1 antibodies or PD-1 binding fragments thereof are an antagonist of the PD-1/PD-L1 interaction, such as an anti-PD-1 antibody, a PD-1 binding fragment thereof, an anti-PD-L1 antibody, or a PD-L1 binding fragment thereof.
  • the first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction comprises an anti-PD-1 antibody or a PD-1 binding fragment thereof.
  • PD-1 is human PD-1, in particular human PD-1 comprising the sequence set forth in SEQ ID NO: 90, 91, or 92.
  • the anti-PD-1 antibody or a PD-1 binding fragment thereof specifically binds to PD-1.
  • the anti-PD-1 antibody or fragment thereof is selected from the group consisting of Pembrolizumab, Nivolumab, Cemiplimab, Dostarlimab, Toripalimab, Retifanlimab, and PD-1 binding fragments thereof.
  • the anti-PD-1 antibody or fragment thereof is a monoclonal antibody or a fragment thereof.
  • the anti-PD-1 antibody or fragment thereof is a humanized antibody. In some embodiments, the anti-PD-1 antibody or fragment thereof is a monoclonal humanized antibody. In some embodiments, the fragment of the anti-PD-1 antibody may be any antigen-binding fragment of the anti-PD-1 antibody, for example a Fab, a Fab', a F (ab') 2, a Fv, a scFv, a Fab'-SH, an sdAb, or a VHH. In some embodiments, the anti-PD-1 antibody or fragment thereof is selected from a full-length anti-PD-1 antibody, an scFv of an anti-PD-1 antibody or a VHH of an anti-PD-1 antibody. In some embodiments, the anti-PD-1 antibody or fragment thereof is an scFv of an anti-PD-1 antibody. In some embodiments, the anti-PD-1 antibody or fragment thereof is a VHH of an anti-PD-1 antibody.
  • the anti-PD-1 antibody or fragment thereof may comprise a variable region that specifically binds to PD-1.
  • the anti-PD-1 antibody or fragment thereof comprises a heavy chain variable region and/or a light chain variable domain.
  • the anti-PD-1 antibody or fragment thereof comprises a heavy chain variable region and a light chain variable domain.
  • the anti-PD-1 antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
  • the anti-PD-1 antibody or fragment thereof comprises a heavy chain and/or a light chain.
  • the anti-PD-1 antibody or fragment thereof comprises a heavy chain and a light chain. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of a heavy chain and a light chain as a single chain (scFv) .
  • anti-PD-1 antibody CDRs variable region sequences, and heavy and light chain sequences are provided in Table 1, e.g., for anti-PD-1 antibodies A1 and A2, respectively.
  • the anti-PD-1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-1 antibody A1 shown in Table 1.
  • a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 45, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 46, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 47, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 48, or a variant thereof having up to three amino acid substitutions, additions or
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 51, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 52, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 53, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 54, or a variant thereof having up to three amino acid substitutions, additions or deletion
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 56, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 56, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively, or (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55 and 50, respectively.
  • amino acid sequence comprises (i) the HCDR1, HCDR2, HCDR
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 56, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively, or (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55 and 50, respectively.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 56.
  • the anti-PD-1 antibody or fragment thereof is an scFv comprising: (i-1a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 45, 46, and 47, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 48, 49, and 50, respectively; (i-1b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 51, 52, and 53, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 54, 55, and 50, respectively; (i-1c) the amino acid sequence of SEQ ID NO: 56 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%,
  • the anti-PD-1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-1 antibody A2 shown in Table 1.
  • a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 57, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 58, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 59 or 60, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system.
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 61, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 62, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 63 or 64, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system.
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 65, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 65, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 65, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 65.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 66, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 66, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 66, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively.
  • the anti-PD-1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 66.
  • the anti-PD-1 antibody or fragment thereof is a VHH comprising: (i-2a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively; (i-2b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively, or a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively; (i-2c) the amino acid sequence of SEQ ID NO: 65 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at
  • the first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction comprises an anti-PD-L1 antibody or a PD-L1 binding fragment thereof.
  • PD-L1 is human PD-L1, in particular human PD-L1 comprising the sequence set forth in SEQ ID NO: 93, 94, 95, or 96.
  • the anti-PD-L1 antibody or a PD-L1 binding fragment thereof specifically binds to PD-L1.
  • the anti-PD-L1 antibody or fragment thereof is selected from the group consisting of Atezolizumab, Durvalumab, Avelumab, and PD-L1 binding fragments thereof.
  • the anti-PD-L1 antibody or fragment thereof is a monoclonal antibody or a fragment thereof.
  • the anti-PD-L1 antibody or fragment thereof is a humanized antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is a monoclonal humanized antibody. In some embodiments, the fragment of the anti-PD-L1 antibody may be any antigen-binding fragment of the anti-PD-L1 antibody, for example a Fab, a Fab', a F (ab') 2, a Fv, a scFv, a Fab'-SH, an sdAb, or a VHH.
  • the anti-PD-L1 antibody or fragment thereof is selected from a full-length anti-PD-L1 antibody, an scFv of an anti-PD-L1 antibody or a VHH of an anti-PD-L1 antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is a VHH of an anti-PD-L1 antibody.
  • the anti-PD-L1 antibody or fragment thereof may comprise a variable region that specifically binds to PD-L1.
  • the anti-PD-L1 antibody or fragment thereof comprises a heavy chain variable region and/or a light chain variable domain.
  • the anti-PD-L1 antibody or fragment thereof comprises a heavy chain variable region and a light chain variable domain.
  • the anti-PD-L1 antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
  • the anti-PD-L1 antibody or fragment thereof comprises a heavy chain and/or a light chain.
  • the anti-PD-L1 antibody or fragment thereof comprises a heavy chain and a light chain. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises of a heavy chain and a light chain as a single chain (scFv) . In some embodiments, the anti-PD-L1 antibody or fragment thereof is a VHH of an anti-PD-L1 antibody.
  • anti-PD-L1 antibody CDRs Exemplary anti-PD-L1 antibody CDRs, variable region sequences, and heavy and light chain sequences are provided in Table 1, e.g., for anti-PD-L1 antibodies A3 and A4, respectively.
  • the anti-PD-L1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-L1 antibody A3 shown in Table 1.
  • a VH of the anti-PD-L1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 18, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 19, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 38, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system.
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • a VH of the anti-PD-L1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 1, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 2, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 3, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system.
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 9, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 9, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 9, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9.
  • the anti-PD-L1 antibody or fragment thereof is a VHH comprising: (i-3a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively; (i-3b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively; or (i-3c) the amino acid sequence of SEQ ID NO: 9 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the amino acid sequence SEQ ID NO: 9, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID
  • the anti-PD-L1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-L1 antibody A4 shown in Table 1.
  • the anti-PD-L1 antibody or fragment thereof comprises (a) a heavy chain variable region (VH) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 73, and/or a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 74.
  • VH heavy chain variable region
  • VL light chain variable region
  • a VH of the anti-PD-L1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 67, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 68, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 69, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 70, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73.
  • the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 73.
  • the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74.
  • the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 74.
  • the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a
  • the anti-PD-L1 antibody or fragment thereof comprises a (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in
  • the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74.
  • the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively.
  • the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 73, and (II) comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 74.
  • the anti-PD-L1 antibody or fragment thereof is a fragment of a humanized anti-PD-L1 antibody comprising: (i-4a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences of SEQ ID NO: 73, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences of SEQ ID NO: 74; (i-4b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively; (i-4c) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at
  • Another component of the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a second binding region which binds to VEGF, VEGFR, or both and which antagonizes the VEGF/VEGFR interaction.
  • this second binding region comprises an antagonist of the VEGF/VEGFR interaction, such as an anti-VEGF antibody, a VEGF binding fragment thereof, a protein comprising a VEGFR domain, an anti-VEGFR antibody, or a VEGF binding fragment thereof.
  • the second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction comprises an anti-VEGF antibody or a VEGF binding fragment thereof, preferably an anti-VEGF-A antibody or a VEGF-A binding fragment thereof.
  • VEGF is human VEGF, in particular human VEGF comprising the sequence set forth in SEQ ID NO: 97 or human VEGF-A comprising the sequence set forth in SEQ ID NO: 98.
  • the anti-VEGF antibody or a VEGF binding fragment thereof specifically binds to VEGF, in particular to VEGF-A.
  • the anti-VEGF antibody or fragment thereof is selected from the group consisting of Bevacizumab, Ramucirumab, Ranibizumab, and VEGF binding fragments thereof.
  • the anti-VEGF antibody or fragment thereof is a monoclonal antibody or a fragment thereof.
  • the anti-VEGF antibody or fragment thereof is a humanized antibody. In some embodiments, the anti-VEGF antibody or fragment thereof is a monoclonal humanized antibody. In some embodiments, the fragment of the anti-VEGF antibody may be any antigen-binding fragment of the anti-VEGF antibody, for example a Fab, a Fab', a F (ab') 2, a Fv, a scFv, a Fab'-SH, an sdAb, or a VHH. In some embodiments, the anti-VEGF antibody or fragment thereof is selected from a full-length anti-VEGF antibody, an scFv of an anti-VEGF antibody, or a VHH of an anti-VEGF antibody. In some embodiments, the anti-VEGF antibody or fragment thereof is a full-length anti-VEGF antibody.
  • the anti-VEGF antibody or fragment thereof may comprise a variable region that specifically binds to VEGF, in particular VEGF-A.
  • the anti-VEGF antibody or fragment thereof comprises a heavy chain variable region and/or a light chain variable domain.
  • the anti-VEGF antibody or fragment thereof comprises a heavy chain variable region and a light chain variable domain.
  • the anti-VEGF antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
  • the anti-VEGF antibody or fragment thereof comprises a heavy chain and/or a light chain.
  • the anti-VEGF antibody or fragment thereof comprises a heavy chain and a light chain.
  • the anti-VEGF antibody or fragment thereof comprises two heavy chains and two light chains.
  • anti-VEGF antibody CDRs variable region sequences, and heavy and light chain sequences are provided in Table 1; cf., e.g., SEQ ID NOs: 39, 40, 41, 42, 43, 8, 4, 5, 6, 7, 44, 8, 10, 11, 12, 13, 15, 80, and 17.
  • the anti-VEGF antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
  • the anti-VEGF antibody or fragment thereof comprises (a) a heavy chain variable region (VH) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 10, and/or a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 11.
  • VH heavy chain variable region
  • VL light chain variable region
  • a VH of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 39, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 40, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 41, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 42, or a variant thereof having up to three amino acid substitutions, additions or deletion
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • a VH of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 5, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 6, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 7, or a variant thereof having up to three amino acid substitutions, additions or deletions; (
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively.
  • the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10.
  • the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively.
  • the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 10.
  • the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
  • VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
  • the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11.
  • the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
  • the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 11.
  • the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-VEGF antibody or fragment thereof comprises a (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof
  • the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11.
  • the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11, wherein either (i) the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or (ii) the VH comprises the
  • the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 10, and (II) comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 11.
  • the anti-VEGF antibody or fragment thereof comprises a CH1 domain, preferably comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 12.
  • the anti-VEGF antibody or fragment thereof comprises an Fc region comprising one or more mutations which reduce or abolish immune effect functions. Suitable mutations are known in the art and may include one or more amino acid substitutions (cf., e.g., Wilkinson, I., et al., 2021. FcPLoS One, 16 (12) , p. e0260954; and Liu, R., et al., 2020. Antibodies, 9 (4) , p. 64) . In some embodiments, the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising a L234A and/or a L235A mutation.
  • the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising L234A and L235A mutations.
  • Amino acids are numbered according to the EU numbering (EU-index) as set forth in Kabat (Kabat, E.A.; National Institutes of Health (U.S. ) Office of the Director. Sequences of Proteins of Immunological Interest, 5th ed.; DIANE Publishing: Collingdale, PA, USA, 1991) . For example, these may correspond to L240A and L241A in SEQ ID NO: 16.
  • the anti-VEGF antibody or fragment thereof comprises a Fc domain comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 13.
  • the anti-VEGF antibody or fragment thereof comprises a CL domain, preferably comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 15.
  • the anti-VEGF antibody or fragment thereof comprises a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • HC heavy chain
  • the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively.
  • the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively.
  • the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80.
  • the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, wherein the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively.
  • the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of the amino acid sequence of SEQ ID NO: 80.
  • the anti-VEGF antibody or fragment thereof comprises a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • LC light chain
  • the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
  • the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
  • the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 44, respectively, or the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
  • the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-VEGF antibody or fragment thereof comprises (I) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions
  • the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, where
  • the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and (II) an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and (II) an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein either (i) the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or (ii) the HC comprises the C
  • the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of the amino acid sequence of SEQ ID NO: 80, and (II) an LC comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-VEGF antibody comprises or consists of two HCs each comprising the amino acid sequence of SEQ ID NO: 80; and two LCs each comprising the amino acid sequence of SEQ ID NO: 17.
  • the anti-VEGF antibody or fragment thereof is a full-length humanized anti-VEGF antibody comprising: (ii-1a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences of SEQ ID NO: 10, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences of SEQ ID NO: 11; (ii-1b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively; (ii-1c) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and a light chain variable region (VL
  • the second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction comprises a VEGFR domain.
  • a protein which comprises a VEGFR domain is Aflibercept. Exemplary sequences of VEGFR domains are provided in Table 1.
  • the second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction comprises at least one VEGFR-1 domain, or at least one VEGFR-2 domain, or at least one VEGFR-3 domain, or at least one VEGFR-1 domain and at least one VEGFR-2 domain, such as two VEGFR-1 domains, or two VEGFR-2 domains, or two VEGFR-1 domains and two VEGFR-2 domains, or two VEGFR-3 domains.
  • the VEGFR domain is VEGFR-1 comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 75, or is an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75.
  • the VEGFR domain is VEGFR-2 comprising or consisting of the amino acid sequence set forth in: SEQ ID NO: 76, or is an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76.
  • the VEGFR domain is VEGFR-3 comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 77, or is an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77.
  • the second binding region comprises at least one VEGFR-1 domain comprising or consisting of at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75.
  • the second binding region comprises at least one VEGFR-2 domain comprising or consisting of at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76.
  • the second binding region comprises at least one VEGFR-3 domain comprising or consisting of at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77.
  • the second binding region comprises (i) at least one VEGFR-1 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75, or at least one VEGFR-3 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76, and (ii) at least one VEGFR-2 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77.
  • the second binding region comprises (i) two VEGFR-1 domains or two amino acid sequences each having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75, (ii) two VEGFR-2 domains or two amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76, (iii) two VEGFR-3 domains or two amino acid sequences each having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77, (iv) a combination of (
  • the second binding region comprises or consists of the amino acid sequence of SEQ ID NO: 78 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 78.
  • the second binding region comprises or consists of (i) at least one VEGFR-1 or VEGFR-3 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75 or 77; (ii) at least one VEGFR-2 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76; and (iii) an Fc part.
  • the second binding region may comprise or consist of the amino acid sequence of SEQ ID NO: 79 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 79.
  • the second binding region comprises: (ii-2a) the amino acid sequence of SEQ ID NO: 75 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75; (ii-2b) the amino acid sequence of SEQ ID NO: 76 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SE Q ID NO: 76; (ii-2c) the amino acid sequence of SEQ ID NO: 77 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75
  • the second binding region comprises two amino acid sequences each comprising or consisting of the amino acid sequence of SEQ ID NO: 79.
  • the resulting second binding region may have the setup of an antibody when it comes to the Fc parts, i.e., the two Fc parts form an Fc domain via at least one disulfide bond, and the two N-terminal regions each comprise or consist of two VEGFR domains.
  • the resulting second binding region may have an Fc domain and two VEGFR-containing arms, wherein each arm comprises two VEGFR domains such that the resulting second binding region overall comprises four VEGFR domains.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises a multispecific antibody, preferably a bispecific antibody.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent can comprise any of the first and second binding regions as outlined above, in any combination.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may comprise (i) a PD-1 binding antibody or PD-1 binding fragment thereof, such as Pembrolizumab, Nivolumab, Cemiplimab, Dostarlimab, Toripalimab, and Retifanlimab, or a fragment of any of the afore-mentioned antibodies, or any of the PD-1 binding sequences as disclosed above; and (ii) a VEGF binding antibody or VEGFR-binding fragment thereof, such as Bevacizumab, Ramucirumab, and Ranibizumab, or a fragment of any of the afore-mentioned antibodies, or any of the VEGF-binding sequences as disclosed above; or a VEGFR domain or a protein comprising the same, such as
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may comprise (i) a PD-L1 binding antibody or PD-L1 binding fragment thereof, such as Atezolizumab, Durvalumab, and Avelumab, or a fragment of any of the afore-mentioned antibodies, or any of the PD-L1 binding sequences as disclosed above; and (ii) a VEGF binding antibody or VEGFR-binding fragment thereof, such as Bevacizumab, Ramucirumab, and Ranibizumab, or a fragment of any of the afore-mentioned antibodies, or any of the VEGF-binding sequences as disclosed above; or a VEGFR domain or a protein comprising the same, such as Aflibercept, or any of the VEGFR sequences as disclosed above.
  • a PD-L1 binding antibody or PD-L1 binding fragment thereof such as Atezolizumab, Durvalumab, and Avelumab
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises (i) the PD-L1 binding sequences, such as the VHH sequences and the anti-PD-L1 HCDRs thereof as disclosed above; and (ii) a VEGF binding antibody or VEGFR-binding fragment thereof as disclosed above, such as Bevacizumab.
  • Exemplary bispecific anti-PD-1/PD-L1xVEGF/VEGFR binding agents include the following: the anti-PD-1/PD-L1xVEGF/VEGFR binding agents C1, C2a, C2b, C2c, C2d, C3a, and C3b provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , SYN-2510 (ImmuneOnco/Instil Bio) , AI-081 (OncoC4) , HB0025 (Huabo Biopharm) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , CVL006 (Convalife) , DR30206 (Zhejiang Doer Bio) , HC
  • the bispecific antibody is selected from the group consisting of Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , CVL006 (Convalife) , DR30206 (Zhejiang Doer Bio) , HC010 (HC Biopharma) , and MHB039A (Minghui Pharma) .
  • Preferred anti-PD-1/PD-L1xVEGF/VEGFR binding agents are the anti-PD-1/PD-L1xVEGF/VEGFR binding agents C1, C2a, C2b, C2c, C2d, C3a, and C3b, in particular, anti-PD-1/PD-L1xVEGF/VEGFR binding agent C1.
  • the antibody comprises any combination of one or more full-length antibodies, such as a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, or a polyclonal antibody, and/or one or more antibody fragments, such as a Fab, a Fab', a F (ab') 2, an Fv, an scFv, a Fab'-SH, an sdAb, or a VHH.
  • the antibody comprises a full-length antibody and an antibody fragment, such as a Fab, a Fab', a F (ab') 2, an Fv, an scFv, a Fab'-SH, an sdAb, or a VHH.
  • the first binding region comprises an anti-PD-1 antibody or fragment thereof, or comprises an antibody fragment selected from an Fab, an Fab', an F (ab') 2, an Fv, an scFv, an Fab'-SH, an sdAb, and a VHH, preferably an scFv or a VHH.
  • the anti-PD-1 antibody or fragment thereof is an anti-PD-1 single-chain variable fragment.
  • the anti-PD-1 antibody or fragment thereof is an anti-PD-1 VHH.
  • the first binding region comprises an anti-PD-L1 antibody or fragment thereof, or comprises an antibody fragment selected from an Fab, an Fab', an F (ab') 2, an Fv, an scFv, an Fab'-SH, an sdAb, and a VHH, preferably a sdAb, most preferably a VHH.
  • the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 single domain antibody.
  • the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 VHH.
  • the second binding region comprises an anti-VEGF antibody or fragment thereof, or comprises a human antibody, a humanized antibody, or a chimeric antibody, preferably a humanized antibody.
  • the anti-VEGF antibody or fragment thereof is a monoclonal antibody, for example a monoclonal human antibody, a monoclonal humanized antibody, or a monoclonal chimeric antibody.
  • the anti-VEGF antibody or fragment thereof is a monoclonal humanized antibody.
  • the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises an anti-VEGF antibody (e.g., anti-VEGF-A antibody) and/or an anti-PD-L1 single domain antibody.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises an anti-VEGF antibody (e.g., anti-VEGF-A antibody) and/or an anti-PD-1 single-chain variable fragment.
  • the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has monovalent binding to VEGF and/or to PD-L1.
  • the bispecific antibody has monovalent binding to VEGF and/or to PD-1.
  • the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and/or to PD-L1.
  • the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and/or to PD-1.
  • the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and to PD-L1. In some preferred embodiments, the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and to PD-1.
  • the anti-VEGF antibody e.g., anti-VEGF-A antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof.
  • the anti-PD-L1 antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof.
  • the anti-PD-1 antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof.
  • the anti-VEGF antibody (e.g., anti-VEGF-A antibody) or fragment thereof is a bivalent anti-VEGF antibody or fragment thereof and/or the anti-PD-L1 antibody or fragment thereof is a bivalent antibody or fragment thereof.
  • the anti-VEGF antibody e.g., anti-VEGF-A antibody or fragment thereof is a bivalent anti-VEGF antibody or fragment thereof and/or the anti-PD-1 antibody or fragment thereof is a bivalent antibody or fragment thereof.
  • the anti-VEGF antibody e.g., anti-VEGF-A antibody
  • the anti-PD-L1 is a monovalent single domain antibody.
  • the anti-VEGF antibody e.g., anti-VEGF-A antibody
  • the anti-PD-1 is a bivalent single-chain variable fragment.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a bispecific anti-PD-L1xVEGF antibody, i.e., a bispecific antibody, wherein the first binding region of the bispecific antibody binds to PD-L1 and the second binding region binds to VEGF, preferably to VEGF-A.
  • An exemplary and preferred bispecific anti-PD-L1xVEGF antibody is the anti-PD-1/PD-L1xVEGF/VEGFR binding agent C1 provided in Table 1.
  • Exemplary bispecific anti-PD-L1xVEGF/VEGFR binding agents include the following: the anti-PD-L1xVEGF/VEGFR binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , DR30206 (Zhejiang Doer Bio) .
  • An exemplary trispecific anti-PD-L1xVEGF/VEGFR binding agent is HC010 (HC Biopharma) , which further binds to TGFbeta.
  • the anti-PD-L1xVEGF antibody comprises an anti-VEGF antibody as described herein and at least one anti-PD-L1 single domain antibody as described herein, preferably an anti-VEGF antibody as described herein and two anti-PD-L1 single domain antibodies as described herein, more preferably an anti-VEGF antibody as described herein and two anti-PD-L1 VHHs as described herein.
  • the at least one anti-PD-L1 single domain antibody may be fused to the anti-VEGF antibody.
  • each anti-PD-L1 single domain antibody is terminally fused to the anti-VEGF antibody.
  • each anti-PD-L1 single domain antibody is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain.
  • each anti-PD-L1 single domain antibody is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain.
  • the anti-PD-L1xVEGF antibody more preferably comprises an anti-VEGF antibody and two anti-PD-L1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-L1 VHH, more preferably at the C-terminus of the heavy chain of the anti-VEGF antibody.
  • the anti-PD-L1 single domain antibody (in particular, VHH) can be fused to the anti-VEGF antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSG (SEQ ID NO: 14) .
  • a linker e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSG (SEQ ID NO: 14) .
  • the anti-PD-L1xVEGF antibody comprises a linker, which more preferably comprises or consists of the amino acid sequence of SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
  • the anti-PD-L1xVEGF antibody comprises (i) an anti-VEGF antibody comprising two heavy chains as described herein; and (ii) two anti-PD-L1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-L1 VHH through a linker comprising the amino acid sequence of SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
  • the anti-PD-L1xVEGF antibody comprises two anti-PD-L1 VHHs as described herein, wherein each VHH is fused to the C-terminus of an anti-VEGF-A antibody as described herein, preferably the C-terminus of each heavy chain of the anti-VEGF-A antibody.
  • the anti-PD-L1xVEGF antibody can therefore comprise an anti-VEGF-A antibody as described herein and two anti-PD-L1 VHHs as described herein, wherein each heavy chain of the anti-VEGF-A antibody is fused to an anti-PD-L1 VHH through a linker, such as a linker comprising the amino acid sequence shown in SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
  • a linker such as a linker comprising the amino acid sequence shown in SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
  • the anti-PD-L1xVEGF antibody comprises (i) an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7, 44 and 8 (IMGT) ) or variants thereof having up to three amino acid substitutions, additions or deletions; and (ii) an anti-PD-L1 single domain comprising the HCDRs as described above (i.e., SEQ ID NOs: 18, 19, and 38 (Kabat) or SEQ ID NOs: 1, 2 and 3 (IMGT) ) , or variants thereof having up to three amino acid substitutions, additions or deletions.
  • an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7,
  • the anti-PD-L1xVEGF antibody comprises (1) at least one anti-PD-L1 single domain antibody comprising or consisting of the amino acid sequence of SEQ ID NO: 9 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 9, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 9 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or the HCDR1, HCDR2, and HCDR3 sequences set forth in: 1, 2, and 3, respectively; and/or (2) an anti-VEGF antibody comprising (i) a heavy chain
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-VEGF antibody comprises: (ii-1d) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11; (ii-1e) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (
  • the anti-PD-L1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 16, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-L1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 16, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively
  • the anti-PD-L1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 16, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively,
  • the anti-PD-L1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-L1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-L1xVEGF antibody comprises (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 16; and (ii) two light chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • anti-PD-L1xVEGF antibody comprising two heavy chain-VHH fusions having the amino acid sequence of SEQ ID NO: 16 and two light chains having the amino acid sequence of SEQ ID NO: 17 is disclosed as compound "Ava-2GS-NSD" in WO 2022/042719. C2.
  • Anti-PD-1xVEGF antibodies comprising two heavy chain-VHH fusions having the amino acid sequence of SEQ ID NO: 16 and two light chains having the amino acid sequence of SEQ ID NO: 17 is disclosed as compound "Ava-2GS-NSD" in WO 2022/042719. C2.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a bispecific anti-PD-1xVEGF antibody, i.e., a bispecific antibody, wherein the first binding region of the bispecific antibody binds to PD-1 and the second binding region binds to VEGF, preferably to VEGF-A.
  • Exemplary bispecific anti-PD-1xVEGF/VEGFR binding agents include the following: the anti-PD-1xVEGF/VEGFR binding agents C2a, C2b, C2c, C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) and MHB039A (Minghui Pharma) .
  • the anti-PD-1xVEGF antibody comprises an anti-VEGF antibody as described herein and at least one anti-PD-1 scFv as described herein, preferably an anti-VEGF antibody as described herein and at least two anti-PD-1 scFvs as described herein, more preferably an anti-VEGF antibody as described herein and two anti-PD-1 scFvs as described herein.
  • the at least one anti-PD-1 scFv may be fused to the anti-VEGF antibody.
  • each anti-PD-1 scFv is terminally fused to the anti-VEGF antibody.
  • each anti-PD-1 scFv is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain.
  • each anti-PD-1 scFv is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain.
  • the anti-PD-1xVEGF antibody more preferably comprises an anti-VEGF antibody and two anti-PD-1 scFvs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 scFv, more preferably at the C-terminus of the heavy chain of the anti-VEGF antibody.
  • the anti-PD-1 scFv can be fused to the anti-VEGF antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 88) .
  • a linker e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 88) .
  • the anti-PD-1xVEGF antibody comprises a linker, which more preferably comprises or consists of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody comprising two heavy chains as described herein; and (ii) two anti-PD-1 scFvs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 scFv through a linker, wherein the linker preferably comprises or consists of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • the anti-PD-1xVEGF antibody comprises two anti-PD-1 scFvs as described herein, wherein each anti-PD-1 scFv is fused to the C-terminus of an anti-VEGF-Aantibody as described herein, preferably the C-terminus of each heavy chain of the anti-VEGF-A antibody.
  • the anti-PD-1xVEGF antibody can therefore comprise an anti-VEGF-A antibody as described herein and two anti-PD-1 scFvs as described herein, wherein each heavy chain of the anti-VEGF-A antibody is fused to an anti-PD-1 scFv through a linker, such as a linker comprising or consisting of the amino acid sequence shown in SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • a linker comprising or consisting of the amino acid sequence shown in SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7, 44, and 8 (IMGT) ) or variants thereof having up to three amino acid substitutions, additions or deletions; and (ii) an anti-PD-1 scFv comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 51, 52, 53, 54, 55, and 50 (Kabat) or SEQ ID NOs: 45, 46, 47, 48, 49, and 50 (IMGT) ) , or variants thereof having up to three amino acid substitutions, additions or deletions.
  • an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42,
  • the anti-PD-1xVEGF antibody comprises (1) at least one anti-PD-1 scFv comprising or consisting of the amino acid sequence of SEQ ID NO: 56 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56 preferably comprises the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55, and 50, respectively, or the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in:
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-VEGF antibody comprises (a) the VH and VL sequences as specified above under (ii-1d) , (ii-1e) , or (ii-1f) , or (b) the HC and LC sequences as specified above under (ii-1g) , (ii-1h) , or (ii-1i) .
  • the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 81 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 81, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 81, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 39
  • the anti-PD-L1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 81, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in:
  • the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 81 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 81; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 81; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises or consists of (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 81; and (ii) two light chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises an anti-VEGF antibody as described herein and at least one anti-PD-1 VHH as described herein, preferably an anti-VEGF antibody as described herein and at least two anti-PD-1 VHHs as described herein, more preferably an anti-VEGF antibody as described herein and two or four anti-PD-1 VHHs as described herein.
  • the at least one anti-PD-1 VHH may be fused to the anti-VEGF antibody.
  • each anti-PD-1 VHH is terminally fused to the anti-VEGF antibody.
  • each anti-PD-1 VHH is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain.
  • each anti-PD-1 VHH is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain.
  • the anti-PD-1xVEGF antibody more preferably comprises an anti-VEGF antibody and two anti-PD-1 VHH, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 VHH, more preferably at the C-terminus of the heavy chain of the anti-VEGF antibody.
  • the anti-PD-1 VHH can be fused to the anti-VEGF antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 88) .
  • the anti-PD-1xVEGF antibody comprises a linker, which more preferably comprises or consists of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody comprising or consisting of two heavy chains as described herein; and (ii) at least two anti-PD-1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 VHH through a linker comprising or consisting of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • the anti-PD-1xVEGF antibody comprises two or four anti-PD-1 VHHs as described herein, wherein each anti-PD-1 VHH is fused to the C-terminus of an anti-VEGF-A antibody as described herein, preferably the C-terminus of each heavy chain of the anti-VEGF-A antibody.
  • the anti-PD-1xVEGF antibody can therefore comprise an anti-VEGF-A antibody as described herein and two anti-PD-1 VHHs as described herein, wherein each heavy chain of the anti-VEGF-A antibody is fused to an anti-PD-1 VHH through a linker, such as a linker comprising or consisting of the amino acid sequence shown in SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • a linker comprising or consisting of the amino acid sequence shown in SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
  • the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7, 44, and 8 (IMGT) ) or variants thereof having up to three amino acid substitutions, additions or deletions; and (ii) an anti-PD-1 VHH comprising the HCDRs as described above (i.e., SEQ ID NOs: 57, 58, 59, or 60 (Kabat) or SEQ ID NOs: 61, 62, 63, or 64 (IMGT) ) , or variants thereof having up to three amino acid substitutions, additions or deletions.
  • an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat)
  • the anti-PD-1xVEGF antibody comprises (1) at least one anti-PD-1 VHH comprising or consisting of the amino acid sequence of SEQ ID NO: 65 or 66 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 65 or 66, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 65 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or the HCDR1, HCDR2, and HCDR3 sequences set forth in: 61, 62, and 63, respectively, and the amino acid sequence having
  • the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
  • the anti-VEGF antibody comprises (a) the VH and VL sequences as specified above under (ii-1d) , (ii-1e) , or (ii-1f) , or (b) the HC and LC sequences as specified above under (ii-1g) , (ii-1h) , or (ii-1i) .
  • the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 82, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 82, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58,
  • the anti-PD-1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 82, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62,
  • the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 82; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 83 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 83, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 83, and (B) a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59,
  • the anti-PD-1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 83, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 83, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61,
  • the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 83 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 83; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 84 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 84, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 84, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58,
  • the anti-PD-1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 84, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62,
  • the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 84 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 84; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82, 83, or 84; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1xVEGF antibody comprises or consists of (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 82, the amino acid sequence of 83, or the amino acid sequence of 84; and (ii) two light chains each comprising comprising or consisting of the amino acid sequence of SEQ ID NO: 17. C3.
  • Anti-PD-L1xVEGFR antibodies comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82, 83, or 84; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a bispecific anti-PD-L1xVEGFR antibody, i.e., a bispecific antibody, wherein the first binding region of the bispecific antibody binds to PD-L1 and the second binding region comprises at least one VEGFR domain.
  • Exemplary bispecific anti-PD-L1xVEGFR antibodies are the anti-PD-1/PD-L1xVEGF/VEGFR binding agents C3a and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , and HB0025 (Huabo Biopharm) .
  • Such antibodies may alternatively be referred to as fusion proteins comprising an anti-PD-1/PD-L1 antibody or a fragment thereof and a VEGFR domain.
  • the bispecific anti-PD-L1xVEGFR antibody comprises any combination of one or more full-length antibodies, such as a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, or a polyclonal antibody, and/or one or more antibody fragments, such as a Fab, a Fab', a F (ab') 2, an Fv, an scFv, a Fab'-SH, an sdAb, or a VHH; and at least one VEGFR domain.
  • the bispecific anti-PD-L1xVEGFR antibody comprises a full-length antibody and at least one VEGFR domain.
  • the anti-PD-L1 antibody or fragment thereof comprises a human antibody, a humanized antibody, or a chimeric antibody, preferably a humanized antibody.
  • the anti-PD-L1 antibody or fragment thereof is a monoclonal antibody, for example a monoclonal human antibody, a monoclonal humanized antibody, or a monoclonal chimeric antibody.
  • Such antibodies or fragments thereof are fused to at least one, preferably at least two VEGFR domains, most preferably two VEGFR domains.
  • the anti-PD-L1 antibody or fragment thereof comprises an antibody fragment selected from a Fab, an Fab', an F (ab') 2, an Fv, an scFv, an Fab'-SH, an sdAb, and a VHH, preferably a sdAb, most preferably a VHH.
  • the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 single domain antibody.
  • the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 VHH.
  • the VEGFR domain comprising protein comprises an Fc part and at least two VEGFR fused to the N-terminus thereof.
  • the bispecific anti-PD-L1xVEGFR antibody has monovalent binding to VEGF and/or to PD-L1. In some embodiments, the bispecific anti-PD-L1xVEGFR antibody has bivalent binding to VEGF and/or to PD-L1. In some preferred embodiments, the bispecific anti-PD-L1xVEGFR antibody has bivalent binding to VEGF and to PD-L1.
  • the bispecific anti-PD-L1xVEGFR antibody comprises an anti-PD-L1 antibody, preferably a humanized anti-PD-L1 antibody, fused to a VEGFR domain. In some embodiments, the bispecific anti-PD-L1xVEGFR antibody comprises a full-length humanized antibody and two VEGFR domains.
  • each VEGFR domain may be fused to the anti-PD-L1 antibody.
  • each VEGFR domain is terminally fused to the anti-PD-L1 antibody.
  • each VEGFR domain is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain.
  • each VEGFR domain is fused to the N-terminus or the C-terminus of an anti-PD-L1 antibody, preferably the N-terminus of a heavy chain or the C-terminus of a heavy chain.
  • the bispecific antibody preferably comprises a humanized anti-PD-L1 antibody and two VEGFR domains, wherein each heavy chain of the anti-PD-L1 antibody is fused to a VEGFR domain, preferably one VEGFR domain.
  • the VEGFR domain can be fused to the anti-PD-L1 antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art.
  • a linker e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art.
  • GS-linker glycine and serine-rich linker
  • An exemplary linker is GGSGGSGGSGGSGGS (SEQ ID NO: 89) .
  • the bispecific anti-PD-L1xVEGFR antibody comprises (a) an anti-PD-L1 antibody, preferably a humanized anti-PD-L1 antibody, comprising a heavy chain variable region, wherein the heavy chain variable region comprises: (i) a HCDR1 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 67, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a HCDR2 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 68, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a HCDR3 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 69, or a variant thereof having up to three amino acid substitutions, additions or deletions; and a light chain variable region, wherein the light chain variable region comprises: (i) a LCDR1 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 67
  • the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 85, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a
  • the anti-PD-L1xVEGFR antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 85, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 86, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 70, 71,
  • the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 85; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86.
  • the anti-PD-L1xVEGFR antibody comprises or consist of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 87 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 87, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions
  • the anti-PD-L1xVEGFR antibody comprises or consists of (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 87, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 86, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO:
  • the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 87 or an amino acid sequence comprising or consisting of at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 87; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86.
  • the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or 87; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86.
  • the anti-PD-L1xVEGFR antibody comprises or consists of (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or the amino acid sequence of SEQ 87; and (ii) two light chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 86.
  • the bispecific antibody comprises an anti-PD-L1 antibody or fragment thereof. In some preferred embodiments, the bispecific antibody comprises an anti-VEGF antibody or fragment thereof, preferably an anti-VEGF-A antibody or fragment thereof.
  • the bispecific antibody comprises a Fab, F’b', F (ab') 2, Fd, Fv, sdAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH.
  • the bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably fused to the N-or C-terminus of the anti-VEGF antibody heavy or light chain. In some embodiments, the bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably fused to the C-terminus of the anti-VEGF antibody heavy chain.
  • the anti-PD-L1 antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof. In some embodiments, the anti-VEGF antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof,
  • the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region
  • the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 2, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3, or a variant thereof having up to three, e.g.
  • IMGT numbering system see Ehrenmann F, Kaas Q, Lefranc M P. IMGT/3Dstructure-DB and IMGT/DomainGapAlign: a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF [J] . Nucleic acids research, 2009; 38 (suppl_1) : D301-D307) .
  • the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region
  • the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 38, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity-determining region 3
  • the CDR variant has up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variant has up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variant has up to three amino acid substitutions. In some embodiments, the CDR variant has up to two amino acid substitutions. In some embodiments, the CDR variant has up to one amino acid substitution.
  • the amino acid sequence of the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9.
  • the amino acid sequence of the anti-PD-L1 single domain antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the bispecific antibody can comprise one anti-VEGF antibody and two anti-PD-L1 single domain antibodies, preferably fused to the C-terminus of the anti-VEGF antibody, wherein each of these anti-PD-L1 single domain antibodies comprises or consists of the amino acid sequence shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9
  • the amino acid sequence of the anti-PD-L1 single domain antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity thereto and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and comprises a heavy chain variable region comprising: a HCDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 1; a HCDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 2; and a HCDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 3.
  • a HCDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 1
  • a HCDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 2
  • a HCDR3 comprising or consisting of the amino
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity thereto and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and comprises a heavy chain variable region comprising: a HCDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 18; a HCDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 19; and a HCDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 38.
  • a HCDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 18
  • a HCDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 19
  • a HCDR3 comprising or consisting of the
  • the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9. In some embodiments, the anti-PD-L1 antibody or fragment thereof consists of the amino acid sequence of SEQ ID NO: 9.
  • the bispecific antibody specifically binds to VEGF-A.
  • the anti-VEGF antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably, the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
  • the anti-VEGF-A antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably, the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
  • the anti-VEGF antibody or fragment thereof comprises an Fc region comprising one or more mutations which reduce or abolish immune effect functions.
  • Suitable mutations are known in the art and may include one or more amino acid substitutions (see e.g. Wilkinson, I., et al., 2021. FcPLoS One, 16 (12) , p. e0260954; and Liu, R., et al., 2020. Antibodies, 9 (4) , p. 64) .
  • the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising a L234A (with the numbering according to the EU-index) and/or a L235A mutation (with the numbering according to the EU-index) .
  • the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising L234A and L235A mutations (with the numbering according to the EU-index) .
  • amino acids are numbered according to the Eu numbering (Eu-index) as set forth in Kabat (Kabat, E.A.; National Institutes of Health (U.S. ) Office of the Director. Sequences of Proteins of Immunological Interest, 5th ed.; DIANE Publishing: Collingdale, PA, USA, 1991) .
  • the anti-VEGF antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO: 13.
  • the anti-VEGF-A antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO: 13 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 13.
  • each anti-PD-L1 single domain antibody may be fused to the anti-VEGF antibody by any suitable method.
  • each anti-PD-L1 single domain antibody is terminally-fused to the anti-VEGF antibody.
  • each anti-PD-L1 single domain antibody is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain.
  • each anti-PD-L1 single domain antibodies is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain.
  • the bispecific antibody preferably comprises an anti-VEGF antibody and two anti-PD-L1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-L1 VHH.
  • the VHH can be fused to the anti-VEGF antibody by a linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art.
  • a linker for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art.
  • An example linker is provided by SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 14.
  • the anti-VEGF antibody or fragment thereof comprises a CL, preferably having the amino acid sequence shown in SEQ ID NO: 15 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 15.
  • the anti-VEGF antibody or fragment thereof comprises a CH1, preferably having the amino acid sequence shown in SEQ ID NO: 12 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 12.
  • the anti-VEGF antibody comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6, or a variant thereof having up to three e.g.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity-determining region 3
  • the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions.
  • LCDR1 complementarity-determining region 1
  • LCDR2 complementarity-determining region 2
  • LCDR3 complementarity-determining region 3
  • IMGT/3Dstructure-DB and IMGT/DomainGapAlign a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF [J] . Nucleic acids research, 2009; 38 (suppl_1) : D301-D307) .
  • the anti-VEGF antibody comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 39, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 40, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 41, or a variant thereof having up to three e.g.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity-determining region 3
  • the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 42, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 43, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions.
  • LCDR1 complementarity-determining region 1
  • LCDR2 complementarity-determining region 2
  • LCDR3 complementarity-determining region 3
  • the CDR variant has up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variant has up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variant has up to three amino acid substitutions. In some embodiments, the CDR variant has up to two amino acid substitutions. In some embodiments, the CDR variant has up to one amino acid substitution.
  • the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody (or anti-VEGF-A antibody) is shown in SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 11; or wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 11.
  • the amino acid sequence of the heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17.
  • the anti-VEGF-A antibody is bevacizumab.
  • the bispecific antibody is encoded by one or more nucleic acid molecules.
  • the bispecific antibody can preferably comprise: a. two anti-PD-L1 single domain antibodies each comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 2, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3; and b.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity-determining region 3
  • an anti-VEGF-A antibody comprising a heavy chain variable region comprising: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and a light chain variable region comprising: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
  • HCDR1 complementarity-determining region 1
  • HCDR2 complementarity-determining region 2
  • HCDR3 complementarity
  • bispecific antibody or bispecific antibody variants are described in WO 2022/042719, which disclosure is hereby incorporated in its entirety.
  • the bispecific antibody can be produced by various methods, see, for example, the method of Songsivilai et al. (Clin. Exp. Immunol., 79: 315-321 (1990) ) , and the method of Kostelny et al. (J. Immunol., 148: 1547-1553 (1992) which disclosures are also hereby incorporated in their entirety.
  • Regimen of the present invention Administration and regimens with a focus on the binding agent
  • the binding agent (including, e.g., the bispecific antibody) and the chemotherapy are separately administered.
  • a dosage of the binding agent (including, e.g., the bispecific antibody) and a dosage of the chemotherapy are separately administered.
  • a dosage of the binding agent (including, e.g., the bispecific antibody) and a dosage of the chemotherapy are administered using a single composition.
  • a dosage of the binding agent including, e.g., the bispecific antibody
  • a dosage of the chemotherapy are administered concurrently or consecutively.
  • the binding agent (including, e.g., the bispecific antibody) and the chemotherapy provided herein can be administered via any suitable enteral route or parenteral route of administration.
  • enteral route refers to the administration via any part of the gastrointestinal tract. Examples of enteral routes include oral, mucosal, buccal, and rectal route, or intragastric route.
  • Parenteral route refers to a route of administration other than enteral route.
  • parenteral routes of administration examples include intravenous, intramuscular, intradermal, intraperitoneal, intratumour, intravesical, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, transtracheal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal, subcutaneous, or topical administration.
  • the binding agent including, e.g., the bispecific antibody
  • the chemotherapy of the disclosure can be administered using any suitable method, such as by oral ingestion, nasogastric tube, gastrostomy tube, injection, infusion, implantable infusion pump, and osmotic pump.
  • treatment regimen ” “dosing protocol, ” and “dosing regimen” are used interchangeably to refer to the dosage and timing of administration of each dosage binding agent (including, e.g., the bispecific antibody) and the chemotherapy in a combination therapy of the disclosure.
  • a parenteral route of administration of the binding agent and the chemotherapy is particularly preferred, wherein an intravenous administration is particularly preferred.
  • the binding agent (including, e.g., the bispecific antibody) and/or the chemotherapy is administered intravenously, preferably wherein the binding agent (including, e.g., the bispecific antibody) and the chemotherapy are administered intravenously.
  • the binding agent (including, e.g., the bispecific antibody) and/or the chemotherapy is administered via an IV injection or IV infusion.
  • the binding agent (including, e.g., the bispecific antibody) and the chemotherapy are administered via an IV injection or IV infusion.
  • the binding agent (including, e.g., the bispecific antibody) and the chemotherapy can be administered concurrently or consecutively via an IV infusion.
  • the binding agent (including, e.g., the bispecific antibody) is administered in a step-up dosing regimen to a subject or patient, e.g., with a first and one or more subsequent dose regimens.
  • the dosage of the binding agent including, e.g., the bispecific antibody
  • the dosage of the binding agent is lower than the dosage of the binding agent (including, e.g., the bispecific antibody) in a second dose regimen.
  • the dosage of the binding agent (including, e.g., the bispecific antibody) in each subsequent dosage regimen is higher compared to the dosage of the binding agent (including, e.g., the bispecific antibody) in the dose regimen that precedes each subsequent dose regimen.
  • the dosage of the chemotherapeutic agent may be the same in the first and one or more subsequent dose regimens. In some embodiments of the afore mentioned examples, in the first dose regimen the dosage of the chemotherapeutic agent may be lower than the dosage of the chemotherapeutic agent in a second dose regimen.
  • the dosage of the chemotherapeutic agent in each subsequent dosage regimen may be higher compared to the dosage of the chemotherapeutic agent in the dose regimen that precedes each subsequent dose regimen.
  • the subject has been determined to have a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 30, preferably ⁇ 20, more preferably ⁇ 10, and most preferably ⁇ 10 or even ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP, preferably TAP.
  • CPS combined positive score
  • the subject has been preferably determined to have a PD-L1 expression score before the treatment as determined by a CPS of up to 20, preferably from 1 to 20 or > 0 to 20, more preferably from 1 to ⁇ 10, such as 1 to 9, or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • the subject has been determined to have a PD-L1 expression score as determined by a CPS from 1 to 9, 1 to 8, 1 to 7, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • the subject has a PD-L1 expression score as determined by a CPS of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, preferably a CPS of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, more preferably a CPS of 1, 2, 3, 4, 5, 6, 7, 8, or 9 or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
  • the subject has been determined to have a PD-L1 expression score as determined by CPS of ⁇ 10, ⁇ 9, ⁇ 8, ⁇ 7, ⁇ 6, ⁇ 5, ⁇ 4, or ⁇ 3 before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
  • the subject has been determined to have a PD-L1 expression score as determined by CPS of > 0 before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
  • the subject is part of a patient group to be treated with the combination therapy disclosed herein.
  • this patient group comprises one or more subjects having a PD-L1 expression score as determined by a CPS of ⁇ 30, preferably ⁇ 20, more preferably ⁇ 10, and most preferably ⁇ 10 or even ⁇ 1, before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
  • This patient group may further comprise one or more subjects having a PD-L1 expression score as determined by a CPS of > 30, preferably >20, more preferably > 10, and most preferably ⁇ 10 or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
  • the binding agent (including, e.g., the bispecific antibody) is administered in a dosage ranging from 0.1 mg/kg to 45 mg/kg body weight, preferably 1 mg/kg to 30 mg/kg body weight per treatment.
  • the treatment comprises repeated treatments, wherein the treatment cycle is repeated at least 1, 2, 3, 4, 5, 6, 7, or 8 times, wherein each cycle preferably has up to 30, preferably 28 days or 21 days. However, the treatment can be continued until disease progression or the initiation of a new anti-tumour treatment.
  • the subject has not been previously treated for cancer, i.e., is treatment naive. In some embodiments, the subject has been previously treated for cancer, in particular the subject has had at least one previous chemotherapy treatment.
  • the subject has not been previously treated with a PD-1/PD-L1 inhibitor, such as pembrolizumab or atezolizumab.
  • a PD-1/PD-L1 inhibitor such as pembrolizumab or atezolizumab.
  • the binding agent (including, e.g., the bispecific antibody) is administered every 6 weeks, preferably every 4 weeks, more preferably every 3 weeks, and most preferably every 2 weeks.
  • the binding agent (including, e.g., the bispecific antibody) is administered every 2 weeks at a dosage ranging from 10 mg/kg to 30 mg/kg, preferably ranging from 15 mg/kg to 25 mg/kg, more preferably being 20 mg/kg.
  • the binding agent (including, e.g., the bispecific antibody) is administered every 3 weeks at a dosage ranging from 20 mg/kg to 40 mg/kg, preferably ranging from 25 mg/kg to 35 mg/kg, more preferably being 30 mg/kg.
  • the chemotherapy is administered once or more within the first 20 days or the first 21 days of each cycle, wherein the chemotherapy is administered twice or more within the first 20 days or the first 21 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 20 days or the first 21 days of each cycle.
  • the chemotherapy is administered once or more within the first 14 days or the first 15 days of each cycle, wherein the chemotherapy is administered twice or more within the first 14 days or the first 15 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 14 days or the first 15 days of each cycle.
  • the chemotherapy can be administered on the 1 st , 8 th , and 15 th day of each cycle, wherein each cycle has 21 days.
  • the treatment method disclosed herein results in increased overall survival in said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and an anti-PD-L1 antibody or chemotherapy and an anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  • the treatment method disclosed herein results in increased median progression-free survival said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or anti-PD-L1 antibody or anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  • the binding agent (including, e.g., the bispecific antibody) is administered concurrently with chemotherapy.
  • the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered within up to 15 days of each other.
  • the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered within about two days of each other.
  • the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered within about one day of each other.
  • the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered concurrently (for example by simultaneous (same day) administration) .
  • the binding agent (including, e.g., the bispecific antibody) is administered on day 1 of the chemotherapy treatment cycle.
  • the observed toxicities of the treatment with the binding agent are similar to those commonly seen with either chemotherapy or immunotherapy alone.
  • the chemotherapy comprises a chemotherapy agent selected from lurbinectedin, topotecan, taxane, such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
  • a chemotherapy agent selected from lurbinectedin, topotecan, taxane, such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
  • the chemotherapy comprises a chemotherapy agent selected from topotecan, taxane, such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
  • a chemotherapy agent selected from topotecan, taxane, such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
  • the platinum-based chemotherapy comprises cisplatin, oxaliplatin or carboplatin.
  • the chemotherapy agent is selected from the group consisting of a (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  • a platinum-based chemotherapy agent preferably carboplatin, cisplatin or oxaliplatin
  • an antimetabolite chemotherapy agent preferably 5-
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with paclitaxel, lurbinectedin, or topotecan. In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with paclitaxel or topotecan.
  • the cancer is small cell lung cancer and, preferably, the method of treatment can be a second line cancer treatment.
  • the method of treatment comprising administering to a patient suffering from advanced small cell lung cancer as second line treatment the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with paclitaxel every 3 weeks at a dosage of 175 mg/m 2 .
  • the combination may be administered for five cycles followed by the administration of the binding agent (including, e.g., the bispecific antibody) only as maintenance therapy.
  • the method of treatment comprising administering to a patient suffering from advanced small cell lung cancer as second line treatment the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with topotecan at a dosage of 1.5 mg/m 2 administered IV or 2.3 mg/m 2 administered orally once per day on days 1 to 5 of every 3 weeks cycle.
  • the patient may have progressed during or after first-line platinum-based therapy (with or without checkpoint inhibitor therapy) .
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin.
  • the cancer is a malignant mesothelioma (including malignant pleural mesothelioma [MPM] and malignant peritoneal mesothelioma [MPeM] ) and, preferably, the method of treatment can be a first line cancer treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin. In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with paclitaxel and carboplatin.
  • the cancer is NSCLC, preferably the method of treatment can be a first line treatment. In one example, the cancer is non-squamous NSCLC. In another example, the cancer is squamous NSCLC.
  • pemetrexed and carboplatin can be administered at a dosage of 500 mg/m 2 and AUC 5 (preferably with a total dose of ⁇ 750 mg) , respectively, every 3 weeks, e.g. for 4 cycles.
  • the cancer is non-squamous NSCLC.
  • paclitaxel and carboplatin can be administered at a dosage of 200 mg/m 2 and AUC 6 (preferably with a total dose of ⁇ 900 mg) , respectively, every 3 weeks, e.g. for 4 cycles.
  • the cancer is squamous NSCLC.
  • the cancer is a solid tumour.
  • the cancer is selected from the group consisting of malignant mesothelioma, hepatocellular cancer, neuroendocrine neoplasm, melanoma, lung cancer, liver cancer, stomach cancer, renal (cell) cancer, urothelial cancer, cervical cancer, colorectal cancer, ovarian cancer, colon cancer, breast cancer, esophagus cancer, head and neck cancer and pancreatic cancer, preferably the cancer is selected from urothelial cancer, breast cancer and esophagus cancer.
  • the cancer can be selected from non-small cell lung cancer (NSCLC) , small cell lung cancer (SCLC) including extensive stage small cell lung cancer and limited stage small cell lung cancer, urothelial carcinoma, esophageal cancer, head and neck squamous cell carcinoma (HNSCC) , triple-negative breast cancer (TNBC) or cervical cancer.
  • the cancer can preferably be TNBC, gastric adenocarcinoma, gastroesophageal junction (GEJ) adenocarcinoma, esophageal squamous-cell carcinoma, cervical cancer, urothelial carcinoma, or HNSCC.
  • NSCLC non-small cell lung cancer
  • SCLC small cell lung cancer
  • HNSCC head and neck squamous cell carcinoma
  • TNBC triple-negative breast cancer
  • the cancer can preferably be TNBC, gastric adenocarcinoma, gastroesophageal junction (GEJ) adenocarcinoma, esophageal squam
  • the cancer is selected from the group consisting of small cell lung cancer, non-small cell lung cancer, triple-negative breast cancer, malignant mesothelioma, hepatocellular cancer, neuroendocrine neoplasm, and colorectal cancer.
  • the cancer is TNBC.
  • the TNBC may be a TNBC of the subtype (1) luminal androgen receptor (LAR) , (2) immunomodulatory (IM) , (3) basal-like immune-suppressed (BLIS) , or (4) mesenchymal-like (MES) according to the Fudan University Shanghai Cancer Center (FUSCC) classification (see Jiang YZ, Ma D, Suo C, et al. Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies. Cancer Cell. 2019; 35 (3) : 428-440. e5) , preferably TNBC of the subtype IM or MES.
  • LAR luminal androgen receptor
  • IM immunomodulatory
  • BLIS basal-like immune-suppressed
  • MES mesenchymal-like
  • the cancer is small cell lung cancer (SCLC) , non-small cell lung cancer (NSCLC) or triple-negative breast cancer (TNBC) , preferably the cancer is triple-negative breast cancer, for example advanced or metastatic triple-negative breast cancer.
  • SCLC small cell lung cancer
  • NSCLC non-small cell lung cancer
  • TNBC triple-negative breast cancer
  • the cancer is triple-negative breast cancer, for example advanced or metastatic triple-negative breast cancer.
  • the NSCLC has a squamous histology. In some embodiments, the NSCLC has a non-squamous histology. In some embodiments, the NCLC is an EGFR mutation-positive NSCLC. Administration and regimens
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin or in combination with a taxane, such as paclitaxel or docetaxel, preferably docetaxel.
  • the cancer is NSCLC, preferably the method of treatment can be a second line treatment.
  • the cancer is an EGFR-mutant advanced non-squamous NSCLC and, preferably, the method of treatment can be a cancer treatment following a failed EGFR-TKI treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin to a patient suffering from NSCLC as second-line treatment, in particular to a patient suffering from NSCLC who progressed after EGFR-TKI treatment.
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent is administered at a dosage of 30 mg/kg plus AUC 5 mg/mL/min carboplatin and 500 mg/m 2 pemetrexed every three weeks, e.g. for 4 cycles.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with a taxane, such as paclitaxel or docetaxel, preferably docetaxel, to a patient suffering from NSCLC, e.g.
  • a taxane such as paclitaxel or docetaxel, preferably docetaxel
  • squamous NSCLC as second-line treatment, e.g., to a patient suffering from second-line stage IV or recurrent NSLC who progressed at least one prior line of an immune-oncology (IO) comprising treatment such as an immune checkpoint inhibitor comprising treatment or chemoimmunotherapy.
  • IO immune-oncology
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent is administered at a dosage range of 1400 mg to 2000 mg (with 1400 mg or 2000 mg being preferred) plus 75 mg/m 2 docetaxel every three weeks.
  • the binding agent may be administered at a dosage of 2000 mg binding agent (including, e.g., the bispecific antibody) plus 75 mg/m 2 docetaxel every three weeks.
  • the binding agent may be administered in a step-up dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is 1400 mg binding agent (including, e.g., the bispecific antibody) plus 60 mg/m 2 docetaxel every three weeks or 1400 mg binding agent (including, e.g., the bispecific antibody) plus 75 mg/m 2 docetaxel every three weeks and the second dose regimen is 2000 mg binding agent (including, e.g., the bispecific antibody) plus 75 mg/m 2 docetaxel every three weeks.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone, nab-paclitaxel, paclitaxel, or gemcitabine and carboplatin, preferably nab-paclitaxel, paclitaxel, gemcitabine and carboplatin or eribulin.
  • the cancer is TNBC (e.g., advanced or metastatic TNBC) and, preferably, the method of treatment can be a first line cancer treatment or second line cancer treatment.
  • nab-paclitaxel can be administered at a dosage of 100 mg/m 2 on the 1 st , 8 th , and 15 th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 15 mg/kg or 20 mg/kg or 1400 mg on the 1 st and 15 th day of a 28-days treatment cycle) .
  • the binding agent including, e.g., the bispecific antibody
  • paclitaxel can be administered at a dosage of 90 mg/m 2 on the 1 st , 8 th , and 15 th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg on the 1 st and 15 th day of a 28-days treatment cycle) .
  • the binding agent including, e.g., the bispecific antibody
  • Gemcitabine and carboplatin can be administered at a dosage of 1000 mg/m 2 and AUC 2 respectively, on the 1 st and 8 th day of a 21-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg on the 1 st day of a 21-days treatment cycle) .
  • Eribulin can be administered at a dosage of 1.4 mg/m 2 on the 1 st and 8 th day of a 21-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg on the first day of a 21-days treatment cycle) .
  • Nab-paclitaxel can be administered at a dosage of 100 mg/m 2 on the 1 st , 8 th , and 15 th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg on the 1 st and 15 th day of a 28-days treatment cycle) .
  • paclitaxel can be administered at a dosage of 90 mg/m 2 on the 1 st , 8 th , and 15 th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg on the 1 st and 15 th day of a 28-days treatment cycle) .
  • gemcitabine can be administered at a dosage of 1000 mg/m 2 and carboplatin can be administered at AUC2 on the 1 st and 8 th , and 15 th day of a 21-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg every 3 weeks) .
  • the patient may have a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 10 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • CPS combined positive score
  • the patient may have a PD-L1 expression before the treatment that provides a score of ⁇ 10 when determined by the combined positive scope (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating score.
  • the treatment may be a first line cancer treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with FOLFIRNOX, preferably comprising folinic acid, oxaliplatin, Irinotecan Hydrochloride, and 5-fluorouracil.
  • FOLFIRNOX folinic acid, oxaliplatin, Irinotecan Hydrochloride, and 5-fluorouracil.
  • the cancer is hepatocellular carcinoma and, preferably, the method of treatment can be a first line cancer treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) , e.g. at a dosage of 20 mg/kg every two weeks, in combination with oxaliplatin, calcium folinate, and 5-fluorouracil (which may also be referred to as FOLFOX4) .
  • the cancer is hepatocellular carcinoma and, preferably, the method of treatment can be a first line cancer treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with irinotecan, 5-fluorouracil, calcium folinate.
  • the cancer is unresectable neuroendocrine neoplasm and, preferably, the method of treatment can be a second line cancer treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with etoposide and platinum.
  • the cancer is small cell lung cancer, e.g., extensive-stage small cell lung cancer or limited-stage small cell lung cancer, and, preferably, the method of treatment can be a first line cancer treatment.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg in combination with etoposide with carboplatin to a subject having small cell lung cancer, e.g., extensive-stage small cell lung cancer or limited-stage small cell lung cancer.
  • the binding agent including, e.g., the bispecific antibody
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (preferably at 20 mg/kg or 30 mg/kg) in combination with paclitaxel, lurbinectedin, or topotecan to a subject having small cell lung cancer, e.g., extensive-stage small cell lung cancer.
  • this method of treatment is a second line treatment, in particular a treatment of SCLC patients who progressed during or after first-line platinum-based therapy (with or without checkpoint inhibitor therapy) .
  • this method of treatment comprises paclitaxel, in particular paclitaxel every 3 weeks at a dosage of 175 mg/m 2 .
  • this method of treatment comprises topotecan, in particular topotecan at a dosage of 1.5 mg/m 2 administered IV or 2.3 mg/m 2 administered orally once per day on days 1 to 5 of every three weeks cycle.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage ranging from 1400 mg to 2000 mg in combination with etoposide with carboplatin to a subject having small cell lung cancer, e.g., extensive-stage small cell lung cancer.
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent including, e.g., the bispecific antibody
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 2 weeks at a dosage ranging from 10 mg/kg to 20 mg/kg, e.g. 15 mg/kg or 20 mg/kg, preferably on the 1 st and 15 th day of a 28 day cycle, in combination with nab-paclitaxel, paclitaxel, gemcitabine with carboplatin, or eribulin to a subject having triple-negative breast cancer.
  • the binding agent including, e.g., the bispecific antibody
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) at a flat dosage, e.g. at a dosage of 500 mg to 3000 mg, 750 mg to 2500 mg, 1000 mg to 2250 mg or 1500 mg to 2000 mg to a subject. 2000 mg may be particularly preferred.
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent is administered every week or every 2, 3 or 4 weeks, preferably every 2 weeks. Preferred is an amount of 2000 mg administered every 3 weeks.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 2 weeks at a dosage of 500 mg to 3000 mg, 750 mg to 2500 mg, 1000 mg to 2250 mg or 1500 mg to 2000 mg to a subject, preferably of 1000 mg or 1400 mg to a subject having triple-negative breast cancer, preferably on the 1 st and 15 th day of a 28-day treatment cycle.
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent including, e.g., the bispecific antibody
  • a dosage of 1400 mg preferably on the 1 st and 15 th day of a 28-days treatment cycle
  • to administer the binding agent including, e.g., the bispecific antibody
  • a dosage of 2000 mg preferably on the 1 st day of a 21-days treatment cycle
  • the binding agent (including, e.g., the bispecific antibody) is administered in combination with a chemotherapy.
  • the chemotherapy is selected from the group consisting of nab-paclitaxel, paclitaxel, or gemcitabine with carboplatin.
  • the chemotherapy is nab-paclitaxel.
  • the chemotherapy is administered on the 1 st , 8 th and 15 th day of a 28-day treatment cycle.
  • the subject has not received prior systemic treatment, preferably not in the advanced setting.
  • the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 2 weeks at a dosage of 500 mg to 3000 mg, 750 mg to 2500 mg, 1000 mg to 2250 mg or 1500 mg to 2000 mg to a subject, preferably of 1000 mg or 1400 mg to a subject having small cell lung cancer, preferably on the 1 st day of a 21-day treatment cycle.
  • the binding agent including, e.g., the bispecific antibody
  • the binding agent (including, e.g., the bispecific antibody) is administered in combination with a chemotherapy, preferably selected from paclitaxel or topotecan.
  • a chemotherapy preferably selected from paclitaxel or topotecan.
  • the chemotherapy is paclitaxel.
  • the paclitaxel is administered on the 1 st day of a 21-day treatment cycle.
  • the chemotherapy is topotecan.
  • the topotecan is administered on the 1 st and 5 th day of a 21-day treatment cycle.
  • the invention further provides a chemotherapy agent for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) ; and b. the chemotherapy agent; optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the chemotherapy agent optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • CPS combined positive score
  • the embodiments disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in a method of treatment of the chemotherapy agent in combination with the binding agent (including, e.g., the bispecific antibody) as disclosed herein.
  • the “binding agent (including, e.g., the bispecific antibody) for use in a method of treating” embodiments equally apply to the chemotherapy agent for use in a method of treating disclosed herein.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with paclitaxel, wherein the antibody is administered every three weeks at a dosage of about 20 mg/kg or about 30 mg/kg and the paclitaxel is administered every three weeks at a dosage of about 175 mg/m 2 .
  • a binding agent preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with paclitaxel,
  • the patient progressed during or after first-line platinum-based therapy (with or without immuno-oncology (IO) , e.g., checkpoint inhibitor therapy) , i.e. the patient failed first-line platinum-based chemotherapy.
  • first-line platinum-based therapy with or without immuno-oncology (IO) , e.g., checkpoint inhibitor therapy
  • IO immuno-oncology
  • the method of the present paragraph is a second-line treatment for a patient suffering from small cell lung cancer.
  • the combination is administered intravenously.
  • the treatment may take place for 5 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 20 mg/kg or about 30 mg/kg every three weeks.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with topotecan, e.g., wherein the antibody is administered every three weeks at a dosage of about 20 mg/kg or about 30 mg/kg and the topotecan is administered every three weeks at days 1 to 5 once daily at a dosage of about 1.5 mg/m 2 IV or about 2.3 mg/m 2 orally.
  • a binding agent preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with small cell lung cancer, the method
  • the patient progressed during or after first-line platinum-based therapy (with or without immuno-oncology (IO) , e.g., checkpoint inhibitor therapy) , i.e. the patient failed first-line platinum-based chemotherapy.
  • first-line platinum-based therapy with or without immuno-oncology (IO) , e.g., checkpoint inhibitor therapy
  • IO immuno-oncology
  • the method of the present paragraph is a second-line treatment for a patient suffering from small cell lung cancer. It is also preferred that the antibody is administered intravenously.
  • systemic chemotherapy in particular with platinum-etoposide
  • the patient has not received systemic treatment for extensive-stage small cell lung cancer prior to the administration, i.e. the treatment is a first-line treatment.
  • the method of the present paragraph is a first-line treatment for a patient suffering from extensive-stage small cell lung cancer.
  • the antibody is administered intravenously. The treatment may take place for 4 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 20 mg/kg or about 30 mg/kg every three weeks.
  • LS-SCLC limited-stage small cell lung cancer
  • the patient had treatment-free interval (TFI) ⁇ 6 months since the last chemotherapy, chemoradiotherapy or radiotherapy.
  • TKI treatment-free interval
  • the antibody is administered intravenously.
  • the treatment may take place for 4 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 20 mg/kg or about 30 mg/kg every three weeks.
  • systemic chemotherapy in particular with platinum-etoposide
  • the patient has not received systemic treatment for extensive-stage small cell lung cancer prior to the administration, i.e. the treatment is a first-line treatment.
  • the method of the present paragraph is a first-line treatment for a patient suffering from extensive-stage small cell lung cancer.
  • the antibody is administered intravenously. The treatment may take place for 4 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 1400 mg or about 2000 mg every three weeks.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with triple negative breast cancer (TNBC) , the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy, wherein the systemic chemotherapy is nab-paclitaxel, which can preferably be administered at a dosage of about 100 mg/m 2 on the 1 st , 8 th , and 15 th day of a 28-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 15 mg/kg or about 20 mg/kg on the 1 st and 15 th day of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is paclitaxel, which can preferably be administered at a dosage of about 90 mg/m 2 on the 1 st and
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with triple negative breast cancer (TNBC) , the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy, wherein the systemic chemotherapy is nab-paclitaxel, which can preferably be administered at a dosage of about 100 mg/m 2 on the 1 st , 8 th , and 15 th day of a 28-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 1400 mg every two weeks of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is paclitaxel, which can preferably be administered at a dosage of about 90 mg/m 2 on the 1 st , 8 th , and 15 th day of
  • the antibody is administered intravenously. It is further preferred that the patient suffering from TNBC has not received systemic treatment for TNBC prior to the administration, i.e., the treatment is a first-line treatment.
  • the TNBC may be locally recurrent inoperable or metastatic TNBC.
  • the present method is in particular applicable for a patient with TNBC whose PD-L1 status is considered negative based on current clinical practice and thus wherein the patient has a PD-L1 expression before the treatment that provides a score of ⁇ 10 when determined by the combined positive score (CPS) (or a score that is indicative of such PD-L1 expression when determined by another integrating scoring algorithm) .
  • CPS combined positive score
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with non-small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular carboplatin and pemetrexed) , wherein the antibody is preferably administered every three weeks at a dosage of about 30 mg/kg together with the chemotherapy (in particular carboplatin and pemetrexed at a dosage of about AUC 5 mg/mL/min carboplatin and about 500 mg/m 2 pemetrexed) .
  • chemotherapy in particular carboplatin and pemetrexed
  • the patient progressed after first-line EGFR-TKI therapy, i.e. the patient failed first-line EGFR-TKI therapy.
  • the method of the present paragraph is a second-line treatment for a patient suffering from non-small cell lung cancer.
  • the patient suffers from a NSCLC with PD-L1 expressed, i.e. with a PD-L1 expression before the treatment that provides a score of > 1%when determined by the tumour proportion score (TPS) (or a score that is indicative of such PD-L1 expression when determined by another integrating scoring algorithm) .
  • TPS tumour proportion score
  • patients with a PD-L1 expression before the treatment that provides a score of ⁇ 1%when determined by the tumour proportion score (TPS) also respond to the method of treatment of the present paragraph such that also a patient suffering from NSCLC with a PD-L1 expression before the treatment that provides a score of ⁇ 1%when determined by the TPS (or a score that is indicative of such PD-L1 expression when determined by another integrating scoring algorithm) can effectively be treated.
  • TPS tumour proportion score
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with non-small cell lung cancer (NSCLC) , e.g., to a patient suffering from NSCLC as second-line treatment, e.g., to a patient suffering from second-line stage IV or recurrent NSCLC or, e.g., to a patient who progressed at least one prior line of an immune-oncology (IO) -comprising treatment (such as an immune checkpoint inhibitor-comprising treatment) , wherein the method comprises administering to the subject the bispecific antibody (preferably at a dosage range of about 1400 mg to about 2000 mg [with about 1400 mg or about 2000 mg being more preferred] every three weeks) and docetaxel (preferably at about 75 mg/m
  • the bispecific antibody may be administered at a dosage of about 2000 mg bispecific antibody plus about 75 mg/m 2 docetaxel every three weeks.
  • the bispecific antibody may be administered in a step-up dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 1400 mg bispecific antibody plus about 60 mg/m 2 docetaxel every three weeks or about 1400 mg bispecific antibody plus about 75 mg/m 2 docetaxel every three weeks or about 2000 mg bispecific antibody plus about 60 mg/m 2 docetaxel every three weeks and the second dose regimen is about 2000 mg bispecific antibody plus about 75 mg/m 2 docetaxel every three weeks.
  • the bispecific antibody may be administered in a step-down dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 2000 mg bispecific antibody plus about 75 mg/m 2 docetaxel every three weeks and the second dose regimen is about 1400 mg bispecific antibody plus about 75 mg/m 2 docetaxel every three weeks or about 2000 mg bispecific antibody plus about 60 mg/m 2 docetaxel every three weeks.
  • Combination (i) may in particular be administered to a patient suffering from non-squamous NSCLC, whereas combination (ii) may in particular be administered to a patient suffering from squamous NSCLC, wherein the maintenance therapy may be at a dosage of about 1400 mg or about 2000 mg bispecific antibody plus optionally pemetrexed (preferably at a dosage of about 500 mg/m 2 ) for patient (i) and a dosage of about 1400 mg or about 200 mg bispecific antibody for patient (ii) , wherein the administration every three weeks is preferably maintained during maintenance therapy.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with hepatocellular cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular FOLFOX4, i.e. oxaliplatin, leucovorin and 5-fluorouracil) , wherein the antibody is administered every two weeks at a dosage of about 20 mg/kg together with the chemotherapy.
  • chemotherapy in particular FOLFOX4, i.e. oxaliplatin, leucovorin and 5-fluorouracil
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with hepatocellular cancer, the method comprising administering to the subject the bispecific antibody (preferably as the sole active agent) , wherein the antibody is administered every two weeks at a dosage of about 20 mg/kg.
  • the patient has not received systemic treatment for hepatocellular cancer prior to the administration, i.e. the treatment is a first-line treatment.
  • the method of the present paragraph is a first-line treatment for a patient suffering from hepatocellular cancer.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with malignant mesothelioma (preferably unresectable malignant mesothelioma) , the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular carboplatin and pemetrexed or cisplatin and pemetrexed) , wherein the antibody is preferably administered every three weeks at a dosage of about 30 mg/kg together with the chemotherapy (in particular carboplatin and pemetrexed at a dosage of about AUC 5 mg/mL/min carboplatin and about 500 mg/m 2 pemetrexed or cisplatin and pemetrexed at a dosage of about 75 mg/m 2 cisplatin and about 500
  • the malignant mesothelioma may be malignant pleural mesothelioma (MPM) or malignant peritoneal mesothelioma (MPeM) .
  • the patient has not received systemic treatment for malignant mesothelioma prior to the administration, i.e. the treatment is a first-line treatment.
  • the method of the present paragraph is a first-line treatment for a patient suffering from malignant mesothelioma.
  • the antibody is administered intravenously. The treatment may take place for 4 to 6 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 30 mg/kg every three weeks.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with neuroendocrine neoplasms (NEN) , the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular FOLFIRI, i.e.
  • the antibody is preferably administered every two weeks at a dosage of about 20 mg/kg together with the chemotherapy (in particular irinotecan at a dosage of about 180 mg/m 2 , leucovorin at a dosage of about 400 mg/m 2 , and 5-fluorouracil at a dosage of about 2800 mg/m 2 ) .
  • the patient has received systemic treatment for NEN prior to the administration, i.e. the treatment is a second-line treatment.
  • the method of the present paragraph is a second-line treatment for a patient suffering from NEN. It is also preferred that the antibody and the chemotherapy is administered intravenously.
  • the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with colorectal cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy.
  • a binding agent preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with colorectal cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy.
  • Ivonescimab may be administered at a dosage of about 2000 mg plus about 75 mg/m 2 docetaxel every three weeks.
  • Ivonescimab may be administered in a step-up dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 1400 mg Ivonescimab plus about 60 mg/m 2 docetaxel every three weeks or about 1400 mg Ivonescimab plus about 75 mg/m 2 docetaxel every three weeks or about 2000 mg Ivonescimab plus about 60 mg/m 2 docetaxel every three weeks and the second dose regimen is about 2000 mg Ivonescimab plus about 75 mg/m 2 docetaxel every three weeks.
  • Ivonescimab may be administered in a step-down dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 2000 mg Ivonescimab plus about 75 mg/m 2 docetaxel every three weeks and the second dose regimen is about 1400 mg Ivonescimab plus about 75 mg/m 2 docetaxel every three weeks or about 2000 mg Ivonescimab plus about 60 mg/m 2 docetaxel every three weeks.
  • the binding agent preferably the bispecific antibody that specifically binds to PD-L1 and VEGF (most preferably PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with (i) etoposide and carboplatin; (ii) paclitaxel; (iii) topotecan; (iv) Nab-paclitaxel; (v) paclitaxel; (vi) gemcitabine; (vii) eribulin; (viii) carboplatin and pemetrexed; (ix) carboplatin and paclitaxel; or (x) docetaxel.
  • the binding agent preferably the bispecific antibody that specifically binds to PD-L1 and VEGF (most preferably PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with (i) etoposide and carboplatin; (ii) paclitaxel; (iii) topot
  • the binding agent i.e., the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may be administered in the form of any suitable pharmaceutical composition as described herein.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered in the form of an injection or infusion.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent can be administered prior to, simultaneously with, or after administration of the chemotherapy agent.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered prior to the administration of the chemotherapy agent.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered after the administration of the chemotherapy agent.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered simultaneously with the chemotherapy agent.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent and the chemotherapy agent may be administered using a composition comprising both drugs.
  • the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may be administered into one extremity of the subject, and the chemotherapy agent may be administered into another extremity of the subject.
  • the invention further provides a method of treating cancer in a subject, the method comprising administering to the subject a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) , in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the embodiments disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in a method of treating cancer in a subject as disclosed herein.
  • the binding agent including, e.g., the bispecific antibody
  • the bispecific antibody for use in a method of treating embodiments equally apply to the method of treatment disclosed herein.
  • the method is a method for increased overall survival in said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or an anti PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-L1 antibody is preferably pembrolizumab.
  • the chemotherapy or the binding agent including, e.g., the bispecific antibody
  • an anti PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody
  • the anti-PD-L1 antibody is preferably atezolizumab
  • the anti-PD-L1 antibody is preferably pembrolizumab.
  • the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b. a chemotherapy, preferably nab-paclitaxel; wherein the subject has a PD-L1 expression score before the treatment similar or identical to a CPS of ⁇ 1 to ⁇ 10 as determined by the TPS scoring algorithm.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy preferably nab-paclitaxel
  • the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b. a chemotherapy, preferably nab-paclitaxel; wherein the subject has a PD-L1 expression score before the treatment similar or identical to a CPS of ⁇ 1 to ⁇ 10 as determined by the TAP scoring algorithm.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy preferably nab-paclitaxel
  • the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b. a chemotherapy, preferably nab-paclitaxel; wherein the subject has a combined positive score (CPS) of ⁇ 1 to ⁇ 10 before the treatment.
  • the CPS is preferably determined with Dako’s PD-L1 IHC 22C3 pharmDx kit (SK006) according to the manufacturer’s TNBC Instructions for Use.
  • the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy preferably a chemotherapy agent
  • the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ⁇ 1 to ⁇ 10 or another integrating scoring algorithm defining a similar PD-L1 expression score, wherein the other integrating scoring algorithm is selected from tumour area positivity (TAP) score and tumour proportion score (TPS) , preferably wherein the other integrating scoring algorithm is TAP score.
  • CPS combined positive score
  • TAP tumour area positivity
  • TPS tumour proportion score
  • the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy preferably a chemotherapy agent
  • the subject has a PD-L1 expression before the treatment that provides a score of ⁇ 1 to ⁇ 10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
  • CPS combined positive score
  • TPS tumour area positivity
  • TPS tumour proportion score
  • the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b. the chemotherapy agent.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the use comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b. a chemotherapy agent.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. the binding agent (including, e.g., the bispecific antibody) ; and b. the chemotherapy agent.
  • a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject: a. a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) ; and b. the chemotherapy agent.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the invention further provides a method for determining whether a cancer in a subject is susceptible to treatment with a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score, wherein the CPS of ⁇ 1 indicates that the subject is susceptible to treatment with the binding agent (including, e.g., the bispecific antibody) and the chemotherapy.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • a chemotherapy agent comprising detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ⁇ 1 or other integrating scoring algorithm defining a similar PD-L1
  • the embodiments for determining PD-L1 expression score in cancer tissue disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in the method for determining whether a cancer in a subject is susceptible to treatment with a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy.
  • a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the “binding agent (including, e.g., the bispecific antibody) for use in a method of treating” embodiments equally apply to the determining method disclosed herein.
  • the cancer is triple negative breast cancer, which means that the method is for determining whether triple negative breast cancer in a subject is susceptible to treatment with a binding agent and a chemotherapy agent, wherein the method comprises the step as outlined above.
  • the treatment corresponds to the combination of a binding agent and a chemotherapy agent as outlined herein in the aspect relating to the treatment of triple negative breast cancer, i.e., the treatment is in particular the combination of a binding agent and a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
  • the method of treatment of a cancer e.g., the method of treating triple negative breast cancer by a combination of a binding agent and a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline
  • a cancer e.g., triple negative breast cancer
  • the method determining whether a cancer in a subject is susceptible to such treatment may be regarded as a step of the corresponding method of treatment.
  • the method for determining whether a cancer in a subject is susceptible to treatment with a binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy is conducted using the IHC assay PD-L1 IHC 22C3 pharmDx in accordance with the manufacturer’s guide.
  • the invention further provides kit-of-parts comprising the binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and the chemotherapy agent disclosed herein.
  • the binding agent including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the chemotherapy agent disclosed herein including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF
  • the embodiments disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in the kit-of-parts disclosed herein.
  • the binding agent including, e.g., the bispecific antibody
  • the bispecific antibody for use in a method of treating embodiments equally apply to the kit-of-parts disclosed herein.
  • the binding agent including, e.g., the bispecific antibody
  • the chemotherapy agent are comprised in separate container.
  • kit-of parts further comprise instructions for use.
  • kits or kit-of-parts of the present invention is a kit or kit-of-part comprising a. a composition comprising a binding agent of the present invention; and b. a composition comprising a chemotherapy agent, wherein the chemotherapy agent may be selected from a group or wherein the chemotherapy agent may be defined as comprising a combination, e.g., a combination of carboplatin and pemetrexed. It is understood that, if more than one chemotherapy agent is comprised in the kit, composition b. may comprise both chemotherapy agents or composition b. comprises the first chemotherapy agent, whereas there is yet a further composition c. comprising the second chemotherapy agent. This applies equally if more than 2 chemotherapy agents are present.
  • an exemplary kit or kit-of-parts of the present invention is a kit or kit-of-part comprising a. a composition comprising a binding agent of the present invention; and b. a composition comprising a first chemotherapy agent, c. a composition comprising a second chemotherapy agent wherein such a kit is suitable if the binding agent is administered in combination with two chemotherapy agents, which are comprised in separate compositions.
  • the agents described herein may be present and/or administered (e.g., as part of a treatment method) as a composition, e.g., each of the agents described herein is contained in separate vials or containers of, e.g., a kit and/or each of the agents described herein is administered separately from the other agents (e.g., each of the agents described herein is administered in separate compositions) .
  • the agents described herein such as binding agent of the present invention, the chemotherapeutic agent, etc.
  • composition may be present and/or administered (e.g., as part of a treatment method) as a single composition, e.g., each of the agents described herein is contained in a single vial or container and/or each of the agents described herein are administered together.
  • the composition may be a pharmaceutical composition.
  • composition relates to a formulation comprising a therapeutically effective agent, preferably together with one or more pharmaceutically acceptable carriers, diluents and/or excipients.
  • pharmaceutically acceptable refers to the non-toxicity of a material which does not interact with the action of the active component of the pharmaceutical composition.
  • compositions according to the present disclosure may contain salts, buffers, stabilizers, preservatives, amino acids and/or surface-active agents.
  • pharmaceutical compositions of the present disclosure comprise one or more pharmaceutically acceptable carriers, diluents, and/or excipients.
  • the bispecific antibody can be linked by various methods, see, for example, the method of Songsivilai et al. (Clin. Exp. Immunol., 79: 315-321 (1990) ) , and the method of Kostelny et al. (J. Immunol., 148: 1547-1553 (1992) which disclosures are also hereby incorporated in their entirety.
  • the bispecific antibody is conjugated with a chemotherapeutical agent to obtain an immunoconjugate.
  • the immunoconjugate contains: a. a bispecific antibody as disclosed herein; and b. a conjugation moiety selected from the group consisting of detectable labels, drugs, toxins, cytokines, radionuclides, or enzymes, gold nanoparticles/nanorods, nanomagnetic particles, viral coat proteins or VLPs, or their combination.
  • the radionuclide includes: a. a diagnostic isotope selected from the group consisting of Tc-99m, Ga-68, F-18, I-123, I- 125, I-131, In-111, Ga-67, Cu-64, Zr-89, C-11, Lu-177, Re-188, or a combination thereof; and/or b.
  • a diagnostic isotope selected from the group consisting of Tc-99m, Ga-68, F-18, I-123, I- 125, I-131, In-111, Ga-67, Cu-64, Zr-89, C-11, Lu-177, Re-188, or a combination thereof.
  • a therapeutic isotope selected from the group consisting of Lu-177, Y-90, Ac-225, As-211, Bi-212, Bi-213, Cs-137, Cr-51, Co-60, Dy-165, Er-169, Fm-255, Au-198, Ho-166, I-125, I-131, Ir-192, Fe-59, Pb-212, Mo-99, Pd-103, P-32, K-42, Re-186, Re-188, Sm-153, Ra223, Ru-106, Na24, Sr89, Tb-149, Th-227, Xe-133Yb-169, Yb-177, or a combination thereof.
  • the coupling moiety is a drug or a toxin.
  • the drug is a cytotoxic drug.
  • the cytotoxic drugs are selected from the group consisting of anti-tubulin drugs, DNA minor groove binding reagents, DNA replication inhibitors, alkylating reagents, antibiotics, folic acid antagonists, antimetabolites, chemotherapy A sensitizer, a topoisomerase inhibitor, a vinca alkaloid, or a combination thereof.
  • particularly useful cytotoxic drugs include, for example, DNA minor groove binding agents, DNA alkylating agents, and tubulin inhibitors.
  • Typical cytotoxic drugs include, for example, auristatins, camptothecins, duocarmycins, etoposides, maytansines and maytansinoids (eg DM1 and DM4) , taxanes (taxanes) , benzodiazepines, or benzodiazepine containing drugs (eg, pyrrolo [1, 4] benzodiazepines (PBDs) , indoline benzodiazepines indolinobenzodiazepines and oxazolidinobenzodiazepines) , vinca alkaloids, or combinations thereof.
  • auristatins camptothecins, duocarmycins, etoposides
  • maytansines and maytansinoids eg DM1 and DM4
  • taxanes taxanes
  • benzodiazepines or benzodiazepine containing drugs (eg, pyrrolo [1, 4] benzodiazepines (PBDs)
  • the toxin is selected from the following group: Auristatins (e.g., auristatin E, auristatin F, MMAE, and MMAF) , chlortetracycline, maytansoid, gamatoxin, gamatoxin A-chain, combretastatin, docarmicin, Lastatin, doxorubicin, daunorubicin, paclitaxel, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, autumn Narcissin, Dihydroxyanthraxdione, Actinomycin, Diphtheria Toxin, Pseudomonas Exotoxin (PE) A, PE40, Acacia toxin, Acacia A chain, Capsule root toxin A chain, ⁇ -Sarcinus, gelonin, mitogellin, ret
  • the present invention also relates to the following embodiments: 1.
  • a method of treating small cell lung cancer in a subject comprising administering to the subject: a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating small cell lung cancer the method comprising administering to the subject: a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use in a method of treating small cell lung cancer in a subject, the method comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent.
  • the bispecific antibody for use according to any one of embodiments 5 to 8 the chemotherapy agent for use according to any one of embodiments 5 to 8, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 8, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg or about 30 mg/kg, preferably intravenously every three weeks at day 1.
  • the treatment is a third-line treatment, preferably wherein the cancer progressed during or after first-line and second-line platinum-based chemotherapy with or without immuno-oncology treatment.
  • the method according to embodiment 12 or 13, the bispecific antibody for use according to embodiment 12 or 13, the chemotherapy agent for use according to embodiment 12 or 13, or the bispecific antibody and the chemotherapy agent for use according to embodiment 12 or 13, wherein the carboplatin is administered at a dosage of about AUC 5 and the etoposide is administered at a dosage of about 100 mg/m 2 , preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and etoposide is administered every three weeks at days 1 to 3. 15.
  • the bispecific antibody for use according to any one of embodiments 12 to 14 the chemotherapy agent for use according to any one of embodiments 12 to 14, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 12 to 14, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg or about 30 mg/kg, preferably intravenously every three weeks at day 1. 16.
  • the bispecific antibody for use according to embodiment 17 or 18, the chemotherapy agent for use according to embodiment 17 or 18, or the bispecific antibody and the chemotherapy agent for use according to embodiment 17 or 18, wherein the carboplatin is administered at a dosage of about AUC 5 and the etoposide is administered at a dosage of about 100 mg/m 2 , preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and etoposide is administered every three weeks at days 1 to 3. 20.
  • the bispecific antibody for use according to any one of embodiments 17 to 19 the chemotherapy agent for use according to any one of embodiments 17 to 19, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 17 to 19, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg or about 30 mg/kg, preferably intravenously every three weeks at day 1. 21.
  • the bispecific antibody for use according to any one of embodiments 17 to 19 the chemotherapy agent for use according to any one of embodiments 17 to 19, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 17 to 19, wherein the bispecific antibody is administered at a dosage of about 1400 mg or about 2000 mg, preferably intravenously every three weeks at day 1. 22.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of paclitaxel, topotecan, and a combination of carboplatin and etoposide.
  • the chemotherapy agent is selected from the group consisting of paclitaxel, topotecan, and a combination of carboplatin and etoposide.
  • a composition comprising a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • kits comprising a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a composition comprising a chemotherapy agent selected from the group consisting of paclitaxel, topotecan, and a combination of carboplatin and etoposide. 26. The composition according to embodiment 24, or the kit according to embodiment 25 for use as medicament.
  • the present invention also relates to the following embodiments: 1.
  • a method of treating small cell lung cancer in a subject comprising administering to the subject: a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
  • a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction
  • a chemotherapy agent comprising (i) a first binding region
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating small cell lung cancer, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating small cell lung cancer the method comprising administering to the subject: a.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating small cell lung cancer in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent. 5.
  • the topoisomerase inhibitor is topotecan, preferably wherein the topotecan is administered intravenously at a dosage of about 1.5 mg/m 2 every three weeks at days 1 to 5 or orally at a dosage of about 2.3 mg/m 2 every three weeks at days 1 to 5.
  • the binding agent for use according to any one of embodiments 5 to 8 the chemotherapy agent for use according to any one of embodiments 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 8, wherein the binding agent is administered at a dosage of about 10 mg/kg to about 100 mg/kg, preferably intravenously every three weeks at day 1. 10.
  • the treatment is a third-line treatment, preferably wherein the cancer progressed during or after first-line and second-line platinum-based chemotherapy with or without immuno-oncology treatment.
  • the binding agent for use according to embodiment 12 or 13, the chemotherapy agent for use according to embodiment 12 or 13, or the binding agent and the chemotherapy agent for use according to embodiment 12 or 13, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC 5 every three weeks at day 1, and the topoisomerase inhibitor is etoposide, preferably wherein the etoposide is administered at a dosage of about 100 mg/m 2 every three weeks at days 1 to 3. 15.
  • a platinum-based chemotherapy agent preferably carboplatin, cisplatin or oxaliplatin
  • a topoisomerase inhibitor preferably topotecan or etoposide.
  • the binding agent for use according to embodiment 17 or 18, the chemotherapy agent for use according to embodiment 17 or 18, or the binding agent and the chemotherapy agent for use according to embodiment 17 or 18, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC 5 every three weeks at day 1, and the topoisomerase inhibitor is etoposide, preferably wherein the etoposide is administered at a dosage of about 100 mg/m 2 every three weeks at days 1 to 3. 20.
  • the binding agent for use according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF. 24.
  • the binding agent for use according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF. 26.
  • the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) ,
  • the binding agent for use according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17. 28.
  • a binding agent binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
  • a taxane preferably paclitaxel, nab-paclitaxel or docetaxel
  • a topoisomerase inhibitor preferably topotecan or etoposide
  • a composition comprising a. a binding agent binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
  • a kit comprising a.
  • composition comprising a binding agent binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • composition comprising a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
  • a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably
  • the chemotherapy agent is selected from the group consisting of (i) paclitaxel, (ii) topotecan, and (iii) a combination of carboplatin and etoposide.
  • the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
  • the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL
  • the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. the chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use in a method of treating hepatocellular cancer in a subject, the method comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent. 5.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of oxaliplatin and 5-fluorouracil, optionally in further combination with leucovorin.
  • a composition comprising a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • a method of treating hepatocellular cancer in a subject comprising administering to the subject: a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
  • a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction
  • a chemotherapy agent comprising (i) a first binding region which binds to PD-1, PD-L
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating hepatocellular cancer, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating hepatocellular cancer the method comprising administering to the subject: a.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating hepatocellular cancer in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent. 5.
  • a platinum-based chemotherapy agent preferably carboplatin, cisplatin or oxaliplatin
  • an antimetabolite chemotherapy agent preferably 5-fluorouracil or gemcitabine
  • the binding agent is administered at a dosage of from 10 mg/kg to about 100 mg/kg, preferably intravenously every two weeks at day 1.
  • the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for hepatocellular cancer.
  • the binding agent for use according to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF. 10.
  • the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma)
  • the binding agent for use according to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF. 12.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin. 15.
  • a composition comprising a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent being a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin. 16.
  • a composition comprising a chemotherapy agent being a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin. 17.
  • a chemotherapy agent being a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin. 17.
  • a method of treating malignant mesothelioma in a subject comprising administering to the subject: a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating malignant mesothelioma, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating malignant mesothelioma the method comprising administering to the subject: a.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. the chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use in a method of treating malignant mesothelioma in a subject, the method comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent. 5.
  • the bispecific antibody for use according to embodiment 5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the carboplatin is administered at a dosage of about AUC 5 and the pemetrexed is administered at a dosage of about 500 mg/m 2 , preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1; or wherein the cisplatin is administered at a dose of about 75 mg/m 2 and the pemetrexed is administered at a dosage of about 500 mg/m 2 , preferably wherein the cisplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1.
  • the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for malignant mesothelioma.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of carboplatin and pemetrexed or cisplatin and pemetrexed.
  • a composition comprising a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • a kit comprising a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a composition comprising a chemotherapy agent being a combination of carboplatin and pemetrexed or cisplatin and pemetrexed. 13.
  • the composition according to embodiment 11, or the kit according to embodiment 12 for use as medicament.
  • a method of treating malignant mesothelioma in a subject comprising administering to the subject: a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
  • a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction
  • a chemotherapy agent comprising (i) a first binding region which binds to PD-1,
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating malignant mesothelioma, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating malignant mesothelioma the method comprising administering to the subject: a.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating malignant mesothelioma in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent. 5.
  • the binding agent is administered at a dosage of from about 10 mg/kg to about 100 mg/kg, preferably intravenously every three weeks at day 1.
  • MPM pleural mesothelioma
  • MPeM malignant peritoneal mesothelioma
  • the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a chemotherapy agent being a combination of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (ii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate. 17.
  • a kit comprising a.
  • a method of treating neuroendocrine neoplasms in a subject comprising administering to the subject: a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating neuroendocrine neoplasms, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating neuroendocrine neoplasms the method comprising administering to the subject: a.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. the chemotherapy agent 4.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use in a method of treating neuroendocrine neoplasms in a subject, the method comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent. 5.
  • the treatment is a second-line treatment, preferably wherein the subject has received prior systemic treatment for neuroendocrine neoplasms.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of irinotecan and 5-fluorouracil, optionally in further combination with leucovorin.
  • a composition comprising a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • a chemotherapy agent being a combination of irinotecan and 5-fluorouracil, optionally in further combination with leucovorin.
  • a kit comprising a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a composition comprising a chemotherapy agent being a combination of irinotecan and 5- fluorouracil, optionally in further combination with leucovorin. 12.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating neuroendocrine neoplasms, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating neuroendocrine neoplasms the method comprising administering to the subject: a.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating neuroendocrine neoplasms in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent. 5.
  • the treatment is a second-line treatment, preferably wherein the subject has received prior systemic treatment for neuroendocrine neoplasms.
  • the binding agent for use according to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF. 10.
  • the binding agent for use according to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF. 12.
  • the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM
  • the binding agent for use according to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17. 14.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
  • a topoisomerase inhibitor preferably an anthracycline, topotecan, irinotecan or etoposide
  • an antimetabolite chemotherapy agent preferably 5-fluorouracil or gem
  • a composition comprising a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • composition comprising a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a composition comprising a chemotherapy agent being a combination of a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin. 17.
  • a chemotherapy agent being a combination of a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin. 17.
  • a chemotherapy agent being a combination of a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide
  • an antimetabolite chemotherapy agent preferably 5-fluorouraci
  • the present invention relates to the following numbered embodiments: 1.
  • a method of treating non-small cell lung cancer in a subject comprising administering to the subject: a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating non-small cell lung cancer, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating non-small cell lung cancer the method comprising administering to the subject: a.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. the chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use in a method of treating non-small cell lung cancer in a subject, the method comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent. 5.
  • the bispecific antibody is administered at a dosage of about 2000 mg, preferably intravenously every three weeks at day 1.
  • the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line chemotherapy with immuno-oncology treatment.
  • the bispecific antibody for use according to embodiment 16, the chemotherapy agent for use according to embodiment 16, or the bispecific antibody and the chemotherapy agent for use according to embodiment 16, wherein the carboplatin is administered at a dosage of about AUC 5 and the pemetrexed is administered at a dosage of about 500 mg/m 2 , preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1. 18.
  • the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for non-small cell lung cancer.
  • the non-small cell lung cancer is squamous non-small cell lung cancer, preferably squamous non-small cell lung cancer of Stage IIIB/IIIC or IV.
  • the method according to embodiment 21, the bispecific antibody for use according to embodiment 21, the chemotherapy agent for use according to embodiment 21, or the bispecific antibody and the chemotherapy agent for use according to embodiment 21, wherein the carboplatin is administered at a dosage of about AUC 6 and the paclitaxel is administered at a dosage of about 200 mg/m 2 , preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and the paclitaxel is administered intravenously every three weeks at day 1. 23.
  • the bispecific antibody for use according to any one of embodiments 20 to 22 the chemotherapy agent for use according to any one of embodiments 20 to 22, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 20 to 22, wherein the bispecific antibody is administered at a dosage of about 2000 mg or 1400 mg, preferably intravenously every three weeks at day 1. 24.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of docetaxel, a combination of carboplatin and pemetrexed, and a combination of carboplatin and paclitaxel.
  • a composition comprising a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • a kit comprising a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a composition comprising a chemotherapy agent selected from the group consisting of docetaxel, a combination of carboplatin and pemetrexed, and a combination of carboplatin and paclitaxel.
  • a method of treating non-small cell lung cancer in a subject comprising administering to the subject: a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
  • a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction
  • a chemotherapy agent comprising (i) a first binding region which binds to PD-1, PD-L
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating non-small cell lung cancer, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating non-small cell lung cancer the method comprising administering to the subject: a.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating non-small cell lung cancer in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent. 5.
  • the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line chemotherapy with immuno-oncology treatment.
  • a platinum-based chemotherapy agent preferably carboplatin, cisplatin or oxaliplatin
  • an antifolate chemotherapy agent preferably pemetrexed or methotrexate.
  • the binding agent for use according to embodiment 9, the chemotherapy agent for use according to embodiment 9, or the binding agent and the chemotherapy agent for use according to embodiment 9, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC 5 every three weeks at day 1, and the antifolate chemotherapy agent is pemetrexed, preferably wherein the pemetrexed is administered intravenously at a dosage of about 500 mg/m 2 every three weeks at day 1. 11.
  • a platinum-based chemotherapy agent preferably carboplatin, cisplatin or oxaliplatin
  • an antifolate chemotherapy agent preferably pemetrexed or methotrexate.
  • the binding agent for use according to embodiment 14, the chemotherapy agent for use according to embodiment 14, or the binding agent and the chemotherapy agent for use according to embodiment 14, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC 5 every three weeks at day 1, and the antifolate chemotherapy agent is pemetrexed, preferably wherein the pemetrexed is administered intravenously at a dosage of about 500 mg/m 2 every three weeks at day 1. 16.
  • the binding agent for use according to embodiment 19, the chemotherapy agent for use according to embodiment 19, or the binding agent and the chemotherapy agent for use according to embodiment 19, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC 6 every three weeks at day 1, and the taxane is paclitaxel, preferably wherein the paclitaxel is administered intravenously at a dosage of about 200 mg/m 2 every three weeks at day 1. 21.
  • the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma)
  • the binding agent for use according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF. 26.
  • the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) ,
  • the binding agent for use according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17. 28.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane
  • a composition comprising a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; an b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; an b.
  • a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
  • a kit comprising a.
  • composition comprising a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • composition comprising a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
  • a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin;
  • the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL
  • the present invention relates to the following numbered embodiments: 1.
  • a method of treating triple-negative breast cancer in a subject comprising administering to the subject: a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. a chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating triple-negative breast cancer, the method comprising administering to the subject: a. the bispecific antibody; and b. a chemotherapy agent.
  • a chemotherapy agent for use in a method of treating triple-negative breast cancer the method comprising administering to the subject: a.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b. the chemotherapy agent.
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use in a method of treating triple-negative breast cancer in a subject, the method comprising administering to the subject: a. the bispecific antibody; and b. the chemotherapy agent. 5.
  • the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for triple-negative breast cancer.
  • the bispecific antibody for use according to embodiment 14, the chemotherapy agent for use according to embodiment 14, or the bispecific antibody and the chemotherapy agent for use according to embodiment 14, wherein the gemcitabine is administered at a dosage of about 1000 mg/m 2 and carboplatin is administered at a dosage of about AUC 2, preferably wherein the gemcitabine is administered intravenously every three weeks at days 1 and 8 and the carboplatin is administered intravenously every three weeks at days 1 and 8. 16.
  • the bispecific antibody for use according to embodiment 16 the chemotherapy agent for use according to embodiment 16, or the bispecific antibody and the chemotherapy agent for use according to embodiment 16, wherein the eribuline is administered at a dosage of about 1.4 mg/m 2 , preferably intravenously every three weeks at days 1 and 8. 18.
  • CPS combined positive score
  • CPS combined positive score
  • the bispecific antibody for use according to any one of embodiments 20 to 22 the chemotherapy agent for use according to any one of embodiments 20 to 22, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 20 to 22, wherein the another integrating scoring algorithm is the tumour area positivity (TAP) score or the tumour proportion score (TPS) . 24.
  • TAP tumour area positivity
  • TPS tumour proportion score
  • the CPS has been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula: wherein (i) the test sample is a cancer tissue sample; and/or (ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour infiltrating) lymphocytes and macrophages; and/or (iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by using the 22C3 antibody) ; and/or (iv) viable tumour cells are determined by sta
  • a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
  • a composition comprising a.
  • a kit comprising a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and b.
  • composition comprising a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
  • a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
  • a method of treating triple-negative breast cancer in a subject comprising administering to the subject: a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. a chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
  • a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction
  • a chemotherapy agent comprising (i) a first binding region which binds to PD-1, PD-L1,
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating triple-negative breast cancer, the method comprising administering to the subject: a. the binding agent; and b. a chemotherapy agent. 3.
  • a chemotherapy agent for use in a method of treating triple-negative breast cancer the method comprising administering to the subject: a.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b. the chemotherapy agent.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating triple-negative breast cancer in a subject, the method comprising administering to the subject: a. the binding agent; and b. the chemotherapy agent. 5.
  • the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for triple-negative breast cancer.
  • the binding agent for use according to any one of embodiments 5, 6 or 10 the chemotherapy agent for use according to any one of embodiments 5, 6 or 10, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5, 6 or 10, wherein the binding agent is administered at a dosage of from about 10 mg/kg to 100 mg/kg, preferably intravenously every four weeks at days 1, 8 and 15;or wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every four weeks at days 1 and 15. 12.
  • the binding agent for use according to embodiment 13, the chemotherapy agent for use according to embodiment 13, or the binding agent and the chemotherapy agent for use according to embodiment 13, wherein the antimetabolite chemotherapy agent is gemcitabine, preferably wherein the gemcitabine is administered intravenously at a dosage of about 1000 mg/m 2 every three weeks at days 1 and 9, and wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC 2 every three weeks at days 1 and 8. 15.
  • the binding agent for use according to any one of embodiments 13 to 16 the chemotherapy agent for use according to any one of embodiments 13 to 16, or the binding agent and the chemotherapy agent for use according to any one of embodiments 13 to 16, wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every three weeks at day 1. 18.
  • CPS combined positive score
  • TAP tumour area positivity
  • TPS tumour proportion score
  • the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma)
  • the binding agent for use according to any one of embodiments 2 and 5 to 25, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 25, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 25, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF. 29.
  • the binding agent for use according to any one of embodiments 2 and 5 to 25, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 25, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 25, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17. 31.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline.
  • the chemotherapy agent is selected from the group consisting of (i) a taxane,
  • a composition comprising a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline.
  • a kit comprising a.
  • composition comprising a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and b.
  • composition comprising a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline. 34.
  • a chemotherapy agent selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline.
  • the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
  • the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
  • SEQ ID NO: 1-3 are exemplary CDR1-3 amino acid sequences of an anti-PD-L1 antibody using the IMGT numbering system.
  • SEQ ID NO: 4-6 are exemplary HCDR1-3 amino acid sequences of an anti-VEGF antibody using the IMGT numbering system.
  • SEQ ID NO: 7 and 8 are exemplary LCDR1 and 3 amino acid sequences of an anti-VEGF antibody using the IMGT numbering system.
  • SEQ ID NO: 9 is an exemplary amino acid sequence of an anti-PD-L1 VHH.
  • SEQ ID NO: 10 is an exemplary VH amino acid sequence of an anti-VEGF antibody.
  • SEQ ID NO: 11 is an exemplary VL amino acid sequence of an anti-VEGF antibody.
  • SEQ ID NO: 12 is an exemplary CH1 amino acid sequence of an anti-VEGF antibody.
  • SEQ ID NO: 13 is an exemplary IgG1 Fc region amino acid sequence of an anti-VEGF antibody.
  • SEQ ID NO: 14 is an exemplary linker amino acid sequence of the bispecific antibody as disclosed herein.
  • SEQ ID NO: 15 is an exemplary CL amino acid sequence of an anti-VEGF antibody.
  • SEQ ID NO: 16 is an exemplary heavy chain amino acid sequence of the bispecific antibody as disclosed herein comprising an exemplary anti-PD-L1 VHH amino acid sequence.
  • SEQ ID NO: 17 is an exemplary light chain amino acid sequence of the bispecific antibody as disclosed herein.
  • SEQ ID NO: 18, 19 and 38 are exemplary CDR1-3 amino acid sequences of an anti-PD-L1 antibody using the Kabat numbering system.
  • SEQ ID NO: 20-25 are exemplary LCDR1-3 and HCDR1-3 amino acid sequences of the 22C3 antibody.
  • SEQ ID NO: 26 is an exemplary VL amino acid sequence of the 22C3 antibody.
  • SEQ ID NO: 29 is an exemplary VL amino acid sequence of the 28-8 antibody.
  • SEQ ID NO: 36 is an exemplary VH amino acid sequence of the SP263 antibody.
  • SEQ ID NO: 37 is an exemplary VL amino acid sequence of the SP263 antibody.
  • SEQ ID NO: 42 and 43 are exemplary LCDR1 and 2 amino acid sequences of an anti-VEGF-Aantibody using the Kabat numbering system.
  • An exemplary anti-VEGF-ALCDR2 is the amino acid sequence FTS referred to herein as SEQ ID NO: 44 using the IGMT numbering system.
  • nucleotide sequences depend on the individual context for cloning and codon-optimization as described herein but still encode the same amino acid sequence.
  • This example is to provide guidelines for evaluating PD-L1 expression on formalin-fixed, 20 paraffin-embedded (FFPE) tumour tissue section with Dako’s PD-L1 IHC 22C3 pharmDx kit (SK006) according to the manufacturer’s TNBC Instructions for Use.
  • This immunohistochemical (IHC) assay has been performed using the Dako Auto-stainer Link 48 automated staining system.
  • PD-L1 IHC 22C3 pharmDx kit which is a qualitative immunohistochemical assay using Monoclonal Mouse Anti-PD-L1, Clone 22C3.
  • This kit is intended for use in the detection of PD-L1 protein in formalin-fixed, paraffm-30 embedded (FFPE) tumour tissue using EnVision FLEX visualization system on Autostainer Link 48.
  • FFPE formalin-fixed, paraffm-30 embedded
  • PD-L1 protein expression was used to determine a Combined Positive Score (CPS) .
  • CPS Combined Positive Score
  • PD-L1 IHC 22C3 pharmDx contains optimized reagents to perform an IHC staining procedure using a linker and a chromogen enhancement reagent. Deparaffinization, rehydration, and target retrieval was performed using a 3-in-1 procedure on PT Link.
  • hematoxylin and eosin (H&E) stain of the tissue specimen were evaluated first to assess tissue histology and preservation quality.
  • PD-L1 IHC 22C3 pharmDx and the H&E staining were performed on serial sections from the same paraffin block of the specimen (sample) . Tissue specimens were intact, well preserved, and confirmed tumour indication.
  • the specimen contained a minimum of 100 viable tumour cells to determine the percentage of positive cells.
  • tissue from a deeper level of the block, or potentially another block presented sufficient number of viable tumour cells for PD-L1 IHC 22C3 pharmDx testing.
  • the PD-L1 IHC 22C3 pharmDx Control Cell Line Slide were examined to determine that reagents are functioning properly. Each slide contained sections of cell pellets with positive and negative PD-L1 expression. The percentage of positive cells, staining intensity, and non-specific staining were assessed in both cell pellets. If any staining of the Control Cell Line Slide was not satisfactory, all results with the subject specimens were considered invalid. The Control Cell Line Slide has not been used as an aid in interpretation of subject results.
  • the overall staining intensity was evaluated using the following guide:
  • the following stainings were acceptable for the PD-L1 positive cell pellet: - Cell membrane staining of ⁇ 70%of cells - ⁇ 2+ average staining intensity - Non-specific staining ⁇ 1+ intensity
  • the following stainings were acceptable: - No specific staining - Non-specific staining ⁇ 1+ intensity. Note that staining of a few cells in the MCF-7 cell pellet may occasionally be observed. The following acceptance criteria are applicable: the presence of ⁇ 10 total cells with distinct plasma membrane staining, or cytoplasmic staining with ⁇ 1+ intensity within the boundaries of the MCF-7 cell pellet are acceptable
  • TNBC Positive and Negative User Control Tissue
  • the TNBC Positive Control Tissue Slides were examined to verify that the fixation method and epitope retrieval process are effective.
  • the Positive Control Tissue Slides were stained with both PD-L1 primary antibody and Negative Control Reagent.
  • the ideal positive control tissue provided a complete dynamic representation of weak-to-moderate staining of tumour cells and tumour-associated mononuclear inflammatory cells (MICs) .
  • MICs tumour-associated mononuclear inflammatory cells
  • Known positive tissue controls were utilized for monitoring the correct performance of processed tissues and test reagents, not as an aid in formulating a specific diagnosis of subject samples. If stainings of positive in-house control tissue were not satisfactory, all results with the subject specimen were considered invalid.
  • - Requirements for slide stained with PD-L1 Presence of brown plasma membrane staining were observed.
  • Non-specific staining ⁇ 1+ - Requirements for slide stained with Negative Control Reagent No membrane staining.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Biochemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Molecular Biology (AREA)
  • Public Health (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR), or both and which antagonizes the VEGF/VEGFR interaction (more particularly to a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) ) for use in the treatment of a subject afflicted with cancer, wherein the subject is administered a combination comprising the binding agent (more particularly the bispecific antibody) and a chemotherapy, optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.

Description

COMBINATION THERAPY COMPRISING A PD-1/PD-L1 AND VEGF/VEGFR BINDING AGENT AND A CHEMOTHERAPY FOR CANCER TREATMENT TECHNICAL FIELD
The invention inter alia relates to methods for treating cancer in a subject using a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction (which can, e.g., be a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) ) in combination with a chemotherapy agent, optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm. The invention further concerns a method for determining whether a cancer in a subject is susceptible to treatment with the above-mentioned binding agent and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject the PD-L1 expression before the treatment by CPS or another integrating scoring algorithm, wherein a CPS of ≥ 1 indicates that the subject is susceptible to the treatment, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
BACKGROUND OF THE INVENTION
The approval of immune checkpoint inhibitors (ICIs) targeting programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) has dramatically changed the treatment landscape for cancer subjects. Immune checkpoint therapies targeting the PD-1: PD-L1 pathway have resulted in groundbreaking improvements in clinical response in multiple human cancers (Brahmer et al., N Engl J Med 2012, 366: 2455-65; Garon et al. N Engl J Med 2015, 372: 2018-28; Hamid et al, N Engl J Med 2013, 369: 134-44; Robert et al, Lancet 2014, 384: 1109-17; Robert et al, N Engl J Med 2015, 372: 2521-32; Robert et al., N Engl J Med 2015 , 372: 320-30; Topalian et al, N Engl J Med 2012, 366: 2443-54; Topalian et al, J Clin Oncol 2014, 32: 1020-30; Wolchok et al, N Engl J Med 2013, 369: 122-33) .
Numerous studies have found higher response rates and more favorable survival outcomes when subjects were treated with ICI therapy in combination with chemotherapy as compared with the conventional chemotherapy alone.
Despite these promising improvements, response to ICI treatment is not guaranteed in all cancer subjects. Predictive biomarkers, such as scoring PD-L1 expression in cancer tissue, are therefore sometimes used to evaluate the likelihood to respond to ICI treatment alone or in combination with chemotherapy, thereby defining treatment-eligible patient groups (Ulas, Ezgi B et al. “Predictive Value of Combined Positive Score and Tumour Proportion Score for Immunotherapy Response in Advanced NSCLC. ” JTO clinical and research reports vol. 4, 9 100532.25 May. 2023, doi: 10.1016/j. jtocrr. 2023.100532) .
Cancer types are scored for PD-L1 expression for example by using the combined positive score (CPS) , which covers the PD-L1 expression on both the tumour cells and the immune cells in the tumour microenvironment. Studies have explored the relationship between the expression of programmed death ligand 1 (PD-L1) and prognosis in cancer, and high PD-L1 expression determined by immunohistochemistry is suggested to be a predictive biomarker of response to immunotherapy in several cancers (Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N Engl J Med. 2022; 387 (3) : 217-226. Doi: 10.1056/NEJMoa2202809, Chen, Xiao-Jiang et al. “Prognostic Significance of PD-L1 Expression in Gastric Cancer Subjects with Peritoneal Metastasis. ” Biomedicines vol. 11, 7 2003.15 Jul. 2023, doi: 10.3390/biomedicines11072003) . High expression of PD-L1 on tumour cells is at the same time, however, associated with poor prognosis in cancer subjects.
Currently used ICI therapy targeting the PD-1: PD-L1 pathway alone or in combination with chemotherapy leads to unsatisfactory responses especially in subjects having a low PD-L1 expression on cancer cells.
For example, the approved anti-PD-L1 antibody pembrolizumab plus chemotherapy regime is currently considered the standard of care (SOC) in the first-line treatment of patients with metastatic TNBC whose tumours express PD-L1 with a CPS score of ≥10; and clinically significant improvements have been observed in progression-free survival (PFS) (9.7 months for pembrolizumab plus chemotherapy versus 5.6 months for chemotherapy alone) and overall survival (OS) (23.0 months for pembrolizumab plus chemotherapy versus 16.1 months for chemotherapy alone) . However, these benefits have not been observed in patients with a CPS of <10. For this patient group, the SOC remains chemotherapy alone, with a median PFS of only about 6 months and a median OS of about 16 months (Cortes et al. 2022) .
There is therefore an unmet need for improved methods for the treatment of cancer and specific cancer types, respectively, on a general level, and in particular for cancers having a low PD-L1 expression score for which the SOC momentarily is chemotherapy alone with associated poor prognosis of PFS.
SUMMARY OF THE INVENTION
Against the aforementioned background, it is an object of the present invention to provide effective pharmacological means to treat cancer, preferably cancer having a low PD-L1 expression (score) in a subject. It is also an object of the present invention to provide pharmacological means to elicit a therapeutically effective immune response for the treatment of cancer, preferably cancer having a low PD-L1 expression (score) .
These and other objects are achieved by the invention set forth in the claims and embodiments explained in more detail below.
The invention provides a method of treating cancer in a subject, the method comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
In an embodiment of the method, the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
The invention further provides a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating cancer, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
In an embodiment of the binding agent for use, the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
The invention further provides a chemotherapy agent for use in a method of treating cancer, the method comprising administering to the subject
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
In an embodiment of the chemotherapy agent for use, the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
The invention further provides a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating cancer in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
In an embodiment of the binding agent and the chemotherapy agent, the binding agent is administered in an effective amount and the chemotherapy agent is administered in an effective amount.
In an embodiment, the invention provides a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy, preferably a chemotherapy agent;
optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a 
combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
In other words, the present application provides in an embodiment a bispecific antibody that specifically binds to programmed death-ligand 1 (PD-L1) and Vascular Endothelial Growth Factor (VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy, preferably a chemotherapy agent.
It can be preferred that the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
Furthermore, the invention concerns in an embodiment a method of treating cancer in a subject, the method comprising administering to the subject a bispecific antibody that specifically binds to PD-L1 and VEGF in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
In other words, the present application provides in an embodiment a method of treating cancer in a subject, the method comprising administering to the subject a bispecific antibody that specifically binds to PD-L1 and VEGF in combination with chemotherapy. It can be preferred that the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
The invention also concerns in an embodiment a chemotherapy agent for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. a bispecific antibody that specifically binds to PD-L1 and VEGF; and
b. the chemotherapy agent;
optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a 
combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
In some embodiments of the invention, there is provided use of a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
In some embodiments of the invention, there is provided use of a bispecific antibody that specifically binds to PD-L1 and VEGF in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
In some embodiments of the invention, there is provided use of a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
In some embodiments of the invention, there is provided use of a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. a bispecific antibody that specifically binds to PD-L1 and VEGF; and
b. the chemotherapy agent.
The invention is concerned with a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
The invention is further concerned with a composition comprising
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent selected from the group consisting of (i) a platinum-based 
chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
In an embodiment, the composition is a pharmaceutical composition.
The invention is further concerned with a kit comprising
a. a composition comprising a binding agent comprising (i) a first binding region which binds 
to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a composition comprising a chemotherapy agent selected from the group consisting of (i) 
a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
In an embodiment, each composition of the kit is a pharmaceutical composition. As noted below, the composition comprising a chemotherapy agent may be more than one composition, namely if the chemotherapy agent is a combination of chemotherapy agents and each chemotherapy agent of the combination may be provided in a separate composition.
Further, the invention also provides in an embodiment a kit of parts comprising the bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy agent.
A kit of the invention typically includes instructions.
The invention also concerns a method for determining whether a cancer in a subject is susceptible to treatment with a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject the PD-L1 expression before the treatment by determining the combined positive scope (CPS) or another score, preferably the tumour area positivity (TAP) score or the tumour proportion score (TPS) , wherein a CPS of ≥ 1 indicates a PD-L1 expression that renders the subject susceptible to treatment with the bispecific antibody and the chemotherapy, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the TAP score or the TPS, more preferably when determined by the TAP score.
The invention also concerns in an embodiment a method for determining whether a cancer in a subject is susceptible to treatment with a bispecific antibody that specifically binds to PD-L1 and VEGF and a chemotherapy, wherein the method comprises detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score, wherein the CPS of ≥ 1 indicates that the subject is susceptible to treatment with the bispecific antibody and the chemotherapy.
BRIEF DESCRIPTION OF THE FIGURES
Fig. 1. (A) - (C) depicts boxplots of AUCtau, ss by 20 mg/kg Q2W (A) versus 1200 mg Q2W (B) or 1500 mg Q2W (C) dosing regimens. N is the number of participants; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of AUCtau, ss, the box represents the inter-quartile distance, and the whiskers represent ≤1.5 times the inter-quartile range (75th -25th quartile) . The number at the top of the figure represents the number of participants included in each category. Abbreviations: AUCtau, ss = area under the curve during the dosing interval at steady-state; N = number; Q2W = every two weeks.
Fig. 2 (A) depicts boxplots of AUCtau, ss for 30 mg/kg Q3W versus 2100 mg Q3W dosing regimens. N is the number of simulated patients in the different body weight ranges as indicated; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of AUCtau, ss, the box represents the inter-quartile distance, and the whiskers represent ≤1.5 times the inter-quartile range (75th -25th quartile) .
Fig. 2 (B) depicts boxplots of cmax, ss for 30 mg/kg Q3W versus 2100 mg Q3W dosing regimens. N is the number of simulated patients in the different body weight ranges as indicated; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of cmax, ss, the box represents the inter-quartile distance, and the whiskers represent ≤1.5 times the inter-quartile range (75th -25th quartile) .
Fig. 2 (C) depicts boxplots of cmin, ss for 30 mg/kg Q3W versus 2100 mg Q3W dosing regimens. N is the number of simulated patients in the different body weight ranges as indicated; the x-axis represents baseline body weight, the horizontal line at the center of the box is the median of cmin, ss, the box represents the inter-quartile distance, and the whiskers represent ≤1.5 times the inter-quartile range (75th -25th quartile) .
DETAILED DESCRIPTION OF THE INVENTION
Although certain embodiments of the present invention are described in detail below, it is to be understood that this invention is not limited to the particular embodiments, methodologies, protocols and reagents described herein as these may vary within the scope set by the claims. It is also to be understood that terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which is defined by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art.
In the following description, certain elements of the present invention will be described. These elements may be discussed with specific embodiments, however, it should be understood that they may be combined in any manner and in any number to create additional embodiments. The variously described examples, features and particular embodiments should not be construed to limit the present invention to only the explicitly described embodiments or to the explicitly described combination of features. This description should be understood to disclose and encompass embodiments which combine the explicitly described embodiments with any number of the disclosed and/or preferred elements. Furthermore, any permutations and combinations of all described elements in this application should be considered disclosed by this description unless the context indicates otherwise.
The above objects are achieved by the following embodiments in accordance with the invention:
1. A bispecific antibody that specifically binds to PD-L1 and VEGF for use in a method of treating a 
subject with cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy, preferably a chemotherapy agent;
optionally wherein the subject has a PD-L1 expression score before the treatment as determined 
by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score; or
optionally wherein the subject has a PD-L1 expression before the treatment that provides a score 
of ≥1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
2. The bispecific antibody for use according to embodiment 1, wherein the other integrating scoring 
algorithms are selected from TAP and TPS; or
wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when 
determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , preferably when determined by the tumour area positivity (TAP) score.
3. The bispecific antibody for use according to embodiment 1 or 2, wherein the CPS has been 
determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages; and/or
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably 
by using the anti-PD-L1 antibody 22C3) ; and/or
(iv) viable tumour cells are determined by staining with a viability dye; and/or
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 
staining lymphocytes and PD-L1 staining macrophages.
4. The bispecific antibody for use according to embodiment 3, wherein the PD-L1 staining cells are 
tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
5. The bispecific antibody for use according to embodiment 3, wherein the PD-L1 staining cells are 
tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
6. The bispecific antibody for use according to any of the preceding embodiments, wherein the PD-
L1 expression score has been determined using a TAP scoring algorithm in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer sample; and/or;
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells; and/or
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 
immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 
positive tumour cells and immune cells; and/or
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
7. The bispecific antibody for use according to embodiment 6, wherein the tumour area is determined 
using a hematoxylin and eosin (H&E) stain.
8. The bispecific antibody for use according to embodiment 6 or 7, wherein the PD-L1 positive tumour 
cells and immune cells are PD-L1 staining cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and immune cells (lymphocytes and macrophages) within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
9. The bispecific antibody for use according to any of the preceding embodiments, wherein the PD-
L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or;
(ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
(iii) viable tumour cells are determined by staining with a viability dye; and/or
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
10. The bispecific antibody for use according to any one of embodiments 3 to 9, wherein the cancer 
tissue sample is a tissue section of a tumour biopsy.
11. The bispecific antibody for use according to any of the preceding embodiments, wherein the PD-
L1 expression is detected by immunohistochemistry (IHC) staining.
12. The bispecific antibody for use according to embodiment 10 or 11, wherein the tissue section is a 
formalin fixed and embedded in paraffin wax (FFPE) tissue section.
13. The bispecific antibody for use according to any of embodiments 10-12, wherein the tissue section 
is stained.
14. The bispecific antibody for use according to embodiment 13, wherein the stain comprises a 
hematoxylin and eosin (H&E) stain, preferably wherein the viability dye is propidium iodide.
15. The bispecific antibody for use according to any of embodiments 3-14, wherein the number of 
viable tumour cells in the tumour tissue sample is determined by flow cytometry.
16. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody and the chemotherapy are separately administered.
17. The bispecific antibody for use according to any of the preceding embodiments, wherein a dosage 
of the bispecific antibody and a dosage of the chemotherapy are administered concurrently or consecutively.
18. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
chemotherapy comprises a platinum-based chemotherapy.
19. The bispecific antibody for use according to any of the preceding embodiments, wherein the subject 
has a PD-L1 expression score as determined by a CPS of up to 20, preferably from 1 to 20, more preferably from 1 to <10, before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score; or
wherein the subject has a PD-L1 expression before the treatment that provides a score of < 20, 
preferably from 1 to 20, more preferably from 1 to <10 when determined by the CPS or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
20. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody dosage is in a range of from 0.1 mg/kg to 45 mg/kg body weight, preferably 1 mg/kg to 30 mg/kg body weight.
21. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody and/or the chemotherapy is administered intravenously, preferably wherein the bispecific antibody and the chemotherapy are administered intravenously.
22. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody and/or the chemotherapy is administered via an IV injection or IV infusion.
23. The bispecific antibody for use according to any of the preceding embodiments, wherein a 
treatment cycle is repeated at least 1, 2, 3, 4, 5, 6, 7, or 8 times.
24. The bispecific antibody for use according to any of the preceding embodiments, wherein each cycle 
has up to 28 days, preferably 28 or 21 days.
25. The bispecific antibody for use according to any of the preceding embodiments, wherein the subject 
has not been previously treated for cancer.
26. The bispecific antibody for use according to any of embodiments 1-24, wherein the subject has 
been previously treated for cancer, preferably wherein the subject had at least one previous chemotherapy treatment.
27. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody is administered every 6 weeks, preferably every 4 weeks, more preferably every 3 weeks or every 2 weeks.
28. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody is administered every 2 weeks at a dosage ranging from 10 mg/kg to 30 mg/kg, preferably ranging from 15 mg/kg to 25 mg/kg, more preferably at a dosage of 20 mg/kg.
29. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody is administered every 3 weeks at a dosage ranging from 20 mg/kg to 40 mg/kg, preferably ranging from 25 mg/kg to 35 mg/kg, more preferably at a dosage of 30 mg/kg.
30. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
chemotherapy is administered once or more within the first 20 days or the first 21 days of each cycle, wherein the chemotherapy is administered twice or more within the first 20 days or the first 21 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 20 days or the first 21 days of each cycle.
31. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
chemotherapy is administered once or more within the first 14 days or the first 15 days of each cycle, wherein the chemotherapy is administered twice or more within the first 14 days or the first 15 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 14 days or the first 15 days of each cycle.
32. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
chemotherapy is administered on the 1st, 8th, and 15th days of each cycle.
33. The bispecific antibody for use according to any of the preceding embodiments, wherein overall 
survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
34. The bispecific antibody for use according to any of the preceding embodiments, wherein median 
progression-free survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
35. The bispecific antibody for use according to any of the preceding embodiments, wherein the cancer 
comprises one or more solid tumours.
36. The bispecific antibody for use according to any of the preceding embodiments, wherein the cancer 
is selected from the group consisting of melanoma, lung, liver, stomach, renal cell, urothelial, cervical, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancers, preferably wherein the cancer is selected from urothelial, breast and esophagus cancer.
37. The bispecific antibody for use according to any of the preceding embodiments, wherein the cancer 
is small cell lung cancer (SCLC) , non-small cell lung cancer (NSCLC) or triple-negative breast cancer (TNBC) , preferably advanced triple-negative breast cancer.
38. The bispecific antibody for use according to embodiment 37, wherein the NSCLC has a squamous 
histology.
39. The bispecific antibody for use according to embodiment 37, wherein the NSCLC has a non-
squamous histology.
40. The bispecific antibody for use according to embodiment 37, wherein the NSCLC is an EGFR 
mutation-positive NSCLC.
41. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody comprises an anti-PD-L1 antibody or fragment thereof.
42. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody comprises an anti-VEGF antibody or fragment thereof.
43. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody comprises a Fab, Fab’, F (ab’) 2, Fd, Fv, dAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH.
44. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably wherein each VHH is fused to the C-terminus of an anti-VEGF antibody.
45. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1 or 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO:2 or 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3 or 38.
46. The bispecific antibody for use according to embodiment 45, wherein the amino acid sequence of 
the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 9.
47. The bispecific antibody for use according to any of embodiments 42-46, wherein the anti-VEGF 
antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
48. The bispecific antibody for use according to any of embodiments 42-47, wherein the anti-VEGF 
antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO: 13 or having at least 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 13.
49. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody specifically binds to VEGF-A.
50. The bispecific antibody for use according to any of embodiments 42-49, wherein a heavy chain 
variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and
wherein a light chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-
determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
51. The bispecific antibody for use according to embodiment 50, wherein the amino acid sequence of 
the heavy chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 11; or
wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody has 
at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 11.
52. The bispecific antibody for use according to any of the preceding embodiments, wherein the amino 
acid sequence of the heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or
wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 95%, 
96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17.
53. The bispecific antibody for use according to any of embodiments 42-52, wherein the anti-VEGF 
antibody is bevacizumab.
54. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
bispecific antibody is encoded by one or more nucleic acid molecules.
55. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
chemotherapy comprises a chemotherapy agent selected from lurbinectedin, topotecan, taxane such as paclitaxel, docetaxel, or nanoparticle albumin-bound paclitaxel (nab-paclitaxel) , pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
56. The bispecific antibody for use according to any of embodiments 18-55, wherein the platinum-
based chemotherapy comprises cisplatin, oxaliplatin or carboplatin.
57. The bispecific antibody for use according to any of the preceding embodiments, wherein the 
method of treatment comprises administering the bispecific antibody in combination with paclitaxel to a subject having small cell lung cancer.
58. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody in combination with pemetrexed and carboplatin (preferably as first line treatment of non-squamous NSCLC) or a taxane (preferably as first line treatment of squamous NSCLC) , such as paclitaxel or docetaxel (preferably as second line treatment of NSCLC) , to a subject having NSCLC, preferably as second line treatment, or wherein the method of treatment comprises administering the bispecific antibody in combination with pemetrexed and carboplatin to a subject having malignant mesothelioma.
59. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody in combination with nab-paclitaxel to a subject having triple-negative breast cancer.
60. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody in combination with oxaliplatin, calcium folinate, and 5-fluorouracil to a subject having hepatocellular carcinoma, preferably wherein the method of treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg being particularly preferred) .
61. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody in combination with irinotecan, 5-fluorouracil, calcium folinate to a subject having unresectable neuroendocrine neoplasm.
62. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody in combination with etoposide and platinum to a subject having small cell lung cancer, preferably as first line treatment and/or extensive-stage small cell lung cancer, most preferably as first line treatment of extensive-stage small cell lung cancer or as second-line treatment of limited-stage small cell lung cancer.
63. The bispecific antibody for use according to embodiment 62, wherein the method of treatment 
comprises administering the bispecific antibody every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg or 30 mg/kg being particularly preferred) or at a dose ranging from 1400 mg to 2000 mg in combination with etoposide (preferably at a dosage of 100 mg/m2) and with carboplatin (preferably at a dosage of AUC=5 (total dose ≤ 750 mg) to a subject having small cell lung cancer, preferably as first line treatment and/or extensive-stage small cell lung cancer, most preferably as first line treatment of extensive-stage small cell lung cancer or as second-line treatment of limited-stage small cell lung cancer.
64. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg or 30 mg/kg being particularly preferred) in combination with paclitaxel, lurbinectedin, or topotecan to a subject having small cell lung cancer, preferably as second line treatment, and/or extensive-stage small cell lung cancer, more preferably wherein the method of treatment comprises administering to a patient suffering from advanced small cell lung cancer as second line treatment the bispecific antibody every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with (i) paclitaxel every 3 weeks at a dosage of 175 mg/m2, optionally for 5 cycles, or (ii) topotecan at a dosage of 1.5 mg/m2 administered IV or 2.3 mg/m2 administered orally once per day on days 1 to 5 of every 3 weeks cycle.
65. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 10 mg/kg to 20 mg/kg in combination with nab-paclitaxel, paclitaxel, or gemcitabine with carboplatin to a subject having triple-negative breast cancer.
66. The bispecific antibody for use according to any of embodiments 1-55, wherein the method of 
treatment comprises administered the bispecific antibody every 2 weeks at a dosage of 1000 mg to 2000 mg to a subject having triple-negative breast cancer, preferably on the 1st and 15th day of a 28-day treatment cycle, preferably wherein the bispecific antibody is administered in combination with a chemotherapy, preferably on the 1st, 8th, and 15th day of a 28-day treatment cycle.
67. A method of treating cancer, preferably triple-negative breast cancer (TNBC) in a subject, the 
method comprising administering to the subject a bispecific antibody that specifically bind to PD-L1 and VEGF in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ≥ 1, preferably from 1 to 20, preferably from 1 to <10, or other integrating scoring algorithm defining a similar PD-L1 expression score; or
optionally wherein the subject has a PD-L1 expression before the treatment that provides a score 
of ≥ 1, preferably from 1 to 20, preferably from 1 to <10, when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
68. The method according to embodiment 67, wherein the method is a method for extending 
progression-free survival in said subject compared to the chemotherapy or an anti-PD-L1 antibody or an anti-PD-1 antibody or the bispecific antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
69. The method according to embodiment 67 or 68, wherein the method is a method for increased 
overall survival in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
70. A method for determining whether a cancer in a subject is susceptible to treatment with a bispecific 
antibody that specifically binds to PD-L1 and VEGF and a chemotherapy, wherein the method comprises detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score, wherein the CPS of ≥ 1 indicates that the subject is susceptible to treatment with the bispecific antibody and the chemotherapy; or
wherein the method comprises detecting in a sample of the subject a PD-L1 expression before the 
treatment by determining the combined positive scope (CPS) or another score, preferably the tumour area positivity (TAP) score or the tumour proportion score (TPS) , wherein a CPS of ≥ 1 indicates a PD-L1 expression that renders the subject susceptible to treatment with the bispecific antibody and the chemotherapy, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the TAP score or the TPS, more preferably when determined by the TAP score.
71. The method according to embodiment 70, wherein the cancer is selected from the group consisting 
of melanoma, lung, liver, stomach, renal cell, urothelial, cervical, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancers, preferably wherein the cancer is selected from urothelial, breast and esophagus cancer.
72. The method according to embodiment 70 or 71, wherein the sample is a cancer tissue sample.
73. The method according to any of embodiments 70-72, wherein the bispecific antibody comprises an 
anti-PD-L1 antibody or fragment thereof.
74. The method according to any of embodiments 70-73, wherein the bispecific antibody comprises an 
anti-VEGF antibody or fragment thereof.
75. The method according to any of embodiments 70-74, wherein the bispecific antibody comprises a 
Fab, Fab', F (ab') 2, Fd, Fv, sdAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH.
76. The method according to any of embodiments 70-75, wherein the bispecific antibody comprises 
two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably each VHH is fused to the C-terminus of the anti-VEGF antibody.
77. The method according to any of embodiments 70-76, wherein the bispecific antibody comprises an 
anti-PD-L1 single domain antibody comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1 or 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 2 or 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3 or 38.
78. The method according to embodiment 77, wherein the amino acid sequence of the anti-PD-L1 
single domain antibody is shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9.
79. The method according to any of embodiments 74-78, wherein the anti-VEGF antibody or fragment 
thereof comprises a constant region preferably derived from a human antibody, preferably, the constant region is selected from the constant region of human IgGl, IgG2, IgG3 or IgG4.
80. The method according to any of embodiments 74-79, wherein the anti-VEGF antibody or fragment 
thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO:13 or having at least 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 13.
81. The method according to any of embodiments 70-80, wherein the bispecific antibody specifically 
binds to VEGF-A.
82. The method according to any of embodiments 74-81, wherein a heavy chain variable region of the 
anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and a light chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
83. The method according to any of embodiments 74-82, wherein the amino acid sequence of the 
heavy chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 11; or
wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody has 
at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 11.
84. The method according to any of embodiments 70-83, wherein the amino acid sequence of the 
heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or
wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 95%, 
96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17.
85. The method according to any of embodiments 70-84, wherein the anti-VEGF antibody is 
bevacizumab.
86. The method according to any of embodiments 70-85, wherein the method comprises the step of 
determining the CPS in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages; and/or
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably 
by using the anti-PD-L1 antibody 22C3) ; and/or
(iv) viable tumour cells are determined by staining with a viability dye; and/or
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 
staining lymphocytes and PD-L1 staining macrophages.
or optionally wherein PD-L1 staining cells are tumour cells with partial or complete linear membrane 
staining that is distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
87. The method according to any one of embodiments 70-85, wherein the method comprises the step 
of determining the TAP score in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer sample; and/or;
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells; and/or
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 
immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 
positive tumour cells and immune cells; and/or
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
88. The method according to any one of embodiments 70-85, wherein the method comprises the step 
of determining the TPS in a test sample of the subject by determining the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or;
(ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
(iii) viable tumour cells are determined by staining with a viability dye; and/or
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
89. The method according to embodiment 86-88, wherein the cancer tissue sample is a tissue section 
of a tumour biopsy, preferably wherein the tissue section is a formalin fixed and embedded in paraffin wax (FFPE) tissue section.
90. The method according to any of embodiments 70-89, wherein PD-L1 expression is detected by 
immunohistochemistry (IHC) staining.
91. The method according to embodiment 89 or 90, wherein the tissue section is stained.
92. The method according to embodiment 91, wherein the stain comprises a hematoxylin and eosin 
(H&E) stain, preferably wherein the viability dye is propidium iodide.
93. The method according to any of embodiments 86-92, wherein the viable tumour cells and the 
number of lymphocytes and macrophages are counted in the tumour nests and the adjacent supporting stroma of the tumour tissue sample.
94. The method according to any of embodiments 86-93, wherein the number of viable tumour cells in 
the tumour tissue sample is determined by flow cytometry.
95. A kit of parts comprising a bispecific antibody that specifically binds to PD-L1 and VEGF and a 
chemotherapy agent.
96. The kit of parts according to embodiment 95, wherein the bispecific antibody and the chemotherapy 
agent are comprised in separate container.
97. The kit of parts according to embodiment 95 or 96, further comprising instructions for use.
98. A chemotherapy agent for use in a method of treating a subject with cancer, the method comprising 
administering to the subject:
a. a bispecific antibody that specifically binds to PD-L1 and VEGF; and
b. the chemotherapy agent;
optionally wherein the subject has a PD-L1 expression score before the treatment as determined 
by a combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score; or
optionally wherein the subject has a PD-L1 expression before the treatment that provides a score 
of ≥ 1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
99. The chemotherapy agent for use according to embodiment 98, wherein the other integrating 
scoring algorithms are selected from TAP and TPS; or
wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when 
determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , preferably when determined by the tumour area positivity (TAP) score.
100. The chemotherapy agent for use according to embodiment 98 or 99, wherein the CPS has 
been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages; and/or
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably 
by using the anti-PD-L1 antibody 22C3) ; and/or
(iv) viable tumour cells are determined by staining with a viability dye; and/or
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 
staining lymphocytes and PD-L1 staining macrophages.
101. The chemotherapy agent for use according to embodiment 100, wherein the PD-L1 staining 
cells are tumour cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
102. The chemotherapy agent for use according to embodiment 100 or 101, wherein the viable 
tumour cells and the number of lymphocytes and macrophages are counted in the tumour nests and the adjacent supporting stroma of the tumour tissue sample.
103. The chemotherapy agent for use according to any of embodiments 98-102, wherein the 
PD-L1 expression score has been determined using a TAP scoring algorithm in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer sample; and/or;
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells; and/or
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 
immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 
positive tumour cells and immune cells; and/or
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
104. The chemotherapy agent for use according to embodiment 103, wherein the tumour area 
is determined using a hematoxylin and eosin (H&E) stain.
105. The chemotherapy agent for use according to embodiment 103 or 104, wherein the PD-L1 
positive tumour cells and immune cells are PD-L1 staining cells with partial or complete linear membrane staining that is perceived distinct from cytoplasmic staining, and immune cells (lymphocytes and macrophages) within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
106. The chemotherapy agent for use according to any of embodiments 98-105, wherein the 
PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or;
(ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
(iii) viable tumour cells are determined by staining with a viability dye; and/or
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
107. The chemotherapy agent for use according to embodiments 100 to 106, wherein the cancer 
tissue sample is a tissue section of a tumour biopsy.
108. The chemotherapy agent for use according to any of embodiments 98-107, wherein the 
PD-L1 expression is detected by immunohistochemistry (IHC) staining.
109. The chemotherapy agent for use according to any of embodiments 107 or 108, wherein the 
tissue section is a formalin fixed and embedded in paraffin wax (FFPE) tissue section.
110. The chemotherapy agent for use according to any of embodiments 107-109, wherein the 
tissue section is stained.
111. The chemotherapy agent for use according to embodiment 110, wherein the stain 
comprises a hematoxylin and eosin (H&E) stain, preferably wherein the viability dye is propidium iodide.
112. The chemotherapy agent for use according to any of embodiments 100-111, wherein the 
number of viable tumour cells in the tumour tissue sample is determined by flow cytometry.
113. The chemotherapy agent for use according to any of embodiments 98-112, wherein the 
bispecific antibody and the chemotherapy are separately administered.
114. The chemotherapy agent for use according to any of embodiments 98-113, wherein a 
dosage of the bispecific antibody and a dosage of the chemotherapy are administered concurrently or consecutively.
115. The chemotherapy agent for use according to any of embodiments 98-114, wherein the 
chemotherapy comprises a platinum-based chemotherapy.
116. The chemotherapy agent for use according to any of embodiments 98-115, wherein the 
subject has a PD-L1 expression score before the treatment as determined by a CPS of up to 20, preferably from 1 to 20, more preferably from 1 to <10, or other integrating scoring algorithm defining a similar PD-L1 expression score; or
wherein the subject has a PD-L1 expression before the treatment that provides a score of < 20, 
preferably from 1 to 20, more preferably from 1 to <10 when determined by the CPS or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
117. The chemotherapy agent for use according to any of embodiments 98-116, wherein the 
bispecific antibody dosage is in a range of from 0.1 mg/kg to 45 mg/kg body weight, preferably 1 mg/kg to 30 mg/kg body weight.
118. The chemotherapy agent for use according to any of embodiments 98-117, wherein the 
bispecific antibody and/or the chemotherapy is administered intravenously, preferably wherein the bispecific antibody and the chemotherapy are administered intravenously.
119. The chemotherapy agent for use according to any of embodiments 98-118, wherein the 
bispecific antibody and/or the chemotherapy is administered via an IV injection or IV infusion.
120. The chemotherapy agent for use according to any of embodiments 98-119, wherein a 
treatment cycle is repeated at least 1, 2, 3, 4, 5, 6, 7 or 8 times.
121. The chemotherapy agent for use according to any of embodiments 98-120, wherein each 
cycle has up to 28 days, preferably 28 or 21 days.
122. The chemotherapy agent for use according to any of embodiments 98-121, wherein the 
subject has not been previously treated for cancer.
123. The chemotherapy agent for use according to any of embodiments 98-121, wherein the 
subject has been previously treated for cancer, preferably wherein the subject had at least one previous chemotherapy treatment.
124. The chemotherapy agent for use according to any of embodiments 98-123, wherein the 
bispecific antibody is administered every 6 weeks, preferably every 4 weeks, more preferably every 3 weeks or every 2 weeks.
125. The chemotherapy agent for use according to any of embodiments 98-124, wherein the 
bispecific antibody is administered every 2 weeks at a dosage ranging from 10 mg/kg to 30 mg/kg, preferably ranging from 15 mg/kg to 25 mg/kg, more preferably at a dosage of 20 mg/kg.
126. The chemotherapy agent for use according to any of embodiments 98-125, wherein the 
bispecific antibody is administered every 3 weeks at a dosage ranging from 20 mg/kg to 40 mg/kg, preferably ranging from 25 mg/kg to 35 mg/kg, more preferably at a dosage of 30 mg/kg.
127. The chemotherapy agent for use according to any of embodiments 98-126, wherein the 
chemotherapy is administered once or more within the first 20 days or the first 21 days of each cycle, wherein the chemotherapy is administered twice or more within the first 20 days or the first 21 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 20 days or the first 21 days of each cycle.
128. The chemotherapy agent for use according to any of embodiments 98-127, wherein the 
chemotherapy is administered once or more within the first 14 days or the first 15 days of each cycle, wherein the chemotherapy is administered twice or more within the first 14 days or the first 15 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 14 days or the first 15 days of each cycle.
129. The chemotherapy agent for use according to any of embodiments 98-128, wherein the 
chemotherapy is administered on the 1st, 8th, and 15th days of each cycle.
130. The chemotherapy agent for use according to any of embodiments 98-129, wherein overall 
survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
131. The chemotherapy agent for use according to any of embodiments 98-130, wherein median 
progression-free survival is increased in said subject compared to the chemotherapy or the bispecific antibody or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
132. The chemotherapy agent for use according to any of embodiments 98-131, wherein the 
cancer comprises one or more solid tumours.
133. The chemotherapy agent for use according to any of embodiments 98-132, wherein the 
cancer is selected from the group consisting of melanoma, lung, liver, stomach, renal cell, urothelial, cervical, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancers, preferably wherein the cancer is selected from urothelial, breast and esophagus cancer.
134. The chemotherapy agent for use according to any of embodiments 98-133, wherein the 
cancer is small cell lung cancer (SCLC) , non-small cell lung cancer (NSCLC) or triple-negative breast cancer (TNBC) , preferably advanced triple-negative breast cancer.
135. The chemotherapy agent for use according to embodiment 134, wherein the NSCLC has 
a squamous histology.
136. The chemotherapy agent for use according to embodiment 134, wherein the NSCLC has 
a non-squamous histology.
137. The chemotherapy agent for use according to embodiment 134, wherein the NSCLC is an 
EGFR mutation-positive NSCLC.
138. The chemotherapy agent for use according to any of embodiments 98-137, wherein the 
bispecific antibody comprises an anti-PD-L1 antibody or fragment thereof.
139. The chemotherapy agent for use according to any of embodiments 98-138, wherein the 
bispecific antibody comprises an anti-VEGF antibody or fragment thereof.
140. The chemotherapy agent for use according to any of embodiments 98-139, wherein the 
bispecific antibody comprises a Fab, Fab’, F (ab’) 2, Fd, Fv, sdAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH.
141. The chemotherapy agent for use according to any of embodiments 98-140, wherein the 
bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably wherein each VHH is fused to the C-terminus of the anti-VEGF antibody.
142. The chemotherapy agent for use according to any of embodiments 98-141, wherein the 
bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1 or 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO:2 or 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3 or 38.
143. The chemotherapy agent for use according to embodiment 142, wherein the amino acid 
sequence of the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 9.
144. The chemotherapy agent for use according to any of embodiments 139-143, wherein the 
anti-VEGF antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
145. The chemotherapy agent for use according to any of embodiments 139-144, wherein the 
anti-VEGF antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO: 13 or having at least 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 13.
146. The chemotherapy agent for use according to any of embodiments 98-145, wherein the 
bispecific antibody specifically binds to VEGF-A.
147. The chemotherapy agent for use according to any of embodiments 139-146, wherein a 
heavy chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and
a light chain variable region of the anti-VEGF antibody comprises: (i) a complementarity-
determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
148. The chemotherapy agent for use according to embodiments 147, wherein the amino acid 
sequence of the heavy chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 11; or
wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody has 
at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody has at least 95%, 96%, 97%, 98%, or 99% identity to SEQ ID NO: 11.
149. The chemotherapy agent for use according to any of embodiments 98-148, wherein the 
amino acid sequence of the heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17.
150. The chemotherapy agent for use according to any of embodiments 139-149, wherein the 
anti-VEGF antibody is bevacizumab.
151. The chemotherapy agent for use according to any of embodiments 98-150, wherein the 
bispecific antibody is encoded by one or more nucleic acid molecules.
152. The chemotherapy agent for use according to any of embodiments 98-151, wherein the 
chemotherapy agent is selected from lurbinectedin, topotecan, taxane such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) , or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
153. The chemotherapy agent for use according to any of embodiments 115-152, wherein the 
platinum-based chemotherapy comprises cisplatin, oxaliplatin or carboplatin.
154. The chemotherapy agent for use according to any of embodiments 98-153, wherein the 
method of treatment comprises administering the bispecific antibody in combination with paclitaxel to a subject having small cell lung cancer.
155. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody in combination with pemetrexed and carboplatin (preferably as first line treatment of non-squamous NSCLC) or a taxane (preferably as first line treatment of squamous NSCLC) , such paclitaxel or docetaxel (preferably as second line treatment of NSCLC) , to a subject having NSCLC, preferably as second line treatment, or wherein the method of treatment comprises administering the bispecific antibody in combination with pemetrexed and carboplatin to a subject having malignant mesothelioma.
156. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody in combination with nab-paclitaxel to a subject having triple-negative breast cancer.
157. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody in combination with oxaliplatin, calcium folinate, and 5-fluorouracil to a subject having hepatocellular carcinoma, preferably wherein the method of treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg being particularly preferred) .
158. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody in combination with irinotecan, 5-fluorouracil, calcium folinate to a subject having unresectable neuroendocrine neoplasm.
159. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody in combination with etoposide and platinum to a subject having small cell lung cancer, preferably as first line treatment and/or extensive-stage small cell lung cancer, most preferably as first line treatment of extensive-stage small cell lung cancer or as second-line treatment of limited-stage small cell lung cancer.
160. The chemotherapy agent for use according to embodiment 159, wherein the method of 
treatment comprises administering the bispecific antibody every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg or 30 mg/kg being particularly preferred) or at a dose ranging from 1400 mg to 2000 mg in combination with etoposide (preferably at a dosage of 100 mg/m2) with carboplatin (preferably at a dosage of AUC=5 (total dose ≤ 750 mg) to a subject having small cell lung cancer, preferably as first line treatment and/or extensive-stage small cell lung cancer, most preferably as first line treatment of extensive-stage small cell lung cancer or as second-line treatment of limited-stage small cell lung cancer.
161. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (with 20 mg/kg or 30 mg/kg being particularly preferred) in combination with paclitaxel, lurbinectedin, or topotecan to a subject having small cell lung cancer, preferably as second line treatment and/or extensive-stage small cell lung cancer, more preferably wherein the method of treatment comprises administering to a patient suffering from advanced small cell lung cancer as second line treatment the bispecific antibody every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with (i) paclitaxel every 3 weeks at a dosage of 175 mg/m2, optionally for 5 cycles, or (ii) topotecan at a dosage of 1.5 mg/m2 administered IV or 2.3 mg/m2 administered orally once per day on days 1 to 5 of every 3 weeks cycle.
162. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administering the bispecific antibody every 2 weeks at a dosage ranging from 10 mg/kg to 20 mg/kg in combination with nab-paclitaxel, paclitaxel, or gemcitabine with carboplatin to a subject having triple-negative breast cancer.
163. The chemotherapy agent for use according to any of embodiments 98-154, wherein the 
method of treatment comprises administered the bispecific antibody every 2 weeks at a dosage of 1000 mg to 2000 mg to a subject having triple-negative breast cancer, preferably on the 1st and 15th day of a 28-day treatment cycle and preferably wherein the bispecific antibody is administered in combination with a chemotherapy, preferably on the 1st, 8th, and 15th day of a 28-day treatment cycle.
DEFINITIONS
The terms indicated for explanation of the invention and the disclosure have the following meaning, unless otherwise indicated in the description or the claims. Additional definitions are set forth throughout the detailed description.
Terms “a” and “an” and “the” and similar reference used in the context of describing the invention (especially in the context of the claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.
The terms “about” or “approximately” as used herein denotes a range of ±10%of a reference value. For examples, “about 10” defines a range of 9 to 11. In general, those skilled in the art, familiar with the context, will appreciate the relevant degree of variance encompassed by “about” or “approximately” in that context.
The term “adjuvant” relates to a compound which prolongs, enhances or accelerates an immune response. Adjuvants comprise a heterogeneous group of compounds such as oil emulsions (e.g., Freund’s adjuvants) , mineral compounds (such as alum) , bacterial products (such as Bordetella pertussis toxin) , or immune-stimulating complexes. Examples of adjuvants include, without limitation, LPS, GP96, CpG oligodeoxynucleotides, growth factors, and cytokines, such as monokines, lymphokines, interleukins, chemokines. The chemokines may be IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-12, INFa, INF-γ, GM-CSF, LT-a. Further known adjuvants are aluminum hydroxide, Freund’s adjuvant or oil such as ISA51. Other suitable adjuvants for use in the present disclosure include lipopeptides, such as Pam3Cys, as well as lipophilic components, such as saponins, trehalose-6, 6-dibehenate (TDB) , monophosphoryl lipid-A (MPL) , monomycoloyl glycerol (MMG) , or glucopyranosyl lipid adjuvant (GLA) .
“Antibodies” are glycoproteins belonging to the immunoglobulin superfamily. The term "full-length antibody" may refer to an immunoglobulin molecule that binds to a target molecule and contains four peptide chains: two heavy chains and two light chains which are connected to each other through disulfide bonds. An antibody may recognise an antigen via the fragment antigen-binding (Fab) variable region. The fragment crystallizable region (Fc region) is the tail region of an antibody that may allow antibodies to activate the immune system. The hinge region is a stretch of heavy chains linking the Fab and Fc regions. The heavy chain and light chain may each comprise a variable region and one or more constant domains. For example, in IgG antibodies, a heavy chain comprises a variable region (VH) and three constant domains (CH1, CH2, and CH3) and a light chain comprises a variable region (VL) and one constant domain (CL) . Examples antibodies include a human antibody, a humanized antibody, a chimeric antibody, a multispecific antibody, a monoclonal antibody, and a polyclonal antibody.
The term “antibody fragment” may refer to a fragment of an antibody, or a genetically engineered product of one of more fragments of an antibody, which fragment is involved in binding with the target molecule. Examples of antibody fragments include an antigen-binding fragment (Fab) , a Fab', a Fab'-SH, a fragment antibody (F (ab’) 2) , a variable region (Fv) , a single chain antibody (scFv) , a single-domain antibody (sdAb) , and a camelid antibody (VHH) . The term “Antigen-binding fragment” or “Fab” refers to a region of an antibody that binds to antigens and is composed of one constant and one variable region of each of the heavy and the light chain. The term “fragment antibody” or “F (ab’) 2” refers to a region of an antibody that remains following digestion of the Fc region while leaving intact some of the hinge region. The term “Fab’ ” refers to a fragment formed by the reduction of a F (ab') 2 fragment. The term “Fab’-SH” refers to a Fab’ fragment with a free sulfhydryl group.
The term "binding agent" in the context of the present disclosure refers to any agent capable of binding to desired antigens. In certain embodiments, the binding agent is an antibody, antibody fragment, or construct thereof. The binding agent may also comprise synthetic, modified or non-naturally occurring moieties, in particular non-peptide moieties. Such moieties may, for example, link desired antigen-binding functionalities or regions such as antibodies or antibody fragments. In one embodiment, the binding agent is a synthetic construct comprising antigen-binding CDRs or variable regions.
As used herein, the terms "binding" or "capable of binding” in the context of the binding of an antibody to a predetermined antigen or epitope typically is a binding with an affinity corresponding to a KD of about 10-7 M or less, such as about 10-8 M or less, such as about 10-9 M or less, about 10-10 M or less, or about 10-11 M or even less, when determined, for instance, using Bio-Layer Interferometry (BLI) or using surface plasmon resonance (SPR) technology in a BIAcore 3000 instrument using the antigen as the ligand and the antibody as the analyte. The antibody binds to the predetermined antigen with an affinity corresponding to a KD that is at least ten-fold lower, such as at least 100-fold lower, for instance at least 1,000-fold lower, such as at least 10,000-fold lower, for instance at least 100,000-fold lower than its KD for binding to a non-specific antigen (e.g., BSA, casein) other than the predetermined antigen or a closely-related antigen. The amount with which the affinity is higher is dependent on the KD of the antibody, so that when the KD of the antibody is very low (that is, the antibody is highly specific) , then the degree to which the affinity for the antigen is lower than the affinity for a non-specific antigen may be at least 10,000-fold. The term "kd" (sec-1) , as used herein, refers to the dissociation rate constant of a particular antibody-antigen interaction. Said value is also referred to as the koff value. The term "KD" (M) , as used herein, refers to the dissociation equilibrium constant of a particular antibody-antigen interaction.
A binding region as defined herein (such as an antibody or a fragment thereof as defined herein) "antagonizes the PD-1/PD-L1 interaction" means that the binding region is antagonistic towards the PD-1/PD-L1 signaling pathway. In particular, such binding region which antagonizes the PD-1/PD-L1 interaction totally or partially inhibits, reduces, interferes with or negatively modulates the PD-1/PD-L1 signaling pathway by binding to PD-1, PD-L1, or both. Preferably, the binding region which antagonizes the PD-1/PD-L1 interaction mediates its antagonistic activity in a manner that totally inhibits (i.e., blocks) or partially reduces the binding of PD-L1 to PD-1.
A binding region as defined herein (such as an antibody or a fragment thereof as defined herein) "antagonizes the VEGF/VEGFR interaction" means that the binding region is antagonistic towards the VEGF/VEGFR signaling pathway. In particular, such binding region which antagonizes the VEGF/VEGFR interaction totally or partially inhibits, reduces, interferes with or negatively modulates the VEGF/VEGFR signaling pathway by binding to VEGF, VEGFR, or both. Preferably, the binding region which antagonizes the VEGF/VEGFR interaction mediates its antagonistic activity in a manner that totally inhibits (i.e., blocks) or partially reduces the binding of VEGF to VEGFR.
As used herein, the term "cancer disease" or "cancer" refers to or describes the physiological condition in an individual that is typically characterized by unregulated cell growth and typically a disease characterized by aberrantly regulated cellular growth, proliferation, differentiation, adhesion, and/or migration. By "cancer cell" is meant an abnormal cell that grows by a rapid, uncontrolled cellular proliferation and continues to grow after the stimuli that initiated the new growth cease. The term "cancer" according to the present disclosure also comprises cancer metastases. By "metastasis" is meant the spread of cancer cells from its original site to another part of the body. The formation of metastasis is a very complex process and depends on detachment of malignant cells from the primary tumor, invasion of the extracellular matrix, penetration of the endothelial basement membranes to enter the body cavity and vessels, and then, after being transported by the blood, infiltration of target organs. Finally, the growth of a new tumor, i.e. a secondary tumor or metastatic tumor, at the target site depends on angiogenesis. Tumor metastasis often occurs even after the removal of the primary tumor because tumor cells or components may remain and develop metastatic potential. In one embodiment, the term "metastasis" according to the present disclosure relates to "distant metastasis" which relates to a metastasis which is remote from the primary tumor and the regional lymph node system.
The term “carrier” refers to a component which may be natural, synthetic, organic, inorganic in which the active component is combined in order to facilitate, enhance or enable administration of the pharmaceutical composition. A carrier as used herein may be one or more compatible solid or liquid fillers, diluents or encapsulating substances, which are suitable for administration to subject. Suitable carriers include, without limitation, sterile water, Ringer, Ringer lactate, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy-propylene copolymers. In some embodiments, the pharmaceutical composition of the present disclosure includes isotonic saline. Pharmaceutically acceptable carriers, excipients or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington’s Pharmaceutical Sciences, Mack Publishing Co. (A. R Gennaro edit. 1985) . Pharmaceutical carriers, excipients or diluents can be selected with regard to the intended route of administration and standard pharmaceutical practice.
“CDR” or “CDRs” means complementarity determining region (s) in an immunoglobulin variable region. The variable regions of the heavy and light chains each contain three CDRs, designated CDR1, CDR2 and CDR3. The precise boundaries of these CDRs can be defined according to various numbering systems known in the art (see e.g. Dondelinger, M., et al., 2018. Frontiers in immunology, 9, p. 2278) , such as the Kabat numbering system (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md., 1991; Kabat et al., 1992, Sequences of Proteins of Immunological Interest, DIANE Publishing: 2719) , the Chothia numbering system (Chothia & Lesk (1987) J. Mol. Biol. 196: 901-917; Chothia et al. (1989) Nature 342: 878-883) or the IMGT numbering system (Lefranc et al., Dev. Comparat. Immunol. 27: 55-77, 2003; Ehrenmann F, Kaas Q, Lefranc M P. IMGT/3Dstructure-DB and IMGT/DomainGapAlign: a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF [J] . Nucleic acids research, 2009; 38 (suppl_1) : D301-D307) . For a given antibody, those skilled in the art will readily identify the CDRs defined by each numbering system. Also, the correspondence between different numbering systems is well known to those skilled in the art (for example, see Lefranc et al., Dev. Comparat. Immunol. 27: 55-77, 2003) .
The terms “chemotherapeutic agent” or “chemotherapeutical agent” or “chemotherapy agent” can be used interchangeably herein. A chemotherapeutic agent is a chemical compound useful in the treatment of cancer. Classes of chemotherapeutic agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytotoxic/antitumour antibiotics, topoisomerase inhibitors, photosensitizers, anti-estrogens and selective estrogen receptor modulators (SERMs) , anti-progesterones, estrogen receptor down-regulators (ERDs) , estrogen receptor antagonists, leutinizing hormone-releasing hormone agonists, anti-androgens, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, anti-sense oligonucleotides that that inhibit expression of genes implicated in abnormal cell proliferation or tumour growth. Chemotherapeutic agents useful in the treatment methods of the present invention include cytostatic and/or cytotoxic agents. Chemotherapeutic agents as used herein do not include antibodies.
“Chimeric antibody” refers to an antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, while the remainder of the chain (s) is identical with or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity.
Unless expressly specified otherwise, the term “comprising” is used in the context of the present disclosure to indicate that further members may optionally be present in addition to the members of the list introduced by “comprising” . It is, however, contemplated as specific embodiments of the present invention that each time the term “comprising” is used, this shall also encompass the possibility of no further members being present, i.e., for the purpose of this embodiment “comprising” can be understood as having the meaning of “consisting of” .
“Combination therapy” refers to the bispecific antibody disclosed herein that specifically binds to PD-L1 and VEGF in combination with the chemotherapy disclosed herein for use in a method of treating a subject with cancer, optionally wherein the subject has been determined to have a combined positive score (CPS) of ≥ 1 before the treatment. Each component of the combination therapy, i.e., the bispecific antibody and the chemotherapy may be administered separately.
The “combined positive score” or “CPS, ” refers to a well-known algorithm for determining a PD-L1 expression score from a tumour sample of a subject (see for example Kulangara, Karina & Hanks, Debra & Waldroup, Stephanie & Peltz, Lindsay & Shah, Supriya & Roach, Charlotte & Juco, Jonathan & Emancipator, Kenneth & Stanforth, Dave. (2017) , Development of the combined positive score (CPS) for the evaluation of PD-L1 in solid tumours with the immunohistochemistry assay PD-L1 IHC 22C3 pharmDx. Journal of Clinical Oncology) . When using CPS, the PD-L1 expression score is determined by taken into account the number of PD-L1 staining cells (i.e., tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages; and/or
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by 
using the anti-PD-L1 antibody 22C3) ; and/or
(iv) viable tumour cells are determined by staining with a viability dye; and/or
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 staining 
lymphocytes and PD-L1 staining macrophages.
It can be preferred that the CPS has been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer tissue sample;
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages;
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by 
using the anti-PD-L1 antibody 22C3) ;
(iv) viable tumour cells are determined by staining with a viability dye; and
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 staining 
lymphocytes and PD-L1 staining macrophages.
The CPS is approved as companion diagnostic for the treatment of cancer using pembrolizumab.
The term “disease control rate” or DCR is the percentage of all subjects in a study or treatment group who have either a complete response, a partial response, or a stable disease to the treatment (CR, PR or SD) . The DCR can be calculated by adding the numbers of subjects having CR, the numbers of subjects having PR, and the numbers of subjects having SD and dividing the resulting sum by the total number of subjects in the treatment group. The DCReval, i.e., the DCR of all evaluable subjects in a study or treatment group, is the percentage of all evaluable subjects in a study or treatment group who have either a complete response, a partial response, or a stable disease to the treatment (CR, PR or SD) .
The term "Duration of response” or DOR only applies to subjects whose confirmed best overall response is CR or PR and is defined as the time from the first documentation of objective tumor response (CR or PR) to the date of first PD or death due to underlying cancer.
The term “diluent” relates to a diluting and/or thinning agent. Moreover, the term “diluent” includes any one or more of fluid, liquid or solid suspension and/or mixing media. Examples of suitable diluents include ethanol, glycerol, and water.
Herein, the term “DNA” relates to a nucleic acid molecule which is entirely or at least substantially composed of deoxyribonucleotide residues. In preferred embodiments, the DNA contains all or a majority of deoxyribonucleotide residues. As used herein, “deoxyribonucleotide” refers to a nucleotide which lacks a hydroxyl group at the 2’-position of a β-D-ribofuranosyl group. DNA encompasses without limitation, double stranded DNA, single stranded DNA, isolated DNA such as partially purified DNA, essentially pure DNA, synthetic DNA, recombinantly produced DNA, as well as modified DNA that differs from naturally occurring DNA by the addition, deletion, substitution and/or alteration of one or more nucleotides. Such alterations may refer to addition of non-nucleotide material to internal DNA nucleotides or to the end (s) of DNA. It is also contemplated herein that nucleotides in DNA may be non-standard nucleotides, such as chemically synthesized nucleotides or ribonucleotides. For the present disclosure, these altered DNAs are considered analogs of naturally-occurring DNA. A molecule contains “a majority of deoxyribonucleotide residues” if the content of deoxyribonucleotide residues in the molecule is more than 50% (such as at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%) , based on the total number of nucleotide residues in the molecule. The total number of nucleotide residues in a molecule is the sum of all nucleotide residues (irrespective of whether the nucleotide residues are standard (i.e., naturally occurring) nucleotide residues or analogs thereof) . DNA may be recombinant DNA and may be obtained by cloning of a nucleic acid, in particular cDNA. The cDNA may be obtained by reverse transcription of RNA.
As used herein, the term “effective amount” refers to an amount of a given substance that is sufficient in quantity to produce a desired effect, including an improvement or remediation of the disease, disorder, or symptoms of the disease or condition. The combination therapy described herein is to be administered to a patient in need therefore in an effective amount.
As used herein, the term “encode” or “encoding” refers to sequence information of a first molecule that guides production of a second molecule having a defined sequence of nucleotides (e.g., mRNA) or a defined sequence of amino acids. For example, a DNA molecule can encode an RNA molecule (e.g., by a transcription process that includes a DNA-dependent RNA polymerase enzyme) . An RNA molecule can encode a polypeptide (e.g., by a translation process) . Thus, a gene, a cDNA, or a single-stranded RNA (e.g., an mRNA) encodes a polypeptide if transcription and translation of mRNA corresponding to that gene produces the polypeptide in a cell or other biological system. In some embodiments, a coding region of a single-stranded RNA encoding a target polypeptide agent refers to a coding strand, the nucleotide sequence of which is identical to the mRNA sequence of such a target polypeptide agent. In some embodiments, a coding region of a single-stranded RNA encoding a target polypeptide agent refers to a non-coding strand of such a target polypeptide agent, which may be used as a template for transcription of a gene or cDNA. As is understood in the art, the phrase “nucleic acid encoding a peptide or protein” means that the polynucleotide, if present in the appropriate environment, for example within a cell and/or in a cell-free translation system, can direct the assembly of amino acids to produce the peptide or protein via a process of translation.
The term “epitope” refers to the part of an antigen that as used herein, refers to an agent that elicits an immune response; and/or (ii) an agent that binds to a T cell receptor (e.g., when presented by an MHC molecule) or to an antibody. For example, epitopes are the discrete, three-dimensional sites on an antigen, which are recognized by the immune system. Epitopes usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three-dimensional structural characteristics, as well as specific charge characteristics. Conformational and non-conformational epitopes are distinguished in that the binding to the former but not the latter is lost in the presence of denaturing solvents.
The term “excipient” as used herein refers to a substance which may be present in a pharmaceutical composition of the present disclosure but is not an active ingredient. Examples of excipients, include without limitation, carriers, binders, diluents, lubricants, thickeners, surface active agents, preservatives, stabilizers, emulsifiers, buffers, flavoring agents, or colorants.
As used herein, the term “gene” refers to a DNA sequence in a chromosome that codes for a protein. In some embodiments, a gene includes coding sequence (i.e., sequence that encodes a particular protein) ; in some embodiments, a gene includes non-coding sequence. In some particular embodiments, a gene may include both coding (e.g., exonic) and non-coding (e.g., intronic) sequences. In some embodiments, a gene may include one or more regulatory elements that, for example, may control or impact one or more aspects of gene expression (e.g., cell-type-specific expression, inducible expression, etc. ) .
“Human antibody” refers to an antibody that comprises human immunoglobulin protein sequences only. A human antibody may contain murine carbohydrate chains if produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse cell. Similarly, “mouse antibody” or “rat antibody” refer to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
“Humanized antibody” refers to forms of antibodies that contain sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain minimal sequence derived from non-human immunoglobulin. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable regions, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin sequence.
The term “immune cell” means any cell of hematopoietic lineage involved in regulating an immune response against an antigen (e.g., a bacterial or viral infection or an auto-antigen) . In typical embodiments, an immune cell is a leukocyte, such as a white blood cell. Immune cells include neutrophils, eosinophils, basophils, lymphocytes, and/or monocytes. Lymphocytes include T lymphocytes and B lymphocytes. Immune cells can also be dendritic cells, natural killer (NK) cells, and/or a mast cell.
The term "immunoglobulin" relates to proteins of the immunoglobulin superfamily, preferably to antigen receptors such as antibodies or the B cell receptor (BCR) . The immunoglobulins are characterized by a structural domain, i.e., the immunoglobulin domain, having a characteristic immunoglobulin (Ig) fold. The term encompasses membrane bound immunoglobulins as well as soluble immunoglobulins. Membrane bound immunoglobulins are also termed surface immunoglobulins or membrane immunoglobulins, which are generally part of the BCR. Soluble immunoglobulins are generally termed antibodies. The structure of immunoglobulins has been well characterized. See, e.g., Fundamental Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N. Y. (1989) ) . Briefly, immunoglobulins generally comprise several chains, typically two identical heavy chains and two identical light chains which are linked via disulfide bonds. These chains are primarily composed of immunoglobulin domains or regions, such as the VL or VL (variable light chain) domain/region, CL or CL (constant light chain) domain/region, VH or VH (variable heavy chain) domain/region, and the CH or CH (constant heavy chain) domains/regions CH1 (CH1) , CH2 (CH2) , CH3 (CH3) , and CH4 (CH4) . The heavy chain constant region typically is comprised of three domains, CH1, CH2, and CH3. The hinge region is the region between the CH1 and CH2 domains of the heavy chain and is highly flexible. Disulfide bonds in the hinge region are part of the interactions between two heavy chains in an IgG molecule. Each light chain typically is comprised of a VL and a CL. The light chain constant region typically is comprised of one domain, CL. The VH and VL regions may be further subdivided into regions of hypervariability (or hypervariable regions which may be hypervariable in sequence and/or form of structurally defined loops) , also termed complementarity determining regions (CDRs) , interspersed with regions that are more conserved, termed framework regions (FRs) . Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 (see also Chothia and Lesk, J. Mol. Biol. 196, 901-917 (1987) ) .
As used herein, an "instructional material" or "instructions" includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the compositions and methods described herein. The instructional material of the kit of the disclosure may, for example, be affixed to a container which contains the compositions of the disclosure or be shipped together with a container which contains the compositions. Alternatively, the instructional material may be shipped separately from the container with the intention that the instructional material and the compositions be used cooperatively by the recipient.
The term “Kabat, ” as used herein, means an immunoglobulin alignment and numbering system pioneered by Elvin a. Kabat ( (1991) Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institutes of Health, Bethesda, Md. ) or Kabat et al., 1992, Sequences of Proteins of Immunological Interest, DIANE Publishing: 2719.
The term “kit” as used herein is synonymous with the term “kit of part” or “kit of parts” and refers to a plurality of compositions that may, e.g., be shipped or stored together, in particular because they are relevant for or required for an administration regimen as disclosed herein.
The term “Mononuclear inflammatory cells” (MIC) as used herein refer to lymphocytes and macrophages.
The term "monoclonal antibody" refers to an antibody obtained from a substantially homogeneous population of antibodies. The individual antibodies composing the population may be identical except for possible naturally occurring mutations, which may be present in minor amounts. Monoclonal antibodies are highly specific and target a single antigenic epitope. In contrast, polyclonal antibody preparations typically include a large number of antibodies which are specific for different epitopes.
The term “nucleoside” relates to compounds which can be thought of as nucleotides without a phosphate group. While a nucleoside is a nucleobase linked to a sugar (e.g., ribose or deoxyribose) , a nucleotide is composed of a nucleoside and one or more phosphate groups. Examples of nucleosides include cytidine, uridine, pseudouridine, adenosine, and guanosine. The five standard nucleosides which usually make up naturally occurring nucleic acids are uridine, adenosine, thymidine, cytidine and guanosine. The five nucleosides are commonly abbreviated to their one letter codes U, A, T, C and G, respectively. However, thymidine is more commonly written as “dT” ( “d” represents “deoxy” ) as it contains a 2’-deoxyribofuranose moiety rather than the ribofuranose ring found in uridine. This is because thymidine is found in deoxyribonucleic acid (DNA) and not ribonucleic acid (RNA) . Conversely, uridine is found in RNA and not DNA. The remaining three nucleosides may be found in both RNA and DNA. In RNA, they would be represented as A, C and G, whereas in DNA they would be represented as dA, dC and dG. A modified purine (A or G) or pyrimidine (C, T, or U) base moiety is, in some embodiments, modified by one or more alkyl groups, e.g., one or more C1-4 alkyl groups, e.g., one or more methyl groups. Particular examples of modified purine or pyrimidine base moieties include N7-alkyl-guanine, N6-alkyl-adenine, 5-alkyl-cytosine, 5-alkyl-uracil, and N (1) -alkyl-uracil, such as N7-C1-4 alkyl-guanine, N6-C1-4 alkyl-adenine, 5-C1-4 alkyl-cytosine, 5-C1-4 alkyl-uracil, and N (1) -C1-4 alkyl-uracil, preferably N7-methyl-guanine, N6-methyl-adenine, 5-methyl-cytosine, 5-methyl-uracil, and N (1) -methyl-uracil.
The term “objective response rate” or ORR is the percentage of all subjects in a study or treatment group who have either a partial or complete response to the treatment. The ORR can be calculated by adding the numbers of subjects having CR and the numbers of subjects having PR and dividing the resulting sum by the total number of subjects in the treatment group. The ORReval, i.e., the ORR of all evaluable subjects in a study or treatment group, is the percentage of all evaluable subjects in a study or treatment group who have either a partial or complete response to the treatment.
The term "Overall survival” or OS is defined as the number of days from Day 1 in Cycle 1 to death due to any cause. If a subject is not known to have died, then OS will be censored at the latest date the subject was known to be alive (on or before the cut-off date) .
A "PD-1 antibody" or "anti-PD-1 antibody" is an antibody as described above, which is directed against the antigen PD-1 and which preferably binds specifically to the antigen PD-1.
A "PD-L1 antibody" or "anti-PD-L1 antibody" is an antibody as described above, which is directed against the antigen PD-L1 and which preferably binds specifically to the antigen PD-L1.
PD-L1 expression score as used herein can be determined using a PD-L1 scoring algorithm such as CPS, TPS, or TAP. As known to the skilled person, the same PD-L1 expression can lead to different numerical values depending on the PD-L1 scoring algorithm.
The term “pharmaceutical composition” relates to a composition comprising a therapeutically effective agent, preferably together with pharmaceutically acceptable carriers, diluents and/or excipients. Said pharmaceutical composition is useful for treating, preventing, or reducing the severity of a disease by administration of said pharmaceutical composition to a subject.
The term “pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” means solvents, dispersion media, coatings, antibacterial agents and antifungal agents, isotonic agents, and absorption delaying agents, and the like, which are compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. In certain embodiments, the pharmaceutically acceptable carrier or excipient is not naturally occurring.
The term “platinum-containing chemotherapy” (also known as platins) refers to the use of chemotherapeutic agent (s) used to treat cancer that are coordination complexes of platinum. Platinum-containing chemotherapeutic agents are alkylating agents that crosslink DNA, resulting in ineffective DNA mismatch repair and generally leading to apoptosis. Examples of platins include cisplatin, carboplatin, and oxaliplatin.
The term “plurality” refers to the state of being plural.
The terms “polynucleotide” and “nucleic acid” can be used interchangeably herein to refer to polymers of nucleotides. The term “polynucleotide” comprises deoxyribonucleic acid (DNA) , ribonucleic acid (RNA) , combinations thereof, and modified forms thereof. The term comprises genomic DNA, cDNA, mRNA, recombinantly produced and chemically synthesized molecules. In some embodiments, a polynucleotide is DNA. In some embodiments, a polynucleotide is RNA. In some embodiments, a polynucleotide is a mixture of DNA and RNA. A polynucleotide may be present as a single-stranded or double-stranded and linear or covalently circularly closed molecule. A polynucleotide can be isolated. The term “isolated polynucleotide “means, according to the present disclosure, that the polynucleotide (i) was amplified in vitro, for example via polymerase chain reaction (PCR) for DNA or in vitro transcription (using, e.g., an RNA polymerase) for RNA, (ii) was produced recombinantly by cloning, (iii) was purified, for example, by cleavage and separation by gel electrophoresis, or (iv) was synthesized, for example, by chemical synthesis.
The terms “polypeptide, ” “peptide, ” and “protein” are used interchangeably herein to refer to polymers of amino acids.
The term "Progression-free survival” or PFS is defined as the number of days from Day 1 in Cycle 1 to the first documented progression or death due to any cause.
The term “recombinant” when used in the context of a polynucleotide means a polynucleotide having nucleotide sequences that are not naturally joined together and can be made by artificially combining two otherwise separated segments of sequence. This artificial combination is often accomplished by chemical synthesis or, more commonly, by the artificial manipulation of isolated segments of nucleic acids, for example, by genetic engineering techniques. Recombinant polynucleotides include vectors comprising an amplified or assembled polynucleotide, which can be used to transform or transfect a suitable host cell. A host cell that comprises the recombinant polynucleotide is referred to as a “recombinant host cell. ” The polynucleotide is then expressed in the recombinant host cell to produce a “recombinant polypeptide. ” A recombinant polynucleotide can also comprise a non-coding function.
A “sequence identity” as used herein is obtained by determining the number of identical positions at which the sequences to be compared correspond, dividing this number by the number of positions compared (e.g., the number of positions in the reference sequence) and multiplying this result by 100.
A “single domain antibody” (sdAb) is an antibody composed of a single variable region (e.g., heavy chain variable region) composed of antibody fragments. Typically, a single domain antibody, domain antibody or nanobody consists of 4 framework regions and 3 complementarity determining regions, the 4 framework regions are respectively FR1-FR4, and the 3 complementarity determining regions are respectively CDR1 -CDR3. In certain embodiments, the single domain antibody of the present application may have a structure of FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4. These antibodies do not require light chain variable regions to bind antigens with high affinity and specificity. Compared with antibodies composed of heavy chain and light chain, single domain antibodies have high solubility, high stability to heat, pH, protease and other deforming agents, and only need single-chain expression to facilitate large-scale production. As used herein, the term “framework region” or “FR” residues refers to those amino acid residues in an antibody variable region other than the CDR residues as defined above.
An antibody that “specifically binds to” a specified target protein is an antibody that exhibits preferential binding to that target as compared to other proteins, but this specificity does not require absolute binding specificity. An antibody is considered “specific” for its intended target if its binding is determinative of the presence of the target protein in a sample, e.g., without producing undesired results such as false positives. Antibodies, or binding fragments thereof, useful in the present invention will bind to the target protein with an affinity that is at least two fold greater, preferably at least ten times greater, more preferably at least 20-times greater, and most preferably at least 100-times greater than the affinity with non-target proteins. As used herein, an antibody is said to bind specifically to a polypeptide comprising a given amino acid sequence, e.g., the amino acid sequence of a mature human PD-L1 molecule, if it binds to polypeptides comprising that sequence but does not bind to proteins lacking that sequence.
As used herein, a “subject” is a human of either gender (a male or a female) . The subject may be of any age. In some embodiments, the subject is female. In another embodiment, the subject is male. In some embodiments, the subject is a subject having cancer, in particular a female subject having cancer and/or a male subject having cancer.
As disclosed in more detail herein below, the term “TAP” score refers to the “Tumour Area Positivity” score. The PD-L1 expression score can be determined using a TAP scoring algorithm by determining in a test sample of the subject the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer sample; and/or
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells; and/or
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 immunohistochemistry 
staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive 
tumour cells and immune cells; and/or
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
It can be preferred that the PD-L1 expression score has been determined using a TAP scoring algorithm in a test sample of the subject by determining the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer sample;
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells;
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1; 
immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ;
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive 
tumour cells and immune cells; and
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
Alternatively, the TAP score might be referred to as total percentage of the tumour area (tumour and any desmoplastic stroma) covered with tumour cells with PD-L1 membranous staining at any intensity and tumour-associated immune cells with any pattern.
As disclosed in more detail herein below, the term “TPS” refers to the “Tumour Proportion Score” . The PD-L1 expression score can be determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

optionally wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
(iii) viable tumour cells are determined by staining with a viability dye; and/or
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
It can be preferred that the PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer tissue sample;
(ii) the cancer tissue sample comprises at least 100 viable tumour cells;
(iii) viable tumour cells are determined by staining with a viability dye; and
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
The term “treating” when used in the context of a disease or disease condition means ameliorating, improving or remedying a disease, disorder, or symptom of a disease or condition associated with the disease, or can mean completely or partially stopping, on a molecular level, the biochemical basis of the disease, etc. It describes an act that leads to the elimination, reduction, alleviation, reversal, or prevention or delay of onset or recurrence of any symptom of a disease.
The term "treatment cycle" is herein defined as the time period, within the effects of separate dosages of the therapeutic agent (e.g., the anti-PD-1/PD-L1xVEGF/VEGFR binding agent or combination partner) cease to exist due to its pharmacodynamics, or in other words the time period after the subject's body is essentially cleared from the administrated therapeutic agent. Multiple small doses in a small time window, e.g. within 2-24 few hours, such as 2-12 hours or on the same day, might be equal to a larger single dose.
The term “triple-negative breast cancer” as used herein is used in the usually sense and refers to the cancer class that tests negative for estrogen receptors and progesterone receptors expression, and HER2 overexpression or gene amplification. TNBC is human epidermal growth factor receptor 2 (HER2) negative and has <1%expression of estrogen receptors (ER) and progesterone receptors (PR) by immunostaining. It is a biologically aggressive tumour, characterized by moderate/high grade and highly proliferative cancer cells, which, together with limited treatment options leads to the poorest prognosis among the breast cancer subtypes.
“Tumour” as it applies to a subject diagnosed with, or suspected of having, a cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumours and secondary neoplasms. A solid tumour is an abnormal growth or mass of tissue that usually does not contain cysts or liquid areas. Different types of solid tumours are named for the type of cells that form them. Examples of solid tumours are sarcomas, carcinomas, and lymphomas. Leukemias (cancers of the blood) generally do not form solid tumours (National Cancer Institute, Dictionary of Cancer Terms) .
“Variable regions” as used herein means the segment of an antibody which contains three CDRs, designated CDR1, CDR2 and CDR3. A “variable region” of an antibody refers to the variable region of the antibody light chain or the variable region of the antibody heavy chain, either alone or in combination. The variable region of the heavy chain may be referred to as “VH. ” The variable region of the light chain may be referred to as “VL. ” Typically, the variable regions of both the heavy and light chains comprise three hypervariable regions, the CDRs, which are located within relatively conserved framework regions (FR) . The CDRs are usually aligned by the framework regions, enabling binding to a specific epitope. In general, from N-terminal to C-terminal, both light and heavy chains variable regions comprise FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated herein, each individual value is incorporated into the specification as if it was individually recited herein. All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g., “such as” ) , provided herein is intended merely to better illustrate the invention and does not pose a limitation on the scope of the invention otherwise claimed. No language in the specification should be construed as indicating any non-claimed element essential to the practice of the invention.
A "VEGF antibody" or "anti-VEGF antibody" is an antibody as described above, which is directed against the antigen VEGF and which preferably binds specifically to the antigen VEGF.
A "VEGFR antibody" or "anti-VEGFR antibody" is an antibody as described above, which is directed against the antigen VEGFR and which preferably binds specifically to the antigen VEGFR.
All patents, patent applications, and other publications cited in this application are incorporated by reference in the entirety for all purposes.
THE COMBINATION THERAPY OF THE INVENTION
The targets
The present invention inter alia provides a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction (including, e.g., a bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably a chemotherapy agent
optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the 
combined positive score (CPS) of < 10, preferably ≥ 1, more preferably 1 to 20, most preferably 1 to <10 or other integrating scoring algorithm defining a similar PD-L1 expression score. It can be preferred that the subject has the afore-mentioned PD-L1 expression score before the treatment.
For example, the present invention provides a bispecific antibody that specifically binds to PD-L1 and VEGF for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy, preferably a chemotherapy agent
optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the 
combined positive score (CPS) of ≥ 1, preferably 1 to 20, more preferably 1 to <10.
As further example, the present invention provides a bispecific antibody that specifically binds to PD-L1 and VEGF for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy, preferably a chemotherapy agent
optionally wherein the subject has a PD-L1 expression score before the treatment as determined by an 
integrating scoring algorithm defining a PD-L1 expression score similar or identical to the CPS of ≥ 1, preferably 1 to 20, more preferably 1 to <10.
In some embodiments, a PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS means an ≥ 70%or ≥ 80%, preferably ≥85%, more preferably ≥90%, most preferably ≥95%average positive agreement (APA) and ≥ 70%or ≥ 80%, preferably ≥85%, more preferably ≥90%, most preferably ≥95%average negative agreement (ANA) , and ≥70%or ≥ 80%, preferably ≥85%, more preferably ≥90%, most preferably ≥95%overall percent agreement (OPA) between and within readers with 95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS. For example, the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ≥70%average positive agreement (APA) and ≥ 70%average negative agreement (ANA) , and ≥ 70%overall percent agreement (OPA) between and within readers with ≥95%confidence intervals (Cis) when comparing the scoring algorithm TAP with the CPS. For example, the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ≥85%average positive agreement (APA) and ≥ 85%average negative agreement (ANA) , and ≥ 85%overall percent agreement (OPA) between and within readers with ≥95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS. In some embodiments, the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ≥90%average positive agreement (APA) and ≥ 90%average negative agreement (ANA) , and ≥ 90%overall percent agreement (OPA) between and within readers with ≥95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS. In some embodiments, the PD-L1 expression score determined by an integrating scoring algorithm and defining a PD-L1 expression score similar to the CPS can mean an ≥95%average positive agreement (APA) and ≥ 95%average negative agreement (ANA) , and ≥ 95%overall percent agreement (OPA) between and within readers with ≥95%confidence intervals (Cis) when comparing the scoring algorithm with the CPS.
In some embodiments, the other integrating scoring algorithms are selected from TAP and TPS. In some preferred embodiments, the other integrating scoring algorithm is TAP.
The integrating scoring algorithm defining a similar PD-L1 expression score preferably is a tumour cell integrating scoring algorithm such TPS or a tumour cell and immune cell integrating scoring algorithm such as CPS or TAP.
Vascular endothelial growth factor (VEGF) , also known as vascular permeability factor (VPF) or vasculotropin, is a highly specific homodimer that promotes the growth of vascular endothelial cells. Body protein. VEGF family proteins include VEGF-A, VEGF-B, VEGF-C, VEGF-D, VEGF-E, VEGF-F and placental growth factor (PIGF) , among which VEGF-Ais involved in the early formation of blood vessels play an important role. In 1983, Senger et al. first isolated liver cancer cells from guinea pigs, which can increase the permeability of venules and venules, promote the division and proliferation of vascular endothelial cells, and induce the formation of blood vessels. Meanwhile, VEGF is involved in the pathogenesis and progression of many angiogenesis-dependent diseases, including cancer, certain inflammatory diseases, and diabetic retinopathy. Therefore, VEGF is an important target in antitumour drug research.
The main receptors of VEGF proteins are VEGFR1, VEGFR2, VEGFR3, NRP1, NRP2 and NRP3. However, the binding of VEGF family protein members to VEGF receptors is selective, and VEGFA can bind to VEGFR1 and VEGFR2, activate endogenous kinase activation, and promote new blood vessels. Blocking the binding of VEGF to the receptor can be applied to the treatment of various cancers, such as breast cancer, colon cancer, lung cancer, colorectal, ovarian cancer, endometrial cancer, mesothelioma (including malignant pleural mesothelioma [MPM] and malignant peritoneal mesothelioma [MPeM] ) , cervical cancer, kidney cancer (Rakesh R. Ramjiawan, Arjan W. Griffioen, and Dan G. Duda, Angiogenesis. 2017 20 (2) : 185–204. ) .
Apart of its angiogenesis-modulating capabilities VEGF, in particular VEGF-A, is associated with a range of immunosuppressive effects at successive steps in the cancer-immunity cycle, such as diminished antigen presentation, T cell priming, T cell trafficking, and T cell tumour infiltration. Hence, the binding agent (including, e.g., the bispecific antibody) binding to VEGF can release immunosuppression. For example, an anti-VEGF antibody treatment alone results in increased gene expression associated with Th1 chemokines involved with T-cell trafficking, tumour MHC-I protein expression and infiltration of tumour-specific T-cell clones demonstrating that an anti-VEGF antibody is capable of inducing anti-tumour immune responses.
By blocking the VEGF-A/VEGFR-2 interaction the binding agent (including, e.g., the bispecific antibody) may reverse VEGF-A mediated inhibition of monocyte-to-dendritic cell maturation thereby increasing numbers of dendritic cells (DCs) , and reverse VEGF-A mediated inhibition of dendritic cell maturation thereby increasing tumour-antigen presentation. The binding agent (including, e.g., the bispecific antibody) may further reverse or reduce VEGF-A mediated enhanced T cell exhaustion, as VEGF-A binding to VEGFR-2 on the surface of CD8+ T cells has been shown in preclinical studies to result in upregulated expression of the immune-checkpoint molecules (and exhaustion markers) PD-1, Cytotoxic T Lymphocyte antigen 4 (CTLA-4) , and T cell immunoglobulin mucin receptor 3 (TIM3) as well as that of lymphocyte activation gene 3 protein (LAG3) . The binding agent (including, e.g., the bispecific antibody) may further reverse or reduce VEGF-A mediated proliferation of Treg cells, and reverse VEGF-A mediated downregulation of adhesion molecules (e.g. ICAM-1, or CD34) to allow for more efficient immune cell infiltration of the tumour. VEGF thus has a major role in the generation of an immunosuppressive tumour microenvironment.
In addition, blocking the VEGF-A/VEGFR-2 interaction can induce physical changes in the tumour vasculature such as vascular normalization, a process whereby hypoxia is transiently alleviated and the tumour vasculature reverts back to resemble that of a nonmalignant tissue (Jain, R. K. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 307, 58–62 (2005) ) .
Programmed death-ligand 1 (PD-L1) , also known as CD274, is a member of the B7 family and is a ligand of PD-1. PD-L1 is a type I transmembrane protein with a total of 290 amino acids, including an IgV-like domain, an IgC-like domain, a transmembrane hydrophobic domain and an intracellular domain consisting of 30 amino acids. Unlike other B7 family molecules, PD-L1 negatively regulates immune responses. Studies have found that PD-L1 is mainly expressed in activated T cells, B cells, macrophages and dendritic cells, etc. In addition to lymphocytes, PD-L1 is also expressed in other tissues such as thymus, heart, placenta, etc. endothelial cells, as well as various non-lymphoid lineages such as melanoma, lung cancer, liver cancer, gastric cancer, renal cell cancer, urothelial cancer, cervical cancer, colorectal cancer, ovarian cancer, colon cancer, breast cancer, esophageal cancer, head and neck cancer, etc. (Akintunde Akinleye&Zoaib Rasool, Journal of Hematology&Oncology volume 12, Article number: 92 (2019) ) . PD-L1 regulates autoreactive T-and B-cells, and immune tolerance, and plays a role in peripheral tissue T-and B-cell responses. Interfering with the PD-1: PD-L1 interaction reinvigorates exhausted T cells, as PD-1 is a major regulator of T-cell exhaustion. Hence blocking the PD-1: PD-L1 pathway restores T-cell function and improves tumour eradication.
High expression of PD-L1 on tumour cells is associated with poor prognosis in cancer subjects. For a better prognosis in cancer subjects, it is therefore highly desired to provide an effective treatment for low PD-L1 expressing cancer.
Surprisingly, the combination therapy disclosed herein comprising the binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction (including, e.g., a bispecific antibody that specifically binds to PD-L1 and VEGF) and the chemotherapy shows particularly encouraging anti-tumour activity even when the subject has a low PD-L1 expression score. This can distinguish it from treatments of the prior art for the same cancer in which only patient groups with higher PD-L1 expression scores could be effectively targeted by immunotherapy. The combination of the PD-L1 and VEGF binding agent (including, e.g., the bispecific antibody) of the invention with the chemotherapy is highly effective in cancer treatment, even in subjects having a low PD-L1 expression score as, e.g., determined by the PD-L1 scoring algorithm CPS of < 10 or ≥ 1. In addition, the afore-mentioned combination shows a good safety profile for cancer subjects.
The release in immunosuppression by blocking VEGF and PD-L1 and the consequential influx of immune cells is contemplated to lead to an increased expression in PD-L1 on tumour cells, further enriching for the binding agent (including, e.g., the bispecific antibody) of the invention in the tumour microenvironment. Hence, the binding agent (including, e.g., the bispecific antibody) is contemplated to transform PD-L1 low-expressing tumours into more inflamed, immune cell infiltrated “hot tumours” .
The combination therapy comprising the binding agent (including, e.g., the bispecific antibody) and chemotherapy can be used in a method of treating cancer in a subject as disclosed herein. Features described herein in more detail in connection with the “binding agent /bispecific antibody for use in a method of treating” embodiments equally apply to the corresponding method of treatment embodiments.
The binding agent (including, e.g., the bispecific antibody) and chemotherapy disclosed herein can provide for substantial improvement in a subject’s overall survival (OS) , progression-free survival (PFS) , objective response rate (ORR) , duration of response, and/or disease control rate. As regards PFS, the binding agent (including, e.g, the bispecific antibody) and chemotherapy disclosed herein can provide for a PFS of at least about 9 months, preferably of at least about 13 months in patients suffering from TNBC (see also example 3 herein) .
Objective Response Rate (ORR) refers to the number (%) of subjects with at least one visit response of Complete Response (CR) or Partial Response (PR) per RECIST 1.1. Duration of Response (DoR) refers to the time from the date of first documented response until the first date of documented progression or death in the absence of disease progression (i.e., date of PFS event or censoring –date of first response + 1) . Disease Control Rate (DCR) refers to the rate of best objective response of CR, PR, or stable disease (SD) according to RECIST 1.1.
Determination of the PD-L1 expression
The PD-L1 expression score as described herein is determined by histologically analyzing a tumour sample and applying a scoring algorithm, the scoring algorithm according to the invention is the combined positive score (CPS) or any other scoring algorithm defining a similar PD-L1 expression score as the CPS. CPS is a well-established scoring algorithm which is approved for the treatment of cancer using pembrolizumab in multiple indications. In some embodiments, the treatment method comprises the step of applying the scoring algorithm to determine the PD-L1 expression score and thereby whether the subject shall be treated with the combination therapy of the invention. In other embodiments, the PD-L1 expression score (e.g., CPS or similar score type) is already known and provided before the start of the method of the invention.
The PD-L1 expression score can be determined in form of the CPS in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

wherein preferably the PD-L1 staining cells are tumour cells with partial or complete linear membrane 
staining that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining. The lymphocytes and macrophages are preferably directly associated with the response against the cancer. For example, the PD-L1 staining cells are tumour cells with convincing partial or complete linear membrane staining (at any intensity) that is perceived distinct from cytoplasmic staining, and lymphocytes and macrophages within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining (at any intensity) . The PD-L1 staining cells are preferably viable PD-L1 staining cells.
Categorization using CPS scoring algorithm is for example described in Kulangara, Karina & Hanks, Debra & Waldroup, Stephanie & Peltz, Lindsay & Shah, Supriya & Roach, Charlotte & Juco, Jonathan & Emancipator, Kenneth & Stanforth, Dave. (2017) , Development of the combined positive score (CPS) for the evaluation of PD-L1 in solid tumours with the immunohistochemistry assay PD-L1 IHC 22C3 pharmDx. Journal of Clinical Oncology, which disclosure is incorporated herein in its entirety.
In some preferred embodiments, the number of viable tumour cells in the tumour tissue sample are determining by flow cytometry. For example, a tissue sample analyzed by flow cytometry can be contacted with a viability dye prior to analysis, e.g., propidium iodide. Any convenient viability stain may be employed, with many examples known in the art. In some embodiments, the tumour proportion score (TPS) (an integrating scoring algorithm) is used instead of the CPS. The TPS can be obtained by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 (PD-L1 staining tumour cells) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:
Categorization using TPS scoring algorithm is described in Roach C, Zhang N, Corigliano E, et al. Development of a Companion Diagnostic PD-L1 Immunohistochemistry Assay for Pembrolizumab Therapy in Non-Small-cell Lung Cancer. Appl Immunohistochem Mol Morphol. 2016; 24 (6) : 392-397 (doi: 10.1097/PAI. 0000000000000408) , which disclosure is incorporated herein in its entirety.
For example, the number of total tumour cells and tumour cells positive for PD-L1 (i.e., PD-L1 staining tumour cells) can be assessed using the PD-L1 IHC 22C3 pharmDx assay (Agilent Technologies, Carpinteria, CA, USA) according to the manufacturer’s Instructions for Use. In some exemplary embodiments, the number of tumour cells can be measured in formalin-fixed tumour samples obtained by core-needle or excisional biopsy of a tumour lesion or from tissue resected at or after the time the cancer was diagnosed. In some such embodiments, for determination of PD-L1 expression, positivity is defined as complete circumferential or partial cell membrane staining of viable tumour cells with 1+ to 3+ intensity. Nonspecific staining can be recorded on a 0 to 3 intensity scale, in 0.25 grade increments. Tumour-associated immune cells are preferably excluded from PD-L1 scoring. Cytoplasmic staining, if present, is preferably excluded from the scoring. Scoring can be recorded as percentage of PD-L1-positive tumour cells over total tumour cells in the denominator (TPS) .
In some embodiments, the TAP scoring algorithm is used as the integrating scoring algorithm instead of CPS. The TAP score can be obtained by determining in a test sample of the subject the number of PD-L1 positive tumour and immune cells (PD-L1 staining tumour and immune cells) and the tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:
The TAP score can be determined as described in Liu, Chunyan et al. “Tumour Area Positivity (TAP) score of programmed death-ligand 1 (PD-L1) : a novel visual estimation method for combined tumour cell and immune cell scoring. ” Diagnostic pathology vol. 18, 1 48.19 Apr. 2023, doi: 10.1186/s13000-023-01318-8, which disclosure is incorporated by reference herein in its entirety.
In some embodiments, the tumour area is determined using the hematoxylin and eosin (H&E) stain. In some embodiments, the PD-L1 positive tumour cells and immune cells are PD-L1 staining cells with partial or complete linear membrane staining that is distinct from cytoplasmic staining, and immune cells (lymphocytes and macrophages) within the tumour nests and/or adjacent supporting stroma with membrane and/or cytoplasmic staining.
For example, to determine the TAP score, a hematoxylin and eosin stained slide can be first examined to identify the tumour area (area occupied by all viable tumour cells (TC) and the tumour-associated stroma containing tumour-associated immune cells (IC) ) . The immune cells measured in the nominator are tumour-associated stroma containing tumour-associated immune cells. If tumour nests are separated by non-neoplastic tissue, they can be included as part of the tumour area as long as the tumour nests are bordered on both sides of a 10x field; the intervening non-neoplastic tissue can also be included in the tumour area (commonly abbreviated as 10x field rule) . Necrosis, crush, and cautery artifacts are preferably excluded from tumour area. Tumour-associated IC are preferably intra-and peri-tumoural, including those present within the tumour proper, between tumour nests, and within any tumour-associated reactive stroma. In lymph nodes with focal or discrete tumour metastases, only IC immediately adjacent to the leading edge of the metastatic tumour nest are preferably defined as tumour-associated IC.
The TAP can be determined on the IHC slide by visually aggregating/estimating the area covered by PD-L1 positive TC (PD-L1 staining TC) and tumour-associated IC relative to the total tumour area. Both circumferential and partial/lateral membrane staining of TC at any intensity can be regarded as positive PD-L1 staining, while cytoplasmic staining of TC can be disregarded; membranous, cytoplasmic, and punctate staining of tumour-associated IC at any intensity can be regarded as PD-L1 positive staining.
Intra-luminal macrophage staining is preferably not included in the TAP score unless the macrophages completely fill the luminal space and are in direct contact with the TC. Staining of multi-nucleated giant cells, granulomas, and IC located within blood vessels and lymphatics are preferably not included in the TAP. Off-target staining (e.g., fibroblasts, endothelial cells, neuroendocrine cells, smooth muscle, and nerves) is preferably not included in the TAP.
In some embodiments, the cancer tissue sample is a tissue section of a tumour biopsy, preferably a solid tumour biopsy. Biopsies of interest can include tumour and/or non-neoplastic biopsies of skin (melanomas, carcinomas, etc. ) , soft tissue, bone, breast, colon, liver, kidney, adrenal, gastrointestinal, pancreatic, gall bladder, salivary gland, cervical, ovary, uterus, testis, prostate, lung, thymus, thyroid, parathyroid, pituitary (adenomas, etc. ) , brain, spinal cord, ocular, nerve, and skeletal muscle, etc. In some embodiments, the subject from which the biopsy is obtained has a malignancy is selected from the list consisting of gastric cancer, esophageal carcinomas, head and neck cancer (e.g., head and neck squamous cell carcinoma, or HNSCC) , renal cell carcinoma, urothelial/bladder carcinoma, colorectal cancer, ovarian carcinoma, myeloma, melanoma, lung cancer, classical Hodgkin lymphoma, and breast cancer (e.g., triple-negative breast cancer, hormone receptor positive (ER and/or PR) and Her2 positive breast cancer) , small cell lung cancer including extensive stage small cell lung cancer and limited stage small cell lung cancer, salivary gland carcinoma, vulvar carcinoma, thyroid carcinoma, anal canal carcinoma, biliary carcinoma, mesothelioma, cervical carcinoma, and neuroendocrine carcinoma. In some embodiments, the subject from which the biopsy is obtained has a malignancy selected from melanoma, liver, stomach, renal cell, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck cancer. In some preferred embodiments, the biopsy of interest can include a tumour and optionally non-neoplastic biopsy of any of a melanoma, liver, stomach, renal cell, colorectal, ovarian, colon, breast, esophagus, mesothelioma, and head and neck solid tumour. In some preferred embodiments, the biopsy is obtained from a subject having urothelial, breast, or esophageal cancer, most preferably breast cancer such as TNBC.
Detecting PD-L1 positive cells in a tumour sample is meanwhile clinical routine and required as companion diagnostic for several approved cancer treatments. It can be done in any convenient manner. In certain embodiments, the CPS is calculated from a stained tumour tissue biopsy section (e.g., on a slide) or serial tumour tissue biopsy sections by immunohistochemistry (IHC) staining, in-situ hybridization (ISH; e.g., fluorescence-in-situ-hybridization, or FISH) , histological stain, and a combination thereof. In certain embodiments, a tumour tissue biopsy section is analyzed by IHC. In certain embodiments, the percentage of viable PD-L1 positive and negative tumour cells and PD-L1 positive mononuclear inflammatory cells (MIC) , i.e., lymphocytes and macrophages, is determined within the tumour nests and the adjacent supporting stroma. In such embodiments, cells are positive for PD-L1 staining if they display partial or complete membrane staining relative to all viable tumour cells present in the sample. In some preferred embodiments, PD-L1 is detected by immunohistochemistry (IHC) staining. In some preferred embodiments, the number of viable tumour cells can be determining by flow cytometry. For example, a tissue sample analyzed by flow cytometry can be contacted with a viability dye prior to analysis, e.g., propidium iodide or any convenient viability stain known in the art.
Interpretation of stained slides can preferably be performed using a light microscope with an objective of 20x magnification. All viable tumour cells on the entire slide are preferably evaluated and included in the CPS or TPS PD-L1 scoring assessment optionally together with tumour-associated PD-L1 positive lymphocytes and macrophages for the CPS. A minimum of 100 viable tumour cells are preferably present in the PD-L1 stained slide to be considered adequate for evaluation. For slides comprising less than 100 viable tumour cells, tissue from a deeper level of the block or potentially another block, could present sufficient tumour cells for PD-L1 evaluation.
In some embodiments, the tumour tissue section is formalin fixed and embedded in paraffin wax (FFPE) . In alternative embodiments, the tissue section has been fixed in a different way, including tissue sections that have been fixed in, e.g., acrolein, glyoxal, osmium tetroxide, carbodiimide, mercuric chloride, zinc salts, picric acid, potassium di chromate, ethanol, methanol, acetone, and/or acetic acid.
In other embodiments, the PD-L1 scoring algorithm (such as CPS, TPS, or TAP) is calculated from a tumour tissue sample that is not a fixed section on a slide. For example, in certain embodiments, the CPS is calculated using flow cytometric analysis of a cell suspension from the tumour tissue sample. In these embodiments, the tumour tissue cell suspension can be stained with a detectable PD-L1 binding agent (e.g., a fluorescently labeled antibody) and analyzed on a flow cytometer for counting the number of tumour cells and MIC cells (i.e., lymphocytes and macrophages) expressing PD-L1. Tumour cells and MIC cells in the sample can be distinguished using any convenient flow cytometric parameter, e.g., forward scatter (FS) , side scatter (SS) , or by the expression of one or more additional markers using corresponding detectable binding agents for the one or more additional markers, e.g., markers specific or MIC or tumour cells. In other embodiments, the cells in the tumour tissue sample can be analyzed on a cell-by-cell basis for mRNA expression of PD-L1 and any other desired target, e.g., using single-cell nucleic acid sequencing methods for gene expression profiling (e.g., next generation sequencing methods) .
In some embodiments, the tissue section is stained. In some embodiments, the stain comprises a hematoxylin and eosin (H&E) stain. Hematoxylin, a basic dye, stains nuclei blue due to an affinity to nucleic acids in the cell nucleus; eosin, an acidic dye, stains the cytoplasm pink. A hematoxylin and eosin (H&E) stained section can be used for the evaluation of an acceptable tumour tissue sample or of a tumour area.
There are many other staining techniques known to those of skill in the art that can be used to selectively stain cells and cellular components that find use in the present disclosure, and as such no limitation in this regard is intended. The staining of a target (e.g., PD-L1) in cells from a tumour tissue biopsy is generally done by contacting the cells with one or more detectable target-specific binding agents under suitable conditions to allow for binding of the target-specific binding agent to its desired target (while minimizing nontarget binding) . The term “target-specific binding agent” means any agent that specifically binds to a target or analyte of interest, e.g., a target of interest that is present in a tissue section as described herein (e.g., a polypeptide or polynucleotide) . In some embodiments, the target-specific binding agent is an antibody (or target-binding fragments thereof) , e.g., as used in IHC and flow cytometry.
Staining may be performed with primary and secondary antibodies or without using secondary antibodies (e.g., where the primary antibody is detectably labeled) . Non-limiting examples of anti-PD-L1 antibodies include, but are not limited to, clone 22C3 (Merck & Co. ) , clone 28-8 (Bristol-Myers Squibb) , clones SP263 (Roche) , SP142 (Spring Biosciences) , and clone E1L3N (Cell Signaling Technology) . A preferred anti-PD-L1 antibody is clone SP263 (Roche) , another preferred anti-PD-L1 antibody is clone E1L3N (Cell Signaling Technology) .
Clone 22C3 is a well-known, commercially available anti-PD-L1 antibody described e.g., in US 9,709,568 B2, which disclosure is incorporated by reference in its entirety.
Clone 22C3 comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 23, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 24, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 25; and the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 20, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 21, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 22. The heavy and light chain variable regions are set forth in SEQ ID NO: 27 and 26.
Clone 28-8 is a well-known commercially available anti-PD-L1 antibody described e.g., in US 9,212,224 B2, which disclosure is incorporated by reference in its entirety.
Clone 28-8 comprises a heavy and light chain variable region (i.e., VH and VL) set forth in SEQ ID NO: 28 and 29.
SP263 is a well-known commercially available anti-PD-L1 antibody described e.g., in US 2010/0343556 A1.
Clone SP263 comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 33, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 34, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 35; and the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 30, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 31, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 32. The heavy and light chain variable region are set forth in SEQ ID NO: 36 and 37.
In certain other embodiments, a target specific binding agent is a nucleic acid or nucleic acid binding agent, e.g., as employed in in situ hybridization (ISH) reactions. For example, the target binding reagent can be a DNA, RNA, DNA/RNA hybrid molecule, peptide nucleic acid (PNA) , and the like. No limitation in the metes and bounds of a target-specific binding agent that finds use in the subject disclosure is intended.
The target-specific binding agent (or any secondary reagent used to detect the target-specific binding agent) may be attached to any suitable detectable label (or chromogen) or enzyme capable of producing a detectable label. Thus, in certain embodiments, the first or second label is produced by an enzymatic reaction, e.g., by the activity of horseradish peroxidase, alkaline phosphatase, and the like. Any convenient enzymatic label/chromogen deposition system can be employed (e.g., as used in standard IHC methods) , and as such, no limitation in this regard is intended. In some embodiments, the detectable label is a fluorescent tag.
In some embodiments, the staining reagents used may include a target-specific antibody (e.g., a PD-L1 specific antibody) . Where an additional target is to be detected, the staining reagents used may include one or more additional antibodies that each bind to a different antigen. For example, a set of antibodies may include a first antibody that binds to a first antigen (e.g., PD-L1) , a second antibody that binds to a second antigen, optionally a third antibody that binds to a third antigen and, optionally a fourth antibody that binds to a fourth antigen and/or further antibodies that bind to further antigens. In some embodiments, the antibody/antibodies used are primary antibodies that are detected by use of a secondary antibody (or other reagent) . The staining steps thus may be done by incubating the cells of the tissue sample, e.g., a tissue section or cell suspension, with the primary antibody/antibodies and then, after the primary antibody has bound to the desired target in/on the cells, incubating the cells with the labeled secondary antibody/antibodies (e.g., as is done in standard IHC protocols) . In some embodiments, each of the primary antibodies for each different target is from a different species (e.g., goat, rabbit, mouse, camel, chicken, donkey, etc. ) and the corresponding secondary antibodies specific for each different primary antibody are distinguishably labeled from each other.
In some preferred embodiments, the viable tumour cells and the number of lymphocytes and macrophages are counted in the tumour nests and the adjacent supporting stroma of the tumour tissue sample.
Although the result of a CPS calculation can exceed 100, the maximum score is preferably defined as 100 and a minimum of 100 viable tumour cells are preferably required to calculate the score. See Park et al. Cancer Res. Treat., 2020; 52 (3) : 661-670 and Yamashita et al., Gastric Cancer, 2020, 23: 95-104.
When an integrating scoring algorithm different than the CPS is used in the context of the methods disclosed herein, an in vitro diagnostic device (IVD) bridging study can be performed to determine whether that scoring algorithm defines a similar PD-L1 expression score as the established CPS. How to conduct such bridging assays is well known in the field of companion diagnostics and described, e.g., in detail in the FDA authorized “Principles for Codevelopment of an In Vitro Companion Diagnostic Device with a Therapeutic Product” issued on July 15, 2016, which disclosure is incorporated by reference in its entirety.
As disclosed therein, a test for determining PD-L1 expression score other than the CPS companion diagnostic (e.g., determining PD-L1 expression score using the IHC assay PD-L1 IHC 22C3 pharmDx in accordance with the manufacturer’s guide) can be used, if it can be demonstrated that the other IVD companion diagnostic has performance characteristics that are very similar to those of the CPS companion diagnostic. This is generally demonstrated through a bridging study between the two tests, using the original clinical trial samples and a pre-specified statistical analysis plan, to show that results with the candidate IVD companion diagnostic are very similar to those of the CPS companion diagnostic. A bridging study can evaluate efficacy of the therapeutic product in subjects whose marker status is determined by the candidate IVD companion diagnostic by assessing both concordance and discordance between the two tests using the same specimens from subjects who were tested for trial eligibility. The analysis needs to consider any potential impact of missing samples not available for the concordance study. The ability of the candidate IVD companion diagnostic to predict the efficacy of the therapeutic product can be supported indirectly by high analytical concordance with the CPS companion diagnostic on a large number of representative samples, including samples from subjects excluded from the trial because they were marker-negative by the CPS companion diagnostic. The assessment of the clinical validity of the candidate IVD companion diagnostic can rely on extrapolating the clinical performance characteristics of the CPS companion diagnostic to the clinical performance characteristics of the candidate IVD companion diagnostic. The ideal bridging study is one in which all samples tested with the trial test are retested with the candidate IVD companion diagnostic and valid test results are obtained and used to assess comparative performance. A bridging study with specimens from an all-comers trial also allows an analysis of efficacy using the results of the candidate IVD companion diagnostic. Note, however, that care should preferably be taken in understanding the analytical performance of the IVD prior to the bridging study because adjustments to the IVD should not be made from results obtained with the clinical trial samples. Whether a clinical trial enrolls subjects irrespective of the test result or enrolls only the subset of subjects identified by the test result, both the test-negative and test-positive clinical trial samples should preferably be included in bridging studies to avoid bias due to prescreening.
It is, however, recognized (e.g., by the FDA) that there are many reasons why all the samples tested with the CPS companion diagnostic may not be available for retesting, including that samples are missing, not accessible, or insufficient in quantity to retest, and it may not be possible to retest all samples. If only a subset of samples is retested, it should preferably be ensured that the characteristics of the subset adequately reflect the characteristics that affect test performance (e.g., tumour size, histology, melanin content, necrotic tissue, resected tissue versus core needle biopsy) and that the characteristics of the subjects that may affect therapeutic product efficacy (e.g., patient demographics, stage of disease, stratification factors) are proportionally preserved in the retest sample set when compared to the samples in the original set. In addressing baseline imbalance between the retested and non-retested analysis sets, FDA recommends that any covariates are identified that can affect the test result and then check for baseline imbalance between the retested and non-retested analysis sets using the set of covariates identified. A re-analysis of the primary outcome data should preferably be made according to the final test results with the retest sample set in order to assure that any reclassification that occurs does not alter conclusions about the safety and efficacy of the therapeutic product in the selected population. When all samples are not retested, a second re-analysis can be conducted in which missing data for the final test are imputed. The nature of the re-analysis will be product-specific and may be discussed with the appropriate IVD review center. Finally, additional analytical validation can potentially be requested to support satisfactory concordance across methods where discordance may arise, e.g., precision, limit of detection, and accuracy. In the event there is discordance in a marker-positive-only trial, it is possible that the candidate IVD companion diagnostic will more accurately predict responders, a difference that would represent an advantage for optimal use of the therapeutic product.
For example, when using a PD-L1 expression score obtained from the TAP scoring algorithm, it can be demonstrated that the TAP has performance characteristics that are very similar to those of the CPS companion diagnostic using the described bridging study. The same holds true for a PD-L1 expression score determined using different anti-PD L1 antibodies, such as SP263 or 22C3 or E1L3N. For example, it can be demonstrated that a PD-L1 expression score obtained from using 22C3 and the CPS scoring algorithm has characteristics that are very similar to those of a PD-L1 expression score obtain from using 28-8 and the CPS scoring algorithm using the described bridging study.
The binding agent of the invention
While the present disclosure might make reference to a bispecific antibody specifically binding to PD-L1 and VEGF, this bispecific antibody may comprise in a general aspect an anti-VEGF antibody (e.g., an anti-VEGF-A antibody) or fragment thereof, and an antagonist of the PD1/PD-L1 interaction and/or a PD-1 or PD-L1 blocking antibody. In some embodiments relating to this general aspect, the bispecific antibody comprises an anti-VEGF antibody (e.g., an anti-VEGF-A antibody) or fragment thereof, and a PD-1 or PD-L1 blocking antibody. In some preferred embodiments relating to this general aspect, the bispecific antibody comprises an anti-VEGF antibody (e.g., an anti-VEGF-A antibody) or fragment thereof, and an anti-PD-1 or anti-PD-L1 antibody or fragment thereof. For example, the anti-PD-1 antibody can be selected from any anti-PD-1 antibody known in the art such as pembrolizumab or nivolumab. An exemplary anti-PD-L1 antibody includes atezolizumab. For example, the anti-VEGF antibody can be selected from any VEGF antibody known in the art such as bevacizumab, ranibizumab or an VEGF binding region derived from the anti-VEGF fusion protein Aflibercept (see, e.g., US 7070959) . An exemplary anti-VEGF antibody includes bevacizumab.
The embodiments as set out herein for the bispecific antibody specifically binding to PD-L1 and VEGF as disclosed herein are generally also applicable for the antibody of the present general aspect, unless the context clearly indicates otherwise.
On the most general level, the present disclosure relates to a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction. This binding agent, which is alternatively referred to herein as “anti-PD-1/PD-L1xVEGF/VEGFR binding agent” or simply as “binding agent” will be discussed in more detail in the following sections.
First binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction
One component of the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction. Thus, this first binding region comprises an antagonist of the PD-1/PD-L1 interaction, such as an anti-PD-1 antibody, a PD-1 binding fragment thereof, an anti-PD-L1 antibody, or a PD-L1 binding fragment thereof.
A1. Anti-PD-1 antibodies or PD-1 binding fragments thereof
In some embodiments, the first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction comprises an anti-PD-1 antibody or a PD-1 binding fragment thereof.
In some embodiments, PD-1 is human PD-1, in particular human PD-1 comprising the sequence set forth in SEQ ID NO: 90, 91, or 92.
In some embodiments, the anti-PD-1 antibody or a PD-1 binding fragment thereof (anti-PD-1 antibody or fragment thereof) specifically binds to PD-1.
In some embodiments, the anti-PD-1 antibody or fragment thereof is selected from the group consisting of Pembrolizumab, Nivolumab, Cemiplimab, Dostarlimab, Toripalimab, Retifanlimab, and PD-1 binding fragments thereof.
In some embodiments, the anti-PD-1 antibody or fragment thereof is a monoclonal antibody or a fragment thereof.
In some embodiments, the anti-PD-1 antibody or fragment thereof is a humanized antibody. In some embodiments, the anti-PD-1 antibody or fragment thereof is a monoclonal humanized antibody. In some embodiments, the fragment of the anti-PD-1 antibody may be any antigen-binding fragment of the anti-PD-1 antibody, for example a Fab, a Fab', a F (ab') 2, a Fv, a scFv, a Fab'-SH, an sdAb, or a VHH. In some embodiments, the anti-PD-1 antibody or fragment thereof is selected from a full-length anti-PD-1 antibody, an scFv of an anti-PD-1 antibody or a VHH of an anti-PD-1 antibody. In some embodiments, the anti-PD-1 antibody or fragment thereof is an scFv of an anti-PD-1 antibody. In some embodiments, the anti-PD-1 antibody or fragment thereof is a VHH of an anti-PD-1 antibody.
In some embodiments, the anti-PD-1 antibody or fragment thereof may comprise a variable region that specifically binds to PD-1. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises a heavy chain variable region and/or a light chain variable domain. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises a heavy chain variable region and a light chain variable domain. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises a heavy chain and/or a light chain. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises a heavy chain and a light chain. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of a heavy chain and a light chain as a single chain (scFv) .
Exemplary anti-PD-1 antibody CDRs, variable region sequences, and heavy and light chain sequences are provided in Table 1, e.g., for anti-PD-1 antibodies A1 and A2, respectively.
In some embodiments, the anti-PD-1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-1 antibody A1 shown in Table 1.
In some embodiments, the CDRs are defined according to the IMGT numbering system. Thus, in some embodiments, (I) a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 45, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 46, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 47, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 48, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 49, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 50, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the CDRs are defined according to the Kabat numbering system. Thus, in some embodiments, (I) a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 51, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 52, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 53, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 54, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 55, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 50, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 56, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 56, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively, or (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55 and 50, respectively. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 56, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively, or (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55 and 50, respectively. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 56.
In some embodiments, the anti-PD-1 antibody or fragment thereof is an scFv comprising:
(i-1a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 45, 46, and 47, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 48, 49, and 50, respectively;
(i-1b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 51, 52, and 53, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 54, 55, and 50, respectively;
(i-1c) the amino acid sequence of SEQ ID NO: 56 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56;
(i-1d) an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 
96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56, and comprising the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively; or
(i-1e) an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 
96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56, and comprising the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55 and 50, , respectively.
In some alternative embodiments, the anti-PD-1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-1 antibody A2 shown in Table 1.
In some embodiments, the CDRs are defined according to the Kabat numbering system. Thus, in some embodiments, a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 57, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 58, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 59 or 60, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the CDRs are defined according to the IMGT numbering system. Thus, in some embodiments, a VH of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 61, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 62, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 63 or 64, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 65, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 65, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 65, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 65.
In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 66, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 66, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 66, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively. In some embodiments, the anti-PD-1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 66.
Thus, in some embodiments, the anti-PD-1 antibody or fragment thereof is a VHH comprising:
(i-2a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 57, 58, and 59, respectively, or a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively;
(i-2b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 61, 62, and 63, respectively, or a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively;
(i-2c) the amino acid sequence of SEQ ID NO: 65 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 65, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 65 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively; or
(i-2d) the amino acid sequence of SEQ ID NO: 66 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 66, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 66 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, or comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively.
A2. Anti-PD-L1 antibodies or PD-L1 binding fragments thereof
In some embodiments, the first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction comprises an anti-PD-L1 antibody or a PD-L1 binding fragment thereof.
In some embodiments, PD-L1 is human PD-L1, in particular human PD-L1 comprising the sequence set forth in SEQ ID NO: 93, 94, 95, or 96.
In some embodiments, the anti-PD-L1 antibody or a PD-L1 binding fragment thereof (anti-PD-L1 antibody or fragment thereof) specifically binds to PD-L1.
In some embodiments, the anti-PD-L1 antibody or fragment thereof is selected from the group consisting of Atezolizumab, Durvalumab, Avelumab, and PD-L1 binding fragments thereof.
In some embodiments, the anti-PD-L1 antibody or fragment thereof is a monoclonal antibody or a fragment thereof.
In some embodiments, the anti-PD-L1 antibody or fragment thereof is a humanized antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is a monoclonal humanized antibody. In some embodiments, the fragment of the anti-PD-L1 antibody may be any antigen-binding fragment of the anti-PD-L1 antibody, for example a Fab, a Fab', a F (ab') 2, a Fv, a scFv, a Fab'-SH, an sdAb, or a VHH. In some embodiments, the anti-PD-L1 antibody or fragment thereof is selected from a full-length anti-PD-L1 antibody, an scFv of an anti-PD-L1 antibody or a VHH of an anti-PD-L1 antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is a VHH of an anti-PD-L1 antibody.
In some embodiments, the anti-PD-L1 antibody or fragment thereof may comprise a variable region that specifically binds to PD-L1. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a heavy chain variable region and/or a light chain variable domain. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a heavy chain variable region and a light chain variable domain. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a heavy chain and/or a light chain. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a heavy chain and a light chain. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises of a heavy chain and a light chain as a single chain (scFv) . In some embodiments, the anti-PD-L1 antibody or fragment thereof is a VHH of an anti-PD-L1 antibody.
Exemplary anti-PD-L1 antibody CDRs, variable region sequences, and heavy and light chain sequences are provided in Table 1, e.g., for anti-PD-L1 antibodies A3 and A4, respectively.
In some embodiments, the anti-PD-L1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-L1 antibody A3 shown in Table 1.
In some embodiments, the CDRs are defined according to the Kabat numbering system. Thus, in some embodiments, a VH of the anti-PD-L1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 18, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 19, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 38, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the CDRs are defined according to the IMGT numbering system. Thus, in some embodiments, a VH of the anti-PD-L1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 1, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 2, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 3, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 9, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 9, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 9, wherein the amino acid sequence comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9.
Thus, in some embodiments, the anti-PD-L1 antibody or fragment thereof is a VHH comprising:
(i-3a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 18, 19, and 38, respectively;
(i-3b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 1, 2, and 3, respectively; or
(i-3c) the amino acid sequence of SEQ ID NO: 9 or an amino acid sequence having at least 80%, at least 
85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the amino acid sequence SEQ ID NO: 9, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 9 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively.
In some alternative embodiments, the anti-PD-L1 antibody or fragment thereof is defined by one or more sequences of the anti-PD-L1 antibody A4 shown in Table 1.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises (a) a heavy chain variable region (VH) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 73, and/or a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 74.
In some embodiments, (I) a VH of the anti-PD-L1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 67, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 68, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 69, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-PD-1 antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 70, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 71, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 72, or a variant thereof having up to three amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 73.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 74.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprisesa (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 74, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively. In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises (I) a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 73, and (II) comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 74.
Thus, in some embodiments, the anti-PD-L1 antibody or fragment thereof is a fragment of a humanized anti-PD-L1 antibody comprising:
(i-4a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences of 
SEQ ID NO: 73, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences of SEQ ID NO: 74;
(i-4b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 67, 68, and 69, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively;
(i-4c) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74; or
(i-4d) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 73, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 74, wherein the VH comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the VL comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively.
Second binding region which binds to VEGF, VEGFR, or both and which antagonizes the VEGF/VEGFR 
interaction
Another component of the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a second binding region which binds to VEGF, VEGFR, or both and which antagonizes the VEGF/VEGFR interaction. Thus, this second binding region comprises an antagonist of the VEGF/VEGFR interaction, such as an anti-VEGF antibody, a VEGF binding fragment thereof, a protein comprising a VEGFR domain, an anti-VEGFR antibody, or a VEGF binding fragment thereof.
B1. Anti-VEGF antibodies or VEGF binding fragments thereof
In some embodiments, the second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction comprises an anti-VEGF antibody or a VEGF binding fragment thereof, preferably an anti-VEGF-A antibody or a VEGF-A binding fragment thereof.
In some embodiments, VEGF is human VEGF, in particular human VEGF comprising the sequence set forth in SEQ ID NO: 97 or human VEGF-A comprising the sequence set forth in SEQ ID NO: 98.
In some embodiments, the anti-VEGF antibody or a VEGF binding fragment thereof (anti-VEGF antibody or fragment thereof) specifically binds to VEGF, in particular to VEGF-A.
In some embodiments, the anti-VEGF antibody or fragment thereof is selected from the group consisting of Bevacizumab, Ramucirumab, Ranibizumab, and VEGF binding fragments thereof.
In some embodiments, the anti-VEGF antibody or fragment thereof is a monoclonal antibody or a fragment thereof.
In some embodiments, the anti-VEGF antibody or fragment thereof is a humanized antibody. In some embodiments, the anti-VEGF antibody or fragment thereof is a monoclonal humanized antibody. In some embodiments, the fragment of the anti-VEGF antibody may be any antigen-binding fragment of the anti-VEGF antibody, for example a Fab, a Fab', a F (ab') 2, a Fv, a scFv, a Fab'-SH, an sdAb, or a VHH. In some embodiments, the anti-VEGF antibody or fragment thereof is selected from a full-length anti-VEGF antibody, an scFv of an anti-VEGF antibody, or a VHH of an anti-VEGF antibody. In some embodiments, the anti-VEGF antibody or fragment thereof is a full-length anti-VEGF antibody.
In some embodiments, the anti-VEGF antibody or fragment thereof may comprise a variable region that specifically binds to VEGF, in particular VEGF-A. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a heavy chain variable region and/or a light chain variable domain. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a heavy chain variable region and a light chain variable domain. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a heavy chain and/or a light chain. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a heavy chain and a light chain. In some embodiments, the anti-VEGF antibody or fragment thereof comprises two heavy chains and two light chains.
Exemplary anti-VEGF antibody CDRs, variable region sequences, and heavy and light chain sequences are provided in Table 1; cf., e.g., SEQ ID NOs: 39, 40, 41, 42, 43, 8, 4, 5, 6, 7, 44, 8, 10, 11, 12, 13, 15, 80, and 17.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a constant region, preferably derived from a human antibody, preferably the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises (a) a heavy chain variable region (VH) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 10, and/or a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences of SEQ ID NO: 11.
In some embodiments, the CDRs are defined according to the Kabat numbering system. Thus, in some embodiments, (I) a VH of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 39, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 40, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 41, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 42, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 43, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 8, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the CDRs are defined according to the IMGT numbering system. Thus, in some embodiments, (I) a VH of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 4, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 5, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 6, or a variant thereof having up to three amino acid substitutions, additions or deletions, and/or (II) a VL of the anti-VEGF antibody or fragment thereof comprises: (i) a CDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 7, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a CDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 44, or a variant thereof having up to one amino acid substitution, addition or deletion; and (iii) a CDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 8, or a variant thereof having up to three amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system. In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, wherein the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 10.
In some embodiment, the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11, wherein the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 11.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein either (i) the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or (ii) the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and (II) a VL comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 11, wherein either (i) the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or (ii) the VH comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the VL comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) a VH comprising or consisting of the amino acid sequence of SEQ ID NO: 10, and (II) comprises a VL comprising or consisting of the amino acid sequence of SEQ ID NO: 11.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a CH1 domain, preferably comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 12.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises an Fc region comprising one or more mutations which reduce or abolish immune effect functions. Suitable mutations are known in the art and may include one or more amino acid substitutions (cf., e.g., Wilkinson, I., et al., 2021. FcPLoS One, 16 (12) , p. e0260954; and Liu, R., et al., 2020. Antibodies, 9 (4) , p. 64) . In some embodiments, the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising a L234A and/or a L235A mutation. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising L234A and L235A mutations. Amino acids are numbered according to the EU numbering (EU-index) as set forth in Kabat (Kabat, E.A.; National Institutes of Health (U.S. ) Office of the Director. Sequences of Proteins of Immunological Interest, 5th ed.; DIANE Publishing: Collingdale, PA, USA, 1991) . For example, these may correspond to L240A and L241A in SEQ ID NO: 16. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a Fc domain comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 13.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a CL domain, preferably comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 15.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, wherein the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, or the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an HC comprising or consisting of the amino acid sequence of SEQ ID NO: 80.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 44, respectively, or the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises an LC comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and (II) an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, wherein either (i) the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or (ii) the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and (II) an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and (II) an LC comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein either (i) the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively, or (ii) the HC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the LC comprises the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some embodiments, the anti-VEGF antibody or fragment thereof comprises (I) an HC comprising or consisting of the amino acid sequence of SEQ ID NO: 80, and (II) an LC comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the anti-VEGF antibody comprises or consists of two HCs each comprising the amino acid sequence of SEQ ID NO: 80; and two LCs each comprising the amino acid sequence of SEQ ID NO: 17.
Thus, in some embodiments, the anti-VEGF antibody or fragment thereof is a full-length humanized anti-VEGF antibody comprising:
(ii-1a) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences of 
SEQ ID NO: 10, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences of SEQ ID NO: 11;
(ii-1b) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 39, 40, and 41, respectively, and a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively;
(ii-1c) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set 
forth in: SEQ ID NO: 4, 5, and 6, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively;
(ii-1d) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11;
(ii-1e) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11, wherein the VH comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the VL comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively;
(ii-1f) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11, wherein the VH comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the VL comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively;
(ii-1g) a heavy chain (HC) comprising an amino acid sequence having at least 80%, at least 85%, at least 
90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80 and a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17;
(ii-1h) a heavy chain (HC) comprising an amino acid sequence having at least 80%, at least 85%, at least 
90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80 and a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively; or
(ii-1i) a heavy chain (HC) comprising an amino acid sequence having at least 80%, at least 85%, at least 
90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80 and a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
B2. VEGFR domains
In some embodiments, the second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction comprises a VEGFR domain. In some embodiments, a protein which comprises a VEGFR domain is Aflibercept. Exemplary sequences of VEGFR domains are provided in Table 1.
In some embodiments, the second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction comprises at least one VEGFR-1 domain, or at least one VEGFR-2 domain, or at least one VEGFR-3 domain, or at least one VEGFR-1 domain and at least one VEGFR-2 domain, such as two VEGFR-1 domains, or two VEGFR-2 domains, or two VEGFR-1 domains and two VEGFR-2 domains, or two VEGFR-3 domains.
In some embodiments, the VEGFR domain is VEGFR-1 comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 75, or is an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75.
In some embodiments, the VEGFR domain is VEGFR-2 comprising or consisting of the amino acid sequence set forth in: SEQ ID NO: 76, or is an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76.
In some embodiments, the VEGFR domain is VEGFR-3 comprising or consisting of the amino acid sequence set forth in SEQ ID NO: 77, or is an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77.
In some embodiments, the second binding region comprises at least one VEGFR-1 domain comprising or consisting of at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75.
In some embodiments, the second binding region comprises at least one VEGFR-2 domain comprising or consisting of at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76.
In some embodiments, the second binding region comprises at least one VEGFR-3 domain comprising or consisting of at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77.
In some preferred embodiments, the second binding region comprises (i) at least one VEGFR-1 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75, or at least one VEGFR-3 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76, and (ii) at least one VEGFR-2 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77. In particularly preferred embodiments, the second binding region comprises (i) two VEGFR-1 domains or two amino acid sequences each having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75, (ii) two VEGFR-2 domains or two amino acid sequences having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76, (iii) two VEGFR-3 domains or two amino acid sequences each having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77, (iv) a combination of (i) and (ii) , or (v) a combination of (ii) and (iii) .
In some preferred embodiments, the second binding region comprises or consists of the amino acid sequence of SEQ ID NO: 78 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 78.
In some embodiments, the second binding region comprises or consists of (i) at least one VEGFR-1 or VEGFR-3 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75 or 77; (ii) at least one VEGFR-2 domain or at least one amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 76; and (iii) an Fc part. For example, the second binding region may comprise or consist of the amino acid sequence of SEQ ID NO: 79 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 79.
In some embodiments, the second binding region comprises:
(ii-2a) the amino acid sequence of SEQ ID NO: 75 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 75;
(ii-2b) the amino acid sequence of SEQ ID NO: 76 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SE Q ID NO: 76;
(ii-2c) the amino acid sequence of SEQ ID NO: 77 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 77;
(ii-2d) the amino acid sequence of SEQ ID NO: 78 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 78; or
(ii-2e) the amino acid sequence of SEQ ID NO: 79 or an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 79.
In some embodiments, the second binding region comprises two amino acid sequences each comprising or consisting of the amino acid sequence of SEQ ID NO: 79. The resulting second binding region may have the setup of an antibody when it comes to the Fc parts, i.e., the two Fc parts form an Fc domain via at least one disulfide bond, and the two N-terminal regions each comprise or consist of two VEGFR domains. Accordingly, the resulting second binding region may have an Fc domain and two VEGFR-containing arms, wherein each arm comprises two VEGFR domains such that the resulting second binding region overall comprises four VEGFR domains.
Specific embodiments of the anti-PD-1/PD-L1xVEGF/VEGFR binding agent
In some embodiments, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises a multispecific antibody, preferably a bispecific antibody.
The anti-PD-1/PD-L1xVEGF/VEGFR binding agent can comprise any of the first and second binding regions as outlined above, in any combination. Thus, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may comprise (i) a PD-1 binding antibody or PD-1 binding fragment thereof, such as Pembrolizumab, Nivolumab, Cemiplimab, Dostarlimab, Toripalimab, and Retifanlimab, or a fragment of any of the afore-mentioned antibodies, or any of the PD-1 binding sequences as disclosed above; and (ii) a VEGF binding antibody or VEGFR-binding fragment thereof, such as Bevacizumab, Ramucirumab, and Ranibizumab, or a fragment of any of the afore-mentioned antibodies, or any of the VEGF-binding sequences as disclosed above; or a VEGFR domain or a protein comprising the same, such as Aflibercept, or any of the VEGFR sequences as disclosed above. Alternatively, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may comprise (i) a PD-L1 binding antibody or PD-L1 binding fragment thereof, such as Atezolizumab, Durvalumab, and Avelumab, or a fragment of any of the afore-mentioned antibodies, or any of the PD-L1 binding sequences as disclosed above; and (ii) a VEGF binding antibody or VEGFR-binding fragment thereof, such as Bevacizumab, Ramucirumab, and Ranibizumab, or a fragment of any of the afore-mentioned antibodies, or any of the VEGF-binding sequences as disclosed above; or a VEGFR domain or a protein comprising the same, such as Aflibercept, or any of the VEGFR sequences as disclosed above.
In a particularly preferred embodiment, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises (i) the PD-L1 binding sequences, such as the VHH sequences and the anti-PD-L1 HCDRs thereof as disclosed above; and (ii) a VEGF binding antibody or VEGFR-binding fragment thereof as disclosed above, such as Bevacizumab.
Exemplary bispecific anti-PD-1/PD-L1xVEGF/VEGFR binding agents include the following: the anti-PD-1/PD-L1xVEGF/VEGFR binding agents C1, C2a, C2b, C2c, C2d, C3a, and C3b provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , SYN-2510 (ImmuneOnco/Instil Bio) , AI-081 (OncoC4) , HB0025 (Huabo Biopharm) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , CVL006 (Convalife) , DR30206 (Zhejiang Doer Bio) , HC010 (HC Biopharma) , and MHB039A (Minghui Pharma) . In one embodiment, the bispecific antibody is selected from the group consisting of Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , CVL006 (Convalife) , DR30206 (Zhejiang Doer Bio) , HC010 (HC Biopharma) , and MHB039A (Minghui Pharma) . Preferred anti-PD-1/PD-L1xVEGF/VEGFR binding agents are the anti-PD-1/PD-L1xVEGF/VEGFR binding agents C1, C2a, C2b, C2c, C2d, C3a, and C3b, in particular, anti-PD-1/PD-L1xVEGF/VEGFR binding agent C1.
In some embodiments, the antibody comprises any combination of one or more full-length antibodies, such as a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, or a polyclonal antibody, and/or one or more antibody fragments, such as a Fab, a Fab', a F (ab') 2, an Fv, an scFv, a Fab'-SH, an sdAb, or a VHH. In some embodiments, the antibody comprises a full-length antibody and an antibody fragment, such as a Fab, a Fab', a F (ab') 2, an Fv, an scFv, a Fab'-SH, an sdAb, or a VHH.
In some embodiments, the first binding region comprises an anti-PD-1 antibody or fragment thereof, or comprises an antibody fragment selected from an Fab, an Fab', an F (ab') 2, an Fv, an scFv, an Fab'-SH, an sdAb, and a VHH, preferably an scFv or a VHH. In some embodiments, the anti-PD-1 antibody or fragment thereof is an anti-PD-1 single-chain variable fragment. In some embodiments, the anti-PD-1 antibody or fragment thereof is an anti-PD-1 VHH.
In some embodiments, the first binding region comprises an anti-PD-L1 antibody or fragment thereof, or comprises an antibody fragment selected from an Fab, an Fab', an F (ab') 2, an Fv, an scFv, an Fab'-SH, an sdAb, and a VHH, preferably a sdAb, most preferably a VHH. In some embodiments, the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 single domain antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 VHH.
In some embodiments, the second binding region comprises an anti-VEGF antibody or fragment thereof, or comprises a human antibody, a humanized antibody, or a chimeric antibody, preferably a humanized antibody. In some embodiments, the anti-VEGF antibody or fragment thereof is a monoclonal antibody, for example a monoclonal human antibody, a monoclonal humanized antibody, or a monoclonal chimeric antibody. In some embodiments, the anti-VEGF antibody or fragment thereof is a monoclonal humanized antibody.
In some embodiments, the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises an anti-VEGF antibody (e.g., anti-VEGF-A antibody) and/or an anti-PD-L1 single domain antibody. In some embodiments, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent comprises an anti-VEGF antibody (e.g., anti-VEGF-A antibody) and/or an anti-PD-1 single-chain variable fragment.
In some embodiments, the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has monovalent binding to VEGF and/or to PD-L1. In some embodiments, the bispecific antibody has monovalent binding to VEGF and/or to PD-1. In some embodiment the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and/or to PD-L1. In some embodiment the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and/or to PD-1. In some preferred embodiments, the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and to PD-L1. In some preferred embodiments, the multispecific (preferably bispecific) anti-PD-1/PD-L1xVEGF/VEGFR binding agent has bivalent binding to VEGF and to PD-1.
In some embodiments, the anti-VEGF antibody (e.g., anti-VEGF-A antibody) or fragment thereof is a monovalent or bivalent antibody or fragment thereof. In some embodiments, the anti-PD-L1 antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof. In some embodiments, the anti-PD-1 antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof. In some preferred embodiments, the anti-VEGF antibody (e.g., anti-VEGF-A antibody) or fragment thereof is a bivalent anti-VEGF antibody or fragment thereof and/or the anti-PD-L1 antibody or fragment thereof is a bivalent antibody or fragment thereof. In some preferred embodiments, the anti-VEGF antibody (e.g., anti-VEGF-A antibody) or fragment thereof is a bivalent anti-VEGF antibody or fragment thereof and/or the anti-PD-1 antibody or fragment thereof is a bivalent antibody or fragment thereof. In some preferred embodiments, the anti-VEGF antibody (e.g., anti-VEGF-A antibody) is a bivalent anti-VEGF antibody and the anti-PD-L1 is a monovalent single domain antibody. In some preferred embodiments, the anti-VEGF antibody (e.g., anti-VEGF-A antibody) is a bivalent anti-VEGF antibody and the anti-PD-1 is a bivalent single-chain variable fragment.
C1. Anti-PD-L1xVEGF antibodies
In some preferred embodiments, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a bispecific anti-PD-L1xVEGF antibody, i.e., a bispecific antibody, wherein the first binding region of the bispecific antibody binds to PD-L1 and the second binding region binds to VEGF, preferably to VEGF-A. An exemplary and preferred bispecific anti-PD-L1xVEGF antibody is the anti-PD-1/PD-L1xVEGF/VEGFR binding agent C1 provided in Table 1.
Exemplary bispecific anti-PD-L1xVEGF/VEGFR binding agents include the following: the anti-PD-L1xVEGF/VEGFR binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , DR30206 (Zhejiang Doer Bio) . An exemplary trispecific anti-PD-L1xVEGF/VEGFR binding agent is HC010 (HC Biopharma) , which further binds to TGFbeta.
In some embodiments, the anti-PD-L1xVEGF antibody comprises an anti-VEGF antibody as described herein and at least one anti-PD-L1 single domain antibody as described herein, preferably an anti-VEGF antibody as described herein and two anti-PD-L1 single domain antibodies as described herein, more preferably an anti-VEGF antibody as described herein and two anti-PD-L1 VHHs as described herein.
In some embodiments of the anti-PD-L1xVEGF antibody, the at least one anti-PD-L1 single domain antibody may be fused to the anti-VEGF antibody. In some embodiments, each anti-PD-L1 single domain antibody is terminally fused to the anti-VEGF antibody. In some preferred embodiments, each anti-PD-L1 single domain antibody is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain. In particularly preferred embodiments, each anti-PD-L1 single domain antibody is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain. The anti-PD-L1xVEGF antibody more preferably comprises an anti-VEGF antibody and two anti-PD-L1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-L1 VHH, more preferably at the C-terminus of the heavy chain of the anti-VEGF antibody. The anti-PD-L1 single domain antibody (in particular, VHH) can be fused to the anti-VEGF antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSG (SEQ ID NO: 14) .
In preferred embodiments, the anti-PD-L1xVEGF antibody comprises a linker, which more preferably comprises or consists of the amino acid sequence of SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
In particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises (i) an anti-VEGF antibody comprising two heavy chains as described herein; and (ii) two anti-PD-L1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-L1 VHH through a linker comprising the amino acid sequence of SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
In particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises two anti-PD-L1 VHHs as described herein, wherein each VHH is fused to the C-terminus of an anti-VEGF-A antibody as described herein, preferably the C-terminus of each heavy chain of the anti-VEGF-A antibody. The anti-PD-L1xVEGF antibody can therefore comprise an anti-VEGF-A antibody as described herein and two anti-PD-L1 VHHs as described herein, wherein each heavy chain of the anti-VEGF-A antibody is fused to an anti-PD-L1 VHH through a linker, such as a linker comprising the amino acid sequence shown in SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 14.
In particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises (i) an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7, 44 and 8 (IMGT) ) or variants thereof having up to three amino acid substitutions, additions or deletions; and (ii) an anti-PD-L1 single domain comprising the HCDRs as described above (i.e., SEQ ID NOs: 18, 19, and 38 (Kabat) or SEQ ID NOs: 1, 2 and 3 (IMGT) ) , or variants thereof having up to three amino acid substitutions, additions or deletions. In some embodiments, the anti-PD-L1xVEGF antibody comprises (1) at least one anti-PD-L1 single domain antibody comprising or consisting of the amino acid sequence of SEQ ID NO: 9 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 9, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 9 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, or the HCDR1, HCDR2, and HCDR3 sequences set forth in: 1, 2, and 3, respectively; and/or (2) an anti-VEGF antibody comprising (i) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively; or (ii) a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and a VL comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-VEGF antibody comprises:
(ii-1d) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11;
(ii-1e) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11, wherein the VH comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the VL comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively;
(ii-1f) a heavy chain variable region (VH) comprising an amino acid sequence having at least 80%, at 
least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 10, and a light chain variable region (VL) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 11, wherein the VH comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the VL comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively;
(ii-1g) a heavy chain (HC) comprising an amino acid sequence having at least 80%, at least 85%, at least 
90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17;
(ii-1h) a heavy chain (HC) comprising an amino acid sequence having at least 80%, at least 85%, at least 
90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively; or
(ii-1i) a heavy chain (HC) comprising an amino acid sequence having at least 80%, at least 85%, at least 
90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 80, and a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
In particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 16, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 16, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 18, 19, and 38, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively. In some alternative embodiments, the anti-PD-L1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 16, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 1, 2, and 3, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
In some particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 16 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
In some particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 16; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17. In some particularly preferred embodiments, the anti-PD-L1xVEGF antibody comprises (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 16; and (ii) two light chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 17. Such an anti-PD-L1xVEGF antibody comprising two heavy chain-VHH fusions having the amino acid sequence of SEQ ID NO: 16 and two light chains having the amino acid sequence of SEQ ID NO: 17 is disclosed as compound "Ava-2GS-NSD" in WO 2022/042719.
C2. Anti-PD-1xVEGF antibodies
In some embodiments, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a bispecific anti-PD-1xVEGF antibody, i.e., a bispecific antibody, wherein the first binding region of the bispecific antibody binds to PD-1 and the second binding region binds to VEGF, preferably to VEGF-A.
Exemplary bispecific anti-PD-1xVEGF/VEGFR binding agents include the following: the anti-PD-1xVEGF/VEGFR binding agents C2a, C2b, C2c, C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) and MHB039A (Minghui Pharma) .
In some embodiments, the anti-PD-1xVEGF antibody comprises an anti-VEGF antibody as described herein and at least one anti-PD-1 scFv as described herein, preferably an anti-VEGF antibody as described herein and at least two anti-PD-1 scFvs as described herein, more preferably an anti-VEGF antibody as described herein and two anti-PD-1 scFvs as described herein.
In some embodiments of the anti-PD-1xVEGF antibody, the at least one anti-PD-1 scFv may be fused to the anti-VEGF antibody. In some embodiments, each anti-PD-1 scFv is terminally fused to the anti-VEGF antibody. In some preferred embodiments, each anti-PD-1 scFv is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain. In particularly preferred embodiments, each anti-PD-1 scFv is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain. The anti-PD-1xVEGF antibody more preferably comprises an anti-VEGF antibody and two anti-PD-1 scFvs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 scFv, more preferably at the C-terminus of the heavy chain of the anti-VEGF antibody. The anti-PD-1 scFv can be fused to the anti-VEGF antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 88) .
In preferred embodiments, the anti-PD-1xVEGF antibody comprises a linker, which more preferably comprises or consists of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody comprising two heavy chains as described herein; and (ii) two anti-PD-1 scFvs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 scFv through a linker, wherein the linker preferably comprises or consists of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises two anti-PD-1 scFvs as described herein, wherein each anti-PD-1 scFv is fused to the C-terminus of an anti-VEGF-Aantibody as described herein, preferably the C-terminus of each heavy chain of the anti-VEGF-A antibody. The anti-PD-1xVEGF antibody can therefore comprise an anti-VEGF-A antibody as described herein and two anti-PD-1 scFvs as described herein, wherein each heavy chain of the anti-VEGF-A antibody is fused to an anti-PD-1 scFv through a linker, such as a linker comprising or consisting of the amino acid sequence shown in SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7, 44, and 8 (IMGT) ) or variants thereof having up to three amino acid substitutions, additions or deletions; and (ii) an anti-PD-1 scFv comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 51, 52, 53, 54, 55, and 50 (Kabat) or SEQ ID NOs: 45, 46, 47, 48, 49, and 50 (IMGT) ) , or variants thereof having up to three amino acid substitutions, additions or deletions. In some embodiments, the anti-PD-1xVEGF antibody comprises (1) at least one anti-PD-1 scFv comprising or consisting of the amino acid sequence of SEQ ID NO: 56 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 56 preferably comprises the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55, and 50, respectively, or the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively; and/or (2) an anti-VEGF antibody comprising (i) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and a light chain variable region (VL) comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively; or (ii) a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and a VL comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-VEGF antibody comprises (a) the VH and VL sequences as specified above under (ii-1d) , (ii-1e) , or (ii-1f) , or (b) the HC and LC sequences as specified above under (ii-1g) , (ii-1h) , or (ii-1i) .
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 81 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 81, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 81, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 51, 52, 53, 54, 55, and 50, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively. In some alternative embodiments, the anti-PD-L1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 81, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 45, 46, 47, 48, 49, and 50, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
In some particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 81 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 81; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
In some particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 81; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17. In some particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 81; and (ii) two light chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the anti-PD-1xVEGF antibody comprises an anti-VEGF antibody as described herein and at least one anti-PD-1 VHH as described herein, preferably an anti-VEGF antibody as described herein and at least two anti-PD-1 VHHs as described herein, more preferably an anti-VEGF antibody as described herein and two or four anti-PD-1 VHHs as described herein.
In some embodiments of the anti-PD-1xVEGF antibody, the at least one anti-PD-1 VHH may be fused to the anti-VEGF antibody. In some embodiments, each anti-PD-1 VHH is terminally fused to the anti-VEGF antibody. In some preferred embodiments, each anti-PD-1 VHH is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain. In particularly preferred embodiments, each anti-PD-1 VHH is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain. The anti-PD-1xVEGF antibody more preferably comprises an anti-VEGF antibody and two anti-PD-1 VHH, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 VHH, more preferably at the C-terminus of the heavy chain of the anti-VEGF antibody. The anti-PD-1 VHH can be fused to the anti-VEGF antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art, such as linker a comprising or consisting of the amino acid sequence GGGGSGGGGSGGGGSGGGGS (SEQ ID NO: 88) .
In preferred embodiments, the anti-PD-1xVEGF antibody comprises a linker, which more preferably comprises or consists of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody comprising or consisting of two heavy chains as described herein; and (ii) at least two anti-PD-1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-1 VHH through a linker comprising or consisting of the amino acid sequence of SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises two or four anti-PD-1 VHHs as described herein, wherein each anti-PD-1 VHH is fused to the C-terminus of an anti-VEGF-A antibody as described herein, preferably the C-terminus of each heavy chain of the anti-VEGF-A antibody. The anti-PD-1xVEGF antibody can therefore comprise an anti-VEGF-A antibody as described herein and two anti-PD-1 VHHs as described herein, wherein each heavy chain of the anti-VEGF-A antibody is fused to an anti-PD-1 VHH through a linker, such as a linker comprising or consisting of the amino acid sequence shown in SEQ ID NO: 88 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 88.
In particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises (i) an anti-VEGF antibody or fragment thereof comprising the HCDRs and LCDRs as described above (i.e., SEQ ID NOs: 39, 40, 41, 42, 43, and 8 (Kabat) or SEQ ID NOs: 4, 5, 6, 7, 44, and 8 (IMGT) ) or variants thereof having up to three amino acid substitutions, additions or deletions; and (ii) an anti-PD-1 VHH comprising the HCDRs as described above (i.e., SEQ ID NOs: 57, 58, 59, or 60 (Kabat) or SEQ ID NOs: 61, 62, 63, or 64 (IMGT) ) , or variants thereof having up to three amino acid substitutions, additions or deletions. In some embodiments, the anti-PD-1xVEGF antibody comprises (1) at least one anti-PD-1 VHH comprising or consisting of the amino acid sequence of SEQ ID NO: 65 or 66 or an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 65 or 66, wherein the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 65 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, or the HCDR1, HCDR2, and HCDR3 sequences set forth in: 61, 62, and 63, respectively, and the amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence SEQ ID NO: 66 preferably comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, or the HCDR1, HCDR2, and HCDR3 sequences set forth in: 61, 62, and 64, respectively; and/or (2) an anti-VEGF antibody comprising (i) a heavy chain variable region (VH) comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and a light chain variable region (VL) comprising the CDR1, CDR2, and CDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively; or (ii) a VH comprising the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and a VL comprising the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively.
In some embodiments, the CDR variants have up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variants have up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variants have up to three amino acid substitutions. In some embodiments, the CDR variants have up to two amino acid substitutions. In some embodiments, the CDR variants have up to one amino acid substitution.
In some embodiments, the anti-VEGF antibody comprises (a) the VH and VL sequences as specified above under (ii-1d) , (ii-1e) , or (ii-1f) , or (b) the HC and LC sequences as specified above under (ii-1g) , (ii-1h) , or (ii-1i) .
In one preferred embodiment, the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 82, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 82, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively. In some alternative embodiments, the anti-PD-1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 82, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 82; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
In a second preferred embodiment, the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 83 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 83, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 83, and (B) a light chain (LC) comprising an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 59, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively. In some alternative embodiments, the anti-PD-1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 83, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 83, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 63, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 83 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 83; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
In a third preferred embodiment, the anti-PD-1xVEGF antibody comprises (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 84 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 84, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-1xVEGF antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 84, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 39, 40, and 41, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 57, 58, and 60, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 42, 43, and 8, respectively. In some alternative embodiments, the anti-PD-1xVEGF antibody comprises (A') a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 84, and (B') a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 17, wherein the HC comprises (i) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 4, 5, and 6, respectively, and (ii) the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 61, 62, and 64, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 7, 44, and 8, respectively. In some preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 84 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 84; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 17.
In some particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 82, 83, or 84; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 17. In some particularly preferred embodiments, the anti-PD-1xVEGF antibody comprises or consists of (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 82, the amino acid sequence of 83, or the amino acid sequence of 84; and (ii) two light chains each comprising comprising or consisting of the amino acid sequence of SEQ ID NO: 17.
C3. Anti-PD-L1xVEGFR antibodies
In some embodiments, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is a bispecific anti-PD-L1xVEGFR antibody, i.e., a bispecific antibody, wherein the first binding region of the bispecific antibody binds to PD-L1 and the second binding region comprises at least one VEGFR domain.
Exemplary bispecific anti-PD-L1xVEGFR antibodies are the anti-PD-1/PD-L1xVEGF/VEGFR binding agents C3a and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , and HB0025 (Huabo Biopharm) . Such antibodies may alternatively be referred to as fusion proteins comprising an anti-PD-1/PD-L1 antibody or a fragment thereof and a VEGFR domain.
In some embodiments, the bispecific anti-PD-L1xVEGFR antibody comprises any combination of one or more full-length antibodies, such as a human antibody, a humanized antibody, a chimeric antibody, a monoclonal antibody, or a polyclonal antibody, and/or one or more antibody fragments, such as a Fab, a Fab', a F (ab') 2, an Fv, an scFv, a Fab'-SH, an sdAb, or a VHH; and at least one VEGFR domain. In some embodiments, the bispecific anti-PD-L1xVEGFR antibody comprises a full-length antibody and at least one VEGFR domain.
In some embodiments of the bispecific anti-PD-L1xVEGFR antibody, the anti-PD-L1 antibody or fragment thereof comprises a human antibody, a humanized antibody, or a chimeric antibody, preferably a humanized antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is a monoclonal antibody, for example a monoclonal human antibody, a monoclonal humanized antibody, or a monoclonal chimeric antibody. Such antibodies or fragments thereof are fused to at least one, preferably at least two VEGFR domains, most preferably two VEGFR domains.
In some embodiments of the bispecific anti-PD-L1xVEGFR antibody, the anti-PD-L1 antibody or fragment thereof comprises an antibody fragment selected from a Fab, an Fab', an F (ab') 2, an Fv, an scFv, an Fab'-SH, an sdAb, and a VHH, preferably a sdAb, most preferably a VHH. In some embodiments, the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 single domain antibody. In some embodiments, the anti-PD-L1 antibody or fragment thereof is an anti-PD-L1 VHH.
In some embodiments of the bispecific anti-PD-L1xVEGFR antibody, the VEGFR domain comprising protein comprises an Fc part and at least two VEGFR fused to the N-terminus thereof.
In some embodiments, the bispecific anti-PD-L1xVEGFR antibody has monovalent binding to VEGF and/or to PD-L1. In some embodiments, the bispecific anti-PD-L1xVEGFR antibody has bivalent binding to VEGF and/or to PD-L1. In some preferred embodiments, the bispecific anti-PD-L1xVEGFR antibody has bivalent binding to VEGF and to PD-L1.
In some embodiments, the bispecific anti-PD-L1xVEGFR antibody comprises an anti-PD-L1 antibody, preferably a humanized anti-PD-L1 antibody, fused to a VEGFR domain. In some embodiments, the bispecific anti-PD-L1xVEGFR antibody comprises a full-length humanized antibody and two VEGFR domains.
The one or more VEGFR domains may be fused to the anti-PD-L1 antibody. In some embodiments, each VEGFR domain is terminally fused to the anti-PD-L1 antibody. In some embodiments, each VEGFR domain is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain. In preferred embodiments, each VEGFR domain is fused to the N-terminus or the C-terminus of an anti-PD-L1 antibody, preferably the N-terminus of a heavy chain or the C-terminus of a heavy chain. The bispecific antibody preferably comprises a humanized anti-PD-L1 antibody and two VEGFR domains, wherein each heavy chain of the anti-PD-L1 antibody is fused to a VEGFR domain, preferably one VEGFR domain.
The VEGFR domain can be fused to the anti-PD-L1 antibody by a linker, e.g., a peptide linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art. An exemplary linker is GGSGGSGGSGGSGGS (SEQ ID NO: 89) .
In some embodiments, the bispecific anti-PD-L1xVEGFR antibody comprises (a) an anti-PD-L1 antibody, preferably a humanized anti-PD-L1 antibody, comprising a heavy chain variable region, wherein the heavy chain variable region comprises: (i) a HCDR1 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 67, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a HCDR2 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 68, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a HCDR3 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 69, or a variant thereof having up to three amino acid substitutions, additions or deletions; and a light chain variable region, wherein the light chain variable region comprises: (i) a LCDR1 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 70, or a variant thereof having up to three amino acid substitutions, additions or deletions; (ii) a LCDR2 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 71, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (iii) a LCDR3 comprising or consisting of the amino acid sequence shown in SEQ ID NO: 72, or a variant thereof having up to three amino acid substitutions, additions or deletions; and (b) a VEGFR domain comprising or consisting of the amino acid sequence shown in SEQ ID NO: 77, or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 77.
In some preferred embodiments, the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 85, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1xVEGFR antibody comprises (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 85, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 86, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively. In some preferred embodiments, the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 85; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86.
In some alternatively preferred embodiments, the anti-PD-L1xVEGFR antibody comprises or consist of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 87 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 87, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86, and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. In some embodiments, the anti-PD-L1xVEGFR antibody comprises or consists of (A) a heavy chain (HC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 87, and (B) a light chain (LC) comprising or consisting of an amino acid sequence having at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%sequence identity to the amino acid sequence of SEQ ID NO: 86, wherein the HC comprises the HCDR1, HCDR2, and HCDR3 sequences set forth in: SEQ ID NO: 67, 68, and 69, respectively, and the LC comprises the LCDR1, LCDR2, and LCDR3 sequences set forth in: SEQ ID NO: 70, 71, and 72, respectively. In some preferred embodiments, the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 87 or an amino acid sequence comprising or consisting of at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 87; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100%identity to the amino acid sequence of SEQ ID NO: 86.
In some particularly preferred embodiments, the anti-PD-L1xVEGFR antibody comprises or consists of (i) a heavy chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or 87; and (ii) a light chain sequence comprising or consisting of the amino acid sequence of SEQ ID NO: 86. In some particularly preferred embodiments, the anti-PD-L1xVEGFR antibody comprises or consists of (i) two heavy chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 85 or the amino acid sequence of SEQ 87; and (ii) two light chains each comprising or consisting of the amino acid sequence of SEQ ID NO: 86.
A preferred bispecific antibody
In some preferred embodiments, the bispecific antibody comprises an anti-PD-L1 antibody or fragment thereof. In some preferred embodiments, the bispecific antibody comprises an anti-VEGF antibody or fragment thereof, preferably an anti-VEGF-A antibody or fragment thereof.
In some embodiments, the bispecific antibody comprises a Fab, F’b', F (ab') 2, Fd, Fv, sdAb, complementarity determining region fragment, single chain antibody, humanized antibody, chimeric antibody or diabody antibody, preferably a single domain antibody, more preferably a VHH.
In some embodiments, the bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably fused to the N-or C-terminus of the anti-VEGF antibody heavy or light chain. In some embodiments, the bispecific antibody comprises two anti-PD-L1 single domain antibodies, preferably two VHHs, preferably fused to the C-terminus of the anti-VEGF antibody heavy chain.
In some embodiments, the anti-PD-L1 antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof. In some embodiments, the anti-VEGF antibody or fragment thereof is a monovalent or bivalent antibody or fragment thereof,
In some preferred embodiments, the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 2, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the IMGT numbering system (see Ehrenmann F, Kaas Q, Lefranc M P. IMGT/3Dstructure-DB and IMGT/DomainGapAlign: a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF [J] . Nucleic acids research, 2009; 38 (suppl_1) : D301-D307) .
In an alternative example, the bispecific antibody comprises an anti-PD-L1 single domain antibody comprising a heavy chain variable region, and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 18, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 19, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 38, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions, wherein the CDRs are defined according to the Kabat numbering system.
In some embodiments, the CDR variant has up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variant has up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variant has up to three amino acid substitutions. In some embodiments, the CDR variant has up to two amino acid substitutions. In some embodiments, the CDR variant has up to one amino acid substitution.
In some embodiments, the amino acid sequence of the anti-PD-L1 single domain antibody is shown in SEQ ID NO: 9. In some embodiments, the amino acid sequence of the anti-PD-L1 single domain antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof. For example, the bispecific antibody can comprise one anti-VEGF antibody and two anti-PD-L1 single domain antibodies, preferably fused to the C-terminus of the anti-VEGF antibody, wherein each of these anti-PD-L1 single domain antibodies comprises or consists of the amino acid sequence shown in SEQ ID NO: 9 or wherein the amino acid sequence of the anti-PD-L1 single domain antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9 In some embodiments, the amino acid sequence of the anti-PD-L1 single domain antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 9 and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity thereto and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and comprises a heavy chain variable region comprising: a HCDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 1; a HCDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 2; and a HCDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 3.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9, or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity thereto and/or one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) amino acid additions, deletions and/or substitutions in a framework region thereof, and comprises a heavy chain variable region comprising: a HCDR1 comprising or consisting of the amino acid sequence of SEQ ID NO: 18; a HCDR2 comprising or consisting of the amino acid sequence of SEQ ID NO: 19; and a HCDR3 comprising or consisting of the amino acid sequence of SEQ ID NO: 38.
In some embodiments, the anti-PD-L1 antibody or fragment thereof comprises or consists of the amino acid sequence of SEQ ID NO: 9. In some embodiments, the anti-PD-L1 antibody or fragment thereof consists of the amino acid sequence of SEQ ID NO: 9.
In some preferred embodiments, the bispecific antibody specifically binds to VEGF-A.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably, the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4. In some embodiments, the anti-VEGF-A antibody or fragment thereof comprises a constant region preferably derived from a human antibody, preferably, the constant region is selected from the constant region of human IgG1, IgG2, IgG3 or IgG4.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises an Fc region comprising one or more mutations which reduce or abolish immune effect functions. Suitable mutations are known in the art and may include one or more amino acid substitutions (see e.g. Wilkinson, I., et al., 2021. FcPLoS One, 16 (12) , p. e0260954; and Liu, R., et al., 2020. Antibodies, 9 (4) , p. 64) . In some embodiments, the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising a L234A (with the numbering according to the EU-index) and/or a L235A mutation (with the numbering according to the EU-index) . In some embodiments, the anti-VEGF antibody or fragment thereof comprises an IgG1 Fc region comprising L234A and L235A mutations (with the numbering according to the EU-index) . As noted, amino acids are numbered according to the Eu numbering (Eu-index) as set forth in Kabat (Kabat, E.A.; National Institutes of Health (U.S. ) Office of the Director. Sequences of Proteins of Immunological Interest, 5th ed.; DIANE Publishing: Collingdale, PA, USA, 1991) .
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO: 13. In some embodiments, the anti-VEGF-A antibody or fragment thereof comprises a IgG1 Fc region, preferably having the amino acid sequence shown in SEQ ID NO: 13 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 13.
The one or more anti-PD-L1 single domain antibodies may be fused to the anti-VEGF antibody by any suitable method. In some embodiments, each anti-PD-L1 single domain antibody is terminally-fused to the anti-VEGF antibody. In some embodiments, each anti-PD-L1 single domain antibody is fused to: (i) the C-terminus of a heavy chain; (ii) the N-terminus of a heavy chain; (iii) the C-terminus of a light chain; or (iv) the N-terminus of a light chain. In preferred embodiments, each anti-PD-L1 single domain antibodies is fused to the C-terminus of an anti-VEGF antibody, preferably the C-terminus of a heavy chain. The bispecific antibody preferably comprises an anti-VEGF antibody and two anti-PD-L1 VHHs, wherein each heavy chain of the anti-VEGF antibody is fused to an anti-PD-L1 VHH.
The VHH can be fused to the anti-VEGF antibody by a linker, for example a glycine and serine-rich linker (GS-linker) or any suitable linker known in the art. An example linker is provided by SEQ ID NO: 14 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 14.
In some embodiments, the anti-VEGF antibody or fragment thereof comprises a CL, preferably having the amino acid sequence shown in SEQ ID NO: 15 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 15. In some embodiments, the anti-VEGF antibody or fragment thereof comprises a CH1, preferably having the amino acid sequence shown in SEQ ID NO: 12 or an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to the amino acid sequence shown in SEQ ID NO: 12.
In some embodiments, the anti-VEGF antibody comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6, or a variant thereof having up to three e.g. one or two amino acid substitutions, additions or deletions; and wherein the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions. These CDRs are defined according to the IMGT numbering system (see Ehrenmann F, Kaas Q, Lefranc M P. IMGT/3Dstructure-DB and IMGT/DomainGapAlign: a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF [J] . Nucleic acids research, 2009; 38 (suppl_1) : D301-D307) .
In some embodiments, the anti-VEGF antibody comprises a heavy and light chain variable region, wherein the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 39, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 40, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 41, or a variant thereof having up to three e.g. one or two amino acid substitutions, additions or deletions; and wherein the light chain variable region comprises: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 42, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 43, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8, or a variant thereof having up to three, e.g. one or two, amino acid substitutions, additions or deletions. These CDRs are defined according to the Kabat numbering system.
In some embodiments, the CDR variant has up to two amino acid substitutions, additions or deletions. In some embodiments, the CDR variant has up to one amino acid substitution, addition or deletion. In some embodiments, the CDR variant has up to three amino acid substitutions. In some embodiments, the CDR variant has up to two amino acid substitutions. In some embodiments, the CDR variant has up to one amino acid substitution.
In some embodiments, the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody (or anti-VEGF-A antibody) is shown in SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody is shown in SEQ ID NO: 11; or wherein the amino acid sequence of the heavy chain variable region of the anti-VEGF antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 10, and the amino acid sequence of the light chain variable region of the anti-VEGF antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 11.
In some embodiments, the amino acid sequence of the heavy chain of the bispecific antibody is shown in SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody is shown in SEQ ID NO: 17; or
wherein the amino acid sequence of the heavy chain of the bispecific antibody has at least 80%, 85%, 90%, 
95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 16, and the amino acid sequence of the light chain variable region of the bispecific antibody has at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99%identity to SEQ ID NO: 17.
In some embodiments, the anti-VEGF-A antibody is bevacizumab.
In some embodiments, the bispecific antibody is encoded by one or more nucleic acid molecules.
The bispecific antibody can preferably comprise:
a. two anti-PD-L1 single domain antibodies each comprising a heavy chain variable region, 
and the heavy chain variable region comprises: (i) a complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 1, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 2, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 3; and
b. an anti-VEGF-A antibody comprising a heavy chain variable region comprising: (i) a 
complementarity-determining region 1 (HCDR1) whose amino acid sequence is shown in SEQ ID NO: 4, (ii) a complementarity-determining region 2 (HCDR2) whose amino acid sequence is shown in SEQ ID NO: 5, and (iii) a complementarity-determining region 3 (HCDR3) whose amino acid sequence is shown in SEQ ID NO: 6; and a light chain variable region comprising: (i) a complementarity-determining region 1 (LCDR1) whose amino acid sequence is shown in SEQ ID NO: 7, (ii) a complementarity-determining region 2 (LCDR2) whose amino acid sequence is shown in SEQ ID NO: 44, and (iii) a complementarity-determining region 3 (LCDR3) whose amino acid sequence is shown in SEQ ID NO: 8.
In particularly preferred embodiments, the bispecific antibody or bispecific antibody variants are described in WO 2022/042719, which disclosure is hereby incorporated in its entirety.
The bispecific antibody can be produced by various methods, see, for example, the method of Songsivilai et al. (Clin. Exp. Immunol., 79: 315-321 (1990) ) , and the method of Kostelny et al. (J. Immunol., 148: 1547-1553 (1992) which disclosures are also hereby incorporated in their entirety.
Regimen of the present invention
Administration and regimens with a focus on the binding agent
In some embodiments, the binding agent (including, e.g., the bispecific antibody) and the chemotherapy are separately administered. In some embodiments, a dosage of the binding agent (including, e.g., the bispecific antibody) and a dosage of the chemotherapy are separately administered. In some embodiments, a dosage of the binding agent (including, e.g., the bispecific antibody) and a dosage of the chemotherapy are administered using a single composition.
In some embodiments, a dosage of the binding agent (including, e.g., the bispecific antibody) and a dosage of the chemotherapy are administered concurrently or consecutively.
The binding agent (including, e.g., the bispecific antibody) and the chemotherapy provided herein can be administered via any suitable enteral route or parenteral route of administration. The term “enteral route” of administration refers to the administration via any part of the gastrointestinal tract. Examples of enteral routes include oral, mucosal, buccal, and rectal route, or intragastric route. “Parenteral route” of administration refers to a route of administration other than enteral route. Examples of parenteral routes of administration include intravenous, intramuscular, intradermal, intraperitoneal, intratumour, intravesical, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, transtracheal, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrastemal, subcutaneous, or topical administration. The binding agent (including, e.g., the bispecific antibody) and the chemotherapy of the disclosure can be administered using any suitable method, such as by oral ingestion, nasogastric tube, gastrostomy tube, injection, infusion, implantable infusion pump, and osmotic pump. The suitable route and method of administration may vary depending on a number of factors such as the specific therapeutic agent being used, the rate of absorption desired, specific formulation or dosage form used, type or severity of the disorder being treated, the specific site of action, and conditions of the subject, and can be readily selected by a person skilled in the art. The terms “treatment regimen, ” “dosing protocol, ” and “dosing regimen” are used interchangeably to refer to the dosage and timing of administration of each dosage binding agent (including, e.g., the bispecific antibody) and the chemotherapy in a combination therapy of the disclosure. A parenteral route of administration of the binding agent and the chemotherapy is particularly preferred, wherein an intravenous administration is particularly preferred.
In some preferred embodiments, the binding agent (including, e.g., the bispecific antibody) and/or the chemotherapy is administered intravenously, preferably wherein the binding agent (including, e.g., the bispecific antibody) and the chemotherapy are administered intravenously. In some embodiments, the binding agent (including, e.g., the bispecific antibody) and/or the chemotherapy is administered via an IV injection or IV infusion. Preferably, the binding agent (including, e.g., the bispecific antibody) and the chemotherapy are administered via an IV injection or IV infusion. For example, the binding agent (including, e.g., the bispecific antibody) and the chemotherapy can be administered concurrently or consecutively via an IV infusion.
In one embodiment, the binding agent (including, e.g., the bispecific antibody) is administered in a step-up dosing regimen to a subject or patient, e.g., with a first and one or more subsequent dose regimens. For example, in the first dose regimen the dosage of the binding agent (including, e.g., the bispecific antibody) is lower than the dosage of the binding agent (including, e.g., the bispecific antibody) in a second dose regimen. If the binding agent (including, e.g., the bispecific antibody) is administered in more than two dose regimens, the dosage of the binding agent (including, e.g., the bispecific antibody) in each subsequent dosage regimen is higher compared to the dosage of the binding agent (including, e.g., the bispecific antibody) in the dose regimen that precedes each subsequent dose regimen. In some embodiments of the afore mentioned examples, the dosage of the chemotherapeutic agent may be the same in the first and one or more subsequent dose regimens. In some embodiments of the afore mentioned examples, in the first dose regimen the dosage of the chemotherapeutic agent may be lower than the dosage of the chemotherapeutic agent in a second dose regimen. If the chemotherapeutic agent is administered in more than two dose regimens in some embodiment of the afore mentioned examples, the dosage of the chemotherapeutic agent in each subsequent dosage regimen may be higher compared to the dosage of the chemotherapeutic agent in the dose regimen that precedes each subsequent dose regimen.
In some embodiments, the subject has been determined to have a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ≤ 30, preferably ≤ 20, more preferably ≤ 10, and most preferably < 10 or even < 1 or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP, preferably TAP. For example, the subject has been preferably determined to have a PD-L1 expression score before the treatment as determined by a CPS of up to 20, preferably from 1 to 20 or > 0 to 20, more preferably from 1 to <10, such as 1 to 9, or other integrating scoring algorithm defining a similar PD-L1 expression score. In some preferred embodiments, the subject has been determined to have a PD-L1 expression score as determined by a CPS from 1 to 9, 1 to 8, 1 to 7, 1 to 6, 1 to 5, 1 to 4, or 1 to 3 or other integrating scoring algorithm defining a similar PD-L1 expression score. For example, the subject has a PD-L1 expression score as determined by a CPS of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20, preferably a CPS of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, more preferably a CPS of 1, 2, 3, 4, 5, 6, 7, 8, or 9 or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP. In some particularly preferred embodiments, the subject has been determined to have a PD-L1 expression score as determined by CPS of < 10, < 9, < 8, < 7, < 6, < 5, < 4, or < 3 before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP. In some particularly preferred embodiments, the subject has been determined to have a PD-L1 expression score as determined by CPS of > 0 before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
In some embodiments, the subject is part of a patient group to be treated with the combination therapy disclosed herein. For example, this patient group comprises one or more subjects having a PD-L1 expression score as determined by a CPS of ≤ 30, preferably ≤ 20, more preferably ≤ 10, and most preferably < 10 or even < 1, before the treatment or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP. This patient group may further comprise one or more subjects having a PD-L1 expression score as determined by a CPS of > 30, preferably >20, more preferably > 10, and most preferably ≥ 10 or other integrating scoring algorithm defining a similar PD-L1 expression score such as TPS or TAP.
In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered in a dosage ranging from 0.1 mg/kg to 45 mg/kg body weight, preferably 1 mg/kg to 30 mg/kg body weight per treatment.
In some embodiments, the treatment comprises repeated treatments, wherein the treatment cycle is repeated at least 1, 2, 3, 4, 5, 6, 7, or 8 times, wherein each cycle preferably has up to 30, preferably 28 days or 21 days. However, the treatment can be continued until disease progression or the initiation of a new anti-tumour treatment.
In some embodiments, the subject has not been previously treated for cancer, i.e., is treatment naive. In some embodiments, the subject has been previously treated for cancer, in particular the subject has had at least one previous chemotherapy treatment.
In some embodiment, the subject has not been previously treated with a PD-1/PD-L1 inhibitor, such as pembrolizumab or atezolizumab.
In some embodiment, the subject had one or more been previous treatments with a PD-1/PD-L1 inhibitor, such as pembrolizumab or atezolizumab.
In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered every 6 weeks, preferably every 4 weeks, more preferably every 3 weeks, and most preferably every 2 weeks.
In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered every 2 weeks at a dosage ranging from 10 mg/kg to 30 mg/kg, preferably ranging from 15 mg/kg to 25 mg/kg, more preferably being 20 mg/kg.
In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered every 3 weeks at a dosage ranging from 20 mg/kg to 40 mg/kg, preferably ranging from 25 mg/kg to 35 mg/kg, more preferably being 30 mg/kg.
In some embodiments, the chemotherapy is administered once or more within the first 20 days or the first 21 days of each cycle, wherein the chemotherapy is administered twice or more within the first 20 days or the first 21 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 20 days or the first 21 days of each cycle.
In some embodiments, the chemotherapy is administered once or more within the first 14 days or the first 15 days of each cycle, wherein the chemotherapy is administered twice or more within the first 14 days or the first 15 days of each cycle, more preferably wherein the chemotherapy is administered at least thrice within the first 14 days or the first 15 days of each cycle. For example, the chemotherapy can be administered on the 1st, 8th, and 15th day of each cycle, wherein each cycle has 21 days.
In some embodiments, the treatment method disclosed herein results in increased overall survival in said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or an anti-PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and an anti-PD-L1 antibody or chemotherapy and an anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab. In some embodiments, the treatment method disclosed herein results in increased median progression-free survival said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or anti-PD-L1 antibody or anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
Administration and regimens with a focus on the chemotherapy
The methods of treating disclosed herein comprise a chemotherapy. In some embodiments, the chemotherapy comprises one or more platinum-based chemotherapeutic agents. In some embodiments, the one or more platinum-based chemotherapeutic agents is carboplatin, cisplatin, oxaliplatin, or combinations thereof. In some embodiments, the chemotherapy comprises one or more antimetabolite-based chemotherapy agents. In some embodiments, the one or more antimetabolite-based chemotherapeutic agent is gemcitabine. In some embodiments, chemotherapy comprises one or more platinum-based chemotherapeutic agents in combination with one or more antimetabolite-based chemotherapy agents. In some embodiments, the chemotherapy comprises cisplatin and gemcitabine.
In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered concurrently with chemotherapy. In some embodiments, the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered within up to 15 days of each other. In some embodiments, the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered within about two days of each other. In some embodiments, the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered within about one day of each other. In some embodiments, the binding agent (including, e.g., the bispecific antibody) and chemotherapy are administered concurrently (for example by simultaneous (same day) administration) . In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered on day 1 of the chemotherapy treatment cycle.
In some embodiments, the observed toxicities of the treatment with the binding agent (including, e.g., the bispecific antibody) and chemotherapy combination are similar to those commonly seen with either chemotherapy or immunotherapy alone.
In some embodiments, the chemotherapy comprises a chemotherapy agent selected from lurbinectedin, topotecan, taxane, such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof. In some embodiments, the chemotherapy comprises a chemotherapy agent selected from topotecan, taxane, such as paclitaxel, nanoparticle albumin-bound paclitaxel (nab-paclitaxel) or docetaxel, pemetrexed, 5-fluoruracil, irinotecan, etoposide, gemcitabine, anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone or combinations thereof.
In some embodiments, the platinum-based chemotherapy comprises cisplatin, oxaliplatin or carboplatin.
In some preferred embodiments, the chemotherapy agent is selected from the group consisting of a (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with paclitaxel, lurbinectedin, or topotecan. In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with paclitaxel or topotecan. In some such embodiments, the cancer is small cell lung cancer and, preferably, the method of treatment can be a second line cancer treatment. In a particularly preferred embodiment, the method of treatment comprising administering to a patient suffering from advanced small cell lung cancer as second line treatment the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with paclitaxel every 3 weeks at a dosage of 175 mg/m2. Further, in this particularly preferred embodiment, the combination may be administered for five cycles followed by the administration of the binding agent (including, e.g., the bispecific antibody) only as maintenance therapy. In another particularly preferred embodiment, the method of treatment comprising administering to a patient suffering from advanced small cell lung cancer as second line treatment the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg in combination with topotecan at a dosage of 1.5 mg/m2 administered IV or 2.3 mg/m2 administered orally once per day on days 1 to 5 of every 3 weeks cycle. In the afore-mentioned two particularly preferred embodiments, the patient may have progressed during or after first-line platinum-based therapy (with or without checkpoint inhibitor therapy) .
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin. In some such embodiments, the cancer is a malignant mesothelioma (including malignant pleural mesothelioma [MPM] and malignant peritoneal mesothelioma [MPeM] ) and, preferably, the method of treatment can be a first line cancer treatment.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin. In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with paclitaxel and carboplatin. In some such embodiments, the cancer is NSCLC, preferably the method of treatment can be a first line treatment. In one example, the cancer is non-squamous NSCLC. In another example, the cancer is squamous NSCLC. In some preferred embodiments, pemetrexed and carboplatin can be administered at a dosage of 500 mg/m2 and AUC 5 (preferably with a total dose of ≤ 750 mg) , respectively, every 3 weeks, e.g. for 4 cycles. In some such embodiments, the cancer is non-squamous NSCLC. In some preferred embodiments, paclitaxel and carboplatin can be administered at a dosage of 200 mg/m2 and AUC 6 (preferably with a total dose of ≤900 mg) , respectively, every 3 weeks, e.g. for 4 cycles. In some such embodiments, the cancer is squamous NSCLC.
The cancer to be treated
In some embodiments, the cancer is a solid tumour.
In some embodiments, the cancer is selected from the group consisting of malignant mesothelioma, hepatocellular cancer, neuroendocrine neoplasm, melanoma, lung cancer, liver cancer, stomach cancer, renal (cell) cancer, urothelial cancer, cervical cancer, colorectal cancer, ovarian cancer, colon cancer, breast cancer, esophagus cancer, head and neck cancer and pancreatic cancer, preferably the cancer is selected from urothelial cancer, breast cancer and esophagus cancer. For example, the cancer can be selected from non-small cell lung cancer (NSCLC) , small cell lung cancer (SCLC) including extensive stage small cell lung cancer and limited stage small cell lung cancer, urothelial carcinoma, esophageal cancer, head and neck squamous cell carcinoma (HNSCC) , triple-negative breast cancer (TNBC) or cervical cancer. The cancer can preferably be TNBC, gastric adenocarcinoma, gastroesophageal junction (GEJ) adenocarcinoma, esophageal squamous-cell carcinoma, cervical cancer, urothelial carcinoma, or HNSCC. Most preferably, the cancer is selected from the group consisting of small cell lung cancer, non-small cell lung cancer, triple-negative breast cancer, malignant mesothelioma, hepatocellular cancer, neuroendocrine neoplasm, and colorectal cancer.
Most preferably, the cancer is TNBC. The TNBC may be a TNBC of the subtype (1) luminal androgen receptor (LAR) , (2) immunomodulatory (IM) , (3) basal-like immune-suppressed (BLIS) , or (4) mesenchymal-like (MES) according to the Fudan University Shanghai Cancer Center (FUSCC) classification (see Jiang YZ, Ma D, Suo C, et al. Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies. Cancer Cell. 2019; 35 (3) : 428-440. e5) , preferably TNBC of the subtype IM or MES.
In some preferred embodiments, the cancer is small cell lung cancer (SCLC) , non-small cell lung cancer (NSCLC) or triple-negative breast cancer (TNBC) , preferably the cancer is triple-negative breast cancer, for example advanced or metastatic triple-negative breast cancer.
In some embodiments, the NSCLC has a squamous histology. In some embodiments, the NSCLC has a non-squamous histology. In some embodiments, the NCLC is an EGFR mutation-positive NSCLC.
Administration and regimens
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin or in combination with a taxane, such as paclitaxel or docetaxel, preferably docetaxel. In some such embodiments, the cancer is NSCLC, preferably the method of treatment can be a second line treatment. For example, the cancer is an EGFR-mutant advanced non-squamous NSCLC and, preferably, the method of treatment can be a cancer treatment following a failed EGFR-TKI treatment. Thus, it can be preferred that the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with pemetrexed and carboplatin to a patient suffering from NSCLC as second-line treatment, in particular to a patient suffering from NSCLC who progressed after EGFR-TKI treatment. In the afore-mentioned embodiment, it is particularly preferred that the binding agent (including, e.g., the bispecific antibody) is administered at a dosage of 30 mg/kg plus AUC 5 mg/mL/min carboplatin and 500 mg/m2 pemetrexed every three weeks, e.g. for 4 cycles. Such a treatment may be followed by a maintenance treatment using 30 mg/kg of the binding agent (including, e.g., the bispecific antibody) in combination with 500 mg/m2 pemetrexed. In another embodiment, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with a taxane, such as paclitaxel or docetaxel, preferably docetaxel, to a patient suffering from NSCLC, e.g. squamous NSCLC, as second-line treatment, e.g., to a patient suffering from second-line stage IV or recurrent NSLC who progressed at least one prior line of an immune-oncology (IO) comprising treatment such as an immune checkpoint inhibitor comprising treatment or chemoimmunotherapy. In the afore-mentioned embodiment, it is particularly preferred that the binding agent (including, e.g., the bispecific antibody) is administered at a dosage range of 1400 mg to 2000 mg (with 1400 mg or 2000 mg being preferred) plus 75 mg/m2 docetaxel every three weeks. For example, the binding agent (including, e.g., the bispecific antibody) may be administered at a dosage of 2000 mg binding agent (including, e.g., the bispecific antibody) plus 75 mg/m2 docetaxel every three weeks. In another example, the binding agent (including, e.g., the bispecific antibody) may be administered in a step-up dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is 1400 mg binding agent (including, e.g., the bispecific antibody) plus 60 mg/m2 docetaxel every three weeks or 1400 mg binding agent (including, e.g., the bispecific antibody) plus 75 mg/m2 docetaxel every three weeks and the second dose regimen is 2000 mg binding agent (including, e.g., the bispecific antibody) plus 75 mg/m2 docetaxel every three weeks.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with anthracyclines, capecitabine, vinorelbine, eribulin, ixabepilone, nab-paclitaxel, paclitaxel, or gemcitabine and carboplatin, preferably nab-paclitaxel, paclitaxel, gemcitabine and carboplatin or eribulin. In some such embodiments, the cancer is TNBC (e.g., advanced or metastatic TNBC) and, preferably, the method of treatment can be a first line cancer treatment or second line cancer treatment. For example, nab-paclitaxel can be administered at a dosage of 100 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 15 mg/kg or 20 mg/kg or 1400 mg on the 1st and 15th day of a 28-days treatment cycle) . For example, paclitaxel can be administered at a dosage of 90 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg on the 1st and 15th day of a 28-days treatment cycle) . Gemcitabine and carboplatin can be administered at a dosage of 1000 mg/m2 and AUC 2 respectively, on the 1st and 8th day of a 21-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg on the 1st day of a 21-days treatment cycle) . Eribulin can be administered at a dosage of 1.4 mg/m2 on the 1st and 8th day of a 21-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg on the first day of a 21-days treatment cycle) . In a preferred example, Nab-paclitaxel can be administered at a dosage of 100 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg on the 1st and 15th day of a 28-days treatment cycle) . In a further preferred example, paclitaxel can be administered at a dosage of 90 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg on the 1st and 15th day of a 28-days treatment cycle) . In a further preferred example, gemcitabine can be administered at a dosage of 1000 mg/m2 and carboplatin can be administered at AUC2 on the 1st and 8th, and 15th day of a 21-days treatment cycle (preferably in combination with the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg every 3 weeks) . In the aforementioned examples, the patient may have a PD-L1 expression score before the treatment as determined by a combined positive score (CPS) of < 10 or other integrating scoring algorithm defining a similar PD-L1 expression score. In other words, the patient may have a PD-L1 expression before the treatment that provides a score of < 10 when determined by the combined positive scope (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating score. In the aforementioned examples, the treatment may be a first line cancer treatment.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with FOLFIRNOX, preferably comprising folinic acid, oxaliplatin, Irinotecan Hydrochloride, and 5-fluorouracil. In some such embodiments, the cancer is hepatocellular carcinoma and, preferably, the method of treatment can be a first line cancer treatment.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) , e.g. at a dosage of 20 mg/kg every two weeks, in combination with oxaliplatin, calcium folinate, and 5-fluorouracil (which may also be referred to as FOLFOX4) . In some such embodiments, the cancer is hepatocellular carcinoma and, preferably, the method of treatment can be a first line cancer treatment.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with irinotecan, 5-fluorouracil, calcium folinate. In some such embodiments, the cancer is unresectable neuroendocrine neoplasm and, preferably, the method of treatment can be a second line cancer treatment.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) in combination with etoposide and platinum. In some such embodiments, the cancer is small cell lung cancer, e.g., extensive-stage small cell lung cancer or limited-stage small cell lung cancer, and, preferably, the method of treatment can be a first line cancer treatment.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg in combination with etoposide with carboplatin to a subject having small cell lung cancer, e.g., extensive-stage small cell lung cancer or limited-stage small cell lung cancer. Preferably this method of treatment is a first line treatment and for extensive-stage small cell lung cancer, wherein the binding agent (including, e.g., the bispecific antibody) is administered every 3 weeks at a dosage of 20 mg/kg or 30 mg/kg together with the etoposide (preferably at a dosage of 100 mg/m2) with carboplatin (preferably at a dosage of AUC=5 (total dose ≤ 750 mg) .
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage ranging from 20 mg/kg to 30 mg/kg (preferably at 20 mg/kg or 30 mg/kg) in combination with paclitaxel, lurbinectedin, or topotecan to a subject having small cell lung cancer, e.g., extensive-stage small cell lung cancer. Preferably this method of treatment is a second line treatment, in particular a treatment of SCLC patients who progressed during or after first-line platinum-based therapy (with or without checkpoint inhibitor therapy) . Preferably this method of treatment comprises paclitaxel, in particular paclitaxel every 3 weeks at a dosage of 175 mg/m2. Preferably this method of treatment comprises topotecan, in particular topotecan at a dosage of 1.5 mg/m2 administered IV or 2.3 mg/m2 administered orally once per day on days 1 to 5 of every three weeks cycle.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 3 weeks at a dosage ranging from 1400 mg to 2000 mg in combination with etoposide with carboplatin to a subject having small cell lung cancer, e.g., extensive-stage small cell lung cancer. Preferably this method of treatment is a first line treatment and for extensive-stage small cell lung cancer, wherein the binding agent (including, e.g., the bispecific antibody) is administered every 3 weeks at a dosage of 1400 mg or 2000 mg together with the etoposide (preferably at a dosage of 100 mg/m2) on days 1-3 and carboplatin (preferably at a dosage of AUC=5 (total dose ≤ 750 mg) on day 1 of a 21 day cycle, preferably for 4 cycles followed by a 1400 mg or 2000 mg dose every 3 weeks, respectively, of the binding agent (including, e.g., the bispecific antibody) .
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 2 weeks at a dosage ranging from 10 mg/kg to 20 mg/kg, e.g. 15 mg/kg or 20 mg/kg, preferably on the 1st and 15th day of a 28 day cycle, in combination with nab-paclitaxel, paclitaxel, gemcitabine with carboplatin, or eribulin to a subject having triple-negative breast cancer.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) at a flat dosage, e.g. at a dosage of 500 mg to 3000 mg, 750 mg to 2500 mg, 1000 mg to 2250 mg or 1500 mg to 2000 mg to a subject. 2000 mg may be particularly preferred. In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered every week or every 2, 3 or 4 weeks, preferably every 2 weeks. Preferred is an amount of 2000 mg administered every 3 weeks. Simulations using integrated PopPK (population pharmacokinetic) analysis surprisingly show that flat dosing and weight-based dosing approaches provide similar results (see, e.g., Figures 1 and 2 as well as example 7 of the present application) . A fixed dosing approach is desirable to facilitate drug preparation without the need to calculate the dosage, per participant, thus lowering the potential risk of dosing errors as compared with body weight-based dosing. In addition, fixed dosing also offers several advantages over weight-based dosing including better convenience and compliance, reduction of preparation time, and a reduced amount of drug waste.
In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 2 weeks at a dosage of 500 mg to 3000 mg, 750 mg to 2500 mg, 1000 mg to 2250 mg or 1500 mg to 2000 mg to a subject, preferably of 1000 mg or 1400 mg to a subject having triple-negative breast cancer, preferably on the 1st and 15th day of a 28-day treatment cycle. It can be preferred to administer the binding agent (including, e.g., the bispecific antibody) at a dosage of 1400 mg (preferably on the 1st and 15th day of a 28-days treatment cycle) or to administer the binding agent (including, e.g., the bispecific antibody) at a dosage of 2000 mg (preferably on the 1st day of a 21-days treatment cycle) to a subject having triple-negative breast cancer.
In some embodiments the binding agent (including, e.g., the bispecific antibody) is administered in combination with a chemotherapy.
In some embodiments, the chemotherapy is selected from the group consisting of nab-paclitaxel, paclitaxel, or gemcitabine with carboplatin. In a preferred embodiment, the chemotherapy is nab-paclitaxel. In some embodiments, the chemotherapy is administered on the 1st, 8th and 15th day of a 28-day treatment cycle. In some embodiments, the subject has not received prior systemic treatment, preferably not in the advanced setting. In some preferred embodiments, the method of treatment comprises administering the binding agent (including, e.g., the bispecific antibody) every 2 weeks at a dosage of 500 mg to 3000 mg, 750 mg to 2500 mg, 1000 mg to 2250 mg or 1500 mg to 2000 mg to a subject, preferably of 1000 mg or 1400 mg to a subject having small cell lung cancer, preferably on the 1st day of a 21-day treatment cycle. In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered in combination with a chemotherapy, preferably selected from paclitaxel, topotecan or lurbinectedin. In some embodiments, the binding agent (including, e.g., the bispecific antibody) is administered in combination with a chemotherapy, preferably selected from paclitaxel or topotecan. In some embodiments, the chemotherapy is paclitaxel. In some embodiments, the paclitaxel is administered on the 1st day of a 21-day treatment cycle. In another embodiment, the chemotherapy is topotecan. In some embodiments, the topotecan is administered on the 1st and 5th day of a 21-day treatment cycle.
The invention further provides a chemotherapy agent for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and 
VEGF) ; and
b. the chemotherapy agent;
optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the 
combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
The embodiments disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in a method of treatment of the chemotherapy agent in combination with the binding agent (including, e.g., the bispecific antibody) as disclosed herein. Features described herein in more detail for the “binding agent (including, e.g., the bispecific antibody) for use in a method of treating” embodiments equally apply to the chemotherapy agent for use in a method of treating disclosed herein.
In an aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with paclitaxel, wherein the antibody is administered every three weeks at a dosage of about 20 mg/kg or about 30 mg/kg and the paclitaxel is administered every three weeks at a dosage of about 175 mg/m2. Preferably, the patient progressed during or after first-line platinum-based therapy (with or without immuno-oncology (IO) , e.g., checkpoint inhibitor therapy) , i.e. the patient failed first-line platinum-based chemotherapy. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a second-line treatment for a patient suffering from small cell lung cancer. It is also preferred that the combination is administered intravenously. The treatment may take place for 5 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 20 mg/kg or about 30 mg/kg every three weeks.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with topotecan, e.g., wherein the antibody is administered every three weeks at a dosage of about 20 mg/kg or about 30 mg/kg and the topotecan is administered every three weeks at days 1 to 5 once daily at a dosage of about 1.5 mg/m2 IV or about 2.3 mg/m2 orally. Preferably, the patient progressed during or after first-line platinum-based therapy (with or without immuno-oncology (IO) , e.g., checkpoint inhibitor therapy) , i.e. the patient failed first-line platinum-based chemotherapy. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a second-line treatment for a patient suffering from small cell lung cancer. It is also preferred that the antibody is administered intravenously.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with extensive-stage small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy (in particular with platinum-etoposide) , wherein the antibody is administered every three weeks at a dosage of about 20 mg/kg or about 30 mg/kg in combination with systemic chemotherapy (in particular with platinum [preferably carboplatin at a dosage of about AUC=5 (total dose ≤ about 750 mg) at day 1 of each cycle] and etoposide [preferably at a dosage of about 100 mg/m2 on days 1 to 3 of each cycle] ) . Preferably, the patient has not received systemic treatment for extensive-stage small cell lung cancer prior to the administration, i.e. the treatment is a first-line treatment. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a first-line treatment for a patient suffering from extensive-stage small cell lung cancer. It is also preferred that the antibody is administered intravenously. The treatment may take place for 4 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 20 mg/kg or about 30 mg/kg every three weeks.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with limited-stage small cell lung cancer (LS-SCLC) , the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy (in particular with platinum-etoposide) , wherein the antibody is administered every three weeks at a dosage of about 20 mg/kg or about 30 mg/kg in combination with systemic chemotherapy (in particular with platinum [preferably carboplatin at a dosage of about AUC=5 (total dose ≤ about 750 mg) at day 1 of each cycle] and etoposide [preferably at a dosage of about 100 mg/m2 on days 1 to 3 of each cycle] ) . Preferably, the patient had treatment-free interval (TFI) ≤ 6 months since the last chemotherapy, chemoradiotherapy or radiotherapy. It is also preferred that the antibody is administered intravenously. The treatment may take place for 4 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 20 mg/kg or about 30 mg/kg every three weeks.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with extensive-stage small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy (in particular with platinum-etoposide) , e.g., wherein the antibody is administered every three weeks at a dosage of about 1400 mg or about 2000 mg in combination with systemic chemotherapy (in particular with platinum [preferably carboplatin at a dosage of about AUC=5 (total dose ≤ about 750 mg) at day 1 of each cycle] and etoposide [preferably at a dosage of about 100 mg/m2 on days 1 to 3 of each cycle] ) . Preferably, the patient has not received systemic treatment for extensive-stage small cell lung cancer prior to the administration, i.e. the treatment is a first-line treatment. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a first-line treatment for a patient suffering from extensive-stage small cell lung cancer. It is also preferred that the antibody is administered intravenously. The treatment may take place for 4 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 1400 mg or about 2000 mg every three weeks.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with triple negative breast cancer (TNBC) , the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy, wherein the systemic chemotherapy is nab-paclitaxel, which can preferably be administered at a dosage of about 100 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 15 mg/kg or about 20 mg/kg on the 1st and 15th day of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is paclitaxel, which can preferably be administered at a dosage of about 90 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 1400 mg or about 15 to about 20 mk/kg every two weeks of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is gemcitabine and carboplatin which can preferably be administered at a dosage of about 1000 mg/m2 and AUC 2 respectively, on the 1st and 8th day of a 21-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 1400 mg to about 2000 mg on the 1st day of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is eribulin, which can preferably be administered at a dosage of about 1.4 mg/m2 on the 1st and 8th day of a 21-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 1400 mg to about 2000 mg on the first day of a 21-days treatment cycle) . It is preferred that the antibody is administered intravenously.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with triple negative breast cancer (TNBC) , the method comprising administering to the subject the bispecific antibody in combination with systemic chemotherapy, wherein the systemic chemotherapy is nab-paclitaxel, which can preferably be administered at a dosage of about 100 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 1400 mg every two weeks of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is paclitaxel, which can preferably be administered at a dosage of about 90 mg/m2 on the 1st, 8th, and 15th day of a 28-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 1400 mg every two weeks of a 28-days treatment cycle) ; or wherein the systemic chemotherapy is gemcitabine and carboplatin which can preferably be administered at a dosage of about 1000 mg/m2 and AUC 2 respectively, on the 1st and 8th day of a 21-days treatment cycle (preferably in combination with the bispecific antibody at a dosage of about 2000 mg every three weeks, preferably on the 1st day of a 21-days treatment cycle) . It is preferred that the antibody is administered intravenously. It is further preferred that the patient suffering from TNBC has not received systemic treatment for TNBC prior to the administration, i.e., the treatment is a first-line treatment. The TNBC may be locally recurrent inoperable or metastatic TNBC. The present method is in particular applicable for a patient with TNBC whose PD-L1 status is considered negative based on current clinical practice and thus wherein the patient has a PD-L1 expression before the treatment that provides a score of < 10 when determined by the combined positive score (CPS) (or a score that is indicative of such PD-L1 expression when determined by another integrating scoring algorithm) .
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with non-small cell lung cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular carboplatin and pemetrexed) , wherein the antibody is preferably administered every three weeks at a dosage of about 30 mg/kg together with the chemotherapy (in particular carboplatin and pemetrexed at a dosage of about AUC 5 mg/mL/min carboplatin and about 500 mg/m2 pemetrexed) . Preferably, the patient progressed after first-line EGFR-TKI therapy, i.e. the patient failed first-line EGFR-TKI therapy. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a second-line treatment for a patient suffering from non-small cell lung cancer. Furthermore, it can be preferred that the patient suffers from a NSCLC with PD-L1 expressed, i.e. with a PD-L1 expression before the treatment that provides a score of > 1%when determined by the tumour proportion score (TPS) (or a score that is indicative of such PD-L1 expression when determined by another integrating scoring algorithm) . It is noted, however, that patients with a PD-L1 expression before the treatment that provides a score of < 1%when determined by the tumour proportion score (TPS) also respond to the method of treatment of the present paragraph such that also a patient suffering from NSCLC with a PD-L1 expression before the treatment that provides a score of < 1%when determined by the TPS (or a score that is indicative of such PD-L1 expression when determined by another integrating scoring algorithm) can effectively be treated.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with non-small cell lung cancer (NSCLC) , e.g., to a patient suffering from NSCLC as second-line treatment, e.g., to a patient suffering from second-line stage IV or recurrent NSCLC or, e.g., to a patient who progressed at least one prior line of an immune-oncology (IO) -comprising treatment (such as an immune checkpoint inhibitor-comprising treatment) , wherein the method comprises administering to the subject the bispecific antibody (preferably at a dosage range of about 1400 mg to about 2000 mg [with about 1400 mg or about 2000 mg being more preferred] every three weeks) and docetaxel (preferably at about 75 mg/m2 every three weeks) . For example, the bispecific antibody may be administered at a dosage of about 2000 mg bispecific antibody plus about 75 mg/m2 docetaxel every three weeks. In another example, the bispecific antibody may be administered in a step-up dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 1400 mg bispecific antibody plus about 60 mg/m2 docetaxel every three weeks or about 1400 mg bispecific antibody plus about 75 mg/m2 docetaxel every three weeks or about 2000 mg bispecific antibody plus about 60 mg/m2 docetaxel every three weeks and the second dose regimen is about 2000 mg bispecific antibody plus about 75 mg/m2 docetaxel every three weeks. In another example, the bispecific antibody may be administered in a step-down dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 2000 mg bispecific antibody plus about 75 mg/m2 docetaxel every three weeks and the second dose regimen is about 1400 mg bispecific antibody plus about 75 mg/m2 docetaxel every three weeks or about 2000 mg bispecific antibody plus about 60 mg/m2 docetaxel every three weeks.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with non-small cell lung cancer (NSCLC) , e.g., to a patient suffering from Stage IIIB/IIIC or IV NSCLC, wherein the method comprises administering to the subject the bispecific antibody (preferably at a dosage range of about 1400 mg to about 2000 mg [with about 1400 mg or about 2000 mg being more preferred] ) and (i) platinum (preferably carboplatin at a dosage of about AUC=5 with the total dose ≤ about 750 mg) in combination with pemetrexed (preferably at a dosage of about 500 mg/m2) or (ii) platinum (preferably carboplatin at a dosage of about AUC=5 with the total dose ≤ about 750 mg or more preferably carboplatin at a dosage of about AUC=6 with the total dose ≤ about 900 mg) in combination with paclitaxel (preferably at a dosage of about 200 mg/m2) , wherein the combination is preferably administered every three weeks, optionally for four cycles. Combination (i) may in particular be administered to a patient suffering from non-squamous NSCLC, whereas combination (ii) may in particular be administered to a patient suffering from squamous NSCLC, wherein the maintenance therapy may be at a dosage of about 1400 mg or about 2000 mg bispecific antibody plus optionally pemetrexed (preferably at a dosage of about 500 mg/m2) for patient (i) and a dosage of about 1400 mg or about 200 mg bispecific antibody for patient (ii) , wherein the administration every three weeks is preferably maintained during maintenance therapy.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with hepatocellular cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular FOLFOX4, i.e. oxaliplatin, leucovorin and 5-fluorouracil) , wherein the antibody is administered every two weeks at a dosage of about 20 mg/kg together with the chemotherapy. Preferably, the patient has not received systemic treatment for hepatocellular cancer prior to the administration, i.e. the treatment is a first-line treatment. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a first-line treatment for a patient suffering from hepatocellular cancer.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with hepatocellular cancer, the method comprising administering to the subject the bispecific antibody (preferably as the sole active agent) , wherein the antibody is administered every two weeks at a dosage of about 20 mg/kg. Preferably, the patient has not received systemic treatment for hepatocellular cancer prior to the administration, i.e. the treatment is a first-line treatment. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a first-line treatment for a patient suffering from hepatocellular cancer.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with malignant mesothelioma (preferably unresectable malignant mesothelioma) , the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular carboplatin and pemetrexed or cisplatin and pemetrexed) , wherein the antibody is preferably administered every three weeks at a dosage of about 30 mg/kg together with the chemotherapy (in particular carboplatin and pemetrexed at a dosage of about AUC 5 mg/mL/min carboplatin and about 500 mg/m2 pemetrexed or cisplatin and pemetrexed at a dosage of about 75 mg/m2 cisplatin and about 500 mg/m2 pemetrexed) . The malignant mesothelioma may be malignant pleural mesothelioma (MPM) or malignant peritoneal mesothelioma (MPeM) . Preferably, the patient has not received systemic treatment for malignant mesothelioma prior to the administration, i.e. the treatment is a first-line treatment. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a first-line treatment for a patient suffering from malignant mesothelioma. It is also preferred that the antibody is administered intravenously. The treatment may take place for 4 to 6 cycles (3 weeks/cycle) , followed by a maintenance therapy by the antibody at a dosage of about 30 mg/kg every three weeks.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with neuroendocrine neoplasms (NEN) , the method comprising administering to the subject the bispecific antibody in combination with chemotherapy (in particular FOLFIRI, i.e. irinotecan, leucovorin and 5-fluorouracil) , wherein the antibody is preferably administered every two weeks at a dosage of about 20 mg/kg together with the chemotherapy (in particular irinotecan at a dosage of about 180 mg/m2, leucovorin at a dosage of about 400 mg/m2, and 5-fluorouracil at a dosage of about 2800 mg/m2) . Preferably, the patient has received systemic treatment for NEN prior to the administration, i.e. the treatment is a second-line treatment. Accordingly, on a more general level, it is preferred that the method of the present paragraph is a second-line treatment for a patient suffering from NEN. It is also preferred that the antibody and the chemotherapy is administered intravenously.
In another aspect, the present application is directed to a binding agent, preferably a bispecific antibody that specifically binds to PD-L1 and VEGF (more preferably PM8002 comprising the heavy and light chain amino acid sequences as set forth in SEQ ID NOs: 16 and 17) for use in a method of treating a subject with colorectal cancer, the method comprising administering to the subject the bispecific antibody in combination with chemotherapy.
In another aspect, the present application is directed to Ivonescimab as the binding agent, for use in a method of treating a subject with non-small cell lung cancer (NSCLC) , e.g., to a patient suffering from NSCLC as second-line treatment, e.g., to a patient suffering from second-line stage IV or recurrent NSCLC or, e.g., to a patient who progressed at least one prior line of an immune-oncology (IO) -comprising treatment (such as an immune checkpoint inhibitor-comprising treatment) , wherein the method comprises administering to the subject Ivonescimab (preferably at a dosage range of about 1400 mg to about 2000 mg [with about 1400 mg or about 2000 mg being more preferred] every three weeks) and docetaxel (preferably at about 75 mg/m2 every three weeks) . For example, Ivonescimab may be administered at a dosage of about 2000 mg plus about 75 mg/m2 docetaxel every three weeks. In another example, Ivonescimab may be administered in a step-up dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 1400 mg Ivonescimab plus about 60 mg/m2 docetaxel every three weeks or about 1400 mg Ivonescimab plus about 75 mg/m2 docetaxel every three weeks or about 2000 mg Ivonescimab plus about 60 mg/m2 docetaxel every three weeks and the second dose regimen is about 2000 mg Ivonescimab plus about 75 mg/m2 docetaxel every three weeks. In another example, Ivonescimab may be administered in a step-down dosing regimen with a first and a second dose regimen, preferably wherein the first dose regimen is about 2000 mg Ivonescimab plus about 75 mg/m2 docetaxel every three weeks and the second dose regimen is about 1400 mg Ivonescimab plus about 75 mg/m2 docetaxel every three weeks or about 2000 mg Ivonescimab plus about 60 mg/m2 docetaxel every three weeks.
In the light of the above-stated further aspects of the combination, it is evident that the following combinations (preferably in the respective dosages as it can be derived from the above-stated further aspects of the combination and preferably administered as separate dosage forms) are particularly preferred: The binding agent, preferably the bispecific antibody that specifically binds to PD-L1 and VEGF (most preferably PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with (i) etoposide and carboplatin; (ii) paclitaxel; (iii) topotecan; (iv) Nab-paclitaxel; (v) paclitaxel; (vi) gemcitabine; (vii) eribulin; (viii) carboplatin and pemetrexed; (ix) carboplatin and paclitaxel; or (x) docetaxel.
The binding agent, i.e., the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may be administered in the form of any suitable pharmaceutical composition as described herein. In a preferred embodiment, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered in the form of an injection or infusion.
The anti-PD-1/PD-L1xVEGF/VEGFR binding agent can be administered prior to, simultaneously with, or after administration of the chemotherapy agent.
In one embodiment, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered prior to the administration of the chemotherapy agent.
In one embodiment, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered after the administration of the chemotherapy agent.
In one embodiment, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent is administered simultaneously with the chemotherapy agent. For example, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent and the chemotherapy agent may be administered using a composition comprising both drugs. Alternatively, the anti-PD-1/PD-L1xVEGF/VEGFR binding agent may be administered into one extremity of the subject, and the chemotherapy agent may be administered into another extremity of the subject.
Method of treatment
The invention further provides a method of treating cancer in a subject, the method comprising administering to the subject a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) , in combination with chemotherapy, optionally wherein the subject has a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ≥ 1 or other integrating scoring algorithm defining a similar PD-L1 expression score.
The embodiments disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in a method of treating cancer in a subject as disclosed herein. Features described herein in more detail for the “binding agent (including, e.g., the bispecific antibody) for use in a method of treating” embodiments equally apply to the method of treatment disclosed herein.
In some embodiments, the method is a method for extending progression-free survival in said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or an anti PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-L1 antibody is preferably pembrolizumab. In some embodiments, the method is a method for increased overall survival in said subject compared to the chemotherapy or the binding agent (including, e.g., the bispecific antibody) or an anti PD-L1 antibody or an anti-PD-1 antibody treatment alone or compared to a standard treatment comprising the chemotherapy and the anti-PD-L1 antibody or the chemotherapy and the anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-L1 antibody is preferably pembrolizumab.
In some preferred embodiments, the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably nab-paclitaxel;
wherein the subject has a PD-L1 expression score before the treatment similar or identical to a CPS of ≥1 
to <10 as determined by the TPS scoring algorithm.
In some preferred embodiments, the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably nab-paclitaxel;
wherein the subject has a PD-L1 expression score before the treatment similar or identical to a CPS of ≥1 
to <10 as determined by the TAP scoring algorithm.
In some particularly preferred embodiments, the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably nab-paclitaxel;
wherein the subject has a combined positive score (CPS) of ≥1 to <10 before the treatment. The CPS is 
preferably determined with Dako’s PD-L1 IHC 22C3 pharmDx kit (SK006) according to the manufacturer’s TNBC Instructions for Use.
In some preferred embodiments, the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably a chemotherapy agent;
wherein the subject has a PD-L1 expression score before the treatment as determined by a combined 
positive score (CPS) of ≥1 to <10 or another integrating scoring algorithm defining a similar PD-L1 expression score, wherein the other integrating scoring algorithm is selected from tumour area positivity (TAP) score and tumour proportion score (TPS) , preferably wherein the other integrating scoring algorithm is TAP score.
In some preferred embodiments, the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with triple-negative breast cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably a chemotherapy agent;
wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 to <10 when 
determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
In some preferred embodiments, the invention provides a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) for use in a method of treating a subject with cancer, the method comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy, preferably a chemotherapy agent;
optionally wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 
when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably when determined by the TAP score.
In some aspects of the invention, there is provided use of a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. the chemotherapy agent.
In some aspects of the invention, there is provided use of a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. a chemotherapy agent.
In some aspects of the invention, there is provided use of a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. the binding agent (including, e.g., the bispecific antibody) ; and
b. the chemotherapy agent.
In some aspects of the invention, there is provided use of a chemotherapy agent in the manufacture of a medicament for treating cancer in a subject, the use comprising administering to the subject:
a. a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and 
VEGF) ; and
b. the chemotherapy agent.
OTHER METHODS
The invention further provides a method for determining whether a cancer in a subject is susceptible to treatment with a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject a PD-L1 expression score before the treatment as determined by the combined positive score (CPS) of ≥1 or other integrating scoring algorithm defining a similar PD-L1 expression score, wherein the CPS of ≥ 1 indicates that the subject is susceptible to treatment with the binding agent (including, e.g., the bispecific antibody) and the chemotherapy.
The embodiments for determining PD-L1 expression score in cancer tissue disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in the method for determining whether a cancer in a subject is susceptible to treatment with a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy.
Features described herein in more detail for the “binding agent (including, e.g., the bispecific antibody) for use in a method of treating” embodiments equally apply to the determining method disclosed herein. This means that, if a specific cancer is concerned, the treatment for this specific cancer as disclosed above corresponds to the treatment, the subject is susceptible to. In a preferred embodiment, the cancer is triple negative breast cancer, which means that the method is for determining whether triple negative breast cancer in a subject is susceptible to treatment with a binding agent and a chemotherapy agent, wherein the method comprises the step as outlined above. The treatment, to which the subject is susceptible to, corresponds to the combination of a binding agent and a chemotherapy agent as outlined herein in the aspect relating to the treatment of triple negative breast cancer, i.e., the treatment is in particular the combination of a binding agent and a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
It is preferred that the method of treatment of a cancer (e.g., the method of treating triple negative breast cancer by a combination of a binding agent and a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline) is directly linked to and follows the method for determining whether a cancer (e.g., triple negative breast cancer) in a subject is susceptible to such treatment. In other words, the method determining whether a cancer in a subject is susceptible to such treatment may be regarded as a step of the corresponding method of treatment.
In some preferred embodiments, the method for determining whether a cancer in a subject is susceptible to treatment with a binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and a chemotherapy is conducted using the IHC assay PD-L1 IHC 22C3 pharmDx in accordance with the manufacturer’s guide.
KITS-OF-PARTS OF THE INVENTION
The invention further provides kit-of-parts comprising the binding agent (including, e.g., the bispecific antibody that specifically binds to PD-L1 and VEGF) and the chemotherapy agent disclosed herein.
The embodiments disclosed herein for the method of treatment comprising the binding agent (including, e.g., the bispecific antibody) in combination with the chemotherapy can be used in the kit-of-parts disclosed herein. Features described herein in more detail for the “binding agent (including, e.g., the bispecific antibody) for use in a method of treating” embodiments equally apply to the kit-of-parts disclosed herein.
In some embodiments, the binding agent (including, e.g., the bispecific antibody) and the chemotherapy agent are comprised in separate container.
In some embodiments, the kit-of parts further comprise instructions for use.
An exemplary kit or kit-of-parts of the present invention is a kit or kit-of-part comprising
a. a composition comprising a binding agent of the present invention; and
b. a composition comprising a chemotherapy agent,
wherein the chemotherapy agent may be selected from a group or wherein the chemotherapy agent may 
be defined as comprising a combination, e.g., a combination of carboplatin and pemetrexed. It is understood that, if more than one chemotherapy agent is comprised in the kit, composition b. may comprise both chemotherapy agents or composition b. comprises the first chemotherapy agent, whereas there is yet a further composition c. comprising the second chemotherapy agent. This applies equally if more than 2 chemotherapy agents are present.
In the light of the above, an exemplary kit or kit-of-parts of the present invention is a kit or kit-of-part comprising
a. a composition comprising a binding agent of the present invention; and
b. a composition comprising a first chemotherapy agent,
c. a composition comprising a second chemotherapy agent
wherein such a kit is suitable if the binding agent is administered in combination with two chemotherapy 
agents, which are comprised in separate compositions.
Accordingly, whenever reference is made in a kit or kit-of-parts to a composition comprising more than one chemotherapy agent, this is understood as covering a first scenario, where the more than one chemotherapy agents are comprised in a single composition, and a second scenario, where the more than one chemotherapy agents are each comprised in a compositions, such that not only two but more than two compositions are overall comprised in the kit or kit-of-parts. The second scenario is preferred in the present invention.
COMPOSITION
In some embodiments, the agents described herein (such as binding agent of the present invention, the chemotherapeutic agent, etc. ) may be present and/or administered (e.g., as part of a treatment method) as a composition, e.g., each of the agents described herein is contained in separate vials or containers of, e.g., a kit and/or each of the agents described herein is administered separately from the other agents (e.g., each of the agents described herein is administered in separate compositions) . In some embodiments, the agents described herein (such as binding agent of the present invention, the chemotherapeutic agent, etc. ) may be present and/or administered (e.g., as part of a treatment method) as a single composition, e.g., each of the agents described herein is contained in a single vial or container and/or each of the agents described herein are administered together. The composition may be a pharmaceutical composition.
The term "pharmaceutical composition" relates to a formulation comprising a therapeutically effective agent, preferably together with one or more pharmaceutically acceptable carriers, diluents and/or excipients.
The term "pharmaceutically acceptable" refers to the non-toxicity of a material which does not interact with the action of the active component of the pharmaceutical composition.
The pharmaceutical compositions according to the present disclosure may contain salts, buffers, stabilizers, preservatives, amino acids and/or surface-active agents. In one embodiment, the pharmaceutical compositions of the present disclosure comprise one or more pharmaceutically acceptable carriers, diluents, and/or excipients.
Pharmaceutically acceptable carriers, excipients, or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R Gennaro edit. 1985) .
In one embodiment, the (pharmaceutical) composition comprising the binding agent of the present invention comprises salt, buffers, amino acids, stabilizers, and surface-active agents. In one embodiment, the (pharmaceutical) composition comprising the binding agent of the present invention comprises no preservative. In a more specific embodiment, the (pharmaceutical) composition comprising the binding agent of the present invention comprises L-histidine, histidine hydrochloride, sucrose, disodium edetate, polysorbate 80 and water for injection. In a more specific embodiment, the (pharmaceutical) composition comprising the binding agent of the present invention is diluted with sodium chloride prior to administration.
CONJUGATES OF THE BISPECIFIC ANTIBODY
The bispecific antibody can be linked by various methods, see, for example, the method of Songsivilai et al. (Clin. Exp. Immunol., 79: 315-321 (1990) ) , and the method of Kostelny et al. (J. Immunol., 148: 1547-1553 (1992) which disclosures are also hereby incorporated in their entirety.
In some embodiments, the bispecific antibody is conjugated with a chemotherapeutical agent to obtain an immunoconjugate. In some embodiments, the immunoconjugate contains:
a. a bispecific antibody as disclosed herein; and
b. a conjugation moiety selected from the group consisting of detectable labels, drugs, toxins, 
cytokines, radionuclides, or enzymes, gold nanoparticles/nanorods, nanomagnetic particles, viral coat proteins or VLPs, or their combination.
In some embodiments, the radionuclide includes:
a. a diagnostic isotope selected from the group consisting of Tc-99m, Ga-68, F-18, I-123, I-
125, I-131, In-111, Ga-67, Cu-64, Zr-89, C-11, Lu-177, Re-188, or a combination thereof; and/or
b. a therapeutic isotope selected from the group consisting of Lu-177, Y-90, Ac-225, As-211, 
Bi-212, Bi-213, Cs-137, Cr-51, Co-60, Dy-165, Er-169, Fm-255, Au-198, Ho-166, I-125, I-131, Ir-192, Fe-59, Pb-212, Mo-99, Pd-103, P-32, K-42, Re-186, Re-188, Sm-153, Ra223, Ru-106, Na24, Sr89, Tb-149, Th-227, Xe-133Yb-169, Yb-177, or a combination thereof.
In some embodiments, the coupling moiety is a drug or a toxin. In another preferred embodiment, the drug is a cytotoxic drug. In some embodiments, the cytotoxic drugs are selected from the group consisting of anti-tubulin drugs, DNA minor groove binding reagents, DNA replication inhibitors, alkylating reagents, antibiotics, folic acid antagonists, antimetabolites, chemotherapy A sensitizer, a topoisomerase inhibitor, a vinca alkaloid, or a combination thereof. Examples of particularly useful cytotoxic drugs include, for example, DNA minor groove binding agents, DNA alkylating agents, and tubulin inhibitors. Typical cytotoxic drugs include, for example, auristatins, camptothecins, duocarmycins, etoposides, maytansines and maytansinoids (eg DM1 and DM4) , taxanes (taxanes) , benzodiazepines, or benzodiazepine containing drugs (eg, pyrrolo [1, 4] benzodiazepines (PBDs) , indoline benzodiazepines indolinobenzodiazepines and oxazolidinobenzodiazepines) , vinca alkaloids, or combinations thereof.
In some embodiments, the toxin is selected from the following group: Auristatins (e.g., auristatin E, auristatin F, MMAE, and MMAF) , chlortetracycline, maytansoid, gamatoxin, gamatoxin A-chain, combretastatin, docarmicin, Lastatin, doxorubicin, daunorubicin, paclitaxel, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, autumn Narcissin, Dihydroxyanthraxdione, Actinomycin, Diphtheria Toxin, Pseudomonas Exotoxin (PE) A, PE40, Acacia toxin, Acacia A chain, Capsule root toxin A chain, α -Sarcinus, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotontoxin, calicheamicin, Sapaonaria officinalis inhibitors, glucocorticoids, or a combination thereof.
NUMBERED EMBODIMENTS
The present invention also relates to the following embodiments:
1. A method of treating small cell lung cancer in a subject, the method comprising administering to 
the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent.
2. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating small cell lung cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating small cell lung cancer, the method 
comprising administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. the chemotherapy agent.
4. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use in a method of treating small cell lung cancer in a subject, the method comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is paclitaxel.
6. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the paclitaxel is administered at a dosage of about 175 mg/m2, preferably intravenously every three weeks at day 1.
7. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is topotecan.
8. The method according to embodiment 7, the bispecific antibody for use according to embodiment 
7, the chemotherapy agent for use according to embodiment 7, or the bispecific antibody and the chemotherapy agent for use according to embodiment 7, wherein the topotecan is administered at a dosage of about 1.5 mg/m2 intravenously or about 2.3 mg/m2 orally, preferably every three weeks at days 1 to 5.
9. The method according to any one of embodiments 5 to 8, the bispecific antibody for use according 
to any one of embodiments 5 to 8, the chemotherapy agent for use according to any one of embodiments 5 to 8, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 8, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg or about 30 mg/kg, preferably intravenously every three weeks at day 1.
10. The method according to any one of embodiments 5 to 9, the bispecific antibody for use according 
to any one of embodiments 5 to 9, the chemotherapy agent for use according to any one of embodiments 5 to 9, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 9, wherein the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line platinum-based chemotherapy with or without immuno-oncology treatment.
11. The method according to any one of embodiments 5 to 9, the bispecific antibody for use according 
to any one of embodiments 5 to 9, the chemotherapy agent for use according to any one of embodiments 5 to 9, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 9, wherein the treatment is a third-line treatment, preferably wherein the cancer progressed during or after first-line and second-line platinum-based chemotherapy with or without immuno-oncology treatment.
12. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the small cell lung cancer is limited-stage small cell lung cancer.
13. The method according to embodiment 12, the bispecific antibody for use according to embodiment 
12, the chemotherapy agent for use according to embodiment 12, or the bispecific antibody and the chemotherapy agent for use according to embodiment 12, wherein the chemotherapy agent is carboplatin and etoposide.
14. The method according to embodiment 12 or 13, the bispecific antibody for use according to 
embodiment 12 or 13, the chemotherapy agent for use according to embodiment 12 or 13, or the bispecific antibody and the chemotherapy agent for use according to embodiment 12 or 13, wherein the carboplatin is administered at a dosage of about AUC=5 and the etoposide is administered at a dosage of about 100 mg/m2, preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and etoposide is administered every three weeks at days 1 to 3.
15. The method according to any one of embodiments 12 to 14, the bispecific antibody for use 
according to any one of embodiments 12 to 14, the chemotherapy agent for use according to any one of embodiments 12 to 14, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 12 to 14, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg or about 30 mg/kg, preferably intravenously every three weeks at day 1.
16. The method according to any one of embodiments 12 to 15, the bispecific antibody for use 
according to any one of embodiments 12 to 15, the chemotherapy agent for use according to any one of embodiments 12 to 15, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 12 to 15, wherein the subject had a treatment-free interval of about ≤ 6 months, preferably since the last chemotherapy, chemoradiotherapy or radiotherapy.
17. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the small cell lung cancer is extensive-stage small cell lung cancer.
18. The method according to embodiment 17, the bispecific antibody for use according to embodiment 
17, the chemotherapy agent for use according to embodiment 17, or the bispecific antibody and the chemotherapy agent for use according to embodiment 17, wherein the chemotherapy agent is carboplatin and etoposide.
19. The method according to embodiment 17 or 18, the bispecific antibody for use according to 
embodiment 17 or 18, the chemotherapy agent for use according to embodiment 17 or 18, or the bispecific antibody and the chemotherapy agent for use according to embodiment 17 or 18, wherein the carboplatin is administered at a dosage of about AUC=5 and the etoposide is administered at a dosage of about 100 mg/m2, preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and etoposide is administered every three weeks at days 1 to 3.
20. The method according to any one of embodiments 17 to 19, the bispecific antibody for use 
according to any one of embodiments 17 to 19, the chemotherapy agent for use according to any one of embodiments 17 to 19, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 17 to 19, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg or about 30 mg/kg, preferably intravenously every three weeks at day 1.
21. The method according to any one of embodiments 17 to 19, the bispecific antibody for use 
according to any one of embodiments 17 to 19, the chemotherapy agent for use according to any one of embodiments 17 to 19, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 17 to 19, wherein the bispecific antibody is administered at a dosage of about 1400 mg or about 2000 mg, preferably intravenously every three weeks at day 1.
22. The method according to any one of embodiments 17 to 21, the bispecific antibody for use 
according to any one of embodiments 17 to 21, the chemotherapy agent for use according to any one of embodiments 17 to 21, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 17 to 21, wherein the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for extensive-stage small cell lung cancer.
23. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of paclitaxel, topotecan, and a combination of carboplatin and etoposide.
24. A composition comprising
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent selected from the group consisting of paclitaxel, topotecan, and a 
combination of carboplatin and etoposide.
25. A kit comprising
a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a 
heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent selected from the group consisting of 
paclitaxel, topotecan, and a combination of carboplatin and etoposide.
26. The composition according to embodiment 24, or the kit according to embodiment 25 for use as 
medicament.
The present invention also relates to the following embodiments:
1. A method of treating small cell lung cancer in a subject, the method comprising administering to 
the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating small cell lung cancer, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating small cell lung cancer, the method 
comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating small cell lung cancer in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
6. The method according to embodiment 5, the binding agent for use according to embodiment 5, the 
chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the taxane is paclitaxel, preferably wherein the paclitaxel is administered intravenously at a dosage of about 175 mg/m2 every three weeks at day 1.
7. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a topoisomerase inhibitor, preferably topotecan or etoposide.
8. The method according to embodiment 7, the binding agent for use according to embodiment 7, the 
chemotherapy agent for use according to embodiment 7, or the binding agent and the chemotherapy agent for use according to embodiment 7, wherein the topoisomerase inhibitor is topotecan, preferably wherein the topotecan is administered intravenously at a dosage of about 1.5 mg/m2 every three weeks at days 1 to 5 or orally at a dosage of about 2.3 mg/m2 every three weeks at days 1 to 5.
9. The method according to any one of embodiments 5 to 8, the binding agent for use according to 
any one of embodiments 5 to 8, the chemotherapy agent for use according to any one of embodiments 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 8, wherein the binding agent is administered at a dosage of about 10 mg/kg to about 100 mg/kg, preferably intravenously every three weeks at day 1.
10. The method according to any one of embodiments 5 to 9, the binding agent for use according to 
any one of embodiments 5 to 9, the chemotherapy agent for use according to any one of embodiments 5 to 9, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 9, wherein the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line platinum-based chemotherapy with or without immuno-oncology treatment.
11. The method according to any one of embodiments 5 to 9, the binding agent for use according to 
any one of embodiments 5 to 9, the chemotherapy agent for use according to any one of embodiments 5 to 9, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 9, wherein the treatment is a third-line treatment, preferably wherein the cancer progressed during or after first-line and second-line platinum-based chemotherapy with or without immuno-oncology treatment.
12. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the small cell lung cancer is limited-stage small cell lung cancer.
13. The method according to embodiment 12, the binding agent for use according to embodiment 12, 
the chemotherapy agent for use according to embodiment 12, or the binding agent and the chemotherapy agent for use according to embodiment 12, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
14. The method according to embodiment 12 or 13, the binding agent for use according to embodiment 
12 or 13, the chemotherapy agent for use according to embodiment 12 or 13, or the binding agent and the chemotherapy agent for use according to embodiment 12 or 13, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=5 every three weeks at day 1, and the topoisomerase inhibitor is etoposide, preferably wherein the etoposide is administered at a dosage of about 100 mg/m2 every three weeks at days 1 to 3.
15. The method according to any one of embodiments 12 to 14, the binding agent for use according to 
any one of embodiments 12 to 14, the chemotherapy agent for use according to any one of embodiments 12 to 14, or the binding agent and the chemotherapy agent for use according to any one of embodiments 12 to 14, wherein the binding agent is administered at a dosage of from about 10 mg/kg to 100 mg/kg, preferably intravenously every three weeks at day 1.
16. The method according to any one of embodiments 12 to 15, the binding agent for use according to 
any one of embodiments 12 to 15, the chemotherapy agent for use according to any one of embodiments 12 to 15, or the binding agent and the chemotherapy agent for use according to any one of embodiments 12 to 15, wherein the subject had a treatment-free interval of about ≤ 6 months, preferably since the last chemotherapy, chemoradiotherapy or radiotherapy.
17. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the small cell lung cancer is extensive-stage small cell lung cancer.
18. The method according to embodiment 17, the binding agent for use according to embodiment 17, 
the chemotherapy agent for use according to embodiment 17, or the binding agent and the chemotherapy agent for use according to embodiment 17, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
19. The method according to embodiment 17 or 18, the binding agent for use according to embodiment 
17 or 18, the chemotherapy agent for use according to embodiment 17 or 18, or the binding agent and the chemotherapy agent for use according to embodiment 17 or 18, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=5 every three weeks at day 1, and the topoisomerase inhibitor is etoposide, preferably wherein the etoposide is administered at a dosage of about 100 mg/m2 every three weeks at days 1 to 3.
20. The method according to any one of embodiments 17 to 19, the binding agent for use according to 
any one of embodiments 17 to 19, the chemotherapy agent for use according to any one of embodiments 17 to 19, or the binding agent and the chemotherapy agent for use according to any one of embodiments 17 to 19, wherein the binding agent is administered at a dosage of from about 10 mg/kg to about 100 mg/kg, preferably intravenously every three weeks at day 1.
21. The method according to any one of embodiments 17 to 19, the binding agent for use according to 
any one of embodiments 17 to 19, the chemotherapy agent for use according to any one of embodiments 17 to 19, or the binding agent and the chemotherapy agent for use according to any one of embodiments 17 to 19, wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every three weeks at day 1.
22. The method according to any one of embodiments 17 to 21, the binding agent for use according to 
any one of embodiments 17 to 21, the chemotherapy agent for use according to any one of embodiments 17 to 21, or the binding agent and the chemotherapy agent for use according to any one of embodiments 17 to 21, wherein the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for extensive-stage small cell lung cancer.
23. The method according to any one of embodiments 1 and 5 to 22, the binding agent for use 
according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
24. The method according to embodiment 23, the binding agent for use according to embodiment 23, 
the chemotherapy agent for use according to embodiment 23, or the binding agent and the chemotherapy agent for use according to embodiment 23, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
25. The method according to any one of embodiments 1 and 5 to 22, the binding agent for use 
according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
26. The method according to embodiment 25, the binding agent for use according to embodiment 25, 
the chemotherapy agent for use according to embodiment 25, or the binding agent and the chemotherapy agent for use according to embodiment 25, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
27. The method according to any one of embodiments 1 and 5 to 22, the binding agent for use 
according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
28. A binding agent binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid 
sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
29. A composition comprising
a. a binding agent binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent selected from the group consisting of (i) a taxane, preferably 
paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
30. A kit comprising
a. a composition comprising a binding agent binding to PD-L1 and VEGF comprising a heavy 
chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent selected from the group consisting of (i) 
a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a topoisomerase inhibitor, preferably topotecan or etoposide; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a topoisomerase inhibitor, preferably topotecan or etoposide.
31. The binding agent and the chemotherapy agent for use according to embodiment 28, the 
composition according to embodiment 29, or the kit according to embodiment 30, wherein the chemotherapy agent is selected from the group consisting of (i) paclitaxel, (ii) topotecan, and (iii) a combination of carboplatin and etoposide.
32. The binding agent and the chemotherapy agent for use according to embodiment 28 or 31, the 
composition according to embodiment 29 or 31, or the kit according to embodiment 30 or 31, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
33. The binding agent and the chemotherapy agent for use according to embodiment 32, the 
composition according to embodiment 32, or the kit according to embodiment 32, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
34. The binding agent and the chemotherapy agent for use according to embodiment 28 or 31, the 
composition according to embodiment 29 or 31, or the kit according to embodiment 30 or 31, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
35. The binding agent and the chemotherapy agent for use according to embodiment 34, the 
composition according to embodiment 34, or the kit according to embodiment 34, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
36. The binding agent and the chemotherapy agent for use according to embodiment 28 or 31, the 
composition according to embodiment 29 or 31, or the kit according to embodiment 30 or 31, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
37. The composition according to any one of embodiments 29 and 31 to 36, or the kit according to any 
one of embodiments 30 to 36 for use as medicament.
The present invention also relates to the following embodiments:
1. A method of treating hepatocellular cancer in a subject, the method comprising administering to 
the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent.
2. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating hepatocellular cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating hepatocellular cancer, the method 
comprising administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. the chemotherapy agent.
4. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use in a method of treating hepatocellular cancer in a subject, the method comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of oxaliplatin and 5-fluorouracil, optionally administered together with leucovorin.
6. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the oxaliplatin is administered at a dosage of about 85 mg/m2, the leucovorin is administered at a dosage of about 200 mg/m2, and the 5-fluorouracil is administered at a dosage of about 1000 mg/m2, preferably wherein oxaliplatin, leucovorin, and 5-fluorouracil are administered intravenously every two weeks at day 1.
7. The method according to embodiment 5 or 6, the bispecific antibody for use according to 
embodiment 5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5 or 6, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg, preferably intravenously every two weeks at day 1.
8. The method according to any one of embodiments 5 to 7, the bispecific antibody for use according 
to any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for hepatocellular cancer.
9. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of oxaliplatin and 5-fluorouracil, optionally in further combination with leucovorin.
10. A composition comprising
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent being a combination of oxaliplatin and 5-fluorouracil, optionally in 
further combination with leucovorin.
11. A kit comprising
a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a 
heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent being a combination of oxaliplatin and 5-
fluorouracil, optionally in further combination with leucovorin.
12. The composition according to embodiment 10, or the kit according to embodiment 11 for use as 
medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating hepatocellular cancer in a subject, the method comprising administering to 
the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating hepatocellular cancer, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating hepatocellular cancer, the method 
comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating hepatocellular cancer in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally administered together with folic acid or a derivative thereof, preferably leucovorin.
6. The method according to embodiment 5, the binding agent for use according to embodiment 5, the 
chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the platinum-based chemotherapy agent is oxaliplatin, the folic acid or derivative thereof is leucovorin, and the antimetabolite chemotherapy agent is 5-fluorouracil, preferably wherein the oxaliplatin is administered at a dosage of about 85 mg/m2, the leucovorin is administered at a dosage of about 200 mg/m2, and the 5-fluorouracil is administered at a dosage of about 1000 mg/m2, wherein oxaliplatin, leucovorin, and 5-fluorouracil are administered intravenously every two weeks at day 1.
7. The method according to embodiment 5 or 6, the binding agent for use according to embodiment 
5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the binding agent and the chemotherapy agent for use according to embodiment 5 or 6, wherein the binding agent is administered at a dosage of from 10 mg/kg to about 100 mg/kg, preferably intravenously every two weeks at day 1.
8. The method according to any one of embodiments 5 to 7, the binding agent for use according to 
any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for hepatocellular cancer.
9. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
10. The method according to embodiment 9, the binding agent for use according to embodiment 9, the 
chemotherapy agent for use according to embodiment 9, or the binding agent and the chemotherapy agent for use according to embodiment 9, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
11. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
12. The method according to embodiment 11, the binding agent for use according to embodiment 11, 
the chemotherapy agent for use according to embodiment 11, or the binding agent and the chemotherapy agent for use according to embodiment 11, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
13. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
14. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
15. A composition comprising
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent being a combination of a platinum-based chemotherapy agent, 
preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
16. A kit comprising
a. a composition comprising a binding agent comprising (i) a first binding region which binds 
to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a composition comprising a chemotherapy agent being a combination of a platinum-based 
chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
17. The binding agent and the chemotherapy agent for use according to embodiment 14, the 
composition according to embodiment 15, or the kit according to embodiment 16, wherein the chemotherapy agent is a combination of oxaliplatin and 5-fluorouracil, optionally further comprising leucovorin.
18. The binding agent and the chemotherapy agent for use according to embodiment 14 or 17, the 
composition according to embodiment 15 or 17, or the kit according to embodiment 16 or 17, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
19. The binding agent and the chemotherapy agent for use according to embodiment 18, the 
composition according to embodiment 18, or the kit according to embodiment 18, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
20. The binding agent and the chemotherapy agent for use according to embodiment 14 or 17, the 
composition according to embodiment 15 or 17, or the kit according to embodiment 16 or 17, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
21. The binding agent and the chemotherapy agent for use according to embodiment 20, the 
composition according to embodiment 20, or the kit according to embodiment 20, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
22. The binding agent and the chemotherapy agent for use according to embodiment 14 or 17, the 
composition according to embodiment 15 or 17, or the kit according to embodiment 16 or 17, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
23. The composition according to any one of embodiments 15 and 17 to 22, or the kit according to any 
one of embodiments 16 to 22 for use as medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating malignant mesothelioma in a subject, the method comprising administering to 
the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent.
2. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating malignant mesothelioma, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating malignant mesothelioma, the method 
comprising administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. the chemotherapy agent.
4. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use in a method of treating malignant mesothelioma in a subject, the method comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of carboplatin and pemetrexed or cisplatin and pemetrexed.
6. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the carboplatin is administered at a dosage of about AUC=5 and the pemetrexed is administered at a dosage of about 500 mg/m2, preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1; or wherein the cisplatin is administered at a dose of about 75 mg/m2 and the pemetrexed is administered at a dosage of about 500 mg/m2, preferably wherein the cisplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1.
7. The method according to embodiment 5 or 6, the bispecific antibody for use according to 
embodiment 5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5 or 6, wherein the bispecific antibody is administered at a dosage of about 30 mg/kg, preferably intravenously every three weeks at day 1.
8. The method according to any one of embodiments 1 and 5 to 7, the bispecific antibody for use 
according to any one of embodiments 2 and 5 to 7, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 7, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 4 to 7, wherein the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for malignant mesothelioma.
9. The method according to any one of embodiments 1 and 5 to 8, the bispecific antibody for use 
according to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the malignant mesothelioma is malignant pleural mesothelioma (MPM) or malignant peritoneal mesothelioma (MPeM) , preferably wherein the malignant mesothelioma is unresectable.
10. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of carboplatin and pemetrexed or cisplatin and pemetrexed.
11. A composition comprising
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent being a combination of carboplatin and pemetrexed or cisplatin and 
pemetrexed.
12. A kit comprising
a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a 
heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent being a combination of carboplatin and 
pemetrexed or cisplatin and pemetrexed.
13. The composition according to embodiment 11, or the kit according to embodiment 12 for use as 
medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating malignant mesothelioma in a subject, the method comprising administering to 
the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating malignant mesothelioma, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating malignant mesothelioma, the method 
comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating malignant mesothelioma in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (ii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate.
6. The method according to embodiment 5, the binding agent for use according to embodiment 5, the 
chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the platinum-based chemotherapy agent is carboplatin or cisplatin and the antifolate chemotherapy agent is pemetrexed, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=5 every three weeks at day 1 and the pemetrexed is administered intravenously at a dosage of about 500 mg/m2 every three weeks at day 1 or wherein the cisplatin is administered intravenously at a dosage of about 75 mg/m2 every three weeks at day 1 and the pemetrexed is administered intravenously at a dosage of about 500 mg/m2 every three weeks at day 1.
7. The method according to embodiment 5 or 6, the binding agent for use according to embodiment 
5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the binding agent and the chemotherapy agent for use according to embodiment 5 or 6, wherein the binding agent is administered at a dosage of from about 10 mg/kg to about 100 mg/kg, preferably intravenously every three weeks at day 1.
8. The method according to any one of embodiments 1 and 5 to 7, the binding agent for use according 
to any one of embodiments 2 and 5 to 7, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 7, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 7, wherein the treatment is a first-line treatment, preferably wherein the subject has not received any prior systemic treatment for malignant mesothelioma.
9. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the malignant mesothelioma is malignant pleural mesothelioma (MPM) or malignant peritoneal mesothelioma (MPeM) , preferably wherein the malignant mesothelioma is unresectable.
10. The method according to any one of embodiments 1 and 5 to 9, the binding agent for use according 
to any one of embodiments 2 and 5 to 9, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 9, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 9, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
11. The method according to embodiment 10, the binding agent for use according to embodiment 10, 
the chemotherapy agent for use according to embodiment 10, or the binding agent and the chemotherapy agent for use according to embodiment 10, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
12. The method according to any one of embodiments 1 and 5 to 9, the binding agent for use according 
to any one of embodiments 2 and 5 to 9, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 9, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 9, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
13. The method according to embodiment 12, the binding agent for use according to embodiment 12, 
the chemotherapy agent for use according to embodiment 12, or the binding agent and the chemotherapy agent for use according to embodiment 12, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
14. The method according to any one of embodiments 1 and 5 to 9, the binding agent for use according 
to any one of embodiments 2 and 5 to 9, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 9, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 9, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
15. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (ii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate.
16. A composition comprising
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent being a combination of (i) a platinum-based chemotherapy agent, 
preferably carboplatin, cisplatin or oxaliplatin; and (ii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate.
17. A kit comprising
a. a composition comprising a binding agent comprising (i) a first binding region which binds 
to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a composition comprising a chemotherapy agent being a combination of (i) a platinum-
based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (ii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate.
18. The binding agent and the chemotherapy agent for use according to embodiment 15, the 
composition according to embodiment 16, or the kit according to embodiment 17, wherein the chemotherapy agent is a combination of carboplatin and pemetrexed or a combination of cisplatin and pemetrexed.
19. The binding agent and the chemotherapy agent for use according to embodiment 15 or 18, the 
composition according to embodiment 16 or 18, or the kit according to embodiment 17 or 18, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
20. The binding agent and the chemotherapy agent for use according to embodiment 19, the 
composition according to embodiment 19, or the kit according to embodiment 19, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
21. The binding agent and the chemotherapy agent for use according to embodiment 15 or 18, the 
composition according to embodiment 16 or 18, or the kit according to embodiment 17 or 18, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
22. The binding agent and the chemotherapy agent for use according to embodiment 21, the 
composition according to embodiment 21, or the kit according to embodiment 21, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
23. The binding agent and the chemotherapy agent for use according to embodiment 15 or 18, the 
composition according to embodiment 16 or 18, or the kit according to embodiment 17 or 18, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
24. The composition according to any one of embodiments 16 and 18 to 23, or the kit according to any 
one of embodiments 17 to 23 for use as medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating neuroendocrine neoplasms in a subject, the method comprising administering 
to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent.
2. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating neuroendocrine neoplasms, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating neuroendocrine neoplasms, the method 
comprising administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. the chemotherapy agent
4. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17, and a chemotherapy agent for use in a method of treating neuroendocrine neoplasms in a subject, the method comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of irinotecan and 5-fluorouracil, optionally administered together with leucovorin.
6. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the irinotecan is administered at a dosage of about 180 mg/m2, the leucovorin is administered at a dosage of about 400 mg/m2, and the 5-fluorouracil is administered at a dosage of about 2800 mg/m2, preferably wherein irinotecan, leucovorin, and 5-fluorouracil are administered intravenously every two weeks at day 1.
7. The method according to embodiment 5 or 6, the bispecific antibody for use according to 
embodiment 5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5 or 6, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg, preferably intravenously every two weeks at day 1.
8. The method according to any one of embodiments 5 to 7, the bispecific antibody for use according 
to any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a second-line treatment, preferably wherein the subject has received prior systemic treatment for neuroendocrine neoplasms.
9. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of irinotecan and 5-fluorouracil, optionally in further combination with leucovorin.
10. A composition comprising
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent being a combination of irinotecan and 5-fluorouracil, optionally in 
further combination with leucovorin.
11. A kit comprising
a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a 
heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent being a combination of irinotecan and 5-
fluorouracil, optionally in further combination with leucovorin.
12. The composition according to embodiment 10, or the kit according to embodiment 11 for use as 
medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating neuroendocrine neoplasms in a subject, the method comprising administering 
to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating neuroendocrine neoplasms, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating neuroendocrine neoplasms, the method 
comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating neuroendocrine neoplasms in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally administered together with folic acid or a derivative thereof, preferably leucovorin.
6. The method according to embodiment 5, the binding agent for use according to embodiment 5, the 
chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the topoisomerase inhibitor is irinotecan, , the folic acid or derivative thereof is leucovorin, and the antimetabolite chemotherapy agent is 5-fluorouracil, preferably wherein the irinotecan is administered at a dosage of about 180 mg/m2, the leucovorin is administered at a dosage of about 400 mg/m2, and the 5-fluorouracil is administered at a dosage of about 2800 mg/m2, wherein irinotecan, leucovorin, and 5-fluorouracil are administered intravenously every two weeks at day 1.
7. The method according to embodiment 5 or 6, the binding agent for use according to embodiment 
5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the binding agent and the chemotherapy agent for use according to embodiment 5 or 6, wherein the binding agent is administered at a dosage of from 10 mg/kg to about 100 mg/kg, preferably intravenously every two weeks at day 1.
8. The method according to any one of embodiments 5 to 7, the binding agent for use according to 
any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a second-line treatment, preferably wherein the subject has received prior systemic treatment for neuroendocrine neoplasms.
9. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
10. The method according to embodiment 9, the binding agent for use according to embodiment 9, the 
chemotherapy agent for use according to embodiment 9, or the binding agent and the chemotherapy agent for use according to embodiment 9, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
11. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
12. The method according to embodiment 11, the binding agent for use according to embodiment 11, 
the chemotherapy agent for use according to embodiment 11, or the binding agent and the chemotherapy agent for use according to embodiment 11, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
13. The method according to any one of embodiments 1 and 5 to 8, the binding agent for use according 
to any one of embodiments 2 and 5 to 8, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 8, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
14. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is a combination of a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
15. A composition comprising
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent being a combination of a topoisomerase inhibitor, preferably an 
anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
16. A kit comprising
a. a composition comprising a binding agent comprising (i) a first binding region which binds 
to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a composition comprising a chemotherapy agent being a combination of a topoisomerase 
inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide, and an antimetabolite chemotherapy agent, preferably 5-fluorouracil or gemcitabine, optionally further comprising folic acid or a derivative thereof, preferably leucovorin.
17. The binding agent and the chemotherapy agent for use according to embodiment 14, the 
composition according to embodiment 15, or the kit according to embodiment 16, wherein the chemotherapy agent is a combination of irinotecan and 5-fluorouracil, optionally further comprising leucovorin.
18. The binding agent and the chemotherapy agent for use according to embodiment 14 or 17, the 
composition according to embodiment 15 or 17, or the kit according to embodiment 16 or 17, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
19. The binding agent and the chemotherapy agent for use according to embodiment 18, the 
composition according to embodiment 18, or the kit according to embodiment 18, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
20. The binding agent and the chemotherapy agent for use according to embodiment 14 or 17, the 
composition according to embodiment 15 or 17, or the kit according to embodiment 16 or 17, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
21. The binding agent and the chemotherapy agent for use according to embodiment 20, the 
composition according to embodiment 20, or the kit according to embodiment 20, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
22. The binding agent and the chemotherapy agent for use according to embodiment 14 or 17, the 
composition according to embodiment 15 or 17, or the kit according to embodiment 16 or 17, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
23. The composition according to any one of embodiments 15 and 17 to 22, or the kit according to any 
one of embodiments 16 to 22 for use as medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating non-small cell lung cancer in a subject, the method comprising administering 
to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent.
2. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating non-small cell lung cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating non-small cell lung cancer, the method 
comprising administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. the chemotherapy agent.
4. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use in a method of treating non-small cell lung cancer in a subject, the method comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is docetaxel.
6. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the docetaxel is administered at a dosage of about 75 mg/m2, preferably intravenously every three weeks at day 1.
7. The method according to embodiment 5 or 6, the bispecific antibody for use according to 
embodiment 5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5 or 6, wherein the bispecific antibody is administered at a dosage of about 2000 mg or 1400 mg, preferably intravenously every three weeks at day 1.
8. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the docetaxel is administered at a dosage of about 60 mg/m2, preferably intravenously every three weeks at day 1.
9. The method according to embodiment 8, the bispecific antibody for use according to embodiment 
8, the chemotherapy agent for use according to embodiment 8, or the bispecific antibody and the chemotherapy agent for use according to embodiment 8, wherein the bispecific antibody is administered at a dosage of about 2000 mg, preferably intravenously every three weeks at day 1.
10. The method according to any one of embodiments 5 to 9, the bispecific antibody for use according 
to any one of embodiments 5 to 9, the chemotherapy agent for use according to any one of embodiments 5 to 9, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 9, wherein the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line chemotherapy with immuno-oncology treatment.
11. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of carboplatin and pemetrexed.
12. The method according to embodiment 11, the bispecific antibody for use according to embodiment 
11, the chemotherapy agent for use according to embodiment 11, or the bispecific antibody and the chemotherapy agent for use according to embodiment 11, wherein the carboplatin is administered at a dosage of about AUC=5 and the pemetrexed is administered at a dosage of about 500 mg/m2, preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1.
13. The method according to embodiment 11 or 12, the bispecific antibody for use according to 
embodiment 11 or 12, the chemotherapy agent for use according to embodiment 11 or 12, or the bispecific antibody and the chemotherapy agent for use according to embodiment 11 or 12, wherein the bispecific antibody is administered at a dosage of about 30 mg/kg, preferably intravenously every three weeks at day 1.
14. The method according to any one of embodiments 11 to 13, the bispecific antibody for use 
according to any one of embodiments 11 to 13, the chemotherapy agent for use according to any one of embodiments 11 to 13, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 11 to 13, wherein the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line treatment with an EGFR tyrosine kinase-inhibitor.
15. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the non-small cell lung cancer is non-squamous non-small cell lung cancer, preferably non-squamous non-small cell lung cancer of Stage IIIB/IIIC or IV.
16. The method according to embodiment 15, the bispecific antibody for use according to embodiment 
15, the chemotherapy agent for use according to embodiment 15, or the bispecific antibody and the chemotherapy agent for use according to embodiment 15, wherein the chemotherapy agent is a combination of carboplatin and pemetrexed.
17. The method according to embodiment 16, the bispecific antibody for use according to embodiment 
16, the chemotherapy agent for use according to embodiment 16, or the bispecific antibody and the chemotherapy agent for use according to embodiment 16, wherein the carboplatin is administered at a dosage of about AUC=5 and the pemetrexed is administered at a dosage of about 500 mg/m2, preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and pemetrexed is administered intravenously every three weeks at day 1.
18. The method according to any one of embodiments 15 to 17, the bispecific antibody for use 
according to any one of embodiments 15 to 17, the chemotherapy agent for use according to any one of embodiments 15 to 17, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 15 to 17, wherein the bispecific antibody is administered at a dosage of about 2000 mg or 1400 mg, preferably intravenously every three weeks at day 1.
19. The method according to any one of embodiments 15 to 18, the bispecific antibody for use 
according to any one of embodiments 15 to 18, the chemotherapy agent for use according to any one of embodiments 15 to 18, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 15 to 18, wherein the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for non-small cell lung cancer.
20. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the non-small cell lung cancer is squamous non-small cell lung cancer, preferably squamous non-small cell lung cancer of Stage IIIB/IIIC or IV.
21. The method according to embodiment 20, the bispecific antibody for use according to embodiment 
20, the chemotherapy agent for use according to embodiment 20, or the bispecific antibody and the chemotherapy agent for use according to embodiment 20, wherein the chemotherapy agent is a combination of carboplatin and paclitaxel.
22. The method according to embodiment 21, the bispecific antibody for use according to embodiment 
21, the chemotherapy agent for use according to embodiment 21, or the bispecific antibody and the chemotherapy agent for use according to embodiment 21, wherein the carboplatin is administered at a dosage of about AUC=6 and the paclitaxel is administered at a dosage of about 200 mg/m2, preferably wherein the carboplatin is administered intravenously every three weeks at day 1 and the paclitaxel is administered intravenously every three weeks at day 1.
23. The method according to any one of embodiments 20 to 22, the bispecific antibody for use 
according to any one of embodiments 20 to 22, the chemotherapy agent for use according to any one of embodiments 20 to 22, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 20 to 22, wherein the bispecific antibody is administered at a dosage of about 2000 mg or 1400 mg, preferably intravenously every three weeks at day 1.
24. The method according to any one of embodiments 20 to 23, the bispecific antibody for use 
according to any one of embodiments 20 to 23, the chemotherapy agent for use according to any one of embodiments 20 to 23, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 20 to 23, wherein the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for non-small cell lung cancer.
25. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of docetaxel, a combination of carboplatin and pemetrexed, and a combination of carboplatin and paclitaxel.
26. A composition comprising
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent selected from the group consisting of docetaxel, a combination of 
carboplatin and pemetrexed, and a combination of carboplatin and paclitaxel.
27. A kit comprising
a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a 
heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent selected from the group consisting of 
docetaxel, a combination of carboplatin and pemetrexed, and a combination of carboplatin and paclitaxel.
28. The composition according to embodiment 26, or the kit according to embodiment 27 for use as 
medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating non-small cell lung cancer in a subject, the method comprising administering 
to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating non-small cell lung cancer, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating non-small cell lung cancer, the method 
comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating non-small cell lung cancer in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
6. The method according to embodiment 5, the binding agent for use according to embodiment 5, the 
chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the taxane is docetaxel, preferably wherein the docetaxel is administered intravenously at a dosage of about 75 mg/m2 or about 60 mg/m2 every three weeks at day 1.
7. The method according to embodiment 5 or 6, the binding agent for use according to embodiment 
5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the binding agent and the chemotherapy agent for use according to embodiment 5 or 6, wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every three weeks at day 1.
8. The method according to any one of embodiments 5 to 7, the binding agent for use according to 
any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line chemotherapy with immuno-oncology treatment.
9. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate.
10. The method according to embodiment 9, the binding agent for use according to embodiment 9, the 
chemotherapy agent for use according to embodiment 9, or the binding agent and the chemotherapy agent for use according to embodiment 9, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=5 every three weeks at day 1, and the antifolate chemotherapy agent is pemetrexed, preferably wherein the pemetrexed is administered intravenously at a dosage of about 500 mg/m2 every three weeks at day 1.
11. The method according to embodiment 9 or 10, the binding agent for use according to embodiment 
9 or 10, the chemotherapy agent for use according to embodiment 9 or 10, or the binding agent and the chemotherapy agent for use according to embodiment 9 or 10, wherein the binding agent is administered at a dosage of from about 10 mg/kg to 100 mg/kg, preferably intravenously every three weeks at day 1.
12. The method according to any one of embodiments 9 to 11, the binding agent for use according to 
any one of embodiments 9 to 11, the chemotherapy agent for use according to any one of embodiments 9 to 11, or the binding agent and the chemotherapy agent for use according to any one of embodiments 9 to 11, wherein the treatment is a second-line treatment, preferably wherein the cancer progressed during or after first-line treatment with an EGFR tyrosine kinase-inhibitor.
13. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the non-small cell lung cancer is non-squamous non-small cell lung cancer, preferably non-squamous non-small cell lung cancer of Stage IIIB/IIIC or IV.
14. The method according to embodiment 13, the binding agent for use according to embodiment 13, 
the chemotherapy agent for use according to embodiment 13, or the binding agent and the chemotherapy agent for use according to embodiment 13, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate.
15. The method according to embodiment 14, the binding agent for use according to embodiment 14, 
the chemotherapy agent for use according to embodiment 14, or the binding agent and the chemotherapy agent for use according to embodiment 14, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=5 every three weeks at day 1, and the antifolate chemotherapy agent is pemetrexed, preferably wherein the pemetrexed is administered intravenously at a dosage of about 500 mg/m2 every three weeks at day 1.
16. The method according to any one of embodiments 13 to 15, the binding agent for use according to 
any one of embodiments 13 to 15, the chemotherapy agent for use according to any one of embodiments 13 to 15, or the binding agent and the chemotherapy agent for use according to any one of embodiments 13 to 15, wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every three weeks at day 1.
17. The method according to any one of embodiments 13 to 16, the binding agent for use according to 
any one of embodiments 13 to 16, the chemotherapy agent for use according to any one of embodiments 13 to 16, or the binding agent and the chemotherapy agent for use according to any one of embodiments 13 to 16, wherein the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for non-small cell lung cancer.
18. The method according to embodiment 1, the binding agent for use according to embodiment 2, the 
chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the non-small cell lung cancer is squamous non-small cell lung cancer, preferably squamous non-small cell lung cancer of Stage IIIB/IIIC or IV.
19. The method according to embodiment 18, the binding agent for use according to embodiment 18, 
the chemotherapy agent for use according to embodiment 18, or the binding agent and the chemotherapy agent for use according to embodiment 18, wherein the chemotherapy agent is a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
20. The method according to embodiment 19, the binding agent for use according to embodiment 19, 
the chemotherapy agent for use according to embodiment 19, or the binding agent and the chemotherapy agent for use according to embodiment 19, wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=6 every three weeks at day 1, and the taxane is paclitaxel, preferably wherein the paclitaxel is administered intravenously at a dosage of about 200 mg/m2 every three weeks at day 1.
21. The method according to any one of embodiments 18 to 20, the binding agent for use according to 
any one of embodiments 18 to 20, the chemotherapy agent for use according to any one of embodiments 18 to 20, or the binding agent and the chemotherapy agent for use according to any one of embodiments 18 to 20, wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every three weeks at day 1.
22. The method according to any one of embodiments 18 to 21, the binding agent for use according to 
any one of embodiments 18 to 21, the chemotherapy agent for use according to any one of embodiments 18 to 21, or the binding agent and the chemotherapy agent for use according to any one of embodiments 18 to 21, wherein the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for non-small cell lung cancer.
23. The method according to any one of embodiments 1 and 5 to 22, the binding agent for use 
according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
24. The method according to embodiment 23, the binding agent for use according to embodiment 23, 
the chemotherapy agent for use according to embodiment 23, or the binding agent and the chemotherapy agent for use according to embodiment 23, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
25. The method according to any one of embodiments 1 and 5 to 22, the binding agent for use 
according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
26. The method according to embodiment 25, the binding agent for use according to embodiment 25, 
the chemotherapy agent for use according to embodiment 25, or the binding agent and the chemotherapy agent for use according to embodiment 25, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
27. The method according to any one of embodiments 1 and 5 to 22, the binding agent for use 
according to any one of embodiments 2 and 5 to 22, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 22, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
28. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which 
antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
29. A composition comprising
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; an
b. a chemotherapy agent selected from the group consisting of (i) a taxane, preferably 
paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
30. A kit comprising
a. a composition comprising a binding agent comprising (i) a first binding region which binds 
to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a composition comprising a chemotherapy agent selected from the group consisting of (i) 
a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; and (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
31. The binding agent and the chemotherapy agent for use according to embodiment 28, the 
composition according to embodiment 29, or the kit according to embodiment 30, wherein the chemotherapy agent is selected from the group consisting of (i) docetaxel, (ii) a combination of carboplatin and pemetrexed, and (iii) a combination of carboplatin and paclitaxel.
32. The binding agent and the chemotherapy agent for use according to embodiment 28 or 31, the 
composition according to embodiment 29 or 31, or the kit according to embodiment 30 or 31, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
33. The binding agent and the chemotherapy agent for use according to embodiment 32, the 
composition according to embodiment 32, or the kit according to embodiment 32, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
34. The binding agent and the chemotherapy agent for use according to embodiment 28 or 31, the 
composition according to embodiment 29 or 31, or the kit according to embodiment 30 or 31, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
35. The binding agent and the chemotherapy agent for use according to embodiment 34, the 
composition according to embodiment 34, or the kit according to embodiment 34, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
36. The binding agent and the chemotherapy agent for use according to embodiment 28 or 31, the 
composition according to embodiment 29 or 31, or the kit according to embodiment 30 or 31, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
37. The composition according to any one of embodiments 29 and 31 to 36, or the kit according to any 
one of embodiments 30 to 36 for use as medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating triple-negative breast cancer in a subject, the method comprising 
administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent.
2. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17 for use in a method of treating triple-negative breast cancer, the method comprising administering to the subject:
a. the bispecific antibody; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating triple-negative breast cancer, the method 
comprising administering to the subject:
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the 
amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. the chemotherapy agent.
4. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use in a method of treating triple-negative breast cancer in a subject, the method comprising administering to the subject:
a. the bispecific antibody; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is nab-paclitaxel.
6. The method according to embodiment 5, the bispecific antibody for use according to embodiment 
5, the chemotherapy agent for use according to embodiment 5, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5, wherein the nab-paclitaxel is administered at a dosage of about 100 mg/m2, preferably intravenously every four weeks at days 1, 8 and 15.
7. The method according to embodiment 5 or 6, the bispecific antibody for use according to 
embodiment 5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the bispecific antibody and the chemotherapy agent for use according to embodiment 5 or 6, wherein the bispecific antibody is administered at a dosage of about 20 mg/kg, preferably intravenously every four weeks at days 1, 8 and 15; or wherein the bispecific antibody is administered at a dosage of about 1400 mg, preferably intravenously every four weeks at days 1 and 15.
8. The method according to any one of embodiments 5 to 7, the bispecific antibody for use according 
to any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for triple-negative breast cancer.
9. The method according to any one of embodiments 5 to 8, the bispecific antibody for use according 
to any one of embodiments 5 to 8, the chemotherapy agent for use according to any one of embodiments 5 to 8, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5 to 8, wherein the subject suffers from locally advanced or metastatic triple-negative breast cancer.
10. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is paclitaxel.
11. The method according to embodiment 10, the bispecific antibody for use according to embodiment 
10, the chemotherapy agent for use according to embodiment 10, or the bispecific antibody and the chemotherapy agent for use according to embodiment 10, wherein the paclitaxel is administered at a dosage of about 90 mg/m2, preferably intravenously every four weeks at days 1, 8 and 15.
12. The method according to any one of embodiments 5, 6, 10 or 11, the bispecific antibody for use 
according to any one of embodiments 5, 6, 10 or 11, the chemotherapy agent for use according to any one of embodiments 5, 6, 10 or 11, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 5, 6, 10 or 11, wherein the bispecific antibody is administered at a dosage of about 15 mg/kg or 20 mg/kg, preferably intravenously every four weeks at days 1, 8 and 15; or wherein the bispecific antibody is administered at a dosage of about 1400 mg, preferably intravenously every four weeks at days 1 and 15.
13. The method according to any one of embodiments 10 to 12, the bispecific antibody for use 
according to any one of embodiments 10 to 12, the chemotherapy agent for use according to any one of embodiments 10 to 12, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 10 to 12, wherein the treatment is a first line-treatment or a second line treatment.
14. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is gemcitabine and carboplatin.
15. The method according to embodiment 14, the bispecific antibody for use according to embodiment 
14, the chemotherapy agent for use according to embodiment 14, or the bispecific antibody and the chemotherapy agent for use according to embodiment 14, wherein the gemcitabine is administered at a dosage of about 1000 mg/m2 and carboplatin is administered at a dosage of about AUC=2, preferably wherein the gemcitabine is administered intravenously every three weeks at days 1 and 8 and the carboplatin is administered intravenously every three weeks at days 1 and 8.
16. The method according to embodiment 1, the bispecific antibody for use according to embodiment 
2, the chemotherapy agent for use according to embodiment 3, or the bispecific antibody and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is eribuline.
17. The method according to embodiment 16, the bispecific antibody for use according to embodiment 
16, the chemotherapy agent for use according to embodiment 16, or the bispecific antibody and the chemotherapy agent for use according to embodiment 16, wherein the eribuline is administered at a dosage of about 1.4 mg/m2, preferably intravenously every three weeks at days 1 and 8.
18. The method according to any one of embodiments 14 to 17, the bispecific antibody for use 
according to any one of embodiments 14 to 17, the chemotherapy agent for use according to any one of embodiments 14 to 17, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 14 to 17, wherein the bispecific antibody is administered at a dosage of about 1000 mg to about 2000 mg, preferably of about 1200 mg or 1400 mg or 1600 mg or 2000 mg, more preferably of about 2000 mg; most preferably intravenously every three weeks at day 1.
19. The method according to any one of embodiments 14 to 18, the bispecific antibody for use 
according to any one of embodiments 14 to 18, the chemotherapy agent for use according to any one of embodiments 14 to 18, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 14 to 18, wherein the treatment is a first line-treatment or a second line treatment.
20. The method according to any one of embodiments 1 and 5 to 19, the bispecific antibody for use 
according to any one of embodiments 2 and 5 to 19, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 19, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 4 to 19, wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
21. The method according to embodiment 20, the bispecific antibody for use according to embodiment 
20, the chemotherapy agent for use according to embodiment 20, or the bispecific antibody and the chemotherapy agent for use according to embodiment 20, wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 and <10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
22. The method according to any one of embodiments 1 and 5 to 19, the bispecific antibody for use 
according to any one of embodiments 2 and 5 to 19, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 19, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 4 to 19, wherein the subject has a PD-L1 expression before the treatment that provides a score of <10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
23. The method according to any one of embodiments 20 to 22, the bispecific antibody for use 
according to any one of embodiments 20 to 22, the chemotherapy agent for use according to any one of embodiments 20 to 22, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 20 to 22, wherein the another integrating scoring algorithm is the tumour area positivity (TAP) score or the tumour proportion score (TPS) .
24. The method according to any one of embodiments 20 to 22, the bispecific antibody for use 
according to any one of embodiments 20 to 22, the chemotherapy agent for use according to any one of embodiments 20 to 22, or the bispecific antibody and the chemotherapy agent for use according to any one of embodiments 20 to 22, wherein the CPS has been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages; and/or
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably 
by using the 22C3 antibody) ; and/or
(iv) viable tumour cells are determined by staining with a viability dye; and/or
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 
staining lymphocytes and PD-L1 staining macrophages.
25. The method according to embodiment 23, the bispecific antibody for use according to 
embodiment 23, the chemotherapy agent for use according to embodiment 23, or the bispecific antibody and the chemotherapy agent for use according to embodiment 23, wherein the PD-L1 expression score has been determined using a TAP scoring algorithm by determining in a test sample of the subject the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer sample; and/or
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells; and/or
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 
immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 
positive tumour cells and immune cells; and/or
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
26. The method according to embodiment 23, the bispecific antibody for use according to 
embodiment 23, the chemotherapy agent for use according to embodiment 23, or the bispecific antibody and the chemotherapy agent for use according to embodiment 23, wherein the PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
(iii) viable tumour cells are determined by staining with a viability dye; and/or
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
27. A bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino 
acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
28. A composition comprising
a. a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising 
the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a chemotherapy agent selected from the group consisting of nab-paclitaxel, paclitaxel, a 
combination of gemcitabine and carboplatin, and eribuline.
29. A kit comprising
a. a composition comprising a bispecific antibody binding to PD-L1 and VEGF comprising a 
heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17; and
b. a composition comprising a chemotherapy agent selected from the group consisting of 
nab-paclitaxel, paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
30. The composition according to embodiment 28, or the kit according to embodiment 29 for use as 
medicament.
Additionally, the present invention relates to the following numbered embodiments:
1. A method of treating triple-negative breast cancer in a subject, the method comprising 
administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent.
2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and 
which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating triple-negative breast cancer, the method comprising administering to the subject:
a. the binding agent; and
b. a chemotherapy agent.
3. A chemotherapy agent for use in a method of treating triple-negative breast cancer, the method 
comprising administering to the subject:
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. the chemotherapy agent.
4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and 
which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating triple-negative breast cancer in a subject, the method comprising administering to the subject:
a. the binding agent; and
b. the chemotherapy agent.
5. The method according to embodiment 1, the binding agent for use according to embodiment 2, 
the chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel.
6. The method according to embodiment 5, the binding agent for use according to embodiment 5, 
the chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the taxane is nab-paclitaxel, preferably wherein the nab-paclitaxel is administered intravenously at a dosage of about 100 mg/m2 every four weeks at days 1, 8 and 15.
7. The method according to embodiment 5 or 6, the binding agent for use according to embodiment 
5 or 6, the chemotherapy agent for use according to embodiment 5 or 6, or the binding agent and the chemotherapy agent for use according to embodiment 5 or 6, wherein the binding agent is administered at a dosage of from about 10 mg/kg to about 100 mg/kg, preferably intravenously every four weeks at days 1, 8 and 15; or wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every four weeks at days 1 and 15.
8. The method according to any one of embodiments 5 to 7, the binding agent for use according to 
any one of embodiments 5 to 7, the chemotherapy agent for use according to any one of embodiments 5 to 7, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 7, wherein the treatment is a first line-treatment, preferably wherein the subject has not received any prior systemic treatment for triple-negative breast cancer.
9. The method according to any one of embodiments 5 to 8, the binding agent for use according to 
any one of embodiments 5 to 8, the chemotherapy agent for use according to any one of embodiments 5 to 8, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5 to 8, wherein the subject suffers from locally advanced or metastatic triple-negative breast cancer.
10. The method according to embodiment 5, the binding agent for use according to embodiment 5, 
the chemotherapy agent for use according to embodiment 5, or the binding agent and the chemotherapy agent for use according to embodiment 5, wherein the taxane is paclitaxel, preferably wherein the paclitaxel is administered at a dosage of about 90 mg/m2 intravenously every four weeks at days 1, 8 and 15.
11. The method according to any one of embodiments 5, 6 or 10, the binding agent for use according 
to any one of embodiments 5, 6 or 10, the chemotherapy agent for use according to any one of embodiments 5, 6 or 10, or the binding agent and the chemotherapy agent for use according to any one of embodiments 5, 6 or 10, wherein the binding agent is administered at a dosage of from about 10 mg/kg to 100 mg/kg, preferably intravenously every four weeks at days 1, 8 and 15;or wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every four weeks at days 1 and 15.
12. The method according to embodiment 10 or 11, the binding agent for use according to 
embodiment 10 or 11, the chemotherapy agent for use according to embodiment 10 or 11, or the binding agent and the chemotherapy agent for use according to embodiment 10 or 11, wherein the treatment is a first line-treatment or a second line treatment.
13. The method according to embodiment 1, the binding agent for use according to embodiment 2, 
the chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil, and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin.
14. The method according to embodiment 13, the binding agent for use according to embodiment 13, 
the chemotherapy agent for use according to embodiment 13, or the binding agent and the chemotherapy agent for use according to embodiment 13, wherein the antimetabolite chemotherapy agent is gemcitabine, preferably wherein the gemcitabine is administered intravenously at a dosage of about 1000 mg/m2 every three weeks at days 1 and 9, and wherein the platinum-based chemotherapy agent is carboplatin, preferably wherein the carboplatin is administered intravenously at a dosage of about AUC=2 every three weeks at days 1 and 8.
15. The method according to embodiment 1, the binding agent for use according to embodiment 2, 
the chemotherapy agent for use according to embodiment 3, or the binding agent and the chemotherapy agent for use according to embodiment 4, wherein the chemotherapy agent is a microtubule inhibitor, preferably eribuline.
16. The method according to embodiment 15, the binding agent for use according to embodiment 15, 
the chemotherapy agent for use according to embodiment 15, or the binding agent and the chemotherapy agent for use according to embodiment 15, wherein the microtubule inhibitor is eribuline, preferably wherein the eribuline is administered intravenously at a dosage of about 1.4 mg/m2 every three weeks at days 1 and 8.
17. The method according to any one of embodiments 13 to 16, the binding agent for use according 
to any one of embodiments 13 to 16, the chemotherapy agent for use according to any one of embodiments 13 to 16, or the binding agent and the chemotherapy agent for use according to any one of embodiments 13 to 16, wherein the binding agent is administered at a dosage of from about 1000 mg to about 2000 mg, preferably intravenously every three weeks at day 1.
18. The method according to any one of embodiments 13 to 17, the binding agent for use according 
to any one of embodiments 13 to 17, the chemotherapy agent for use according to any one of embodiments 13 to 17, or the binding agent and the chemotherapy agent for use according to any one of embodiments 13 to 17, wherein the treatment is a first line-treatment or a second line
19. The method according to any one of embodiments 1 and 5 to 18, the binding agent for use 
according to any one of embodiments 2 and 5 to 18, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 18, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 18, wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
20. The method according to embodiment 19, the binding agent for use according to embodiment 19, 
the chemotherapy agent for use according to embodiment 19, or the binding agent and the chemotherapy agent for use according to embodiment 19, wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 and <10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
21. The method according to any one of embodiments 1 and 5 to 18, the binding agent for use 
according to any one of embodiments 2 and 5 to 18, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 18, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 18, wherein the subject has a PD-L1 expression before the treatment that provides a score of <10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
22. The method according to any one of embodiments 19 to 21, the binding agent for use according 
to any one of embodiments 19 to 21, the chemotherapy agent for use according to any one of embodiments 19 to 21, or the binding agent and the chemotherapy agent for use according to any one of embodiments 19 to 21, wherein the another integrating scoring algorithm is the tumour area positivity (TAP) score or the tumour proportion score (TPS) .
23. The method according to any one of embodiments 19 to 21, the binding agent for use according 
to any one of embodiments 19 to 21, the chemotherapy agent for use according to any one of embodiments 19 to 21, or the binding agent and the chemotherapy agent for use according to any one of embodiments 19 to 21, wherein the CPS has been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour 
infiltrating) lymphocytes and macrophages; and/or
(iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably 
by using the 22C3 antibody) ; and/or
(iv) viable tumour cells are determined by staining with a viability dye; and/or
(v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 
staining lymphocytes and PD-L1 staining macrophages.
24. The method according to embodiment 22, the binding agent for use according to embodiment 22, 
the chemotherapy agent for use according to embodiment 22, or the binding agent and the chemotherapy agent for use according to embodiment 22, wherein the PD-L1 expression score has been determined using a TAP scoring algorithm by determining in a test sample of the subject the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer sample; and/or
(ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated 
stroma containing tumour-associated immune cells; and/or
(iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 
immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
(iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 
positive tumour cells and immune cells; and/or
(v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 
positive tumour cells and immune cells relative to the tumour area expressed as %.
25. The method according to embodiment 22, the binding agent for use according to embodiment 22, 
the chemotherapy agent for use according to embodiment 22, or the binding agent and the chemotherapy agent for use according to embodiment 22, wherein the PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:

wherein
(i) the test sample is a cancer tissue sample; and/or
(ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
(iii) viable tumour cells are determined by staining with a viability dye; and/or
(iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
26. The method according to any one of embodiments 1 and 5 to 25, the binding agent for use 
according to any one of embodiments 2 and 5 to 25, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 25, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 25, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
27. The method according to embodiment 26, the binding agent for use according to embodiment 26, 
the chemotherapy agent for use according to embodiment 26, or the binding agent and the chemotherapy agent for use according to embodiment 26, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
28. The method according to any one of embodiments 1 and 5 to 25, the binding agent for use 
according to any one of embodiments 2 and 5 to 25, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 25, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 25, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
29. The method according to embodiment 28, the binding agent for use according to embodiment 28, 
the chemotherapy agent for use according to embodiment 28, or the binding agent and the chemotherapy agent for use according to embodiment 28, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
30. The method according to any one of embodiments 1 and 5 to 25, the binding agent for use 
according to any one of embodiments 2 and 5 to 25, the chemotherapy agent for use according to any one of embodiments 3 and 5 to 25, or the binding agent and the chemotherapy agent for use according to any one of embodiments 4 to 25, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
31. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and 
which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline.
32. A composition comprising
a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both 
and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a chemotherapy agent selected from the group consisting of (i) a taxane, preferably 
paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline.
33. A kit comprising
a. a composition comprising a binding agent comprising (i) a first binding region which binds 
to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
b. a composition comprising a chemotherapy agent selected from the group consisting of (i) 
a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine or 5-fluorouracil; and a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin; and (iii) a microtubule inhibitor, preferably eribuline.
34. The binding agent and the chemotherapy agent for use according to embodiment 31, the 
composition according to embodiment 32, or the kit according to embodiment 33, wherein the chemotherapy agent is selected from the group consisting of paclitaxel, nab-paclitaxel, a combination of gemcitabine and carboplatin, and eribuline.
35. The binding agent and the chemotherapy agent for use according to embodiment 31 or 34, the 
composition according to embodiment 32 or 34, or the kit according to embodiment 33 or 34, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
36. The binding agent and the chemotherapy agent for use according to embodiment 35, the 
composition according to embodiment 35, or the kit according to embodiment 35, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
37. The binding agent and the chemotherapy agent for use according to embodiment 31 or 34, the 
composition according to embodiment 32 or 34, or the kit according to embodiment 33 or 34, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
38. The binding agent and the chemotherapy agent for use according to embodiment 37, the 
composition according to embodiment 37, or the kit according to embodiment 37, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
39. The binding agent and the chemotherapy agent for use according to embodiment 31 or 34, the 
composition according to embodiment 32 or 34, or the kit according to embodiment 33 or 34, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
40. The composition according to any one of embodiments 32 and 34 to 39, or the kit according to 
any one of embodiments 33 to 39 for use as medicament.
Sequence listing
This application contains a Sequence Listing which has been submitted electronically and is hereby incorporated by reference in its entirety. Said Sequence Listing file is named “Sequence Listing B19455WO. xml” and is 92 KB in size.
SEQ ID NO: 1-3 are exemplary CDR1-3 amino acid sequences of an anti-PD-L1 antibody using the IMGT numbering system.
SEQ ID NO: 4-6 are exemplary HCDR1-3 amino acid sequences of an anti-VEGF antibody using the IMGT numbering system.
SEQ ID NO: 7 and 8 are exemplary LCDR1 and 3 amino acid sequences of an anti-VEGF antibody using the IMGT numbering system.
SEQ ID NO: 9 is an exemplary amino acid sequence of an anti-PD-L1 VHH.
SEQ ID NO: 10 is an exemplary VH amino acid sequence of an anti-VEGF antibody.
SEQ ID NO: 11 is an exemplary VL amino acid sequence of an anti-VEGF antibody.
SEQ ID NO: 12 is an exemplary CH1 amino acid sequence of an anti-VEGF antibody.
SEQ ID NO: 13 is an exemplary IgG1 Fc region amino acid sequence of an anti-VEGF antibody.
SEQ ID NO: 14 is an exemplary linker amino acid sequence of the bispecific antibody as disclosed herein.
SEQ ID NO: 15 is an exemplary CL amino acid sequence of an anti-VEGF antibody.
SEQ ID NO: 16 is an exemplary heavy chain amino acid sequence of the bispecific antibody as disclosed herein comprising an exemplary anti-PD-L1 VHH amino acid sequence.
SEQ ID NO: 17 is an exemplary light chain amino acid sequence of the bispecific antibody as disclosed herein.
SEQ ID NO: 18, 19 and 38 are exemplary CDR1-3 amino acid sequences of an anti-PD-L1 antibody using the Kabat numbering system.
SEQ ID NO: 20-25 are exemplary LCDR1-3 and HCDR1-3 amino acid sequences of the 22C3 antibody.
SEQ ID NO: 26 is an exemplary VL amino acid sequence of the 22C3 antibody.
SEQ ID NO: 27 is an exemplary VH amino acid sequence of the 22C3 antibody.
SEQ ID NO: 28 is an exemplary VH amino acid sequence of the 28-8 antibody.
SEQ ID NO: 29 is an exemplary VL amino acid sequence of the 28-8 antibody.
SEQ ID NO: 30-35 are exemplary LCDR1-3 and HCDR1-3 amino acid sequences of the SP263 antibody.
SEQ ID NO: 36 is an exemplary VH amino acid sequence of the SP263 antibody.
SEQ ID NO: 37 is an exemplary VL amino acid sequence of the SP263 antibody.
SEQ ID NO: 39-41 are exemplary HCDR1-3 amino acid sequences of an anti-VEGF-A antibody using the Kabat numbering system.
SEQ ID NO: 42 and 43 are exemplary LCDR1 and 2 amino acid sequences of an anti-VEGF-Aantibody using the Kabat numbering system.
An exemplary anti-VEGF-ALCDR2 is the amino acid sequence FTS referred to herein as SEQ ID NO: 44 using the IGMT numbering system.
The sequences of SEQ ID NO: 1-44 are shown in the table below.












It is evident for the skilled person that the nucleotide sequences depend on the individual context for cloning and codon-optimization as described herein but still encode the same amino acid sequence.
Examples
The following Examples are merely illustrative and shall describe the present invention in a further way. These Examples shall not be construed to limit the present invention thereto.
Example 1: Determining the CPS of a cancer tissue sample using Dako’s PD-L1 IHC 22C3 pharmDx kit in 
combination with anti-PD-L1 antibody clone 22C3
This example is to provide guidelines for evaluating PD-L1 expression on formalin-fixed, 20 paraffin-embedded (FFPE) tumour tissue section with Dako’s PD-L1 IHC 22C3 pharmDx kit (SK006) according to the manufacturer’s TNBC Instructions for Use. This immunohistochemical (IHC) assay has been performed using the Dako Auto-stainer Link 48 automated staining system.
The embodiments below are described with respect to the use of the PD-L1 IHC 22C3 pharmDx kit, which is a qualitative immunohistochemical assay using Monoclonal Mouse Anti-PD-L1, Clone 22C3. This kit is intended for use in the detection of PD-L1 protein in formalin-fixed, paraffm-30 embedded (FFPE) tumour tissue using EnVision FLEX visualization system on Autostainer Link 48. Here, PD-L1 protein expression was used to determine a Combined Positive Score (CPS) .
PD-L1 IHC 22C3 pharmDx contains optimized reagents to perform an IHC staining procedure using a linker and a chromogen enhancement reagent. Deparaffinization, rehydration, and target retrieval was performed using a 3-in-1 procedure on PT Link.
Following peroxidase block, specimens were incubated with the monoclonal mouse primary antibody to PD-L1 or the Negative Control Reagent. Specimens were then incubated with a Mouse LINKER, followed by incubation with a ready-to-use Visualization Reagent consisting of secondary antibody molecules and horseradish peroxidase molecules coupled to a dextran polymer backbone. The enzymatic conversion of the subsequently added chromogen resulted in precipitation of a visible reaction product at the site of the antigen. The color of the chromogenic reaction was modified by a chromogen enhancement reagent. The specimen was counterstained and coverslipped and results were interpreted using a light microscope.
Clinical Interpretation Guidelines for PD-L1 IHC 22C3 pharmDx in Tumour Tissue Specimen  Criteria
A hematoxylin and eosin (H&E) stain of the tissue specimen were evaluated first to assess tissue histology and preservation quality. PD-L1 IHC 22C3 pharmDx and the H&E staining were performed on serial sections from the same paraffin block of the specimen (sample) . Tissue specimens were intact, well preserved, and confirmed tumour indication.
The specimen contained a minimum of 100 viable tumour cells to determine the percentage of positive cells. For specimens with less than 100 viable tumour cells, tissue from a deeper level of the block, or potentially another block, presented sufficient number of viable tumour cells for PD-L1 IHC 22C3 pharmDx testing.
PD-L1 IHC 22C3 pharmDx Control Cell Line Slide
The PD-L1 IHC 22C3 pharmDx Control Cell Line Slide were examined to determine that reagents are functioning properly. Each slide contained sections of cell pellets with positive and negative PD-L1 expression. The percentage of positive cells, staining intensity, and non-specific staining were assessed in both cell pellets. If any staining of the Control Cell Line Slide was not satisfactory, all results with the subject specimens were considered invalid. The Control Cell Line Slide has not been used as an aid in interpretation of subject results.
The overall staining intensity was evaluated using the following guide:
Positive Control Cell Pellet:
The following stainings were acceptable for the PD-L1 positive cell pellet:
- Cell membrane staining of ≥70%of cells
- ≥2+ average staining intensity
- Non-specific staining < 1+ intensity
Negative Control Cell Pellet:
For the PD-L1 negative cell pellet, the following stainings were acceptable:
- No specific staining
- Non-specific staining < 1+ intensity. Note that staining of a few cells in the MCF-7 cell pellet may 
occasionally be observed. The following acceptance criteria are applicable: the presence of ≤10 total cells with distinct plasma membrane staining, or cytoplasmic staining with ≥1+ intensity within the boundaries of the MCF-7 cell pellet are acceptable
Positive and Negative User Control Tissue (TNBC) :
The TNBC Positive Control Tissue Slides were examined to verify that the fixation method and epitope retrieval process are effective. The Positive Control Tissue Slides were stained with both PD-L1 primary antibody and Negative Control Reagent. The ideal positive control tissue provided a complete dynamic representation of weak-to-moderate staining of tumour cells and tumour-associated mononuclear inflammatory cells (MICs) . Known positive tissue controls were utilized for monitoring the correct performance of processed tissues and test reagents, not as an aid in formulating a specific diagnosis of subject samples. If stainings of positive in-house control tissue were not satisfactory, all results with the subject specimen were considered invalid.
- Requirements for slide stained with PD-L1: Presence of brown plasma membrane staining were 
observed. Non-specific staining ≤1+
- Requirements for slide stained with Negative Control Reagent: No membrane staining. Non-
specific staining ≤1+
Optional Control Tissue:
In addition to the Control Cell Line Slide and in-house control tissues, FFPE tonsil were used as an optional control specimen. Tonsil stained with PD-L1 exhibited strong membrane staining in portions of the crypt epithelium and weak-to-moderate membrane staining of the follicular macrophages in the germinal centers.
PD-L1 expression of the endothelium, fibroblasts, and the surface epithelium was absent.
Negative Control Reagent (NCR) :
The slides stained with the NCR were examined to identify non-specific background staining that may interfere with PD-L1 staining interpretation, making the specimen non-evaluable. Satisfactory performance was indicated by 0 specific staining and ≤1+ non-specific staining.
The subject specimens stained were examined with the NCR to determine if there is any non-specific staining that may interfere with interpreting the PD-L1 stained slide. Non-specific staining was ≤1+.
NCR-stained slides indicated non-specific background staining and allowed for better interpretation of subject specimens stained with the primary antibody.
Scoring Guidelines
PD-L1 expression score in TNBC was determined by using combined positive score (CPS) , which is the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) divided by the total number of viable tumour cells, multiplied by 100. Although the result of the calculation can exceed 100, the maximum score was defined as CPS 100.
Any perceptible and convincing partial or complete linear membrane staining (≥1+) of viable tumour cells that was perceived as distinct from cytoplasmic staining was considered PD-L1 staining and was included in the scoring. Any membrane and/or cytoplasmic staining (≥1+) of lymphocytes and macrophages (mononuclear inflammatory cells, MICs) within tumour nests and/or adjacent supporting stroma was considered PD-L1 staining and included in the CPS numerator. Only MICs directly associated with the response against the tumour were scored. Additional CPS inclusion/exclusion criteria is summarized in tables 1 and 2.
- At lower magnifications, all well-preserved tumour areas were examined. Overall areas of PD-L1 
staining and non-staining tumour cells were evaluated, keeping in mind that partial membrane staining or 1+ membrane staining could be difficult to see at low magnifications. There were at least 100 viable tumour cells in the sample
○ A minimum of 100 viable tumour cells must be present in the PD-L1 stained slide (biopsy 
and resection) for the specimen to be considered adequate for evaluation
- For specimens with less than 100 viable tumour cells, tissue from a deeper level of the block or 
potentially another block has a sufficient number of tumour cells for evaluation of PD-L1 expression
- At higher magnification (20×) , PD-L1 expression was evaluated and CPS calculated:
○ The total number of viable tumour cells was determined, both PD-L1 staining and non-
staining (CPS denominator)
○ The number of PD-L1 staining cells was determined (tumour cells, lymphocytes, 
macrophages) (CPS numerator; see Tables 1 and 2 for additional CPS inclusion/exclusion criteria)
○ CPS was calculated
- Evaluation of membrane staining was performed at no higher than 20× magnification.
Table 2: CPS Numerator Inclusion/Exclusion Criteria for TNBC

*In MICs, membrane and cytoplasmic staining can be indistinguishable due to high nuclear to cytoplasmic 
ratio. Therefore, membrane and/or cytoplasmic staining of MICs was included in the score.
Adjacent MICs were defined as being within the same 20× field as the tumour. However, MICs that were 
NOT directly associated with the response against the tumour were excluded.
Macrophages and histiocytes have been considered the same cells.
Table 3: CPS Denominator Inclusion/Exclusion Criteria for TNBC


Example 2: Determining the CPS of a cancer tissue sample using PD-L1 IVD Kit (MEDx Inc. ) in 
combination with anti-PD-L1 antibody clone E1L3N.
The PD-L1 expression score was determined using the PD-L1 IVD Kit (MEDx Inc. ) and clone E1L3N according to the Manufacturer’s Instructions for Use. CPS definition and scoring process has been conducted according to a similar method as the PD-L1 IHC 22C3 pharmDx’s Instructions for Use described in example 1.
Example 3: Efficacy of a First-Line therapy of a bispecific anti VEGF-A anti PD-L1 antibody (PM8002) in 
Combination with Chemotherapy for Subjects with Triple-Negative Breast Cancers (TNBC) .
This is a Phase Ib/II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with nab-paclitaxel in subjects with locally advanced or metastatic triple-negative breast cancer without previous systematic treatment. This study determines the efficacy and safety of first-line treatment with PM8002 in combination with nab-paclitaxel in subjects with previously untreated, unresectable locally advanced or metastatic TNBC.
Approximately 42 subjects (intention-to-treat population (ITT) ) with previously unresectable locally advanced or metastatic TNBC were treated. TNBC is confirmed by histology or cytology, i.e., ER, PR, HER-2 are all negative. Negative ER and PR are defined as: IHCER < 1%, IHCPR < 1%. HER-2 negative is defined as: IHCHER-2 (-) or (1+) , HER-2 (2+) must be tested by FISH and the result is negative.
Subjects who had not received systemic treatment for advanced TNBC in the past were allowed to use taxane anti-tumour therapy in the previous neoadjuvant and/or adjuvant treatment stage, but had to meet the end time of taxane neoadjuvant and/or adjuvant treatment recurrence/metastasis interval ≥ 12 months.
Study arm: PM8002 at 20 mg/kg (Q2W) and nab-paclitaxel at 100 mg/m2 on the 1st, 8th, and 15th days of each cycle until unacceptable toxicity or disease progression were observed. Each cycle contains 28 days.
The primary endpoint of the study was the Objective Response Rate (ORR) assessed by investigators per RECIST v1.1, the incidence and severity of Treatment-Related Adverse Events (TRAEs) graded according to NCI-CTCAE v5.0. Objective response rate (ORR) is the proportion of subjects with complete response (CR) or partial response (PR) , based on RECIST v1.1.
The secondary endpoint of the study was the Progression-free survival (PFS) and the Disease Control rate (DCR) based on Investigator assessments per RECIST v1.1.
The study population included subjects aged 18 to 75 (including boundary value) . Each subject met all of the inclusion criteria and none of the exclusion criteria for this study in order to be randomized to a study intervention.
Inclusion Criteria:
a. Ability to understand and willing to provide written informed consent and to comply with 
scheduled visits and study procedures.
b. Female or male, ages 18 to 75 years.
c. Histologically or cytologically confirmed unresectable locally advanced or metastatic breast 
cancer with negative ER (estrogen receptor, ER <1%tumour cells) , PR (progesterone receptor, PR <1%tumour cells) and HER-2 (IHC 0/IHC 1+/IHC 2+/FISH not-amplified) . Testing results for all three markers conducted within 36 months prior to the initiation of the study by a local facility accredited by clinical research center are acceptable. If deemed necessary by the investigator during screening, subjects may provide additional biopsy to confirm the latest pathological.
d. Subjects who have not received prior systemic treatment for advanced TNBC are eligible 
for the study. Subjects who have received taxane-based chemotherapy or pembrolizumab during the neoadjuvant and/or adjuvant treatment phase are eligible if the occurrence of relapse or metastasis was more than 12 months after the end of such treatment (s) .
e. Subjects should provide a fresh tumour biopsy during the screening period (bone biopsies, 
fine-needle aspiration biopsies, and samples from pleural or peritoneal fluid are not acceptable, subjects with only one target lesion are not eligible to provide biopsy) . Sufficient qualified tumour tissue specimens should be obtained for biomarker analysis including PD-L1 expression levels. If a subject is unable to provide a fresh biopsy, a recent tumour sample (up to a maximum of 24 months prior to the start of the study) processed through formalin-fixed paraffin embedding (FFPE) or unstained slides (3-5 μm) is acceptable for the corresponding biomarker analysis. If a subject is unable to provide specimens that meet the aforementioned requirements, she/he may still participate in the screening process with the consent of the Sponsor.
f. Adequate organ function, as defined below:
i. Hematology (RBC or platelet transfusions, granulocyte colony-stimulating factors 
treatment, or other medical supports are not allowed within 14 days prior to the study treatment initiation) :
1. Neutrophil count (ANC) ≥1.5 × 109/L.
2. Platelet count (PLT) ≥100 × 109/L.
3. Hemoglobin (Hb) ≥90 g/L.
ii. Liver function:
1. Total bilirubin (TBIL) ≤1.5 × upper limit of normal (ULN) , objects with liver 
metastasis ≤2 × ULN, subjects with a previous diagnosis of Gilbert's syndrome ≤3 × ULN.
2. Aspartate aminotransferase (AST) or alanine aminotransferase (ALT) ≤2.5 
× ULN, objects with liver metastasis ≤5 × ULN.
iii. Renal function:
1. Serum creatinine ≤1.5 × ULN or Creatinine Clearance (CrCl) ≥50 ml/min 
{Cockcroft-Gault formula: [ (140 -age) × weight (kg) × (0.85, for women only) ] / [72 × creatinine (mg/dL) ] (conversion of creatinine unit: 1 mg/dL = 88.4 μmol/L) } .
2. Qualitative urine protein ≤1+; If qualitative urine protein ≥ 2+, 24 h urine 
protein quantitative test is required, if the result is < 1g, it is acceptable.
iv. Coagulation function: International normalized ratio (INR) ≤1.5, activated partial 
thromboplastin time (APTT) ≤1.5 × ULN, objects with liver metastasis INR and APTT ≤2 × ULN.
g. Performance status as assessed by the Eastern Cooperative Oncology Group (ECOG) : 0-
1.
h. Life expectancy ≥12 weeks.
i. According to RECIST 1.1, the subject has at least 1 measurable lesion as the targeted 
lesion (a measurable lesion at the previously irradiated radiation field or other local treatment area should not be selected as targeted lesion, the only bone metastasis or the only central nervous system metastasis should not be considered as a measurable lesion) .
j. Female subjects of childbearing potential have a negative blood pregnancy test result 
within 7 days prior to the study treatment and are willing to follow medically approved highly effective contraceptive measures (such as intrauterine device and condom) from signing the informed consent form to until 6 months after the last dosage of treatment.
k. Male subjects are willing to follow medically approved highly effective contraceptive 
measures from signing the informed consent form to until 6 months after the last dosage, and do not donate sperm during this period.
Exclusion Criteria:
a. History of severe allergic disease, severe drug allergy (including allergy to any 
investigational products) or known allergy to any component of the investigational product, or its excipients.
b. Adverse events resulting from prior anti-tumour therapies should be assessed and graded 
according to the CTCAE 5.0 criteria, subjects whose AEs have not returned to Grade 1 or below (unless the investigator determines that the current AEs pose no safety risk to the patients, such as hair loss or stable hypothyroidism under hormone replacement therapy) are not eligible for the study.
c. Those who have previously received any antibody-or inhibitor-based therapy targeting PD-
1/PD-L1 or VEGF.
d. Those with a history of pulmonary fibrosis, or currently diagnosed with severe lung 
diseases such as interstitial pneumonia, pneumoconiosis, chemical pneumonitis, or any other condition resulting in significant impairment in lung function. Exception: asymptomatic interstitial changes caused by previous radiation therapy, chemotherapy, or other factors such as smoking are acceptable.
e. Those who have received any of the following therapies or drugs prior to study initiation:
i. Have received immunotherapy, nitrosoureas, or mitomycin C within 6 weeks prior 
to the initiation of the study treatment, or have received oral fluoropyrimidine-based chemotherapy or small molecule targeted therapy within 2 weeks prior to the start of the study treatment (or within 5 half-lives of the drugs, whichever is longer) , or have received palliative radiotherapy within 7 days prior to the initiation of the study treatment, or have received any other chemotherapy, curative/palliative radiotherapy, endocrine therapy, biologic therapy (including tumour vaccines, cytokines, or growth factors for tumour control) or any experimental anti-tumour drugs within 28 days prior to the initiation of the study treatment.
ii. Have undergone major organ surgery (excluding needle biopsies) within 28 days 
prior to the study treatment or need to undergo elective surgery during the trial.
iii. Have clinically significant unhealed wounds, ulcers, or fractures.
iv. Have received systemic corticosteroids (at a dosage greater than 10 mg/day of 
prednisone or an equivalent dosage of other corticosteroids) within 14 days prior to the initiation of the study treatment. Have received other systemic immunostimulatory agents or immunosuppressive therapies (such as IFN-α, IL-2, or methotrexate) within 4 weeks prior to the initiation of the study treatment or are within 5 half-lives of the treatment drug (whichever is longer) . Exception: excluding local, intranasal, intraocular, intra-articular or inhaled corticosteroids, short-term use (≤7 days) of corticosteroids for prophylaxis (e.g., prevention of contrast agent allergy) or treatment of non-autoimmune conditions (e.g., delayed hypersensitivity reactions caused by exposure to allergens) .
v. Have been vaccinated with live attenuated vaccine (s) within 28 days prior to the 
study treatment.
vi. Have used systemic broad-spectrum antibiotics for ≥7 days within 14 days prior to 
the study treatment.
f. Those who have meningeal metastases, uncontrolled or symptomatic central nervous 
system (CNS) metastases.
g. Those who have active infections requiring intravenous antibiotic therapy at the time of 
study.
h. Those who had or have active autoimmune disease or a history of autoimmune diseases 
with anticipated relapse (such as systemic lupus erythematosus, rheumatoid arthritis, vasculitis, etc. ) , except for those with clinically stable autoimmune thyroid disease or type 1 diabetes.
i. Those who have had other active malignant tumours within 5 years prior to the study 
treatment, except for those that can be treated locally and have been cured (such as basal cell or squamous cell carcinoma of the skin, superficial or non-invasive bladder cancer, carcinoma in situ of the cervix, and papillary carcinoma of thyroid) .
j. Those with any of the following conditions within 6 months prior to the study treatment:
i. Acute coronary syndrome, coronary artery bypass grafting, congestive heart 
failure (CHF) , aortic dissection, stroke, or other grade 3 and above cardiovascular and cerebrovascular events.
ii. New York Heart Association (NYHA) functional classification ≥II heart failure (as 
defined by the NYHA) , or left ventricular ejection fraction (LVEF) < 50%.
iii. Those who have ventricular arrhythmias requiring clinical intervention, second-to 
third-degree atrioventricular block, or congenital long QT syndrome.
iv. Mean QT interval corrected by Fridericia’s method (QTcF) > 450 ms for males and 
> 470 ms for females.
v. Use of cardiac pacemaker.
vi. Cardiac troponin (cTn) I or N > 2 × ULN.
k. Those who have uncontrollable pleural, pericardial, or abdominal effusions.
l. Those with fever of unknown origin > 38.5 ℃prior to the study treatment (subjects with 
fever caused by tumours can be enrolled as judged by the investigator) .
m. Presence any of the following conditions prior to study treatment:
i. Poorly controlled diabetes (fasting blood glucose ≥13.3 mmol/L) .
ii. Poorly controlled hypertension (systolic blood pressure ≥140 mmHg and/or 
diastolic blood pressure ≥90 mmHg) .
iii. Those with a history of hypertensive crisis or hypertensive encephalopathy.
iv. Those with a history of abdominal fistula, tracheoesophageal fistula, 
gastrointestinal perforation, or intra-abdominal abscess within the last 6 months prior to the start of the study treatment.
v. Those who have undergone core needle biopsy or other minor surgical procedures 
within the last 7 days prior to the start of the study treatment (excluding placement of vascular infusion devices) .
n. Those with uncontrolled tumour-related pain requiring analgesic treatment should have a 
stable analgesic regimen at screening. For asymptomatic metastatic lesion, if its growth may cause dysfunction or intractable pain (e.g., current epidural metastasis unrelated to spinal cord compression) , local treatment should be considered before screening, if appropriate.
o. Those who present with a history of major bleeding diathesis or other significant risk of 
bleeding such as:
i. History of intracranial hemorrhage or intraspinal hemorrhage.
ii. With tumour lesions invading large blood vessels and are at significant risk of 
bleeding.
iii. Have had thrombosis or embolism (except intramuscular venous thrombosis that 
is asymptomatic and does not require treatment) within 6 months prior to the study treatment.
iv. Have had clinically significant hemoptysis or tumour hemorrhage of any cause 
within 1 month prior to the study treatment.
v. Have had anticoagulant therapy of therapeutic purposes (except low molecular 
weight heparin for prophylaxis) within 14 days prior to the study treatment.
vi. Have used antiplatelet drugs including but not limited to aspirin (≥100 mg/day) , 
clopidogrel (> 75 mg/day) , dipyridamole, ticlopidine or cilostazol, within 10 days prior to the study treatment, or those requiring long-term antiplatelet therapy.
p. Those who have received allogeneic hematopoietic stem cell transplantation or organ 
transplantation.
q. Those who are known to have a history of alcoholic abuse, psychotropic drug abuse, or 
illicit drug addiction.
r. Those with a documented history of neurologic or mental disorders, such as epilepsy, 
dementia, schizophrenia and so on.
s. Presence of human immunodeficiency virus (HIV) infection or known acquired 
immunodeficiency syndrome (AIDS) .
t. Those with positive result in non-specific treponemal antibody tests for syphilis (such as 
TRUST, PRP) or positive results in specific treponemal antibody tests (such as TPPA) are not eligible for the study, except those who have a positive result in specific treponemal antibody tests but have consistently negative results in non-specific treponemal antibody tests for one year or longer.
u. Those with active tuberculosis or a history of tuberculosis infection that was not 
successfully controlled.
v. Those with active hepatitis B (positive for HBsAg and HBV-DNA ≥1000 IU/ml) are not 
eligible for the study, unless their viral load is controlled (HBV-DNA < 1000 IU/ml) with antiviral medication. Those with active hepatitis C (HCV-RNA > lower limit of detection as determined by the research center) are also not eligible for the study.
w. Subject’s underlying condition may increase the risk of research treatment or complicate 
the interpretation of toxicities and adverse events, as judged by the investigator.
x. Those who are expected to require other anti-tumour drug therapy during the trial.
y. Pregnant or lactating women.
z. Other situations in which investigators consider the patient unsuitable for participation in 
this study.
Before treatment, the CPS of each patient has been determined as described in example 2 using the anti-PD-L1 antibody clone E1L3N and the PD-L1 IVD Kit (MEDx Inc. ) .
Subgroups of patients having a CPS of <1, ≥1, and ≥10 have been formed. The study results are summarized in table 4. As shown in this table, the combination therapy led to an increased median progression-free survival in the CPS subgroup ≥1. Median progression-free survival (95%CI) was 9.2 months with the addition of the bispecific antibody targeting specifically PD-L1 and VEGF-A combined with chemotherapy in subjects having a CPS of ≥1, identical to the median progression-free survival of subjects having a higher PS of ≥10. Thus, contrary to the SOC treatment comprising pembrolizumab and chemotherapy, the combination treatment disclosed herein has a beneficial effect in PD-L1 low expressing cancers (CPS subgroup ≥1) similar to PD-L1 high expressing cancers (CPS subgroup ≥10) . The ORR in the ITT population was 78.6%, including 1 complete response and 32 partial responses, the confirmed ORR was 73.8%with the disease control rate of 95.2%. The ORR in the CPS<1 population was 76.9%, including 10 partial responses, the confirmed ORR was 76.9%with the disease control rate of 100%. The ORR in the CPS≥10 population was 100%, including 9 partial responses, the confirmed ORR was 100%with the disease control rate of 100%. The ORR in the CPS≥1 population was 80.0%, the confirmed ORR was 72.0%. The ORR in the CPS<1 population was 76.9%, the confirmed ORR was 76.2%. with the disease control rate of 100%. Patients were continued to be treated after obtaining the results discussed above and shown below in Tables 4 and 5, and, as of 5 July 2024, the median PFS for the ITT population was 13.5 months (compared to 9.2 months as shown in Table 4) , the median time to response was 1.9 months, and the median duration of response was 11.7 months.
Table 4: Efficacy Outcomes measured by median PFS, 95%confidence interval (CI) , in Subgroups 
according to CPS Status at Baseline

*Unknown CPS due to lack of tissue sections
In the CPS≥1 subgroup, a further subgroup was formed, namely the subgroup of 1≤CPS<10, wherein the results are shown in Table 4a.
Table 4a: Efficacy Outcomes measured by median PFS, 95%confidence interval (CI) , in a further subgroup 
according to CPS Status at Baseline
The ORR in the ITT population 1≤CPS<10 was 68.8%, including 1 complete response and 10 partial responses, the confirmed ORR was 56.3%with the disease control rate of 93.8%.
The combination therapy disclosed herein shows an improved median progression-free survival compared to the standard treatment involving chemotherapy alone or chemotherapy in combination with the anti-PD-L1 antibody pembrolizumab as shown in Table 5.
Table 5: Comparison of the current treatment of care for triple-negative breast cancer with the combination 
therapy disclosed herein


*data derived from Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus Chemotherapy in Advanced 
Triple-Negative Breast Cancer. N Engl J Med. 2022; 387 (3) : 217-226. doi: 10.1056/NEJMoa2202809.
Surprisingly, the combination therapy of the bispecific antibody targeting specifically PD-L1 and VEGF-A combined with chemotherapy showed encouraging anti-tumour activity regardless of PD-L1 status and good safety profile as a first-line therapy for TNBC cancer having a low CPS of ≥1.
The combination therapy therefore meets the high unmet need for a breakthrough therapy designation for subject afflicted with cancer and having a low CPS of ≥1.
The patients of the present study were also classified according to the Fudan University Shanghai Cancer Center (FUSCC) classification (see Jiang YZ, Ma D, Suo C, et al. Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies. Cancer Cell. 2019; 35 (3) : 428-440. e5) , which classifies TNBCs into four transcriptome-based subtypes with distinct molecular features: (1) luminal androgen receptor (LAR) , (2) immunomodulatory (IM) , (3) basal-like immune-suppressed (BLIS) , and (4) mesenchymal-like (MES) . 31 of the 42 patients could be classified according to these four transcriptome-based subtypes, and the results were as shown in Table 6.
Table 6: Efficacy in the FUDAN-subtypes

ORR, objective response rate; cORR, confirmed ORR; disease control rate, DCR; basal-like immune-
suppressed, BLIS; immunomodulatory, IM; luminal androgen receptor (LAR) ; mesenchymal-like, MES.
It is noteworthy that both in the IM and MES subtypes, the ORR was 100%.
All patients experienced treatment-related adverse events (TRAEs) , 54.8%were Grade 3 or 4 and no Grade 5 TRAEs was observed. The most common TRAEs included neutropenia, leukocytopenia, anemia, proteinuria, alopecia and epistaxis. 35.7%patients experienced immune-related adverse events (irAEs) with Grade 3 or 4 of 9.5%, which included hyperthyroidism, hypothyroidism and rash, etc. The most common AEs related to VEGF were hypertension and proteinuria which most were Grade 1-2.
In view of the above results, PM8002 combined with nab-paclitaxel showed encouraging antitumor activity and good safety as first-line therapy for locally advanced and metastatic TNBC.
Example 4: Safety and Efficacy of PM8002 (anti-PD-L1 x VEGF-A bispecific) Combined with Paclitaxel as 
a Second-Line Therapy for Small Cell Lung Cancer
The present example is a Phase II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with paclitaxel as a second-line therapy for patients suffering from small cell lung cancer (SCLC) to assess the safety and efficacy of this treatment.
Patients with advanced SCLC who failed first-line platinum-based chemotherapy (i.e., who progressed during or after first-line platinum therapy) with or without checkpoint inhibitor therapy were enrolled in the present study. There is an unmet medical need for such patients.
Primary endpoints included the objective response rate (ORR) and safety, with progression free survival (PFS) and overall survival (OS) as the secondary endpoints. The endpoints were assessed by investigators according to commonly and routinely used standardized procedures.
All patients received PM8002 at 30 mg/kg (Q3W) and paclitaxel at 175 mg/m2 (Q3W) for 5 cycles (PM8002 and paclitaxel were administered IV) , followed by PM8002 maintenance at 30 mg/kg (Q3W) until unacceptable toxicity or disease progression.
Results: As of April 12, 2024, 60 patients have been enrolled. Among 26 patients, 22 patients completed at least one tumor assessment. 5 patients were still on treatment. Median PFS was 5.9 months and median OS was 14.4 months, which were still maturing. The ORR was 72.7%(16/22) with a disease control rate (DCR) of 81.8% (18/22) , and the median duration of response (DOR) was 11.5 months. Among 34 immunotherapy-treated patients, 29 patients completed at least one tumor assessment. 12 patients were still on treatment. The median PFS was 6.2 months which was still maturing. The ORR was 44.8% (13/29) with a DCR of 89.7% (26/29) . Any-grade treatment-related adverse events (TEAEs) occurred in 95% (57/60) of patients. Grade ≥ 3 TEAEs related to the combination regimen occurred in 70% (42/60) of patients, with the most common grade ≥ 3 TRAEs observed being neutropenia (56.7%, 34/60) and leukopenia (31.7%, 19/60) . Three patients discontinued PM8002 and/or paclitaxel administration due to TRAEs. Any-grade immune-related adverse events (irAEs) occurred in 30% (18/60) of patients, and grade ≥ 3 irAEs occurred in 3.3% (2/60) of patients.
Summarizing the above, PM8002, in combination with paclitaxel, showed encouraging antitumor activity and an acceptable safety profile for patients with advanced SCLC who failed first-line platinum-based chemotherapy with or without immunotherapy.
Example 5: Safety and Efficacy of PM8002 (anti-PD-L1 x VEGF-A bispecific) Combined with Systemic 
Chemotherapy as a First-Line Therapy for Extensive-Stage Small Cell Lung Cancer
The present example is an open label, single arm, multicenter Phase II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with systemic chemotherapy (in particular platinum-etoposide) as a first-line therapy for patients suffering from extensive-stage small cell lung cancer (ES-SCLC) to assess the safety and efficacy of this treatment.
Patients with ES-SCLC have a poor prognosis and there is an unmet medical need for such patients. The patients of the present study suffered from ES-SCLC (histologically or cytologically confirmed) and had not received systemic treatment for ES-SCLC. Further, patients were eligible for the present study if they were equal to or older than 18 years with an Eastern Cooperative Oncology Group Performance Status (ECOG PS) from 0 to 1.
Primary endpoints included the objective response rate (ORR) and safety, with progression free survival (PFS) and overall survival (OS) as the secondary endpoints. The endpoints were assessed by investigators according to RECIST v1.1 and commonly and routinely used standardized procedures as regards the TRAEs.
All patients received PM8002 at 30 mg/kg (Q3W) and platinum-etoposide (with carboplatin at AUC=5 (total dose ≤ 750 mg) on day 1 of each cycle and etoposide at 100 mg/m2 on days 1 to 3 of each cycle) every three weeks for 4 cycles, followed by PM8002 maintenance (at 30 mg/kg) every three weeks until disease progression or unacceptable toxicity.
Results: As of April 12, 2024, 50 patients were enrolled in this study and 48 patients completed at least one tumor evaluation. The median age was 60 years, with a range from 46 to 75 years. A partial response (PR) was observed in 42 patients with a best overall objective response rate (ORR) of 87.50%, Confirmed ORR of 81.25%. The median best percentage tumor volume shrinkage from baseline was 58.48%. The disease control rate (DCR) was 100% (48/48) , and the median progression free survival (PFS) was 6.93 months (95%CI: 4.27, 8.48) . 26 patients were still on treatment. The censored rate of PFS was 47.92%. Median overall survival (OS) had not yet been reached. Any-Grade treatment-related adverse events (TRAEs) occurred in all patients. Grade ≥ 3 TRAEs occurred in 42 patients (84%) . The most commonly observed AE at any Grade was neutrophil count decrease (90%, 45/50) , white blood cell count decrease (76%, 38/50) , and platelet count decrease (60%, 30/50) . Immune-related adverse events (irAEs) occurred in 34%of patients (17/50) with 4% (2/50) Grade ≥ 3. Two patients (4%) discontinued treatment due to TRAEs.
Summarizing the above, PM8002 plus platinum-based chemotherapy as a first-line treatment for ES-SCLC showed some efficacy and acceptable tolerability.
Example 6: Safety and Efficacy of PM8002 (anti-PD-L1 x VEGF-A bispecific) Combined with 
Chemotherapy in patients with EGFR-mutated Non-Small Cell Lung Cancer (NSCLC)
The present example is a Phase II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with chemotherapy (in particular carboplatin and pemetrexed) as a therapy for patients suffering from EGFR-mutated NSCLC who progressed after EGFR-tyrosine kinase inhibitors (TKIs) treatment to assess the safety and efficacy of this treatment. Furthermore, the correlation between tumor PD-L1 expression and clinical response was investigated.
PD-L1 expression was determined by immunohistochemistry (IHC; using TPS) from patients (biopsy at baseline) after progression from EGFR-TKI therapy as disclosed above in Example 2. Patients were divided into 3 groups based on these TPS results (<1%defined as negative, 1 to 49%defined as low expression, and ≥50%defined as high expression, using clone E1L3N as the detection antibody) . The primary endpoint of this study was the objective response rate (ORR) assessed by investigators according to per RECIST v1.1.
Patients received 30 mg/kg PM8002 plus AUC 5 mg/mL/min carboplatin and 500mg/m2 pemetrexed every three weeks (Q3W) for 4 cycles, followed by 30 mg/kg PM8002 and 500mg/m2 pemetrexed as maintenance therapy.
Results: As of 24 July 2024, 64 patients were tested for PD-L1 TPS: 28 (43.8%) were negative, 23 (35.9%) had low expression, and 13 (20.3%) had high expression. All patients were evaluable for safety and efficacy. Overall confirmed ORR was 57.8% (37/64) and DCR was 95.3% (61/64) . In the PD-L1 negative group, the confirmed ORR was 39.3%and DCR was 92.9%. In the PD-L1 low expression group, the confirmed ORR was 60.9%and DCR was 100%. In the PD-L1 high expression group, the confirmed ORR was 92.3%and DCR was 92.3%. Any-grade TRAEs occurred in 98.4% (63/64) and grade ≥ 3 TRAEs occurred in 60.9% (39/64) patients. Any-grade immune-related adverse events (irAEs) occurred in 40.6%(26/64) and grade ≥ 3 irAEs occurred in 6.3% (4/64) patients. 5 patients discontinued PM8002 administration due to TRAEs with 1 TRAE-related death (pneumonia) .
Summarizing the above, PM8002 in combination with chemotherapy showed encouraging antitumor activity and acceptable tolerability in EGFR-mutated NSCLC patients that progressed after prior EGFR-TKI therapy. The anti-tumor activity of PM8002 therapy is positively correlated with tumor PD-L1 expression level.
Example 7: Safety and Efficacy of PM8002 (anti-PD-L1 x VEGF-A bispecific) as a Single Agent or 
Combined with Chemotherapy for the treatment of Hepatocellular Carcinoma (HCC)
The present example is a Phase I/IIa study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) as a single agent for treating HCC. Furthermore, the present example is a Phase II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with chemotherapy (in particular FOLFOX4 [oxaliplatin, leucovorin (which may also be referred to as calcium folinate) and 5-fluorouracil] ) for treating HCC. Both studies assessed the safety and efficacy of the respective treatments as first-line treatments for HCC.
Patients suffering from HCC who had not been treated previously with systemic therapy were included in the two studies. Tumor responses were assessed every 6 weeks for the monotherapy study and every 8 weeks for the combination study. The primary endpoint for both studies was the objective response rate (ORR) based on RECIST v1.1, and a further endpoint was safety assessed by commonly and routinely used standardized procedures as regards the TRAEs.
In the single-agent study, patients received 20 mg/kg PM8002 every two weeks (Q2W) , whereas patients in the combination study received 20 mg/kg PM8002 every two weeks (Q2W) in combination with FOLFOX4 (day 1: oxaliplatin [85 mg/m2, 2-h infusion] plus leucovorin [200 mg/m2, 2-h infusion] , followed by 5-fluorouracil [400 mg/m2, intravenous bolus at hour 2; then 600 mg/m2 for a22-h infusion] ; day 2: leucovorin [200 mg/m2, 2-h infusion] , followed by 5-fluorouracil [400 mg/m2, intravenous bolus at hour 2; then 600 mg/m2 for a 22-h infusion] .
Results: As of April 15, 2024, a total of 46 patients participated in the two studies. 11 patients received at least one dose of PM8002 monotherapy, while 35 patients received at least one dose of the combination therapy. Two patients did not undergo tumor assessment. The ORR for the monotherapy was 27.3%, and 39.4%for the combination therapy. The disease control rates were 100%and 90.9%, respectively. The median progression-free survival was 15.1 months for the monotherapy and 5.8 months for the combination therapy. The median overall survival was 15.4 months for the combination therapy study and not reached for the monotherapy study. Regarding safety, 90.9%of the patients in the monotherapy study and 97.1%of the patients in the combination study experienced treatment-related adverse events (TRAE) , with Grade ≥ 3 adverse events reported in 36.4%and 51.4%of cases for each study, respectively. The most common TRAEs in the monotherapy study were hypertension (45.5%) , proteinuria (45.5%) , and decreased platelet count (36.4%) , while in the combination study, the most common TRAEs were decreased platelet count (71.4%) , decreased neutrophil count (68.6%) , decreased white blood cell count (68.6%) , and anemia (40.0%) .
Summarizing the above, PM8002 as monotherapy and in combination with chemotherapy has shown promising anti-tumor activity and manageable safety for the first line treatment of HCC.
Example 8: Dose simulations based on pharmacokinetic (PK) observations in patients
Overall, 3973 PK observations were collected in 387 Asian patients suffering from advanced solid tumors, SCLC and TNBC, who were treated with PM8002 (comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) on a body-weight-based regimen (up to 45 mg/kg Q2W) . These patients participated in clinical trials for PM8002. The treatment with PM8002 on a body-weight-based regimen resulted in exposures, which have demonstrated an acceptable safety profile with good anti-tumor activity. The main goal of the present simulations was to ensure that the expected flat dose exposures, in particular a flat dose of 2000 mg Q3W, are within the previously established exposures such that it can be concluded that the flat dose exposures also result in an acceptable safety profile with good anti-tumor activity.
Based on the above-mentioned 3973 PK observations, a two-compartment model with linear elimination was developed and this model well described the PK profile in the afore-mentioned patients. The population pharmacokinetic (PopPK) model showed that baseline body weight (BBWT) , albumin (ALB) , alkaline phosphatase (ALP) , estimated glomerular filtration rate (eGFR) , and sex were identified as covariates on the clearance in central compartment. Clearance (CL) , BBWT and sex were identified as covariates on the central volume (Vc) . Although identified as a covariate, the incidence of treatment-emergent antidrug antibodies (ADA) positivity to PM8002 was low. No other covariates (e.g., tumor type, combination therapy) were identified as significant covariates on the PK parameters of PM8002. Based on the comparison of exposure parameters with typical values of the model, the covariates identified in the population PK model (BBWT, ALB, ALP, eGFR, and sex) had no clinically meaningful effect on PM8002 exposures, and no dose adjustment is required based on the above covariates.
To simulate PM8002 exposures in a Western population, the demographic information was adopted from a published article of pembrolizumab in 2841 patients with melanoma or NSCLC, where the body weight in patients with cancer was 77.2±18.9 kg (mean±SD) (Li H, Yu J, Liu C, et al. Time dependent pharmacokinetics of pembrolizumab in patients with solid tumor and its correlation with best overall response. J Pharmacokinet Pharmacodyn 2017; 44 (5) : 403-14) . To avoid extreme values of body weight during the simulation, the body weight range was set at 40–120 kg, which was used as the basis to generate the random values of individual body weights for this population. Based on available clinical data collected in Asian patients, the eGFR distribution was 92.3±26.5 mL/min (mean±SD) . The baseline ALB distribution was 42.2±4.11 g/L (mean±SD) . The baseline ALP distribution was 106.9±55.4 IU/L (mean±SD) . Using body weight information obtained from the Li et al. 2017 publication as well as eGFR, baseline ALB, and baseline ALP from the clinical data, 1000 patients (male: female = 1: 1) were simulated. The sampling times were set at pre-dose, immediately after the end of infusion, 2 h, 6 h, 24 h, 48 h, 168 h, 336 h after the start of infusion, and PK profiles were simulated for 100 times in all patients following administration of weight-based dosing (20 mg/kg Q2W/30 mg/kg Q3W) and flat dosing regimens (1400 mg Q2W/2100 mg Q3W, converted using a body weight of 70 kg) for 20 weeks, with 1.2 h (the mean of actual infusion time) infusion time, and the steady-state exposure parameters (area under the curve during the dosing interval at steady-state [AUCtau, ss] , maximum observed concentration at steady-state [Cmax, ss] and minimum observed concentration at steady-state [Cmin, ss] ) were calculated.
Boxplots of AUCtau, ss at 30 mg/kg Q3W vs. 2100 mg Q3W dosing regimens are presented in Figure 2(A) , and boxplots of corresponding Cmax, ss and Cmin, ss are presented in Figure 2 (B) and 2 (C) , respectively. The calculated AUCtau, ss based on the simulated data following Q2W administration of different weight-based dosing vs. flat dosing regimens are summarized in Table 7, and the calculated Cmax, ss and Cmin, ss are summarized in Table 8 and Table 9, respectively.
Table 7: Summary of the calculated AUCtau, ss (μg. day/mL) based on the simulated data following Q2W/Q3W 
administration of different weight-based dosing vs. flat dosing regimens.

Data are presented as median (25th -75th quartile) .
Table 8: Summary of the calculated cmax, ss (μg/mL) based on the simulated data following Q2W/Q3W 
administration of different weight-based dosing vs. flat dosing regimens.

Data are presented as median (25th -75th quartile) .
Table 9: Summary of the calculated cmin, ss (μg/mL) based on the simulated data following Q2W/Q3W 
administration of different weight-based dosing vs. flat dosing regimens.

Data are presented as median (25th -75th quartile) .
Based on these results, the steady-state exposure parameters are similar in the majority of the population (body weight range of 60–100 kg) . The exposures were slightly higher in participants with body weight at the higher extremity when administrated on a weight-based basis, and the exposures were higher in participants with body weight at the lower extremity when administrated on a flat dose basis. Overall, given that the simulations show that a 2100 mg Q3W PM8002 administration regimen results in exposures that are within the previously established exposures (shown to be safe and effective) , any regimen based on a (slightly) lower dose, such as in particular a 2000 mg Q3W PM8002 administration regimen, is within the previously established exposures by consequence. Furthermore, an amount of 2100 mg is typically rounded down to 2000 mg /vial because of the vial size.
Separately, the two-compartment model with linear elimination was used to simulate PM8002 exposure in a Western population, wherein 460 patients were simulated. PK profiles were simulated in all patients following administration of weight-based dosing (20 mg/kg Q2W) and flat dosing regimens (1200 mg Q2W/1500 mg Q2W, converted using a body weight of 60 kg and 75 kg, respectively) , and the steady-state exposure parameters were calculated. Boxplots of AUCtau, ss at 20 mg/kg Q2W vs. 1200 mg Q2W and 1500 mg Q2W dosing regimens are presented in Figures 1 (A) to (C) .
Example 9: Efficacy and safety of PM8002 (anti-PD-L1 x VEGF-A bispecific) combined with 
chemotherapy in patients with unresectable malignant mesothelioma
The present example is a phase II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) in combination with chemotherapy (in particular carboplatin and pemetrexed) as a therapy for patients suffering from unresectable malignant mesothelioma (malignant pleural mesothelioma [MPM] and malignant peritoneal mesothelioma [MPeM] ) as a first-line treatment to assess the safety and efficacy of this treatment.
The primary endpoints of this study were the objective response rate (ORR) assessed by investigators according to per RECIST v1.1 for MPeM and mRECIST 1.1 for MPM, and safety assessed by the investigators according to Common Terminology Criteria for Adverse Events Version 5.0 (CTCAE V5.0) .
Patients received 30 mg/kg PM8002 IV plus AUC 5 mg/mL/min carboplatin and 500 mg/m2 pemetrexed or 75 mg/m2 cisplatin and 500 mg/m2 pemetrexed every three weeks (Q3W) for 4 to 6 cycles, followed by 30 mg/kg PM8002 as maintenance therapy.
Results: As of 20 December 2024, 31 patients (median age of 58 years [ranging from 43 to 71 years] , 80.6% ECOG PS 1 and 83.9%with metastatic disease, 23 suffering from MPM, 8 from MPeM) had been enrolled and were treated, with the median exposure duration at the cutoff date of 20 December 2024 of 16.0 months (95%CI 8.1, 19.5) and median follow-up time of 19.3 months (95%CI 17.3, 20.9) .
In 23 patients with MPM, one patient had a complete response (CR) and nine patients had partial responses (PRs) as best overall response (BOR) , resulting in a confirmed ORR (cORR) of 43.5%. Ten patients had stable disease (SD) and one patient non-CR/non-PD, giving a disease control rate (DCR) of 87.0%. Median progression-free survival (mPFS) was 11.8 months, and median duration of response (mDOR) was 11.8 months. The 12 months overall survival (OS) rate was 82.6% (95%CI 60.1, 93.1) , with median OS not yet reached. Among 13 patients with MPM of epithelioid histology, cORR was 30.8%, DCR was 84.6%and mPFS was 16.6 months. Among eight patients with MPeM, six patients had PRs as BOR, leading to a cORR of 75.0%. Two patients had SD, resulting in a DCR of 100%; median DOR was 16.3 months. Median PFS and OS were not yet reached, with an OS rate of 62.5% (95%CI 22.9, 86.1) at 12 months. Six patients with MPeM of epithelioid histology displayed a cORR of 83.3%, DCR of 100%and mPFS of 19.5 months.
All patients experienced treatment-related adverse events (TRAEs) , 93.5%of patients (29/31) of Grade (G) 3-4. Five patients (16.1%) had G 3-4 treatment-related serious adverse events (SAEs) . Five patients (16.1%) experienced an immune-related adverse event (irAE) , one patient (3.2%) of G 3-4. The most common TRAEs were decreased neutrophil count (27 patients, 87.1%) , decreased white blood cell count (26 patients, 83.9%) , proteinuria (24 patients, 77.4%) , anemia (23 patients, 74.2%) , decreased platelet count (19 patients, 61.3%) , and nausea (16 patients, 51.6%) . Six patients discontinued treatment due to TRAEs, no treatment-related deaths occurred.
Summarizing the above, PM8002 in combination with chemotherapy as first line treatment for mesothelioma showed encouraging efficacy, including in tumors of epithelioid histology. Adverse events were consistent with those expected for the treatment regimen.
Example 10: Safety and Efficacy of PM8002 (anti-PD-L1 x VEGF-A bispecific) Combined with 
Chemotherapy for the second-line treatment of neuroendocrine neoplasms (NEN)
The present example is a Phase II study of a bispecific anti-VEGF-A anti-PD-L1 antibody (i.e., PM8002 comprising the heavy and light chain amino acid sequence set forth in SEQ ID NO: 16 and 17) combined with chemotherapy (in particular FOLFIRI [irinotecan, leucovorin (which may also be referred to as calcium folinate) and 5-fluorouracil] ) for treating NEN. The study assessed the safety and efficacy of second-line treatment for NEN.
Patients suffering from NEN who had been treated previously with systemic therapy were included into the study. Tumor responses were assessed every 6 weeks for the participants. The primary endpoint was the objective response rate (ORR) based on RECIST v1.1, and a further endpoint was safety assessed by commonly and routinely used standardized procedures as regards the TRAEs.
In the study, patients received 20 mg/kg PM8002 every two weeks (Q2W) , in combination with FOLFIRI (irinotecan, 180 mg/m2, intravenous infusion, D1, Q2W; calcium folinate, 400 mg/m2, intravenous infusion, D1, Q2W; 5-fluorouracil, 400 mg/m2 IV push, D1, followed by continuous IV infusion of 1200 mg/m2/day D1, 2 (total 2400 mg/m2, 46-48 h, Q2W) .
Results: As of January 17, 2025, a total of 15 patients participated in the study. Four patients did not undergo tumor assessment. The ORR for trial treatment was 81.8%. The disease control rate was 90.9%. The median progression-free survival was not reached for the combination therapy. The median overall survival was 7.69 months for the study. Regarding safety, 86.7%of the patients in the study experienced treatment-related adverse events (TRAE) , with Grade ≥ 3 adverse events reported in 60%of cases for in the study. The most common TRAEs in the study were neutropenia (68.8%) , leucopenia (60%) , and weight loss (40%) .
Summarizing the above, PM8002 in combination with chemotherapy has shown promising anti-tumor activity and manageable safety for the second-line treatment of NEN.

Claims (49)

  1. A method of treating cancer in a subject, the method comprising administering to the subject:
    a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
    b. a chemotherapy agent.
  2. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction for use in a method of treating cancer, the method comprising administering to the subject:
    a. the binding agent; and
    b. a chemotherapy agent.
  3. A chemotherapy agent for use in a method of treating cancer, the method comprising administering to the subject:
    a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
    b. the chemotherapy agent.
  4. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use in a method of treating cancer in a subject, the method comprising administering to the subject:
    a. the binding agent; and
    b. the chemotherapy agent.
  5. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is selected from the group consisting of non-small cell lung cancer, small cell lung cancer, triple-negative breast cancer, malignant mesothelioma, hepatocellular cancer, neuroendocrine neoplasm, melanoma, liver cancer, stomach cancer, renal cancer, urothelial cancer, cervical cancer, colorectal cancer, ovarian cancer, colon cancer, esophageal cancer, head and neck cancer and pancreatic cancer.
  6. The method according to claim 1 or 5, the binding agent for use according to claim 2 or 5, the chemotherapy agent for use according to claim 3 or 5, or the binding agent and the chemotherapy agent for use according to claim 4 or 5, wherein the chemotherapy agent is selected from the group consisting of a (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  7. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is small cell lung cancer and the chemotherapy agent is (i) a taxane, preferably paclitaxel; (ii) a topoisomerase inhibitor, preferably topotecan; or (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin; and a topoisomerase inhibitor, preferably etoposide.
  8. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is non-small cell lung cancer and the chemotherapy agent is (i) a taxane, preferably docetaxel; (ii) a combination of a platinum-based chemotherapy agent, preferably carboplatin; and an antifolate chemotherapy agent, preferably pemetrexed; or (iii) a combination of a platinum-based chemotherapy agent, preferably carboplatin; and a taxane, preferably paclitaxel.
  9. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is triple-negative breast cancer and the chemotherapy agent is (i) a taxane, preferably paclitaxel or nab-paclitaxel; (ii) a combination of an antimetabolite chemotherapy agent, preferably gemcitabine, and a platinum-based chemotherapy agent, preferably carboplatin; or (iii) a microtubule inhibitor, preferably eribuline.
  10. The method according to claim 9, the bispecific antibody for use according to claim 9, the chemotherapy agent for use according to claim 9, or the binding agent and the chemotherapy agent for use according to claim 9, wherein the subject has a PD-L1 expression before the treatment that provides a score of ≥1 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
  11. The method according to claim 10, the bispecific antibody for use according to claim 10, the chemotherapy agent for use according to claim 10, or the binding agent and the chemotherapy agent for use according to claim 10, wherein the subject has a PD-L1 expression before the treatment that provides a score of from 1 to <10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
  12. The method according to claim 9, the bispecific antibody for use according to claim 9, the chemotherapy agent for use according to claim 9, or the binding agent and the chemotherapy agent for use according to claim 9, wherein the subject has a PD-L1 expression before the treatment that provides a score of <10 when determined by the combined positive score (CPS) or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm.
  13. The method according to any one of claims 10 to 12, the bispecific antibody for use according to any one of claims 10 to 12, the chemotherapy agent for use according to any one of claims 10 to 12, or the binding agent and the chemotherapy agent for use according to any one of claims 10 to 12, wherein the another integrating scoring algorithm is the tumour area positivity (TAP) score or the tumour proportion score (TPS) .
  14. The method according to any one of claims 10 to 12, the bispecific antibody for use according to any one of claims 10 to 12, the chemotherapy agent for use according to any one of claims 10 to 12, or the binding agent and the chemotherapy agent for use according to any one of claims 10 to 12, wherein the CPS has been determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:
    wherein
    (i) the test sample is a cancer tissue sample; and/or
    (ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour infiltrating) lymphocytes and macrophages; and/or
    (iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by using the 22C3 antibody) ; and/or
    (iv) viable tumour cells are determined by staining with a viability dye; and/or
    (v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 staining lymphocytes and PD-L1 staining macrophages
  15. The method according to claim 13, the bispecific antibody for use according to claim 13, the chemotherapy agent for use according to claim 13, or the binding agent and the chemotherapy agent for use according to claim 13, wherein the PD-L1 expression score has been determined using a TAP scoring algorithm by determining in a test sample of the subject the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:
    wherein
    (i) the test sample is a cancer sample; and/or
    (ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated stroma containing tumour-associated immune cells; and/or
    (iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
    (iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive tumour cells and immune cells; and/or
    (v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 positive tumour cells and immune cells relative to the tumour area expressed as %.
  16. The method according to claim 13, the bispecific antibody for use according to claim 13, the chemotherapy agent for use according to claim 13, or the binding agent and the chemotherapy agent for use according to claim 13, wherein the PD-L1 expression score has been determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:
    wherein
    (i) the test sample is a cancer tissue sample; and/or
    (ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
    (iii) viable tumour cells are determined by staining with a viability dye; and/or
    (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
  17. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is malignant mesothelioma and the chemotherapy agent is a combination of (i) a platinum-based chemotherapy agent, preferably carboplatin, and (ii) an antifolate chemotherapy agent, preferably pemetrexed.
  18. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is hepatocellular cancer and the chemotherapy agent is a combination of (i) a platinum-based chemotherapy agent, preferably oxaliplatin and (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, optionally administered together with folic acid or a derivative thereof, preferably leucovorin.
  19. The method according to claim 1, the binding agent for use according to claim 2, the chemotherapy agent for use according to claim 3, or the binding agent and the chemotherapy agent for use according to claim 4, wherein the cancer is neuroendocrine neoplasm and the chemotherapy agent is a combination of (i) a topoisomerase inhibitor, preferably irinotecan, and (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, optionally administered together with folic acid or a derivative thereof, preferably leucovorin.
  20. The method according to any one of claims 1 and 5 to 19, the binding agent for use according to any one of claims 2 and 5 to 19, the chemotherapy agent for use according to any one of claims 3 and 5 to 19, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 19, wherein overall survival is increased in the subject compared to (a) (i) the chemotherapy agent or (a) (ii) the binding agent or (a) (iii) an anti-PD-L1 antibody or (a) (iv) an anti-PD-1 antibody treatment alone or compared to (b) (i) a standard treatment comprising the chemotherapy agent and an anti-PD-L1 antibody or (b) (ii) the chemotherapy agent and an anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  21. The method according to any one of claims 1 and 5 to 20, the bispecific antibody for use according to any one of claims 2 and 5 to 20, the chemotherapy agent for use according to any one of claims 3 and 5 to 20, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 20, wherein median progression-free survival is increased in the subject compared to (a)(i) the chemotherapy agent or (a) (ii) the binding agent or (a) (iii) an anti-PD-L1 antibody or (a) (iv) an anti-PD-1 antibody treatment alone or compared to (b) (i) a standard treatment comprising the chemotherapy agent and an anti-PD-L1 antibody or (b) (ii) the chemotherapy agent and an anti-PD-1 antibody, wherein the anti-PD-L1 antibody is preferably atezolizumab and the anti-PD-1 antibody is preferably pembrolizumab.
  22. The method according to any one of claims 1 and 5 to 21, the binding agent for use according to any one of claims 2 and 5 to 21, the chemotherapy agent for use according to any one of claims 3 and 5 to 21, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 21, wherein the binding agent and the chemotherapy agent are administered separately.
  23. The method according to any one of claims 1 and 5 to 22, the binding agent for use according to any one of claims 2 and 5 to 22, the chemotherapy agent for use according to any one of claims 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 22, wherein the binding agent is administered at a dosage of from about 1 mg/kg to 100 mg/kg or at a dosage of from about 1000 mg to 2000 mg.
  24. The method according to any one of claims 1 and 5 to 22, the binding agent for use according to any one of claims 2 and 5 to 22, the chemotherapy agent for use according to any one of claims 3 and 5 to 22, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 22, wherein the binding agent and/or the chemotherapy agent is/are administered intravenously, preferably wherein the binding agent and the chemotherapy agent are administered intravenously.
  25. The method according to any one of claims 1 and 5 to 24, the binding agent for use according to any one of claims 2 and 5 to 24, the chemotherapy agent for use according to any one of claims 3 and 5 to 24, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 24, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
  26. The method according to claim 25, the binding agent for use according to claim 25, the chemotherapy agent for use according to claim 25, or the binding agent and the chemotherapy agent for use according to claim 25, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
  27. The method according to any one of claims 1 and 5 to 24, the binding agent for use according to any one of claims 2 and 5 to 24, the chemotherapy agent for use according to any one of claims 3 and 5 to 24, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 24, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
  28. The method according to claim 27, the binding agent for use according to claim 27, the chemotherapy agent for use according to claim 27, or the binding agent and the chemotherapy agent for use according to claim 27, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
  29. The method according to any one of claims 1 and 5 to 24, the binding agent for use according to any one of claims 2 and 5 to 24, the chemotherapy agent for use according to any one of claims 3 and 5 to 24, or the binding agent and the chemotherapy agent for use according to any one of claims 4 to 24, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
  30. A binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent for use as medicament, wherein the chemotherapy agent is selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  31. A composition comprising
    a. a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
    b. a chemotherapy agent selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  32. A kit comprising
    a. a composition comprising a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and
    b. a composition comprising a chemotherapy agent selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  33. The binding agent and the chemotherapy agent for use according to claim 30, the composition according to claim 31, or the kit according to claim 32, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
  34. The binding agent and the chemotherapy agent according to claim 33, the composition according to claim 33, or the kit according to claim 33, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
  35. The binding agent and the chemotherapy agent for use according to claim 30, the composition according to claim 31, or the kit according to claim 32, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
  36. The binding agent and the chemotherapy agent according to claim 35, the composition according to claim 35, or the kit according to claim 35, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
  37. The binding agent and the chemotherapy agent according to claim 30, the composition according to claim 31, or the kit according to claim 32, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
  38. The composition according to any one of claims 31 and 33 to 37, or the kit according to any one of claims 32 to 37 for use as medicament.
  39. A method for determining whether a cancer in a subject is susceptible to treatment with a binding agent comprising (i) a first binding region which binds to PD-1, PD-L1, or both and which antagonizes the PD-1/PD-L1 interaction; and (ii) a second binding region which binds to VEGF, VEGF receptor (VEGFR) , or both and which antagonizes the VEGF/VEGFR interaction; and a chemotherapy agent, wherein the method comprises detecting in a sample of the subject the PD-L1 expression before the treatment by determining the combined positive scope (CPS) or another score, preferably the tumour area positivity (TAP) score or the tumour proportion score (TPS) , more preferably the TAP score, wherein a CPS of ≥ 1 indicates a PD-L1 expression that renders the subject susceptible to treatment with the bispecific antibody and the chemotherapy, or a score that is indicative of such a PD-L1 expression when determined by another integrating scoring algorithm, preferably when determined by the TAP score or the TPS, more preferably when determined by the TAP score.
  40. The method according to claim 39, wherein the cancer is selected from the group consisting of small cell lung cancer, non-small cell lung cancer, triple-negative breast cancer, malignant mesothelioma, hepatocellular cancer, neuroendocrine neoplasm, melanoma, liver cancer, stomach cancer, renal cancer, urothelial cancer, cervical cancer, colorectal cancer, ovarian cancer, colon cancer, esophageal cancer, head and neck cancer and pancreatic cancer; preferably wherein the cancer is triple-negative breast cancer.
  41. The method according to claim 39 or 40, wherein the chemotherapy agent is selected from the group consisting of (i) a platinum-based chemotherapy agent, preferably carboplatin, cisplatin or oxaliplatin, (ii) an antimetabolite chemotherapy agent, preferably 5-fluorouracil, capecitabine or gemcitabine, (iii) an antifolate chemotherapy agent, preferably pemetrexed or methotrexate; (iv) a taxane, preferably paclitaxel, nab-paclitaxel or docetaxel; (v) a topoisomerase inhibitor, preferably an anthracycline, topotecan, irinotecan or etoposide; (vi) a microtubule inhibitor, preferably vinorelbine, ixabepilone or eribuline; and (vii) combinations thereof.
  42. The method according to any one of claims 39 to 41, wherein the CPS is determined in a test sample of the subject by determining the number of PD-L1 staining cells (tumour cells, lymphocytes, macrophages) and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the CPS for the cancer tissue sample using the formula:
    wherein
    (i) the test sample is a cancer tissue sample; and/or
    (ii) the cancer tissue sample comprises at least 100 viable tumour cells as well as (tumour infiltrating) lymphocytes and macrophages; and/or
    (iii) PD-L1 staining cells are determined by PD-L1 immunohistochemistry staining (preferably by using the 22C3 antibody) ; and/or
    (iv) viable tumour cells are determined by staining with a viability dye; and/or
    (v) the number of PD-L1 staining cells is the sum of PD-L1 staining tumour cells, PD-L1 staining lymphocytes and PD-L1 staining macrophages
  43. The method according to any one of claims 39 to 41, wherein the PD-L1 expression is determined using a TAP scoring algorithm by determining in a test sample of the subject the percentage of PD-L1 positive tumour cells and immune cells per tumour area in a cancer tissue sample from the subject; and calculating the TAP for the cancer tissue sample using the formula:
    wherein
    (i) the test sample is a cancer sample; and/or
    (ii) the tumour area is the area occupied by all viable tumour cells and the tumour-associated stroma containing tumour-associated immune cells; and/or
    (iii) PD-L1 positive tumour cells and immune cells are determined by PD-L1 immunohistochemistry staining (preferably by using the anti-PD-L1 antibody SP263) ; and/or
    (iv) PD-L1 positive tumour cells and immune cells means the area covered by such PD-L1 positive tumour cells and immune cells; and/or
    (v) %PD-L1 positive tumour cells and immune cells /tumour area means the area of PD-L1 positive tumour cells and immune cells relative to the tumour area expressed as %.
  44. The method according to any one of claims 39 to 41, wherein the PD-L1 expression is determined using a TPS scoring algorithm by determining in a test sample of the subject the number of viable tumour cells positive for PD-L1 and the total number of viable tumour cells in a cancer tissue sample from the subject; and calculating the TPS for the cancer tissue sample using the formula:
    wherein
    (i) the test sample is a cancer tissue sample; and/or
    (ii) the cancer tissue sample comprises at least 100 viable tumour cells; and/or
    (iii) viable tumour cells are determined by staining with a viability dye; and/or
    (iv) PD-L1 positive cells are determined by PD-L1 immunohistochemistry staining.
  45. The method according to any one of claims 39 to 44, wherein the binding agent comprises (i) a first binding region which binds to PD-L1 and (ii) a second binding region which binds to VEGF.
  46. The method according to claim 45, wherein the binding agent is selected from the group consisting of the binding agents C1, C3a, and C3b provided in Table 1, SYN-2510 (ImmuneOnco/Instil Bio) , HB0025 (Huabo Biopharm) , SG1408 (Hangzhou Sumgen) , B1962 (AP Biosciences /Tasly Pharma) , CVL006 (Convalife) , HC010 (HC Biopharma) , and DR30206 (Zhejiang Doer Bio) .
  47. The method according to any one of claims 39 to 44, wherein the binding agent comprises (i) a first binding region which binds to PD-1 and (ii) a second binding region which binds to VEGF.
  48. The method according to claim 47, wherein the binding agent is selected from the group consisting of the binding agents C2a, C2b, C2c, and C2d provided in Table 1, Ivonescimab/AK112 (AkesoBio/Summit Therapeutics) , AI-081 (OncoC4) , SSGJ-707 (3SBio) , LM-299 (LaNova Medicines/Merck) , JS-207 (Junshi Biosciences) , SCTB14 (Sinocelltech) , and MHB039A (Minghui Pharma) .
  49. The method according to any one of claims 39 to 44, wherein the binding agent is a bispecific antibody binding to PD-L1 and VEGF comprising a heavy chain comprising the amino acid sequence as set forth in SEQ ID NO: 16 and a light chain comprising the amino acid sequence as set forth in SEQ ID NO: 17.
PCT/CN2025/081278 2024-03-07 2025-03-07 Combination therapy comprising a pd-1/pd-l1 and vegf/vegfr binding agent and a chemotherapy for cancer treatment Pending WO2025185733A1 (en)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
PCT/CN2024/080506 WO2025184858A1 (en) 2024-03-07 2024-03-07 Combination therapy comprising a bispecific anti-vegf-a and anti-pd-l1 antibody and a chemotherapy for cancer treatment
CNPCT/CN2024/080506 2024-03-07
CNPCT/CN2024/096309 2024-05-30
CNPCT/CN2024/096438 2024-05-30
CN2024096438 2024-05-30
CN2024096309 2024-05-30
CN2024112562 2024-08-16
CNPCT/CN2024/112562 2024-08-16
ARP240102655A AR134001A1 (en) 2024-03-07 2024-09-30 COMBINATIONAL THERAPY COMPRISING A BISPECIFIC ANTI-VEGF-A AND ANTI-PD-L1 ANTIBODY, AND CHEMOTHERAPY FOR THE TREATMENT OF CANCER
ARP240102655 2024-09-30
CNPCT/CN2024/124994 2024-10-15
PCT/CN2024/124994 WO2025185161A1 (en) 2024-03-07 2024-10-15 Combination therapy comprising a bispecific anti-vegf-a and anti-pd-l1 antibody and a chemotherapy for cancer treatment

Publications (2)

Publication Number Publication Date
WO2025185733A1 true WO2025185733A1 (en) 2025-09-12
WO2025185733A8 WO2025185733A8 (en) 2025-10-02

Family

ID=95154284

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2025/081278 Pending WO2025185733A1 (en) 2024-03-07 2025-03-07 Combination therapy comprising a pd-1/pd-l1 and vegf/vegfr binding agent and a chemotherapy for cancer treatment

Country Status (1)

Country Link
WO (1) WO2025185733A1 (en)

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7070959B1 (en) 1999-06-08 2006-07-04 Regeneron Pharmaceuticals, Inc. Modified chimeric polypeptides with improved pharmacokinetic properties
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
US9709568B2 (en) 2012-12-21 2017-07-18 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (PD-L1)
US20220002418A1 (en) * 2019-04-01 2022-01-06 Huabo Biopharm (Shanghai) Co., Ltd. Anti-pd-l1/vegf bifunctional antibody and use thereof
WO2022042719A1 (en) 2020-08-31 2022-03-03 普米斯生物技术(珠海)有限公司 Anti-vegf-anti-pd-l1 bispecific antibody, pharmaceutical composition of same, and uses thereof
WO2022111476A1 (en) * 2020-11-24 2022-06-02 普米斯生物技术(珠海)有限公司 ANTI-PD-L1-ANTI-VEGF-ANTI-TGF-β MULTISPECIFIC ANTIBODY, PHARMACEUTICAL COMPOSITION AND USE THEREOF
EP3589754B1 (en) * 2017-03-01 2023-06-28 F. Hoffmann-La Roche AG Diagnostic and therapeutic methods for cancer
WO2023165516A1 (en) * 2022-03-02 2023-09-07 三优生物医药(上海)有限公司 Anti-pd-l1 and vegf bispecific antibody and use thereof
WO2024032664A1 (en) * 2022-08-09 2024-02-15 上海济煜医药科技有限公司 Antibody targeting pd-l1 and vegf and use thereof

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7070959B1 (en) 1999-06-08 2006-07-04 Regeneron Pharmaceuticals, Inc. Modified chimeric polypeptides with improved pharmacokinetic properties
US9212224B2 (en) 2012-05-15 2015-12-15 Bristol-Myers Squibb Company Antibodies that bind PD-L1 and uses thereof
US9709568B2 (en) 2012-12-21 2017-07-18 Merck Sharp & Dohme Corp. Antibodies that bind to human programmed death ligand 1 (PD-L1)
EP3589754B1 (en) * 2017-03-01 2023-06-28 F. Hoffmann-La Roche AG Diagnostic and therapeutic methods for cancer
US20220002418A1 (en) * 2019-04-01 2022-01-06 Huabo Biopharm (Shanghai) Co., Ltd. Anti-pd-l1/vegf bifunctional antibody and use thereof
WO2022042719A1 (en) 2020-08-31 2022-03-03 普米斯生物技术(珠海)有限公司 Anti-vegf-anti-pd-l1 bispecific antibody, pharmaceutical composition of same, and uses thereof
EP4209513A1 (en) * 2020-08-31 2023-07-12 Biotheus Inc. Anti-vegf-anti-pd-l1 bispecific antibody, pharmaceutical composition of same, and uses thereof
WO2022111476A1 (en) * 2020-11-24 2022-06-02 普米斯生物技术(珠海)有限公司 ANTI-PD-L1-ANTI-VEGF-ANTI-TGF-β MULTISPECIFIC ANTIBODY, PHARMACEUTICAL COMPOSITION AND USE THEREOF
WO2023165516A1 (en) * 2022-03-02 2023-09-07 三优生物医药(上海)有限公司 Anti-pd-l1 and vegf bispecific antibody and use thereof
WO2024032664A1 (en) * 2022-08-09 2024-02-15 上海济煜医药科技有限公司 Antibody targeting pd-l1 and vegf and use thereof

Non-Patent Citations (33)

* Cited by examiner, † Cited by third party
Title
"Fundamental Immunology", 1989, RAVEN PRESS
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING CO.
AKINLEYEZOAIB RASOOL, JOURNAL OF HEMATOLOGY&ONCOLOGY, vol. 12, 2019
CHEN, XIAO-JIANG ET AL.: "Prognostic Significance of PD-L1 Expression in Gastric Cancer Subjects with Peritoneal Metastasis", BIOMEDICINES, vol. 11, no. 7, 15 July 2023 (2023-07-15), pages 2003
CHOTHIA ET AL., NATURE, vol. 342, 1989, pages 878 - 883
CHOTHIALESK, J. MOL. BIOL., vol. 196, 1987, pages 901 - 917
CORTES JRUGO HSCESCON DW ET AL.: "Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer", N ENGL J MED., vol. 387, no. 3, 2022, pages 217 - 226, XP093162423, DOI: 10.1056/NEJMoa2202809
DONDELINGER, M. ET AL., FRONTIERS IN IMMUNOLOGY, vol. 9, 2018, pages 2278
EHRENMANN FKAAS QLEFRANC M P: "IMGT/3Dstructure-DB and IMGT/DomainGapAlign: a database and a tool for immunoglobulins or antibodies, T cell receptors, MHC, IgSF and MhcSF[J", NUCLEIC ACIDS RESEARCH, vol. 38, 2009, pages D301 - D307, XP055247165, DOI: 10.1093/nar/gkp946
ELVIN A. KABAT: "Sequences of Proteins of Immunological Interest", vol. 2719, 1991, NATIONAL INSTITUTES OF HEALTH, article "National Institutes of Health (U.S.) Office of the Director"
JAIN, R. K.: "Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy", SCIENCE, vol. 307, 2005, pages 58 - 62, XP002503618, DOI: 10.1126/SCIENCE.1104819
JIANG YZMA DSUO C ET AL.: "Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies", CANCER CELL, vol. 35, no. 3, 2019, pages 428 - 440
JIANG YZMA DSUO C ET AL.: "Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies", CANCER CELL., vol. 35, no. 3, 2019, pages 428 - 440
KOSTELNY ET AL., J. IMMUNOL., vol. 148, 1992, pages 1547 - 1553
KULANGARA, KARINAHANKS, DEBRAWALDROUP, STEPHANIEPELTZ, LINDSAYSHAH, SUPRIYAROACH, CHARLOTTEJUCO, JONATHANEMANCIPATOR, KENNETHSTANF: "Development of the combined positive score (CPS) for the evaluation of PD-L1 in solid tumours with the immunohistochemistry assay PD-L1 IHC 22C3 pharmDx", JOURNAL OF CLINICAL ONCOLOGY, 2017
LEFRANC ET AL., DEV. COMPARAT. IMMUNOL., vol. 27, 2003, pages 55 - 77
LEFRANC ET AL., DEV.COMPARAT.IMMUNOL., vol. 27, 2003, pages 55 - 77
LI HYU JLIU C ET AL.: "Time dependent pharmacokinetics of pembrolizumab in patients with solid tumor and its correlation with best overall response", J PHARMACOKINET PHARMACODYN, vol. 44, no. 5, 2017, pages 403 - 14, XP036333210, DOI: 10.1007/s10928-017-9528-y
LIU, CHUNYAN ET AL.: "Tumour Area Positivity (TAP) score of programmed death-ligand 1 (PD-L1): a novel visual estimation method for combined tumour cell and immune cell scoring", DIAGNOSTIC PATHOLOGY, vol. 18, no. 1, 19 April 2023 (2023-04-19), pages 48
LIU, R. ET AL., ANTIBODIES, vol. 9, no. 4, 2020, pages 64
PARK ET AL., CANCER RES. TREAT., vol. 52, no. 3, 2020, pages 661 - 670
PRINCIPLES FOR CODEVELOPMENT OF AN IN VITRO COMPANION DIAGNOSTIC DEVICE WITH A THERAPEUTIC PRODUCT, 15 July 2016 (2016-07-15)
RAKESH R. RAMJIAWANARJAN W. GRIFFIOENDAN G. DUDA, ANGIOGENESIS, vol. 20, no. 2, 2017, pages 185 - 204
ROACH CZHANG NCORIGLIANO E ET AL.: "Development of a Companion Diagnostic PD-L1 Immunohistochemistry Assay for Pembrolizumab Therapy in Non-Small-cell Lung Cancer", APPL IMMUNOHISTOCHEM MOL MORPHOL., vol. 24, no. 6, 2016, pages 392 - 397, XP055867103, DOI: 10.1097/PAI.0000000000000408
ROBERT ET AL., LANCET, vol. 384, 2014, pages 1109 - 17
ROBERT ET AL., N ENGL J MED, vol. 372, 2015, pages 2521 - 30
SONGSIVILAI ET AL., CLIN. EXP. IMMUNOL., vol. 79, 1990, pages 315 - 321
TOPALIAN ET AL., J CLIN ONCOL, vol. 32, 2014, pages 1020 - 30
TOPALIAN ET AL., N ENGL J MED, vol. 366, 2012, pages 2443 - 54
ULAS, EZGI B ET AL.: "Predictive Value of Combined Positive Score and Tumour Proportion Score for Immunotherapy Response in Advanced NSCLC", JTO CLINICAL AND RESEARCH REPORTS, vol. 4, no. 9, 25 May 2023 (2023-05-25), pages 100532
WILKINSON, I. ET AL., FCPLOS ONE, vol. 16, no. 12, 2021, pages 0260954
WOLCHOK ET AL., N ENGL J MED, vol. 369, 2013, pages 122 - 33
YAMASHITA ET AL., GASTRIC CANCER, vol. 23, 2020, pages 95 - 104

Also Published As

Publication number Publication date
WO2025185733A8 (en) 2025-10-02

Similar Documents

Publication Publication Date Title
JP6894952B2 (en) Combination of PD-1 antagonist and VEGFR inhibitor for cancer treatment
JP6876629B2 (en) PD-L1 antagonist combination therapy
JP6788600B2 (en) A combination of PD-1 antagonist and VEGFR / FGFR / RET tyrosine kinase inhibitor for the treatment of cancer
JP6840127B2 (en) Combination of anti-PD-1 antibody and anti-M-CSF antibody in the treatment of cancer
TWI457135B (en) Combination of carboplatin,gemcitabine and anti-vegf antibody for the treatment of recurrent platinum-sensitive advanced epithelial ovarian,fallopian tube or primary peritoneal cancer
JP2025029025A (en) Methods for treating lung cancer using a combination of anti-PD-1 and anti-CTLA-4 antibodies
KR20160108568A (en) Combination of a pd-1 antagonist and an ido1 inhibitor for treating cancer
KR20150094617A (en) Anti-folr1 antibody
WO2023088221A1 (en) Combination therapy of claudin 18.2 antagonist and pd-1/pd-l1 axis inhibitor
JP2019517498A (en) Anti-PD-1 antibody for use in a method of treating recurrent small cell lung cancer
KR20230118713A (en) Anti-pd-1 antibody for use in a method of treating a tumor
JP2024028805A (en) Combination therapy using anti-IL-8 and anti-PD-1 antibodies for cancer treatment
JP2022507294A (en) CDCP1-Targeted therapy
WO2020211804A1 (en) Use of anti-pd-1 antibody in preparation of medicament for treating solid tumors
JP2020536923A (en) Compositions and Methods for Treating Diffuse Large B-Cell Lymphoma
US20240218066A1 (en) Use of anti-pd-1 antibody in combination with first-line chemotherapy in treatment of advanced non-small cell lung cancer
JP2022512866A (en) Anti-LAG3 antibody dosing regimen for treating cancer and combination therapy with anti-PD-1 antibody
KR20210034622A (en) Lung cancer treatment method using PD-1 axis binding antagonist, anti-metabolite, and platinum agent
JP2021533090A (en) Combination therapy
WO2025185733A1 (en) Combination therapy comprising a pd-1/pd-l1 and vegf/vegfr binding agent and a chemotherapy for cancer treatment
WO2025185161A1 (en) Combination therapy comprising a bispecific anti-vegf-a and anti-pd-l1 antibody and a chemotherapy for cancer treatment
US20250044295A1 (en) Biomarkers for predicting eligibility for an anti-ilt4 and anti-pd-1 combination therapy
WO2025184858A1 (en) Combination therapy comprising a bispecific anti-vegf-a and anti-pd-l1 antibody and a chemotherapy for cancer treatment
TW202535957A (en) Combination therapy comprising a bispecific anti-vegf-a and anti-pd-l1 antibody and a chemotherapy for cancer treatment
WO2024251051A1 (en) Combination therapies involving claudin 18.2 antagonists for treatment of cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 25714465

Country of ref document: EP

Kind code of ref document: A1