[go: up one dir, main page]

HK40081400A - Antibodies and fusion proteins that bind to ccr8 and uses thereof - Google Patents

Antibodies and fusion proteins that bind to ccr8 and uses thereof Download PDF

Info

Publication number
HK40081400A
HK40081400A HK62023069692.9A HK62023069692A HK40081400A HK 40081400 A HK40081400 A HK 40081400A HK 62023069692 A HK62023069692 A HK 62023069692A HK 40081400 A HK40081400 A HK 40081400A
Authority
HK
Hong Kong
Prior art keywords
antibody
seq
amino acid
acid sequence
cancer
Prior art date
Application number
HK62023069692.9A
Other languages
Chinese (zh)
Inventor
L‧L‧麦格拉思
F‧德皮斯
C‧胡
L‧G‧普雷斯塔
J‧A‧巴奇
Original Assignee
Gilead Sciences, Inc.
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
Application filed by Gilead Sciences, Inc. filed Critical Gilead Sciences, Inc.
Publication of HK40081400A publication Critical patent/HK40081400A/en

Links

Description

Antibodies and fusion proteins binding to CCR8 and uses thereof
Cross-reference declaration
Priority of U.S. provisional application 62/976,869 filed on 2/14/2020 and U.S. provisional application 63/130,157 filed on 12/23/2020 is claimed in this application under 35 U.S.C. § 119 (e). The entire contents of both provisional applications are incorporated herein by reference.
Sequence listing
This application contains a sequence listing that has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. The ASCII copy was created at 2021, 2, month, 7, under the name 119835-0178_ SL.txt, and has a size of 157 KB.
Technical Field
Antibodies and fusion proteins that bind to CCR8 are provided. Also provided are therapeutic methods comprising administering such antibodies and/or fusion proteins.
Background
Chemokine (C-C motif) receptor 8(CCR8) belongs to the family of G Protein Coupled Receptors (GPCRs). CCR8 is expressed primarily on tumor regulatory t (Treg) cells, a class of immunosuppressive cells found in the tumor microenvironment.
There remains a need for antibodies and fusion proteins that bind CCR8 for use in the treatment of cancer and other diseases and disorders.
Disclosure of Invention
The present disclosure describes an isolated antibody that binds to human CCR8, wherein the antibody comprises:
a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 12, HCDR2 comprising the amino acid sequence of SEQ ID NO. 13, HCDR3 comprising the amino acid sequence of SEQ ID NO. 14, LCDR1 comprising the amino acid sequence of SEQ ID NO. 15, LCDR2 comprising the amino acid sequence of SEQ ID NO. 16, and LCDR3 comprising the amino acid sequence of SEQ ID NO. 17;
b) HCDR1 comprising the amino acid sequence of SEQ ID NO. 24, HCDR2 comprising the amino acid sequence of SEQ ID NO. 25, HCDR3 comprising the amino acid sequence of SEQ ID NO. 26, LCDR1 comprising the amino acid sequence of SEQ ID NO. 27, LCDR2 comprising the amino acid sequence of SEQ ID NO. 28, and LCDR3 comprising the amino acid sequence of SEQ ID NO. 29;
c) HCDR1 comprising the amino acid sequence of SEQ ID NO. 36, HCDR2 comprising the amino acid sequence of SEQ ID NO. 37, HCDR3 comprising the amino acid sequence of SEQ ID NO. 38, LCDR1 comprising the amino acid sequence of SEQ ID NO. 39, LCDR2 comprising the amino acid sequence of SEQ ID NO. 40, and LCDR3 comprising the amino acid sequence of SEQ ID NO. 41;
d) HCDR1 comprising the amino acid sequence of SEQ ID NO. 48, HCDR2 comprising the amino acid sequence of SEQ ID NO. 49, HCDR3 comprising the amino acid sequence of SEQ ID NO. 50, LCDR1 comprising the amino acid sequence of SEQ ID NO. 51, LCDR2 comprising the amino acid sequence of SEQ ID NO. 52, and LCDR3 comprising the amino acid sequence of SEQ ID NO. 53; or
e) HCDR1 comprising the amino acid sequence of SEQ ID NO. 60, HCDR2 comprising the amino acid sequence of SEQ ID NO. 61, 72 or 78, HCDR3 comprising the amino acid sequence of SEQ ID NO. 62, 73 or 79, LCDR1 comprising the amino acid sequence of SEQ ID NO. 63, LCDR2 comprising the amino acid sequence of SEQ ID NO. 64 and LCDR3 comprising the amino acid sequence of SEQ ID NO. 65; or
f) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100, HCDR2 comprising the amino acid sequence of SEQ ID NO 85, HCDR3 comprising the amino acid sequence of SEQ ID NO 86, LCDR1 comprising the amino acid sequence of SEQ ID NO 87, LCDR2 comprising the amino acid sequence of SEQ ID NO 88 and LCDR3 comprising the amino acid sequence of SEQ ID NO 89.
The present application also describes an isolated antibody, wherein the antibody comprises:
a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 60, HCDR2 comprising the amino acid sequence of SEQ ID NO. 61, 72 or 78, HCDR3 comprising the amino acid sequence of SEQ ID NO. 62, 73 or 79, LCDR1 comprising the amino acid sequence of SEQ ID NO. 63, LCDR2 comprising the amino acid sequence of SEQ ID NO. 64 and LCDR3 comprising the amino acid sequence of SEQ ID NO. 65; or
b) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100, HCDR2 comprising the amino acid sequence of SEQ ID NO 85, HCDR3 comprising the amino acid sequence of SEQ ID NO 86, LCDR1 comprising the amino acid sequence of SEQ ID NO 87, LCDR2 comprising the amino acid sequence of SEQ ID NO 88 and LCDR3 comprising the amino acid sequence of SEQ ID NO 89.
In some embodiments, the antibody comprises:
a) a heavy chain variable region (VH) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 68 or 74 and a light chain variable region (VL) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 69 or 75; or
b) A heavy chain variable region (VH) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 92 or 96, and a light chain variable region (VL) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 93 or 97.
In some embodiments, the antibody comprises:
a) a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO 68 or 74 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO 69 or 75; or
b) A heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO:92 or 96, and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO:93 or 97.
In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody is a full-length antibody. In some embodiments, the antibody is an IgG1 or IgG3 antibody.
In some embodiments, the antibody comprises:
a) a Heavy Chain (HC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 70 or 76, and a Light Chain (LC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 71 or 77; or
b) A Heavy Chain (HC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 94 or 98, and a Light Chain (LC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 95 or 99.
In some embodiments, the antibody comprises a Heavy Chain (HC) comprising the amino acid sequence of SEQ ID NO 70 or 76 and a Light Chain (LC) comprising the amino acid sequence of SEQ ID NO 71 or 77; or a Heavy Chain (HC) comprising the amino acid sequence of SEQ ID NO:94 or 98 and a Light Chain (LC) comprising the amino acid sequence of SEQ ID NO:95 or 99.
In some embodiments, the antibody comprises at least one modification that enhances cell killing. In some embodiments, the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC). In some embodiments, the at least one modification is afucosylation. In some embodiments, the at least one modification is one or more heavy chain constant region mutations at one or more positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334 and P396. In some embodiments, the one or more heavy chain constant region mutations are one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L. In some embodiments, the one or more heavy chain constant region mutations are selected from the group consisting of: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E. In some embodiments, the at least one modification is galactosylation.
In some embodiments, the antibody has an affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75pM, or less than 50pM, or less than 25pM D ) (as determined by kinetic exclusion assay (i.e., KinExA)) binds to human CCR 8. In some embodiments, the antibody has an on-cell affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75, or less than 50pM, or less than 25pM D ) Binds human CCR8 as determined by, for example, a kinetic exclusion assay (i.e., KinExA).
The disclosure also describes isolated nucleic acids encoding the antibodies. In some embodiments, the vector comprises an isolated nucleic acid. In some embodiments, the host cell comprises a nucleic acid or vector. In some embodiments, the host cell expresses the antibody. In some embodiments, the host cell is engineered to produce an afucosylated antibody.
In some embodiments, a method of producing an antibody that binds CCR8 includes culturing a host cell under conditions suitable for expression of the antibody. In some embodiments, the method further comprises isolating the antibody.
In some embodiments, a fusion protein comprises (a) CCL1, or an active fragment thereof, or MC148, or an active fragment thereof, and (b) an Fc region. In some embodiments, the Fc region is an IgG1 or IgG3Fc region. In some embodiments, the Fc region comprises the amino acid sequence of SEQ ID NO 4.
In some embodiments, the Fc region comprises at least one modification that enhances cell killing. In some embodiments, the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC). In some embodiments, the at least one modification is afucosylation. In some embodiments, the at least one modification is one or more Fc region mutations at one or more positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334, and P396. In some embodiments, the one or more Fc region mutations are one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L. In some embodiments, the one or more Fc region mutations are selected from: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E. In some embodiments, the at least one modification is galactosylation. In some embodiments, the fusion protein comprises CCL1, or an active fragment thereof. In some embodiments, CCL1, or an active fragment thereof, comprises the amino acid sequence of SEQ ID No. 2 or amino acids 24-96 of SEQ ID No. 2. In some embodiments, the fusion protein comprises the amino acid sequence of SEQ ID NO. 1 or amino acids 24-316 of SEQ ID NO. 1. In some embodiments, the fusion protein comprises MC148 or an active fragment thereof. In some embodiments, MC148 or an active fragment thereof comprises the amino acid sequence of SEQ ID NO 6. In some embodiments, the fusion protein comprises the amino acid sequence of SEQ ID NO 5.
In some embodiments, the isolated nucleic acid encodes a fusion protein. In some embodiments, the vector comprises an isolated nucleic acid. In some embodiments, the host cell comprises a nucleic acid or vector. In some embodiments, the host cell expresses the fusion protein. In some embodiments, the host cell is engineered to produce an afucosylated antibody.
In some embodiments, the method of producing a fusion protein comprises culturing a host cell under conditions suitable for expression of the fusion protein. In some embodiments, the method further comprises isolating the fusion protein.
In some embodiments, a method of treating cancer comprises administering to a subject having cancer an effective amount of an antibody provided herein (e.g., 7-B16, 1-K17, etc.). In some embodiments, a method of treating cancer comprises administering to a subject having cancer an effective amount of a fusion protein provided herein that binds to CCR 8.
In some embodiments, the cancer comprises tumor-infiltrating Treg cells. In some embodiments, the cancer comprises cells that express CCR8 (as determined by, e.g., immunohistochemistry, Fluorescence Activated Cell Sorting (FACS), gene expression analysis (such as Q-PCR or RT-PCR), Western blot, ELISA, and the like). In some embodiments, the CCR 8-expressing cell is a Treg cell. In some embodiments, the Treg cells are T cell subsets that are immunosuppressive and that generally inhibit or down-regulate the induction and proliferation of effector T cells. In some embodiments, the Treg cells express CD4, FOXP3, and CD25(IL-2 receptor alpha chain). In some embodiments, CCR8 is expressed on the surface of Treg cells in less than 10,000 copies per cell, as can be determined by, for example, Fluorescence Activated Cell Sorting (FACS) and flow cytometry. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs express less than 10,000 copies of CCR 8/cell on the cell surface. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of tumor-infiltrating tregs in a sample obtained from a tumor express less than 10,000 copies of CCR 8/cell. In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer is breast cancer (e.g., triple negative breast cancer).
In some embodiments, the cancer comprises cells that express CCR8 (as determined by, for example, immunohistochemistry, Fluorescence Activated Cell Sorting (FACS), gene expression analysis (such as Q-PCR or RT-PCR), Western blot, ELISA, and the like). In some embodiments, the CCR 8-expressing cell is a Treg cell. In some embodiments, the Treg cells are a T cell subset that is immunosuppressive and generally inhibits or downregulates the induction and proliferation of effector T cells. In some embodiments, the Treg cells express CD4, FOXP3, and CD25(IL-2 receptor alpha chain). In some embodiments, the cancer is a solid cancer. In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer is breast cancer (e.g., triple negative breast cancer). In some embodiments, a method of treating a solid cancer comprises administering to a subject having a solid cancer an effective amount of an antibody that binds human CCR8 (e.g., 7-B16, 1-K17, etc.). In some embodiments, the antibody inhibits the binding of CCL1 to CCR 8. In some embodiments, the cancer is a hematologic cancer. In some embodiments, a method of treating a hematologic cancer comprises administering to a subject having a hematologic cancer an effective amount of an antibody that binds human CCR 8. In some embodiments, the antibody inhibits the binding of CCL1 to CCR 8. In some embodiments, the hematologic cancer expresses CCR 8. In some embodiments, the subject has previously received checkpoint inhibitor (CPI) treatment, and optionally wherein the cancer is resistant to the CPI. In some embodiments, the cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab (pembrolizumab), nivolumab (nivolumab), attentizumab (atezolizumab), avizumab (avelumab), duvatuzumab (durvalumab), cimiral mab (cemipimab-rwlc), and sepiolimab (zimberlimab); the anti-CLTA 4 antibody is ipilimumab (ipilimumab) or tremelimumab (tremelimumab); or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan (tiragolumab), viborolizumab (vibostolimab), domavalizumab (domonalizab), AB308, BMS-986207, and brivarezumab.
In some embodiments, a method of selecting a subject with a solid cancer for treatment with an antibody that binds human CCR8 comprises detecting CCR8 expression in a sample from the subject. In some embodiments, the methods further comprise administering an effective amount of an antibody that binds human CCR 8.
In some embodiments, a method of selecting a subject with a hematologic cancer for treatment with an antibody that binds human CCR8 comprises detecting CCR8 expression in a sample from the subject. In some embodiments, the methods further comprise administering an effective amount of an antibody that binds human CCR 8.
In some embodiments, a method of treating a hematologic cancer comprises administering to a subject having a hematologic cancer an effective amount of a fusion protein provided herein that binds to CCR 8. In some embodiments, a method of selecting a subject with a hematologic cancer for treatment with a fusion protein comprising (a) CCL1 or an active fragment thereof, or MC148 or an active fragment thereof, and (b) an Fc region, comprises detecting CCR8 expression in a sample from the subject. In some embodiments, the method further comprises administering an effective amount of a fusion protein comprising (a) CCL1, or an active fragment thereof, or MC148, or an active fragment thereof, and (b) an Fc region. In some embodiments, the hematologic cancer is T cell adult acute lymphocytic leukemia, T cell childhood acute lymphocytic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T cell lymphoma, T cell acute lymphocytic leukemia, adult T cell leukemia/lymphoma, T cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
In one aspect, the disclosure provides an isolated antibody that binds human CCR8, wherein the antibody comprises HCDR3 and the HCDR3 comprises SEQ ID NO:86 or a variant of SEQ ID NO:86 containing 1, 2, or 3 mutations, and wherein the antibody binds human CCR8 and has ADCC activity. In some embodiments, the mutation is a substitution (e.g., a conservative and/or non-conservative substitution), a deletion, or an insertion. In some embodiments, the 1, 2, or 3 mutations are at least one of amino acid positions 1-4, 6, 7, or 12 of SEQ ID No. 86. In some embodiments, the substitution is a conservative substitution. In some embodiments, the conservative substitution is at amino acid position 1, 4, or 12 of SEQ ID NO 86. In some embodiments, the substitution is a non-conservative substitution. In some embodiments, the non-conservative substitution is at amino acid position 7 of SEQ ID NO 86. In some embodiments, the antibody comprises at least 2 hits in HCDR3And (4) generation. In some embodiments, the at least 2 substitutions are at least one of amino acid positions 1-4, 6, 7, or 12 of SEQ ID No. 86. In some embodiments, the at least 2 substitutions are conservative substitutions. In some embodiments, at least one conservative substitution is at amino acid position 1, 4, or 12 of SEQ ID No. 86. In some embodiments, the at least 2 substitutions are non-conservative substitutions. In some embodiments, the at least one non-conservative substitution is at amino acid position 7 of SEQ ID No. 86. In some embodiments, when more than one substitution mutation is present, the mutation comprises a conservative substitution and a non-conservative substitution. In another aspect, the disclosure provides an isolated antibody that binds to human CCR8, wherein the antibody comprises HCDR3 that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID No. 86, and wherein the antibody binds to human CCR8 and has ADCC activity. In some embodiments, HCDR3 comprises an amino acid sequence selected from any one of SEQ ID NO 86 and SEQ ID NO 104-119. In some embodiments, the antibody comprises HCDR1 comprising SEQ ID NO 84 or SEQ ID NO 123. In some embodiments, the antibody comprises HCDR2 comprising SEQ ID NO:85 or SEQ ID NO: 124. In some embodiments, the antibody comprises LCDR1 comprising SEQ ID No. 87 or SEQ ID No. 120. In some embodiments, the antibody comprises LCDR2 comprising SEQ ID No. 88 or SEQ ID No. 121. In some embodiments, the antibody comprises LCDR3 comprising SEQ ID No. 89 or SEQ ID No. 122. In some embodiments, the ADCC activity comprises an EC50 value of less than 200ng/ml, 175ng/ml, 150ng/ml, 125ng/ml, 100ng/ml, 75ng/ml, 50ng/ml, 25ng/ml, 20ng/ml, 15ng/ml, 10ng/ml, 9ng/ml, 8ng/ml, 7ng/ml, 6ng/ml, 5ng/ml, 4ng/ml, 3ng/ml, 2ng/ml, or 1ng/ml, as measured by an ADCC reporter mechanism of action (MOA) based bioassay. In some embodiments, the ADCC activity is more potent than the 7-B16 antibody. In some embodiments, the ADCC activity is at least as effective as the 7-B16 antibody. In some embodiments, the antibody is to K of human CCR8 D Equal to or less than 7-B16 antibody (e.g., as determined by kinetic exclusion assay (i.e., KinExA)). In some embodiments, the antibody is K on a cell of human CCR8 D Equal to or lower than the 7-B16 antibody (e.g., as determined by kinetic exclusion assay (i.e., KinExA)). In some embodiments, the antibody comprises at least one modification that enhances cell killing. In some embodiments, the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC). In some embodiments, the at least one modification is afucosylation. In some embodiments, the at least one modification is one or more heavy chain constant region mutations at one or more positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334, and P396. In some embodiments, the one or more heavy chain constant region mutations are one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L. In some embodiments, the one or more heavy chain constant region mutations are selected from the group consisting of: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E. In some embodiments, the at least one modification is galactosylation. In some embodiments, the antibody has an affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75pM, or less than 50pM, or less than 25pM D ) (as determined by kinetic exclusion assay (i.e., KinExA)) binds to human CCR 8. In some embodiments, the antibody has an on-cell affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75, or less than 50pM, or less than 25pM D ) Binds human CCR8 as determined by, for example, a kinetic exclusion assay (i.e., KinExA). In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a human or humanized antibody. In some embodiments, the antibody is a full length antibody. In some embodiments, the antibody is an IgG1 or IgG3 antibody.
The present disclosure also provides an isolated antibody or fusion protein of any aspect or embodiment described herein for use in treating cancer. In some embodiments, the cancer is a hematologic cancer or a solid cancer/solid tumor. In some embodiments, the cancer expresses CCR 8. In some embodiments, the hematologic cancer is T cell adult acute lymphoblastic leukemia, T cell childhood acute lymphoblastic leukemia, lymphoblastic lymphoma, acute lymphoblastic leukemia, cutaneous T cell lymphoma, T cell acute lymphoblastic leukemia, adult T cell leukemia/lymphoma, T cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma. In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer is breast cancer (e.g., triple negative breast cancer). In some embodiments, the method of treatment may further comprise administering to the subject one or more additional therapeutic agents (e.g., other anti-cancer agents). In some embodiments, the one or more additional therapeutic agents are selected from anti-Trop-2 antibodies (e.g., sabituzumab govitecan), SKB-264, JS-108(DAC-002), dapatozumab derustecan (dapopotab deruxtecan), BAT-8003), anti-CD 47 antibodies or CD47 blockers (e.g., mololizumab (magrolimab), DSP-107, AO-176, ALX-148, IBI-188, lemzopralimab (lemzopralimab), TTI-621, TTI-622), anti-sirpa antibodies (e.g., GS-0189), FLT3L-Fc fusion proteins (e.g., GS-3583), anti-PD-1 antibodies (pabulimab, nivolumab, serolimab), small molecule PD-L1 inhibitors (e.g., PD-L3824), anti-L4238 antibodies (e.g., anti-1, acilizumab), a small molecule MCL1 inhibitor (e.g., GS-9716), a small molecule HPK1 inhibitor (e.g., GS-6451), an HPK1 degrader (PROTAC; e.g., ARV-766), small molecule DGKa inhibitors, small molecule CD73 inhibitors (e.g., AB680), anti-CD 73 antibodies (e.g., olelumab), dual a 2a /A 2b Adenosine receptor antagonists (e.g., etrumadenant (AB928)), anti-TIGIT antibodies (e.g., tiryleigh Eusumab, Venbrizumab, granolizumabMonoclonal antibody, AB308), anti-TREM 1 antibody (e.g., PY159), anti-TREM 2 antibody (e.g., PY314), TGF β -capture agent (e.g., bintrafusip alpha, AGEN-1423), anti-TGF β 1 antibody (e.g., SRK-181), and CAR-T cell therapy (e.g., aliskiren (axicabtagene ciloleucel), brevianobarex (brexurabagene autoleuecel), tesseram (tisagenllectel)). In some embodiments, the one or more additional therapeutic agents are selected from the group consisting of shacetuzumab gavatica-hziy (sacituzumab govitecan-hziy), molucizumab, GS-0189, GS-3583, saprolizumab, GS-4224, GS-9716, GS-6451, AB680, itraconazole (AB), donaldicamab, AB308, PY159, PY314, SRK-181, aliskiren, and breuioleracene. In some embodiments, the administration of the one or more additional therapeutic agents is concurrent with the administration of the antibody, while in some embodiments, the administration of the one or more additional therapeutic agents is performed before or after the administration of the antibody. In some embodiments, the cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, durolizumab, cimiciprizumab, and saprolizumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
The present disclosure also provides a nucleic acid encoding an antibody of any one of the aspects or embodiments described herein.
The present disclosure also provides a method of treating cancer, the method comprising administering to a subject having cancer an antibody according to any one of the aspects or embodiments described herein. In some embodiments, the cancer is a hematologic cancer or a solid cancer/solid tumor. In some embodiments, the cancer expresses CCR 8. In some embodiments, the hematologic cancer is T cell adult acute lymphoblastic leukemia, T cell childhood acute lymphoblastic leukemiaA leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T-cell lymphoma, T-cell acute lymphocytic leukemia, adult T-cell leukemia/lymphoma, T-cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma. In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer is breast cancer (e.g., triple negative breast cancer). In some embodiments, the method of treatment may further comprise administering to the subject one or more additional therapeutic agents (e.g., other anti-cancer agents). In some embodiments, the administration of the one or more additional therapeutic agents is concurrent with the administration of the antibody, while in some embodiments, the administration of the one or more additional therapeutic agents is performed before or after the administration of the antibody. In some embodiments, the one or more additional therapeutic agents are selected from anti-Trop-2 antibodies (e.g., saritumumab govikang, SKB-264, JS-108(DAC-002), daptomab delutene, BAT-8003), anti-CD 47 antibodies or CD47 blockers (e.g., mololizumab, DSP-107, AO-176, ALX-148, IBI-188, rizolizumab, TTI-621, TTI-622), anti-sirpa antibodies (e.g., GS-0189), FLT3L-Fc fusion proteins (e.g., GS-3583), anti-PD-1 antibodies (pabulizumab, nivolumab, saprolizumab), small molecule PD-L1 inhibitors (e.g., GS-4224), anti-PD-L1 antibodies (e.g., avilizumab), small molecule MCL1 inhibitors (e.g., GS-9716), small molecule HPK1 inhibitors (e.g., GS-6451), HPK1 degrader (procac; e.g., ARV-766), small molecule DGKa inhibitors, small molecule CD73 inhibitors (e.g., AB680), anti-CD 73 antibodies (e.g., olaruzumab), dual a 2a /A 2b Adenosine receptor antagonists (e.g., itraconazole (AB928)), anti-TIGIT antibodies (e.g., tegraleigh mab, vibralizumab, donralizumab, AB308), anti-TREM 1 antibodies (e.g., PY159), anti-TREM 2 antibodies (e.g., PY314), TGF β -capturing agents (e.g., bintrafusip alpha, age-1423), anti-TGF β 1 antibodies (e.g., SRK-181), and CAR-T cell therapy (e.g., aliskiren, brij oritem, tesseram). At one endIn some embodiments, the one or more additional therapeutic agents are selected from the group consisting of saritumumab gavatinib-hziy, mololizumab, GS-0189, GS-3583, seraprimab, GS-4224, GS-9716, GS-6451, AB680, eltromazine (AB928), donalizumab, AB308, PY159, PY314, SRK-181, aliskiren, and briguerite.
The present disclosure also provides a use of treating cancer, the use comprising administering to a subject having cancer an antibody according to any aspect or embodiment described herein. In some embodiments, the cancer is a hematologic cancer or a solid cancer/solid tumor. In some embodiments, the cancer expresses CCR 8. In some embodiments, the hematologic cancer is T cell adult acute lymphoblastic leukemia, T cell childhood acute lymphoblastic leukemia, lymphoblastic lymphoma, acute lymphoblastic leukemia, cutaneous T cell lymphoma, T cell acute lymphoblastic leukemia, adult T cell leukemia/lymphoma, T cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma. In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer is breast cancer (e.g., triple negative breast cancer). In some embodiments, the method of treatment may further comprise administering to the subject one or more additional therapeutic agents (e.g., other anti-cancer agents). In some embodiments, the administration of the one or more additional therapeutic agents is concurrent with the administration of the antibody, while in some embodiments, the administration of the one or more additional therapeutic agents is performed before or after the administration of the antibody. In some embodiments, the cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprizumab, and cepacilinumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
The present disclosure also provides a method of treating a refractory cancer or a cancer that is resistant to a checkpoint inhibitor (CPI), the method comprising administering to a subject having a cancer an isolated antibody according to any one of the preceding aspects or embodiments or a fusion protein according to any one of the preceding aspects or embodiments, wherein the subject has been previously treated with chemotherapy or CPI therapy and is not responsive to the chemotherapy or CPI therapy. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprizumab, and cepacilinumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab. In some embodiments, the cancer is a hematologic cancer or a solid cancer. In some embodiments, the cancer expresses CCR 8. In some embodiments, the cancer is T-cell adult acute lymphoblastic leukemia, T-cell childhood acute lymphoblastic leukemia, lymphoblastic lymphoma, acute lymphoblastic leukemia, cutaneous T-cell lymphoma, T-cell acute lymphoblastic leukemia, adult T-cell leukemia/lymphoma, T-cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma. In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer is breast cancer (e.g., triple negative breast cancer). In some embodiments, the method may further comprise administering one or more additional therapeutic agents to the subject. In some embodiments, the one or more additional therapeutic agents are selected from an anti-Trop-2 antibody (e.g., saritumumab gavatinib, SKB-264, JS-108(DAC-002), daptomab delutecan, BAT-8003), an anti-CD 47 antibody, or a CD47 blocker (e.g., mololizumab, DSP-107, a, AO-176, ALX-148, IBI-188, letuzumab, TTI-621, TTI-622), an anti-sirpa antibody (e.g., GS-0189), FLT3L-Fc fusion protein (e.g., GS-3583), an anti-PD-1 antibody (palboclizumab, nivolumab, saprolizumab), a small molecule PD-L1 inhibitor (e.g., GS-4224), an anti-PD-L1 antibody (e.g., avilizumab), a small molecule MCL1 inhibitor (e.g., GS-9716), a small molecule HPK1 inhibitor (e.g., GS-6451), an HPK1 degradant (PROTAC; e.g., ARV-766), small molecule DGKa inhibitors, small molecule CD73 inhibitors (e.g., AB680), anti-CD 73 antibodies (e.g., olaruzumab), dual a 2a /A 2b Adenosine receptor antagonists (e.g., itraconazole (AB928)), anti-TIGIT antibodies (e.g., tegraleigh mab, vibralizumab, donralizumab, AB308), anti-TREM 1 antibodies (e.g., PY159), anti-TREM 2 antibodies (e.g., PY314), TGF β -capturing agents (e.g., bintrafusip alpha, age-1423), anti-TGF β 1 antibodies (e.g., SRK-181), and CAR-T cell therapy (e.g., aliskiren, brij oritem, tesseram). In some embodiments, the one or more additional therapeutic agents are selected from the group consisting of safirtuzumab gavitegaze-hziy, mololizumab, GS-0189, GS-3583, saprolizumab, GS-4224, GS-9716, GS-6451, AB680, itraconazole (AB928), donalizumab, AB308, PY159, PY314, SRK-181, aliskiren and briguerite. In some embodiments, the administration of the one or more additional therapeutic agents is concurrent with the administration of the antibody. In some embodiments, the administration of the one or more additional therapeutic agents is performed before or after the administration of the antibody.
Drawings
FIG. 1 shows a scatter plot of CCR8 mRNA expression levels determined by microarray. On the y-axis is CCR8 gene expression measured by fluorescence intensity (AU). The x-axis represents leukemic sub-type or healthy bone marrow. For each group on the x-axis, the straight line represents the median CCR8 expression level for that leukemic sub-type. The leukemia subtypes shown include Acute Lymphocytic Leukemia (ALL), T-cell acute lymphocytic leukemia (TALL), B-cell acute lymphocytic leukemia (BALL), Acute Myeloid Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Chronic Myeloid Leukemia (CML), myelodysplastic syndrome (MDS).
Fig. 2 shows a boxplot showing CCR8 mRNA expression levels determined by RNA sequencing. On the y-axis is CCR8 gene expression as measured by number of Transcripts Per Million (TPM) of log-transform. On the x-axis is a lymphoma subtype, leukemia subtype or healthy blood or tissue.
Fig. 3A and 3B show that human Tumor Infiltrating (TIL) tregs preferentially express CCR8, but have a low receptor number on a per cell basis. An exemplary flow cytometry plot (gated on tumor T CD4+ cells) (a) and quantification plot (B) are shown.
FIGS. 4A-4B show the binding of anti-CCR 8 monoclonal antibody to CHO-S cells expressing human CCR8 (A) and CHO-S cells expressing CCR4 (B).
FIG. 5 shows antagonist activity of anti-CCR 8 chimeric antibodies using the DiscoverX assay (Eurofins).
Figure 6 shows Antibody Dependent Cellular Cytotoxicity (ADCC) of certain anti-CCR 8 chimeric antibodies using ADCC reporter bioassay (Promega).
Fig. 7A-7B show that afucosylation of human IgG1 anti-CCR 8 antibody is necessary to kill target cells expressing low levels of CCR 8. Fold change in ADCC reporter activity against titrated human IgG1(a) and afucosylated human IgG1(B) versions of anti-human CCR8 clone 1K 17. CHO target cells express 300,000 (black filled circles) or 10,000 (white filled circles) cells surface human CCR8 molecules per cell.
Fig. 8A-8C show that Fc effector function of anti-mouse CCR8mAb is required for efficient killing of tumor-infiltrating (TIL) tregs in vivo. Fc activity (mouse IgG2a, white filled circles) anti-mouse CCR8 mIgG2a antibody was able to deplete TIL tregs in vivo in the mouse MC38 syngeneic model, whereas Fc inactivity (mouse IgG1, black filled circles) anti-mouse CCR8 mIgG1 antibody was not. Exemplary flow cytometry plots (gating tumor T CD4+ cells) (a) and quantification plots ((B) and (C)) show the frequency of TIL tregs three days after injection of 200 μ g of the indicated antibody (n-4).
Fig. 9A-9B show that Fc-active (mouse IgG2a, white filled circles) anti-mouse CCR8 mIgG2a antibody reduces tumor growth (a) and increases survival (B) in the MC38 syngeneic mouse model, whereas Fc-inactive (mouse IgG1, black filled circles) anti-mouse CCR8 mIgG1 antibody does not. Tumor growth curves (a) and survival curves (B) of MC38 mice are shown after therapeutic treatment with isotype control, Fc activity (mouse IgG2a) or Fc inactivity (mouse IgG1) anti-mouse CCR8 antibody (n ═ 10-15).
FIG. 10 shows the expression of CCR8 mRNA in various hematological malignancies compared to normal healthy blood. Indicates cancer types with FDR-modulated p-value < 0.05. N indicates the number of samples in the cancer type. The cancers include Acute Lymphoblastic Leukemia (ALL), Acute Myeloid Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), Chronic Myeloid Leukemia (CML), Mantle Cell Lymphoma (MCL), angioimmunoblastic T-cell lymphoma (ATCL), hepatosplenic T-cell lymphoma (HTCL), peripheral T-cell lymphoma (PTCL NOS), Burkitt's Lymphoma (BL), adult T-cell leukemia/lymphoma (ATLL), Anaplastic Large Cell Lymphoma (ALCL), chronic myelomonocytic leukemia (CMML), Follicular Lymphoma (FL), T-cell lymphoblastic leukemia/lymphoma (TLLL), extranodal NK/T-cell lymphoma (NKTCL), Primary Effusion Lymphoma (PEL), acute lymphoblastic leukemia/acute myeloid leukemia (ALL), Acute Lymphoblastic Leukemia (ALL), chronic lymphocytic leukemia (ALL), and chronic myelogenous leukemia (HTL), AML), Histiocytic Lymphoma (HL), cutaneous T-cell lymphoma (CTCL) and Marginal Zone Lymphoma (MZL).
Figure 11 shows Antibody Dependent Cellular Cytotoxicity (ADCC) of CDR H3 variants of the anti-CCR 8 chimeric antibody 7-B16 determined using ADCC reporter bioassay (Promega).
Detailed Description
Antibodies that bind to CCR8 are provided. Antibody heavy and light chains capable of forming antibodies that bind CCR8 are also provided. In addition, antibodies, heavy and light chains comprising one or more specific Complementarity Determining Regions (CDRs) are provided. Polynucleotides encoding anti-CCR 8 antibodies are provided. Polynucleotides encoding antibody heavy or light chains are also provided. Methods of producing and/or purifying anti-CCR 8 antibodies are provided. Fusion proteins comprising CCL1 or MC148 and an Fc region are also provided. Polynucleotides encoding such fusion proteins are also provided. Methods of producing and/or purifying the fusion proteins are provided. Methods of treatment using the antibodies and/or fusion proteins are provided. Such methods include, but are not limited to, methods of treating cancer. Methods for detecting CCR8 are provided. Such methods include methods of identifying an individual who may benefit from treatment with an antibody or fusion protein provided herein, methods of monitoring treatment of an individual with an antibody or fusion protein provided herein, and methods of improving the efficacy of treatment of an antibody or fusion protein provided herein in an individual.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.
All references cited herein, including patent applications, patent publications, and Genbank accession numbers, are incorporated by reference herein as if each individual reference were specifically and individually indicated to be incorporated by reference in its entirety.
The techniques and procedures described or referenced herein are generally well understood and commonly employed by those skilled in the art using conventional methodologies, such as those described in the following references: sambrook et al, Molecular Cloning A Laboratory Manual 3 rd edition (2001) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.CURRENT PROTOCOLS IN MOLECULAR BIOLOGY (edited by F.M.Ausubel et al, (2003)); the series METHODS IN ENZYMOLOGY (Academic Press, Inc.: PCR 2: A PRACTICAL APPROACH (M.J. MacPherson, B.D. Hames and G.R. Taylor eds (1995)), Harlow and Lane eds (1988) ANTIBODIES, ALABORATORY MANL, and ANIMAL CELL CURTURE (R.I. Freshney eds (1987)); oligonucleotide Synthesis (m.j. gait editors, 1984); methods in Molecular Biology, Humana Press; cell Biology A Laboratory Notebook (edited by J.E.Cellis, 1998) Academic Press; animal Cell Culture (r.i. freshney, eds., 1987); introduction to Cell and Tissue Culture (J.P.Mather and P.E.Roberts,1998) Plenum Press; cell and Tissue Culture Laboratory Procedures (A.doyle, J.B.Griffiths and D.G.Newell, eds., 1993-8) J.Wiley and Sons; handbook of Experimental Immunology (edited by d.m. weir and c.c. blackwell); gene Transfer Vectors for mammlian Cells (edited by J.M.Miller and M.P.Calos, 1987); PCR The Polymerase Chain Reaction (edited by Mullis et al, 1994); current Protocols in Immunology (edited by J.E. Coligan et al, 1991); short Protocols in Molecular Biology (Wiley and Sons, 1999); immunobiology (c.a. janeway and p.travers, 1997); antibodies (p.finch, 1997); antibodies A Practical Approach (D.Catty. eds., IRL Press, 1988-; monoclonal Antibodies A Practical Approach (edited by P.Shepherd and C.dean, Oxford University Press, 2000); a Laboratory Manual (E.Harlow and D.Lane, Cold Spring Harbor Laboratory Press, 1999); the Antibodies (edited by M.Zantetti and J.D.Capra, Harwood Academic Publishers, 1995); and Cancer: Principles and Practice of Oncology (V.T. Devita et al, J.B. Lippincott Company, 1993); and updated versions thereof.
I. Definition of
Unless defined otherwise, scientific and technical terms used in connection with the present disclosure shall have the meanings that are commonly understood by one of ordinary skill in the art. Furthermore, unless the context requires otherwise or clearly dictates otherwise, singular terms shall include the plural and plural terms shall include the singular. For any conflict in definition between various sources or references, the definitions provided herein control.
It is to be understood that the embodiments of the invention described herein include embodiments "consisting of …" and/or "consisting essentially of …". As used herein, the singular forms "a", "an" and "the" include plural referents unless otherwise indicated. The use of the term "or" herein is not meant to imply that alternatives are mutually exclusive.
The use of "or" in this application means "and/or" unless explicitly stated or understood by one of ordinary skill in the art. Use of "or" in the context of multiple dependent claims refers to more than one of the preceding independent or dependent claims.
As understood by those of skill in the art, reference herein to a "value or parameter" about "includes (and describes) embodiments directed to that value or parameter itself. For example, a description referring to "about X" includes a description of "X".
The terms "nucleic acid molecule," "nucleic acid," and "polynucleotide" are used interchangeably and refer to a polymer of nucleotides. Such nucleotide polymers may contain natural and/or non-natural nucleotides and include, but are not limited to, DNA, RNA, and PNA. "nucleic acid sequence" refers to a linear nucleotide sequence comprising a nucleic acid molecule or polynucleotide.
The terms "polypeptide" and "protein" are used interchangeably to refer to a polymer of amino acid residues and are not limited to a minimum length. Such amino acid residue polymers can contain natural or unnatural amino acid residues and include, but are not limited to, peptides, oligopeptides, dimers, trimers, and multimers of amino acid residues. This definition encompasses both full-length proteins and fragments thereof. The term also includes post-expression modifications of the polypeptide, such as glycosylation, sialylation, acetylation, phosphorylation, and the like. Furthermore, for the purposes of this disclosure, "polypeptide" refers to a protein that contains modifications to the native sequence, such as deletions, additions, and substitutions (typically conservative in nature), so long as the protein retains the desired activity. These modifications may be deliberate (e.g.by site-directed mutagenesis) or accidental, such as by mutation of the host producing the protein or by error due to PCR amplification.
As used herein, "CCR 8" and "C-C chemokine receptor type 8" and "chemokine receptor 8" refer to any native CCR8 that results from the expression and processing of CCR8 in a cell. Unless otherwise indicated, the term includes CCR8 from any vertebrate source, including mammals such as primates (e.g., humans and cynomolgus monkeys) and rodents (e.g., mice and rats). The term also includes naturally occurring variants of CCR8, such as splice variants or allelic variants. The amino acid sequence of an exemplary human CCR8 protein is shown in SEQ ID NO 101(UniProt identifier P51685). The amino acid sequence of an exemplary mouse CCR8 protein is shown in SEQ ID NO 102(UniProt identifier P56484). The amino acid sequence of an exemplary cynomolgus monkey CCR8 protein is shown in SEQ ID NO 103(UniProt identifier G7NYJ 2).
As used herein, "CCL 1" and "C-C motif chemokine 1" refer to any native CCR1 that results from the expression and processing of CCR1 in a cell. Unless otherwise indicated, the term includes CCR1 from any vertebrate source, including mammals such as primates (e.g., humans and cynomolgus monkeys) and rodents (e.g., mice and rats). The term also includes naturally occurring variants of CCR1, such as splice variants or allelic variants. The amino acid sequence of an exemplary human CCR1 protein is shown in SEQ ID NO 2(UniProt identifier P22362.1). An exemplary mature CCR1 protein comprises amino acids 24-96 of SEQ ID NO. 2.
As used herein, "MC 148" refers to any native MC148 that results from the expression and processing of MC148 in a cell. Unless otherwise indicated, the term includes MC148 from any viral source, including molluscum contagiosum virus (subtype 1 or subtype 2). The term also includes naturally occurring variants of MC148, such as truncated variants or allelic variants. The amino acid sequence of an exemplary MC148 protein is shown in SEQ ID NO 6 (amino acids 25-104 of UniProt identifier Q98314.1).
As used herein, a "7-B16 antibody" is understood to be any antibody that binds to CCR8 and comprises: (i) a heavy chain comprising SEQ ID NO:82 and a light chain comprising SEQ ID NO:83, (ii) a heavy chain variable region comprising SEQ ID NO:80 and a light chain variable region comprising SEQ ID NO:81, or (iii) HCDR1, HCDR2, and HCDR3 comprising SEQ ID NO:84, 85, and 86, respectively, and LCDR1, LCDR2, and LCDR3 comprising SEQ ID NO:87, 88, and 89, respectively; and chimeric, human or humanized versions of any of the foregoing (i), (ii) or (iii). In some embodiments, the "7-B16 antibody" can be used to specifically refer to an antibody comprising the heavy chain of SEQ ID NO. 82 and the light chain of SEQ ID NO. 83.
The term "specifically binds" to an antigen or epitope is a term well known in the art, and methods of determining such specific binding are also well known in the art. A molecule is said to exhibit "specific binding" or "preferential binding" if it reacts or associates more frequently, more rapidly, with a longer duration, and/or with greater affinity with a particular cell or substance than it does with a replacement cell or substance. An antibody "specifically binds" or "preferentially binds" to a target if it binds to the target with greater affinity, avidity, more readily, and/or for a longer duration than it binds to other substances. For example, an antibody that specifically or preferentially binds to a CCR8 epitope is one that binds that epitope with greater affinity, avidity, more readily, and/or for a longer duration than it binds to other CCR8 epitopes or non-CCR 8 epitopes. It is also understood by reading this definition that, for example, an antibody (or portion or epitope) that specifically or preferentially binds a first target may or may not specifically or preferentially bind a second target. Thus, "specific binding" or "preferential binding" does not necessarily require (although it may include) exclusive binding. Typically, but not necessarily, reference to binding means preferential binding. "specificity" refers to the ability of a binding protein to selectively bind to an antigen.
As used herein, "substantially pure" refers to a material that is at least 50% pure (i.e., free of contaminants), more preferably at least 90% pure, more preferably at least 95% pure, still more preferably at least 98% pure, and most preferably at least 99% pure.
As used herein, the term "epitope" refers to a site on a target molecule (e.g., an antigen, such as a protein, nucleic acid, carbohydrate, or lipid) to which an antigen-binding molecule (e.g., an antibody, antibody fragment, or scaffold protein containing an antibody binding region) binds. Epitopes generally comprise chemically active surface groups of molecules such as amino acids, polypeptides or sugar side chains and have specific three-dimensional structural characteristics as well as specific charge characteristics. Epitopes can be formed from both contiguous residues and/or juxtaposed non-contiguous residues (e.g., amino acids, nucleotides, sugars, lipid moieties) of the target molecule. Epitopes formed from contiguous residues (e.g., amino acids, nucleotides, sugars, lipid moieties) are typically retained upon exposure to denaturing solvents, whereas epitopes formed from tertiary folding are typically lost upon treatment with denaturing solvents. Epitopes can include, but are not limited to, at least 3, at least 5, or 8-10 residues (e.g., amino acids or nucleotides). In some examples, the epitope is less than 20 residues (e.g., amino acids or nucleotides), less than 15 residues, or less than 12 residues in length. Two antibodies can bind to the same epitope within an antigen if they exhibit competitive binding to the antigen. In some embodiments, an epitope may be identified by a certain minimum distance from a CDR residue on the antigen binding molecule. In some embodiments, epitopes can be identified by the above-described distances and are further limited to those residues that participate in the bonds (e.g., hydrogen bonds) between antibody residues and antigen residues. Epitopes can also be identified by various scans, for example, an alanine or arginine scan can indicate one or more residues with which the antigen binding molecule can interact. Unless explicitly indicated, a group of residues that are epitopes does not exclude other residues from being part of the epitope for a particular antibody. Rather, the presence of such a group specifies a minimal series (or set of species) of epitopes. Thus, in some embodiments, a group of residues identified as an epitope designates the smallest epitope associated with the antigen, rather than a unique list of residues for the epitope on the antigen.
A "nonlinear epitope" or "conformational epitope" comprises a non-continuous polypeptide, amino acids, and/or sugars within an antigenic protein to which an antibody specific for the epitope binds. In some embodiments, at least one of the residues will not be contiguous with other residues of interest of the epitope; however, one or more of the residues may also be contiguous with other residues.
A "linear epitope" comprises a contiguous polypeptide, amino acids and/or sugars within the antigenic protein to which an antibody specific for that epitope binds. It should be noted that in some embodiments, not every residue within a linear epitope needs to bind directly to (or participate in a bond with) an antibody. In some embodiments, the linear epitope may result from immunization with a peptide consisting effectively of the sequence of the linear epitope, or from a structural portion of the protein that is relatively separated from the rest of the protein (such that the antibody may at least first interact only with that sequence portion).
The term "antibody" herein is used in the broadest sense and encompasses a variety of antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g., bispecific (such as bispecific T cell engagers) and trispecific antibodies), and antibody fragments so long as they exhibit the desired antigen binding activity.
The term antibody includes, but is not limited to, fragments capable of binding antigen, such as Fv, single chain Fv (scFv), Fab ', double scFv, sdAb (single domain antibody), and (Fab') 2 (including chemically linked F (ab') 2 ). Papain digestion of antibodies produces two identical antigen-binding fragments (called "Fab" fragments, each with a single antigen-binding site), as well as a residual "Fc" fragment (the name of which reflects the ability to crystallize readily). Pepsin treatment produces F (ab') which has two antigen binding sites and is still capable of cross-linking the antigen 2 And (3) fragment. The term antibody also includes, but is not limited to, chimeric antibodies, humanized antibodies, and antibodies of various species (such as mouse, human, cynomolgus monkey, etc.). Furthermore, variants having sequences from other organisms are also contemplated for all of the antibody constructs provided herein. Thus, if a human version of an antibody is disclosed, one skilled in the art would understand how to convert an antibody based on human sequences into mouse, rat, cat, dog, horse, etc. sequences. Antibody fragments also include any orientation of single chain scFv, tandem bis scFv, diabodies, tandem trisdcfv, minibodies, and the like. Antibody fragments also include nanobodies (sdabs, an antibody having a single monomer domain, such as a pair of heavy chain variable domains, without a light chain). In some embodiments, an antibody fragment may be referred to as being of a particular species (e.g., a human scFv or a mouse scFv). This indicates at least part of the sequence of the non-CDR region, not the origin of the construct.
The term "monoclonal antibody" refers to an antibody of a substantially homogeneous population of antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to polyclonal antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. Thus, a monoclonal antibody sample can bind to the same epitope on an antigen. The modifier "monoclonal" indicates the character of the antibody as obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. For example, monoclonal antibodies can be prepared by the hybridoma method first described by Kohler and Milstein,1975, Nature 256:495, or can be prepared by recombinant DNA methods such as those described in U.S. Pat. No. 4,816,567. Monoclonal antibodies can also be isolated from phage libraries generated using techniques described, for example, in McCafferty et al, 1990, Nature 348: 552-.
The term "CDR" denotes a complementarity determining region defined by at least one means of identification by those skilled in the art. In some embodiments, a CDR can be defined according to any Chothia numbering scheme, Kabat numbering scheme, a combination of Kabat and Chothia, AbM definitions, Contact definitions, and/or a combination of Kabat, Chothia, AbM, and/or Contact definitions. Exemplary CDRs (CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3) occur at amino acid residues 24-34 of L1, amino acid residues 50-56 of L2, amino acid residues 89-97 of L3, amino acid residues 50-65 of amino acid residues 31-35B, H2 of H1 and amino acid residues 95-102 of H3. (Kabat et al, Sequences of Proteins of Immunological Interest, 5 th edition Public Health Service, National Institutes of Health, Bethesda, MD (1991)). AbM definitions may include, for example, CDRs at amino acid residues 24-34 of L1, amino acid residues 50-56 of L2, amino acid residues 89-97 of L3, amino acid residues H26-H35B of H1, amino acid residues 50-58 of H2, and amino acid residues 95-102 of H3 (CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2, and CDR-H3). The Contact definition may include, for example, CDRs (CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3) at amino acid residues 30-36 of L1, amino acid residues 46-55 of L2, amino acid residues 89-96 of L3, amino acid residues 30-35 of H1, amino acid residues 47-58 of H2 and amino acid residues 93-101 of H3. Chothia definitions may include, for example, CDRs at amino acid residues 24-34 of L1, amino acid residues 50-56 of L2, amino acid residues 89-97 of L3, amino acid residues 26-32 … 34 of H1, amino acid residues 52-56 of H2, and amino acid residues 95-102 of H3 (CDR-L1, CDR, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3). CDRs as shown in any one or more of the figures may also be provided. Except for V H In addition to CDR1, the CDR typically comprises amino acid residues that form a hypervariable loop. Various CDRs within an antibody may be named by their appropriate number and chain type, including but not limited to: a) CDR-L1, CDR-L2, CDR-L3, CDR-H1, CDR-H2 and CDR-H3; b) CDRL1, CDRL2, CDRL3, CDRH1, CDRH2, and CDRH 3; c) LCDR-1, LCDR-2, LCDR-3, HCDR-1, HCDR-2, and HCDR-3; or d) LCDR1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR 3; and so on. The term "CDR" as used herein also encompasses HVRs or "hypervariable regions", including hypervariable loops. Exemplary hypervariable loops occur at amino acid residues 26-32(L1), 50-52(L2), 91-96(L3), 26-32(H1), 53-55(H2) and 96-101 (H3). (Chothia and Lesk, J.mol.biol.196:901-917 (1987))
The term "heavy chain variable region" as used herein refers to a region comprising at least three heavy chain CDRs. In some embodiments, the heavy chain variable region comprises three CDRs and at least FR2 and FR 3. In some embodiments, the heavy chain variable region comprises at least heavy chain HCDR1, Framework (FR)2, HCDR2, FR3, and HCDR 3. In some embodiments, the heavy chain variable region further comprises at least a portion of FR1 and/or at least a portion of FR 4.
The term "heavy chain constant region" as used herein refers to a region comprising at least three heavy chain constant regions C H 1、C H 2 and C H 3, in the region of the first image. Of course, non-functional alterations within the domains are absent and altered within the scope of the term "heavy chain constant region" unless otherwise indicated. Non-limiting exemplary heavy chain constant regions include γ, δ, and α. Non-limiting exemplary heavy chain constant regions also include epsilon and mu. Each heavy chain constant region corresponds to one antibody isotype. For example, an antibody comprising a gamma constant region is an IgG antibody, an antibody comprising a delta constant region is an IgD antibody, and an antibody comprising an alpha constant region is an IgA antibody. Furthermore, the antibody comprising a mu constant region is an IgM antibody, and the antibody comprising an epsilon constant region is an IgE antibody. Certain isoforms may also be subdivided into subclasses. For example, IgG antibodies include, but are not limited to, IgG1 (comprising γ) 1 Constant region), IgG2 (comprising γ) 2 A constant region),IgG3 (containing gamma) 3 Constant region) and IgG4 (comprising γ) 4 Constant region) antibody; IgA antibodies include, but are not limited to, IgA1 (comprising alpha) 1 Constant region) and IgA2 (comprising. alpha.) 2 Constant region) antibodies; and IgM antibodies include, but are not limited to, IgM1 and IgM 2.
The term "heavy chain" as used herein refers to a polypeptide comprising at least one heavy chain variable region, with or without a leader sequence. In some embodiments, the heavy chain comprises at least a portion of a heavy chain constant region. The term "full-length heavy chain" as used herein refers to a polypeptide comprising a heavy chain variable region and a heavy chain constant region, with or without a leader sequence.
The term "light chain variable region" as used herein refers to a region comprising at least three light chain CDRs. In some embodiments, the light chain variable region comprises three CDRs and at least FR2 and FR 3. In some embodiments, the light chain variable region comprises at least light chain LCDR1, Framework (FR)2, LCDR2, FR3, and LCDR 3. For example, the light chain variable region may comprise light chain CDR1, Framework (FR)2, CDR2, FR3, and CDR 3. In some embodiments, the light chain variable region further comprises at least a portion of FR1 and/or at least a portion of FR 4.
The term "light chain constant region" as used herein refers to a region comprising a light chain constant domain C L Of the area (c). Non-limiting exemplary light chain constant regions include λ and κ. Of course, non-functional alterations in the domains are absent and altered within the scope of the term "light chain constant region" unless otherwise indicated.
The term "light chain" as used herein refers to a polypeptide comprising at least one light chain variable region, with or without a leader sequence. In some embodiments, the light chain comprises at least a portion of a light chain constant region. The term "full length light chain" as used herein refers to a polypeptide comprising a light chain variable region and a light chain constant region, with or without a leader sequence.
An "acceptor human framework" for the purposes herein is a light chain variable domain (V) comprising a derivative human immunoglobulin framework or a human consensus framework as defined below L ) Framework or heavy chain variable domains (V) H ) Amino acid sequence of the frameworkThe frame of the column. Acceptor human frameworks derived from human immunoglobulin frameworks or human consensus frameworks may comprise their same amino acid sequence, or the acceptor human frameworks may contain amino acid sequence variations. In some embodiments, the number of amino acid changes is 10 or less, 9 or less, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less. In some embodiments, V L The recipient human framework is aligned in sequence with V L The human immunoglobulin framework sequences or human consensus framework sequences are identical.
"affinity" refers to the strength of the sum of non-covalent interactions between a single binding site of a molecule (e.g., an antibody) and its binding partner (e.g., an antigen). The affinity of a molecule X for its partner Y can generally be determined by the dissociation constant (K) D ) And (4) showing. Can be prepared by conventional methods known in the art (e.g., ELISA K) D KinExA, bio-layer interferometry (BLI), and/or surface plasmon resonance devices (such as Devices), including those described herein).
The term "K" as used herein D "refers to the equilibrium dissociation constant of an antibody-antigen interaction.
In some embodiments, the use is by surface plasmon resonance assay-2000 or"K" of antibody was measured at 25 ℃ using an immobilized antigen CM5 chip of about 10 Response Units (RU) at 3000(BIAcore, Inc., Piscataway, N.J.) D ”、“K d "," Kd ", or" Kd value ". Briefly, carboxymethylated dextran biosensor Chips (CM) were activated with N-ethyl-N '- (3-dimethylaminopropyl) -carbodiimide hydrochloride (EDC) and N-hydroxysuccinimide (NHS) according to the supplier's instructions5, BIACORE, Inc.). Antigen was diluted to 5 μ g/ml (about 0.2 μ M) with 10mM sodium acetate (pH 4.8) and then injected at a flow rate of 5 μ L/min to achieve approximately 10 Response Units (RU) of conjugated protein. After injection of antigen, 1M ethanolamine was injected to block unreacted groups. For kinetic measurements, at 25 ℃ with a flow rate of about 25. mu.L/min with 0.05% TWEEN-20 TM Surfactant (PBST) in PBS is injected a serial dilution of a polypeptide (e.g., a full-length antibody). Using a simple one-to-one Langmuir binding model: ( Evaluation software version 3.2) association rates (k) were calculated by simultaneous fitting of association and dissociation sensorgrams on ) And dissociation Rate (k) off ). Will equilibrate the dissociation constant (K) d ) Is calculated as k off /k on A ratio. See, e.g., Chen et al, J.mol.biol.293:865-881 (1999). If the binding rate exceeds 10 as determined by the above surface plasmon resonance 6 M -1 s -1 The binding rate can then be determined by using a fluorescence quenching technique that measures the increase or decrease in fluorescence emission intensity (excitation 295 nM; emission 340nM, 16nM bandpass) at 25 ℃ of 20nM anti-antigen antibody in PBS at pH 7.2 in the presence of increasing concentrations of antigen, as in a spectrometer such as an 8000-series SLM-AMINCO equipped with stop flow spectrophotometers (Aviv Instruments) or with a stirred cuvette TM Measured in a spectrophotometer (thermospectonic)).
In some embodiments, the two values (e.g., K) d Values), such as less than about 50%, less than about 40%, less than about 30%, less than about 20%, and/or less than about 10% as a function of the reference/comparison value.
In some embodiments, the two values (e.g., K) d Values), e.g., greater than about 10%, greater than about 20%, greater than about 30%, greater than about 40%, and/or greater than about 50% as a function of the value of the reference/comparator molecule.
"surface plasmon resonance" refers to an optical phenomenonLike, it allows to use for example BIAcore TM The system (BIAcore International AB (a company of GE Healthcare group), Uppsala, Sweden and Piscataway, n.j.) detects changes in protein concentration within a biosensor matrix to analyze real-time biospecific interactions. For further description see Jonsson et al (1993) Ann.biol.Clin.51: 19-26.
"biolayer interferometry" refers to an optical analysis technique that analyzes the interference pattern of light reflected from a fixed protein layer and an internal reference layer on the biosensor tip. The variation in the number of molecules bound to the biosensor tip causes a shift in the interference pattern that can be measured in real time. A non-limiting exemplary apparatus for bio-layer interferometry is ForteBioRED96 system (Pall Corporation). See, e.g., Abdiche et al, 2008, anal. biochem.377: 209-glass 277.
The term "k" as used herein on "refers to the rate constant at which an antibody associates with an antigen. Specifically, the rate constant (k) on And k off ) And equilibrium dissociation constants were measured using IgG (bivalent) with a monovalent CCR8 antigen. ' K on ”、“k on "," association rate constant "or" ka "are used interchangeably herein. This value indicates the rate of binding of the binding protein to its target antigen or the rate of complex formation between the antibody and antigen, as shown in the following equation: antibody ("Ab") + antigen ("Ag") → Ab-Ag.
The term "k" as used herein off "refers to the rate constant at which an antibody dissociates from an antibody/antigen complex. k is a radical of off Is also denoted as "K off "or" dissociation rate constant ". This value indicates the off-rate of an antibody from its target antigen or the off-rate of separation of an Ab-Ag complex into free antibody and antigen over time, as shown in the following equation:
Ab+Ag←Ab-Ag。
the term "biological activity" refers to any one or more biological property of a molecule (whether naturally occurring as found in vivo, or provided or enabled by recombinant means). Biological properties include, but are not limited to, binding to cytokines, inducing cell proliferation, inhibiting cell growth, inducing other cytokines, inducing apoptosis, and enzymatic activity. In some embodiments, the biological activities of CCR8 include anti-apoptotic activity, cellular chemotaxis, immunosuppressive function, and the ability to polarize cells into various cellular differentiation pathways.
An "affinity matured" antibody is one that has one or more alterations in one or more CDRs as compared to a parent antibody without the alterations, such alterations resulting in an improvement in the affinity of the antibody for the antigen.
As used herein, "chimeric antibody" refers to an antibody in which a portion of the heavy and/or light chain is derived from a particular source or species, while at least a portion of the remainder of the heavy and/or light chain is derived from a different source or species. In some embodiments, a chimeric antibody refers to an antibody comprising at least one variable region from a first species (such as mouse, rat, cynomolgus monkey, etc.) and at least one constant region from a second species (such as human, cynomolgus monkey, etc.). In some embodiments, a chimeric antibody comprises at least one mouse variable region and at least one human constant region. In some embodiments, the chimeric antibody comprises at least one cynomolgus monkey variable region and at least one human constant region. In some embodiments, all of the variable regions of the chimeric antibody are from a first species and all of the constant regions of the chimeric antibody are from a second species. As mentioned above, the chimeric construct may also be a functional fragment.
As used herein, a "humanized antibody" refers to an antibody in which at least one amino acid in the framework region of the non-human variable region has been replaced with a corresponding amino acid from the human variable region. In some embodiments, the humanized antibody comprises at least one human constant region or fragment thereof. In some embodiments, the humanized antibody is an antibody fragment, such as Fab, scFv, (Fab') 2 And the like. The term humanized also denotes a chimeric immunoglobulin, immunoglobulin chain or fragment thereof (such as Fv, Fab ', F (ab') 2 Or other antigen-binding subsequence of antibody)Human (e.g., murine) antibody formats. Humanized antibodies include human immunoglobulins (recipient antibody) in which residues from a Complementarity Determining Region (CDR) of the recipient are substituted by residues from a CDR of a non-human species, such as mouse, rat or rabbit (donor antibody), having the desired specificity, affinity and capacity. In some cases, Fv Framework Region (FR) residues of the human immunoglobulin are replaced with corresponding non-human residues. In addition, humanized antibodies may contain residues that are not present in either the recipient antibody or the imported CDR or framework sequences, but which are included to further improve and optimize antibody performance. Generally, a humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the Framework (FR) regions are those of a human immunoglobulin consensus sequence. In some embodiments, the humanized antibody may further comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Other forms of humanized antibodies have one or more CDRs (CDR L1, CDR L2, CDR L3, CDR H1, CDR H2 and/or CDR H3) that are altered relative to the original antibody, which are also referred to as one or more CDRs "derived" from "the one or more CDRs from the original antibody. It is understood that a humanized sequence may be identified by its primary sequence and does not necessarily represent a process of producing an antibody.
As used herein, "CDR-grafted antibody" refers to a humanized antibody in which one or more Complementarity Determining Regions (CDRs) of a first (non-human) species have been grafted onto Framework Regions (FRs) of a second (human) species.
"human antibody" as used herein encompasses antibodies produced in humans, non-human animals (such as those containing human immunoglobulin genes)Mouse) and antibodies selected using ex vivo methods such as phage display (Vaughan et al, 1996, Nature Biotechnology,14: 309-; sheets et al, 1998, Proc. Natl. AcadSci (USA)95: 6157-; hoogenboom and Winter,1991, J.mol.biol.,227: 381; marks et al, 1991, J.mol.biol.,222:581), wherein the antibody repertoire (antibody repertoire) is based on human immunoglobulin sequences. The term "human antibody" refers to the genus of sequences that are human sequences. Thus, the term does not refer to the method of producing the antibody, but to the genus of related sequences.
A "functional Fc region" has the "effector functions" of a native sequence Fc region. Exemplary "effector functions" include Fc receptor binding; a C1q binding; CDC; ADCC; phagocytosis; downregulation of cell surface receptors (e.g., B cell receptors; BCR), and the like. Such effector functions typically require an Fc region to be combined with a binding domain (e.g., an antibody variable domain), and can be evaluated using various assays.
A "native sequence Fc region" comprises an amino acid sequence that is identical to the amino acid sequence of a naturally occurring Fc region. Native sequence human Fc regions include native sequence human IgG1Fc regions (non-a and a allotypes); a native sequence human IgG2Fc region; a native sequence human IgG3Fc region; and the native sequence human IgG4Fc region and naturally occurring variants thereof.
A "variant Fc region" comprises an amino acid sequence that differs from the amino acid sequence of a native sequence Fc region due to at least one amino acid modification. In some embodiments, a "variant Fc region" comprises an amino acid sequence that differs from the amino acid sequence of a native sequence Fc region due to at least one amino acid modification but retains at least one effector function of the native sequence Fc region. In some embodiments, the variant Fc region has at least one amino acid substitution as compared to the native sequence Fc region or the Fc region of the parent polypeptide, e.g., from about 1 to about 10 amino acid substitutions, preferably from about 1 to about 5 amino acid substitutions in the native sequence Fc region or the Fc region of the parent polypeptide. In some embodiments, the variant Fc region herein will have at least about 80% sequence identity with a native sequence Fc region and/or an Fc region of a parent polypeptide, at least about 90% sequence identity therewith, at least about 95%, at least about 96%, at least about 97%, at least about 98%, or at least about 99% sequence identity therewith.
"Fc receptor" or "FcR" describes a receptor that binds to the Fc region of an antibody. In some embodiments, the Fc γ R is a native human FcR. In some embodiments, the FcR is one that binds an IgG antibody (gamma receptor) and includes receptors of the Fc γ RI, Fc γ RII, and Fc γ RIII subclasses, including allelic variants and alternatively spliced forms of those receptors. Fc γ RII receptors include Fc γ RIIA ("activating receptor") and Fc γ RIIB ("inhibiting receptor"), which have similar amino acid sequences that differ primarily in their cytoplasmic domains. The activating receptor Fc γ RIIA contains an immunoreceptor tyrosine-based activation motif (ITAM) in its cytoplasmic domain. The inhibitory receptor Fc γ RIIB contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic domain. (see, e.g., Daeron, Annu. Rev. Immunol.15:203-234 (1997)). FcR is described, for example, in Ravetch and Kinet, Annu.Rev.Immunol 9:457-92 (1991); capel et al, immunolmethods 4:25-34 (1994); and de Haas et al, J.Lab.Clin.Med.126:330-41 (1995). The term "FcR" herein encompasses other fcrs, including those to be identified in the future.
The term "Fc receptor" or "FcR" also includes the neonatal receptor FcRn, which is responsible for the transfer of maternal IgG to the fetus (Guyer et al, J.Immunol.117:587(1976) and Kim et al, J.Immunol.24:249(1994)) and regulation of immunoglobulin homeostasis. Methods for measuring binding to FcRn are known (see, e.g., Ghetie and Ward., Immunol. today 18(12):592-598 (1997); Ghetie et al, Nature Biotechnology,15(7):637-640 (1997); Hinton et al, J.biol. chem.279(8):6213-6216 (2004); WO 2004/92219(Hinton et al)).
"Effector function" refers to a biological activity attributable to the Fc region of an antibody that varies with antibody isotype. Examples of antibody effector functions include: clq binding and Complement Dependent Cytotoxicity (CDC); fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down-regulation of cell surface receptors (e.g., B cell receptors); and B cell activation.
"human effector cells" are leukocytes which express one or more fcrs and perform effector functions. In some embodiments, the cells express at least Fc γ RIII and perform ADCC effector function. Examples of human leukocytes that mediate ADCC include Peripheral Blood Mononuclear Cells (PBMCs), Natural Killer (NK) cells, monocytes, cytotoxic T cells, and neutrophils. The effector cells may be isolated from a natural source, for example from blood.
"antibody-dependent cell-mediated cytotoxicity" or "ADCC" refers to a cytotoxic form in which secreted Ig binds to Fc receptors (FcRs) present on certain cytotoxic cells (e.g., NK cells, neutrophils, and macrophages) enabling these cytotoxic effector cells to specifically bind to antigen-bearing target cells, followed by killing of the target cells with cytotoxins. Primary cells NK cells used to mediate ADCC express Fc γ RIII only, whereas monocytes express Fc γ RI, Fc γ RII and Fc γ RIII. The expression of FcR on hematopoietic cells is summarized in Table 3 at page 464 of ravatch and Kinet, Annu.Rev.Immunol 9:457-92 (1991). To assess ADCC activity of a molecule of interest, an in vitro ADCC assay may be performed, such as the assays described in U.S. Pat. No. 5,500,362 or 5,821,337 or U.S. Pat. No. 6,737,056 (Presta). Effector cells useful in such assays include PBMC and NK cells. Alternatively or in addition, the ADCC activity of the molecule of interest may be assessed in vivo, for example in an animal model such as that disclosed in Clynes et al proc.natl.acad.sci. (USA)95: 652-. Additional polypeptide variants having altered Fc region amino acid sequences (polypeptides having variant Fc regions) and increased or decreased ADCC activity are described, for example, in U.S. patent No. 7,923,538 and U.S. patent No. 7,994,290.
"complement-dependent cytotoxicity" or "CDC" refers to the lysis of target cells in the presence of complement. Activation of the classical complement pathway is initiated by the binding of the first component of the complement system (C1q) to antibodies (of the appropriate subclass) that bind to their cognate antigen. To assess complement activation, CDC assays may be performed, for example, as described in Gazzano-Santoro et al, j.immunol.methods 202:163 (1996). Polypeptide variants having altered Fc region amino acid sequences (polypeptides having variant Fc regions) and increased or decreased C1q binding ability are described, for example, in U.S. Pat. No. 6,194,551B1, U.S. Pat. No. 7,923,538, U.S. Pat. No. 7,994,290, and WO 1999/51642. See also, e.g., Gazzano-Santoro et al, J.Immunol.164: 4178-.
A polypeptide variant having "altered" FcR binding affinity or ADCC activity is a polypeptide variant having enhanced or reduced FcR binding activity and/or ADCC activity as compared to the parent polypeptide or as compared to a polypeptide comprising a native sequence Fc region. A polypeptide variant that "exhibits" increased binding to an FcR "binds at least one FcR with higher affinity than the parent polypeptide. A polypeptide variant that "exhibits" reduced binding to an FcR "binds at least one FcR with lower affinity than the parent polypeptide. Such variants that exhibit reduced binding to an FcR may have little or no significant binding to an FcR compared to a native sequence IgG Fc region, e.g., 0-20% binding to an FcR compared to a native sequence IgG Fc region.
A polypeptide variant that "mediates antibody-dependent cell-mediated cytotoxicity (ADCC) in the presence of human effector cells" more effectively than a parent antibody is one that mediates ADCC in vitro or in vivo more effectively when substantially the same amount of polypeptide variant and parent antibody is used in the assay. Typically, such variants will be identified using an in vitro ADCC assay as disclosed herein, although other assays or methods for determining ADCC activity, e.g. in animal models and the like, are also contemplated.
As used herein, the term "substantially similar" or "substantially the same" means a sufficiently high degree of similarity between two or more numerical values such that one of skill in the art would consider the difference between the two or more values to have little or no biological and/or statistical significance within the context of the biological feature measured by the values. In some embodiments, two or more substantially similar values differ by no more than about any of 5%, 10%, 15%, 20%, 25%, or 50%.
As used herein, the phrase "substantially different" means a sufficiently high degree of difference between two numerical values such that one of skill in the art would consider the difference between the two values to be statistically significant within the context of the biological feature measured by the values. In some embodiments, two substantially different values differ by greater than about any of 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or 100%.
The phrase "significantly reduced" as used herein means a sufficiently high degree of reduction between a numerical value and a reference numerical value such that one of skill in the art would consider the difference between the two values to be statistically significant within the context of the biological feature measured by the value. In some embodiments, the significantly reduced value is reduced by greater than any of about 10%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, or 100% compared to the reference value.
The term "leader sequence" refers to a sequence of amino acid residues located at the N-terminus of a polypeptide that facilitates secretion of the polypeptide from mammalian cells. The leader sequence may be cleaved upon export of the polypeptide from the mammalian cell, thereby forming the mature protein. The leader sequences may be natural or synthetic, and they may be heterologous or homologous to the protein to which they are attached.
A "native sequence" polypeptide comprises a polypeptide having the same amino acid sequence as a naturally occurring polypeptide. Thus, a native sequence polypeptide can have the amino acid sequence of a naturally occurring polypeptide from any mammal. Such native sequence polypeptides may be isolated from nature or may be produced by recombinant or synthetic methods. The term "native sequence" polypeptide specifically encompasses naturally occurring truncated or secreted forms of the polypeptide (e.g., an extracellular domain sequence), naturally occurring variant forms of the polypeptide (e.g., alternatively spliced forms), and naturally occurring allelic variants of the polypeptide.
Polypeptide "variant" means a biologically active polypeptide that has at least about 80% amino acid sequence identity to the native sequence polypeptide after aligning the sequences and introducing gaps, if necessary, to obtain the maximum percent sequence identity and not considering any conservative substitutions as part of the sequence identity. Such variants include, for example, polypeptides in which one or more amino acid residues are added or deleted at the N-terminus or C-terminus of the polypeptide. In some embodiments, variants will have at least about 80% amino acid sequence identity. In some embodiments, variants will have at least about 90% amino acid sequence identity. In some embodiments, the variant has at least about 95% amino acid sequence identity to the native sequence polypeptide.
As used herein, "percent (%) amino acid sequence identity" and "homology" with respect to a peptide, polypeptide, or antibody sequence is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in the particular peptide or polypeptide sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity and not considering any conservative substitutions as part of the sequence identity. Alignments for determining percent amino acid sequence identity can be performed in various ways within the skill in the art, for example, using publicly available computer software such as BLAST, BLAST-2, ALIGN, or MEGALIGN TM (DNASTAR) software. One skilled in the art can determine appropriate parameters for measuring alignment, including any algorithms necessary to achieve maximum alignment over the full length of the sequences being compared.
Amino acid substitutions may include, but are not limited to, the substitution of one amino acid for another in a polypeptide. Exemplary conservative substitutions are shown in table 1. Amino acid substitutions may be introduced into the antibody of interest and the product screened for a desired activity (e.g., retained/improved antigen binding, reduced immunogenicity, or improved ADCC or CDC).
TABLE 1
Original residue Exemplary substitutions
Ala(A) Val;Leu;Ile
Arg(R) Lys;Gln;Asn
Asn(N) Gln;His;Asp、Lys;Arg
Asp(D) Glu;Asn
Cys(C) Ser;Ala
Gln(Q) Asn;Glu
Glu(E) Asp;Gln
Gly(G) Ala
His(H) Asn;Gln;Lys;Arg
Ile(I) Leu; val; met; ala; phe; norleucine
Leu(L) Norleucine; ile; val; met; ala; phe (Phe)
Lys(K) Arg;Gln;Asn
Met(M) Leu;Phe;Ile
Phe(F) Trp;Leu;Val;Ile;Ala;Tyr
Pro(P) Ala
Ser(S) Thr
Thr(T) Val;Ser
Trp(W) Tyr;Phe
Tyr(Y) Trp;Phe;Thr;Ser
Val(V) Ile; leu; met; phe; ala; norleucine
Amino acids can be grouped according to common side chain traits:
(1) hydrophobicity: norleucine, Met, Ala, Val, Leu, Ile;
(2) neutral hydrophilicity: cys, Ser, Thr, Asn, Gln;
(3) acidity: asp and Glu;
(4) alkalinity: his, Lys, Arg;
(5) residues affecting chain orientation: gly, Pro;
(6) aromatic compounds: trp, Tyr, Phe.
Non-conservative substitutions would require conversion of members of one of these classes to another.
The term "vector" is used to describe a polynucleotide that can be engineered to contain a cloned polynucleotide or a polynucleotide that can be propagated in a host cell. The carrier may comprise one or more of the following elements: an origin of replication, one or more regulatory sequences (e.g., promoters and/or enhancers) that regulate the expression of the polypeptide of interest, and/or one or more selectable marker genes (e.g., antibiotic resistance genes and genes that can be used in colorimetric assays, such as beta galactosidase). The term "expression vector" refers to a vector for expressing a polypeptide of interest in a host cell.
"host cell" refers to a cell that may be or has been a recipient for a vector or isolated polynucleotide. The host cell may be a prokaryotic cell or a eukaryotic cell. Exemplary eukaryotic cells include mammalian cells, such as primate or non-primate cells; fungal cells, such as yeast; a plant cell; and insect cells. Non-limiting exemplary mammalian cells include, but are not limited to, NSO cells,Cells (Crucell) and 293 and CHO cells, and their derivatives, such as 293-6E and DG44 cells, respectively. Host cells include progeny of a single host cell, and the progeny may not necessarily be identical (in morphology or genomic DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation. Host cells include cells transfected in vivo with a polynucleotide provided herein.
As used herein, the term "isolated" refers to a molecule that has been separated from at least some components that are typically found or produced in nature. For example, a polypeptide is said to be "isolated" when it is separated from at least some components of the cell from which it was produced. Physically separating the supernatant containing the polypeptide from the cell in which it is produced is considered to "isolate" the polypeptide when it is secreted by the cell after expression. Similarly, a polynucleotide is said to be "isolated" when it is not part of a larger polynucleotide that it normally exists in nature (e.g., genomic or mitochondrial DNA in the case of a DNA polynucleotide), or is isolated from at least some of the components of the cell in which it is produced (e.g., in the case of an RNA polynucleotide). Thus, a DNA polynucleotide contained in a vector within a host cell may be referred to as "isolated.
The terms "individual" or "subject" are used interchangeably herein to refer to an animal; such as mammals. In some embodiments, methods of treating mammals are provided, including, but not limited to, humans, rodents, apes, felines, canines, equines, bovines, porcines, ovines, antelopes, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets. In some examples, "individual" or "subject" refers to an individual or subject in need of treatment for a disease or disorder. In some embodiments, the subject receiving treatment may be a patient, specifying the fact that the subject has been identified as having a treatment-related disorder or as being at sufficient risk of developing the disorder.
As used herein, "disease" or "disorder" refers to a condition for which treatment is needed and/or desired.
As used herein, "cancer" and "tumor" refer to interchangeable terms for any abnormal cell or tissue growth or proliferation in an animal. As used herein, the terms "cancer" and "tumor" encompass solid cancers and hematologic/lymphoid cancers, and also encompass malignant, premalignant, and benign growths, such as dysplasias. Examples of cancer include, but are not limited to, carcinoma, lymphoma, blastoma, sarcoma, and leukemia. More specific non-limiting examples of such cancers include squamous cell cancer, small-cell lung cancer, pituitary cancer, esophageal cancer, astrocytoma, soft tissue sarcoma, non-small cell lung cancer, adenocarcinoma of the lung, squamous carcinoma of the lung, cancer of the peritoneum, hepatocellular cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, cervical cancer, ovarian cancer, liver cancer, bladder cancer, hepatoma, breast cancer, colon cancer, colorectal cancer, endometrial or uterine carcinoma, salivary gland carcinoma, kidney cancer (kidney/renal cancer), liver cancer, prostate cancer, vulval cancer, thyroid cancer, liver cancer, brain cancer, endometrial cancer, testicular cancer, cholangiocarcinoma, gallbladder cancer, gastric cancer, melanoma, mesothelioma, and various types of head and neck cancer. In some embodiments, the hematologic/lymphoid cancer is referred to as "hematologic cancer. Non-limiting exemplary hematologic cancers include B cell and T cell mixed leukemia, B cell lymphoma, Chronic Myeloid Leukemia (CML), chronic myelomonocytic leukemia, diffuse large B cell lymphoma (DLBC), lymphoma, Mantle Cell Lymphoma (MCL), multiple myeloma, myelodysplastic syndrome (MDS), myeloproliferative disorders, peripheral T cell lymphoma, T cell leukemia, Acute Myeloid Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic Lymphoma (SLL), CLL/SLL, mature T cell and NK cell lymphoma, follicular lymphoma, Acute Lymphocytic Leukemia (ALL), T cell acute lymphocytic leukemia (TALL), T cell adult acute lymphocytic leukemia, T cell childhood acute lymphocytic leukemia, lymphoblastic lymphoma, lymphoblastic leukemia, and lymphoblastic leukemia, and myeloproliferative disorders, lymphoblastic leukemia, and myeloproliferative disorders, lymphoblastic leukemia, and myeloproliferative disorders, lymphoblastic leukemia, and myeloproliferative disorders, Cutaneous T Cell Lymphoma (CTCL), adult T cell leukemia/lymphoma (ATLL), T cell lymphoblastic leukemia/lymphoma (TLLL), Angioimmunoblastic T Cell Lymphoma (ATCL), Hepatosplenic T Cell Lymphoma (HTCL), peripheral T cell lymphoma not otherwise specified (PTCL NOS), Burkitt Lymphoma (BL), chronic myelomonocytic leukemia (CMML), extranodal NK/T cell lymphoma (NKTCL), Primary Effusion Lymphoma (PEL), acute lymphocytic leukemia/acute myeloid leukemia (ALL, AML), Histiocytic Lymphoma (HL), Marginal Zone Lymphoma (MZL), B cell acute lymphocytic leukemia, and Anaplastic Large Cell Lymphoma (ALCL).
As used herein, "treatment" is a method for obtaining a beneficial or desired clinical result. "treatment" as used herein encompasses any administration or use of a therapeutic agent for a disease in a mammal (including a human). For purposes of this disclosure, beneficial or desired clinical results include, but are not limited to, any one or more of the following: alleviating one or more symptoms, alleviating the extent of disease, preventing or delaying the spread of disease (e.g., metastasis, e.g., to the lung or lymph nodes), preventing or delaying the recurrence of disease, delaying or slowing the progression of disease, ameliorating the disease state, inhibiting the progression of disease or disease, inhibiting or slowing the progression of disease or disease, arresting its development, and alleviating (whether in part or in whole). "treating" also encompasses reducing the pathological consequences of a proliferative disease. The methods provided herein encompass any one or more of these therapeutic aspects. Consistent with the above, the term treatment does not require 100% removal of all aspects of the disorder.
By "ameliorating" is meant the reduction or amelioration of one or more symptoms as compared to the absence of administration of an anti-CCR 8 antibody. "improving" also includes shortening or reducing the duration of symptoms.
In the context of cancer, the term "treatment" includes any or all of the following: inhibiting cancer cell growth, inhibiting cancer cell replication, reducing overall tumor burden, and ameliorating one or more symptoms associated with the disease.
As used herein, the term "regulatory T cells" (also referred to as "tregs" or "Treg cells" or "suppressor T cells") are T cell subsets that are immunosuppressive and that typically suppress or down-regulate the induction and proliferation of effector T cells. Tregs express CD4, FOXP3 and CD25(IL-2 receptor alpha chain). Human Foxp3+ CD4+ T cells were divided into three subfractions based on the expression levels of Foxp3 and the cell surface molecules CD25 and CD45 RA. The Foxp3hiCD 45-CD 45RA-CD25hi and Foxp3loCD45RA + CD25lo phenotypes corresponded to suppressive Treg cells, whereas the Foxp3loCD45RA-CD25lo fraction labeled non-suppressive active T-effector (Teff) cells. Furthermore, Treg cells from cancer patients are often characterized by a unique expression profile of chemokine receptors such as CCR4, CXCR4, and CCR5 that promotes their migration into tumors in response to corresponding chemokine ligands from the tumor microenvironment, as compared to Treg cells in healthy subjects. See, e.g., Liu et al, FEBS J. (2016)283(14):2731-48, and Miyara et al, Immunity (2009)30, 899-.
"conventional T cells" or "Tconv" are populations of T cells that are generally CD4 positive (i.e., CD4+), but differ from tregs in that Tconv is generally FoxP3 negative (i.e., FoxP 3-).
The term "biological sample" means an amount of material from or previously living. Such substances include, but are not limited to, blood (e.g., whole blood), plasma, serum, urine, amniotic fluid, synovial fluid, endothelial cells, leukocytes, monocytes, other cells, organs, tissues, bone marrow, lymph nodes, and spleen.
The term "control" refers to a composition that is known to contain no analyte ("negative control") or to contain an analyte ("positive control"). The positive control may comprise a known concentration of analyte. "control," "positive control," and "calibrator" are used interchangeably herein to refer to a composition comprising a known concentration of an analyte. A "positive control" can be used to establish an assay performance characteristic and is a useful indicator of the integrity of a reagent (e.g., an analyte).
"predetermined cut-off value" and "predetermined level" generally refer to a measured cut-off value used to assess the diagnostic/prognostic/therapeutic efficacy outcome by comparing the measured outcome to a predetermined cut-off value/level that has been correlated or correlated with various clinical parameters (e.g., disease severity, progression/non-progression/improvement, etc.). While the present disclosure may provide exemplary predetermined levels, it is well known that the cut-off value may vary depending on the nature of the immunoassay (e.g., the antibody used, etc.). Further, it is within the skill of one of ordinary skill in the art to apply the disclosure herein to other immunoassays to obtain immunoassay-specific cut-offs for those other immunoassays based on the present disclosure. While the exact value of the predetermined cut-off/level may vary between assays, correlation, if any, as described herein is generally applicable.
The term "inhibition" or "inhibition" refers to the reduction or cessation of any phenotypic characteristic or the reduction or cessation of the incidence, extent or likelihood of that characteristic. "reduce" or "inhibit" refers to reduce, reduce or prevent activity, function and/or amount as compared to a reference. In some embodiments, "reduce" or "inhibit" means the ability to cause an overall reduction of 20% or greater. In some embodiments, "reduce" or "inhibit" means the ability to cause an overall reduction of 50% or greater. In some embodiments, "reduce" or "inhibit" means the ability to cause an overall reduction of 75%, 85%, 90%, 95%, or greater. In some embodiments, the amount is inhibited or reduced over a period of time relative to a control dose (such as a placebo) over the same period of time. As used herein, "reference" refers to any sample, standard, or level used for comparison purposes. The reference may be obtained from a healthy and/or non-diseased sample. In some examples, the reference may be obtained from an untreated sample. In some examples, the reference is obtained from non-diseased and untreated samples of the subject individual. In some embodiments, the reference is obtained from one or more healthy individuals that are not the subject or patient.
As used herein, "delaying the progression of a disease" means delaying, hindering, slowing, delaying, stabilizing, suppressing and/or delaying the progression of a disease, such as cancer. The delay may have different lengths of time depending on the history of the disease and/or the individual to be treated. It will be apparent to those skilled in the art that a sufficient or significant delay may actually encompass prevention, as the individual does not develop the disease. For example, the development of advanced cancers, such as metastases, may be delayed.
As used herein, "preventing" includes providing prophylaxis regarding the occurrence or recurrence of a disease in a subject who may be predisposed to the disease but has not yet been diagnosed with the disease. Unless otherwise indicated, the terms "reduce", "inhibit" or "prevent" do not denote or require complete prevention at all times.
As used herein, "inhibiting" a function or activity refers to decreasing the function or activity when compared to an otherwise identical condition or alternatively to another condition except for the condition or parameter of interest. For example, an antibody that inhibits tumor growth reduces the rate of tumor growth compared to the rate of tumor growth in the absence of the antibody.
The "therapeutically effective amount" of a substance/molecule, agonist or antagonist may vary depending on factors such as the disease state, age, sex and weight of the individual, and the ability of the substance/molecule, agonist or antagonist to elicit a desired response in the individual. A therapeutically effective amount is also an amount that outweighs any toxic or detrimental effects of the substance/molecule, agonist or antagonist. A therapeutically effective amount may be delivered in one or more administrations. A therapeutically effective amount is an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic and/or prophylactic result.
A prophylactically effective amount is an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, but not necessarily, the prophylactically effective amount will be less than the therapeutically effective amount due to the use of a prophylactic dose in the subject prior to or at an early stage of the disease.
The terms "pharmaceutical formulation" and "pharmaceutical composition" refer to a formulation that is present in a form that allows the biological activity of the active ingredient to be effective and that does not contain additional components that have unacceptable toxicity to the subject to which the formulation is to be administered. Such formulations may be sterile.
By "pharmaceutically acceptable carrier" is meant a non-toxic solid, semi-solid, or liquid filler, diluent, encapsulating material, formulation aid, or carrier conventional in the art, used with a therapeutic agent, which together comprise a "pharmaceutical composition" for administration to a subject. Pharmaceutically acceptable carriers are non-toxic to recipients at the dosages and concentrations employed, and are compatible with other ingredients of the formulation. Pharmaceutically acceptable carriers are suitable for the formulation used.
"sterile" preparations are sterile or substantially free of living microorganisms and spores thereof.
The term "IDO inhibitor" refers to an agent that is capable of inhibiting the activity of indoleamine 2, 3-dioxygenase (IDO) and thereby reversing IDO-mediated immunosuppression. IDO inhibitors may inhibit IDO1 and/or IDO2(INDOL 1). The IDO inhibitor may be a reversible or irreversible IDO inhibitor. A "reversible IDO inhibitor" is a compound that reversibly inhibits IDO enzyme activity at a catalytic site or at a non-catalytic site, and an "irreversible IDO inhibitor" is a compound that irreversibly inhibits IDO enzyme activity by forming a covalent bond with an enzyme. Non-limiting exemplary IDO inhibitors include indoximod (New Link Genetics), INCB024360(Incyte Corp.), 1-methyl-D-tryptophan (New Link Genetics), and GDC-0919(Genentech, Inc.).
"chimeric antigen receptor T cell therapy" or "CAR-T therapy" refers to a therapeutic agent comprising T cells genetically modified to express a receptor that recognizes an antigen expressed by a tumor cell. The antigen may be an antigen specifically expressed by a tumor or an antigen expressed by both cancer cells and healthy tissue. In some embodiments, the CAR-T therapy is an adoptive CAR-T therapy in which patient T cells are removed and modified to express a chimeric antigen receptor and then returned to the patient. See, e.g., Dai et al, 2016, J Natl Cancer Inst,108(7): djv439, doi:10.1093/jnci/djv 439; gill et al 2015, Blood Rev, pii: S0268-960X (15)00080-6, doi: 10.1016/j.blre.2015.10.003; gill et al 2015, Immunol Rev 263(1):68-89.doi 10.1111/imr.12243.
Administration "in combination with" one or more additional therapeutic agents "includes simultaneous (concurrent) and sequential or sequential administration in any order.
The term "simultaneous" is used herein to refer to the administration of two or more therapeutic agents, wherein at least some of the administrations overlap in time or wherein the administration of one therapeutic agent falls within a short period of time relative to the administration of the other therapeutic agent. For example, the two or more therapeutic agents are administered at intervals not exceeding about the specified number of minutes.
The term "sequentially" is used herein to refer to the administration of two or more therapeutic agents, wherein the administration of one or more agents continues after the administration of one or more other agents is discontinued, or wherein the administration of one or more agents begins before the administration of one or more other agents. For example, administration of two or more therapeutic agents is administered at intervals exceeding about the specified number of minutes.
As used herein, "binding" refers to administering one form of treatment in addition to another form of treatment. Thus, "in conjunction with" refers to the administration of one form of treatment before, during, or after the administration of the other form of treatment to the individual.
The term "package insert" is used to refer to an insert typically included in commercial packaging for a therapeutic product that contains information about the indication, usage, dosage, administration, combination therapy, contraindications, and/or warnings regarding the use of such therapeutic products.
An "article of manufacture" is any article of manufacture (e.g., a package or container) or kit comprising at least one reagent, e.g., a drug for treating a disease or disorder (e.g., cancer) or a probe for specifically detecting a biomarker described herein. In some embodiments, the article of manufacture or kit is promoted, distributed, or sold as a means for performing the methods described herein.
The terms "label" and "detectable label" mean a moiety that is attached to an antibody or analyte thereof such that a reaction (e.g., binding) between the members of a specific binding pair is detectable. The labeled member of the specific binding pair is referred to as "detectably labeled". Thus, the term "labeled binding protein" refers to a protein that incorporates a label that provides identification of the binding protein. In some embodiments, the label is a detectable marker that can produce a signal that can be detected by visual or instrumental means, such as a polypeptide that incorporates a radiolabeled amino acid or is linked to a biotin moiety that can be detected by labeled avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or colorimetric methods). Examples of labels for polypeptides include, but are not limited to, the following: a radioisotope or radionuclide (e.g., 3 H、 14 C、 35 S、 90 Y、 99 Tc、 111 In、 125 I、 131 I、 177 Lu、 166 ho or 153 Sm); chromogens, fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzyme labels (e.g., horseradish peroxidase, luciferase, alkaline phosphatase); a chemiluminescent marker; a biotin group; a predetermined polypeptide epitope recognized by the second reporter (e.g., leucine zipper pair sequence, binding site of a second antibody, metal binding domain, epitope tag); and magnetic agents such as gadolinium chelates. Representative examples of labels commonly used in immunoassays include the generation of A light-generating moiety (e.g., an acridinium compound) and a fluorescent moiety (e.g., fluorescein). In this regard, the moiety may not be detectably labeled by itself, but may become detectable upon reaction with another moiety.
The term "conjugate" refers to an antibody chemically linked to a second chemical moiety, such as a therapeutic or cytotoxic agent. The term "agent" includes chemical compounds, mixtures of chemical compounds, biological macromolecules or extracts made from biological materials. In some embodiments, the therapeutic or cytotoxic agent includes, but is not limited to, pertussis toxin, paclitaxel, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxy anthrax dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin, and analogs or homologs thereof. When used in the context of an immunoassay, the conjugate antibody may be an antibody that serves as a detectable label for the detection antibody.
anti-CCR 8 antibodies
Novel antibodies against CCR8 are provided. anti-CCR 8 antibodies include, but are not limited to, humanized antibodies, chimeric antibodies, mouse antibodies, human antibodies, and antibodies comprising the heavy and/or light chain CDRs discussed herein. In some embodiments, isolated antibodies that bind CCR8 are provided. In some embodiments, monoclonal antibodies that bind to CCR8 are provided. In some embodiments, the anti-CCR 8 antibody is an antagonist anti-CCR 8 antibody. In some embodiments, an anti-CCR 8 antibody provided herein inhibits the binding of CCR8 to CCL 1. In some embodiments, administration of an anti-CCR 8 antibody described herein reduces infiltrating Treg cells in the cancer of the subject. In some embodiments, the anti-CCR 8 antibodies herein are administered to treat hematologic cancers that express CCR 8.
In some embodiments, the anti-CCR 8 antibody comprises at least one, two, three, four, five, or six CDRs selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 12; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 13; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 14; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 15; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 16; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the anti-CCR 8 antibody comprises at least one, two, three, four, five, or six CDRs selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 24; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 25; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 26; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 27; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 28; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO. 29.
In some embodiments, the anti-CCR 8 antibody comprises at least one, two, three, four, five, or six CDRs selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 36; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 37; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 38; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 39; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 40; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 41.
In some embodiments, the anti-CCR 8 antibody comprises at least one, two, three, four, five, or six CDRs selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 48; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 49; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 50; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 51; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 52; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 53.
In some embodiments, the anti-CCR 8 antibody comprises at least one, two, three, four, five, or six CDRs selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 63; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65.
In some embodiments, the anti-CCR 8 antibody comprises at least one, two, three, four, five, or six CDRs selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 86; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 87; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 88; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, the anti-CCR 8 antibody comprises a heavy chain variable region and a light chain variable region. In some embodiments, the anti-CCR 8 antibody comprises at least one heavy chain comprising a heavy chain variable region and at least a portion of a heavy chain constant region and at least one light chain comprising a light chain variable region and at least a portion of a light chain constant region. In some embodiments, the anti-CCR 8 antibody comprises two heavy chains and two light chains, wherein each heavy chain comprises a heavy chain variable region and at least a portion of a heavy chain constant region, wherein each light chain comprises a light chain variable region and at least a portion of a light chain constant region. As used herein, a single chain fv (scfv), or any other antibody comprising a single polypeptide chain, e.g., containing all six CDRs (three heavy chain CDRs and three light chain CDRs), is considered to have a heavy chain and a light chain. In some embodiments, the heavy chain is a region of an anti-CCR 8 antibody comprising three heavy chain CDRs. In some embodiments, the light chain is a region of an anti-CCR 8 antibody comprising three light chain CDRs.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs comprising (a) HCDR1 comprising the amino acid sequence of SEQ ID NO: 12; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 13; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 14; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 15; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 16; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO. 17.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs comprising (a) HCDR1 comprising the amino acid sequence of SEQ ID NO: 24; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 25; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 26; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 27; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 28; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO. 29.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs, including (a) HCDR1 comprising the amino acid sequence of SEQ ID NO: 36; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 37; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 38; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 39; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 40; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 41.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs comprising (a) HCDR1 comprising the amino acid sequence of SEQ ID NO: 48; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 49; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 50; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 51; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 52; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 53.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs comprising (a) HCDR1 comprising the amino acid sequence of SEQ ID NO: 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 63; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs comprising (a) HCDR1 comprising the amino acid sequence of SEQ ID NO:84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 86; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 87; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO: 88; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, the anti-CCR 8 antibody comprises six CDRs as described above and binds CCR 8. In some embodiments, the anti-CCR 8 antibody comprises six CDRs as described above, binds CCR8, and inhibits binding of CCR8 to CCL 1. In some embodiments, the anti-CCR 8 antibody comprises six CDRs as described above, binds CCR8, and enhances the immune response in the subject, and/or increases the activation of T cells in the subject upon administration of the antibody to the subject.
In some embodiments, anti-CCR 8 antibodies are provided that compete with the anti-CCR 8 antibodies described herein for binding to CCR 8. In some embodiments, antibodies can be made and/or used that compete for binding with any of the antibodies provided herein.
In some embodiments, an anti-CCR 8 antibody comprises at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 12; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 13; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 14.
In some embodiments, an anti-CCR 8 antibody comprises at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 24; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 25; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 26.
In some embodiments, the anti-CCR 8 antibody comprises at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 36; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 37; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 38.
In some embodiments, the anti-CCR 8 antibody comprises at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 48; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 49; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 50.
In some embodiments, the anti-CCR 8 antibody comprises at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79.
In some embodiments, an anti-CCR 8 antibody comprises at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; and (c) HCDR3 comprising the amino acid sequence of SEQ ID NO: 86.
In some embodiments, an anti-CCR 8 antibody comprises at least one, at least two, or all three VL CDR sequences selected from: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO. 15; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 16; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 17.
In some embodiments, an anti-CCR 8 antibody comprises at least one, at least two, or all three VL CDR sequences selected from: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO. 27; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 28; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 29.
In some embodiments, an anti-CCR 8 antibody comprises at least one, at least two, or all three VL CDR sequences selected from: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO: 39; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 40; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 41.
In some embodiments, the anti-CCR 8 antibody comprises at least one, at least two, or all three VL CDR sequences selected from: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO. 51; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 52; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 53.
In some embodiments, the anti-CCR 8 antibody comprises at least one, at least two, or all three VL CDR sequences selected from: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO. 63; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65.
In some embodiments, the anti-CCR 8 antibody comprises at least one, at least two, or all three VL CDR sequences selected from: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO. 87; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 88; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, any of the six CDRs provided herein can be combined as subparts with any of the other CDRs provided herein, such that there are a total of six CDRs in the construct. Thus, in some embodiments, two CDRs from a first antibody (e.g., HCDR1 and HCDR2) may be combined with four CDRs from a second antibody (HCDR3, LCDR1, LCDR2, and LCDR 3). In some embodiments, two or fewer residues in one or more CDRs may be substituted to obtain variants thereof. In some embodiments, two or fewer residues in 1, 2, 3, 4, 5, or 6 CDRs may be substituted.
In some embodiments, the anti-CCR 8 antibody comprises (I) a VH domain comprising at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 12; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 13; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 14; and (II) a VL domain comprising at least one, at least two, or all three VL CDR sequences selected from: (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 15; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 16; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 17.
In some embodiments, the anti-CCR 8 antibody comprises (I) a VH domain comprising at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 24; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 25; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 26; and (II) a VL domain comprising at least one, at least two, or all three VL CDR sequences selected from: (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 27; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 28; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO. 29.
In some embodiments, an anti-CCR 8 antibody comprises (I) a VH domain comprising at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 36; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 37; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 38; and (II) a VL domain comprising at least one, at least two, or all three VL CDR sequences selected from: (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 39; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 40; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 41.
In some embodiments, an anti-CCR 8 antibody comprises (I) a VH domain comprising at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 48; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 49; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO. 50; and (II) a VL domain comprising at least one, at least two, or all three VL CDR sequences selected from: (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 51; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 52; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 53.
In some embodiments, an anti-CCR 8 antibody comprises (I) a VH domain comprising at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79; and (II) a VL domain comprising at least one, at least two, or all three VL CDR sequences selected from: (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 63; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65.
In some embodiments, the anti-CCR 8 antibody comprises (I) a VH domain comprising at least one, at least two, or all three VH CDR sequences selected from: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 86; and (II) a VL domain comprising at least one, at least two, or all three VL CDR sequences selected from: (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 87; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 88; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, the anti-CCR 8 antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:68 or 74. In some embodiments, VH sequences having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contain substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-CCR 8 antibody comprising the sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:68 or 74. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). Optionally, the anti-CCR 8 antibody comprises the VH sequence in SEQ ID NO:68 or 74, including post-translational modifications of that sequence.
In some embodiments, the VH comprises: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79.
In some embodiments, an anti-CCR 8 antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:92 or 96. In some embodiments, VH sequences having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contain substitutions (e.g., conservative substitutions), insertions, or deletions relative to the reference sequence, but an anti-CCR 8 antibody comprising the sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:92 or 96. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). Optionally, the anti-CCR 8 antibody comprises the VH sequence of SEQ ID NO 92 or 96, including post-translational modifications of that sequence.
In some embodiments, the VH comprises: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 86.
In some embodiments, anti-CCR 8 antibodies are provided, wherein the antibodies comprise a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:69 or 75. In some embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to a reference sequence, but an anti-CCR 8 antibody comprising that sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID No. 69 or 75. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). Optionally, the anti-CCR 8 antibody comprises the VL sequence of SEQ ID NO:69 or 75, including post-translational modifications of this sequence.
In some embodiments, the VL comprises: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO: 63; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65.
In some embodiments, anti-CCR 8 antibodies are provided, wherein the antibodies comprise a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% sequence identity to the amino acid sequence of SEQ ID NO:93 or 97. In some embodiments, a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains substitutions (e.g., conservative substitutions), insertions, or deletions relative to a reference sequence, but an anti-CCR 8 antibody comprising that sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:93 or 97. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). Optionally, the anti-CCR 8 antibody comprises the VL sequence of SEQ ID NO:93 or 97, including post-translational modifications of that sequence.
In some embodiments, the VL comprises: (a) LCDR1 comprising the amino acid sequence of SEQ ID NO: 87; (b) LCDR2 comprising the amino acid sequence of SEQ ID NO. 88; and (c) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, an anti-CCR 8 antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID No. 68 or 74 and a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID No. 69 or 75. In some embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, and a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, but an anti-CCR 8 antibody comprising the sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:68 or 74. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:69 or 75. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). In some embodiments, the anti-CCR 8 antibody comprises: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO. 63; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65.
In some embodiments, the anti-CCR 8 antibody comprises a VH sequence in SEQ ID NO:68 or 74, including post-translational modifications of one or both sequences, and comprises a VL sequence in SEQ ID NO:69 or 75, including post-translational modifications of one or both sequences.
In some embodiments, an anti-CCR 8 antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:92 or 96 and a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:93 or 97. In some embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, and a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, but an anti-CCR 8 antibody comprising the sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:92 or 96. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:93 or 97. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). In some embodiments, the anti-CCR 8 antibody comprises: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 86; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 87; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO: 88; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, the anti-CCR 8 antibody comprises a VH sequence in SEQ ID NO:92 or 96, including post-translational modifications of one or both sequences, and comprises a VL sequence in SEQ ID NO:93 or 97, including post-translational modifications of one or both sequences.
In some embodiments, the anti-CCR 8 antibody comprises a VH as in any embodiment provided herein and a VL as in any embodiment provided herein. In some embodiments, the antibody comprises the VH and VL sequences in SEQ ID NO:68 or 74 and SEQ ID NO:69 or 75, respectively, including post-translational modifications of those sequences. In some embodiments, the antibody comprises the VH and VL sequences in SEQ ID NOs 92 or 96 and 93 or 97, respectively, including post-translational modifications of those sequences.
In some embodiments, anti-CCR 8 antibodies are provided, wherein the antibodies comprise a Heavy Chain (HC) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID No. 70 or 76. Optionally, the anti-CCR 8 antibody comprises the HC sequence of SEQ ID NO 70 or 76, including post-translational modifications.
In some embodiments, anti-CCR 8 antibodies are provided, wherein the antibodies comprise HCs having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:94 or 98. Optionally, the anti-CCR 8 antibody comprises the HC sequence of SEQ ID NO 94 or 98, including post-translational modifications.
In some embodiments, anti-CCR 8 antibodies are provided, wherein the antibodies comprise a Light Chain (LC) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:71 or 77. Optionally, the anti-CCR 8 antibody comprises the LC sequence of SEQ ID NO 71 or 77, including post-translational modifications.
In some embodiments, anti-CCR 8 antibodies are provided, wherein the antibodies comprise LCs having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO 95 or 99. Optionally, the anti-CCR 8 antibody comprises the LC sequence in SEQ ID NO 95 or 99, including post-translational modifications.
In some embodiments, the anti-CCR 8 antibody comprises a HC as in any embodiment provided herein and a LC as in any embodiment provided herein. In some embodiments, the antibody comprises HC and LC sequences in SEQ ID NOs 70 or 76 and 71 or 77, respectively, including post-translational modifications of those sequences. In some embodiments, the antibody comprises HC and LC sequences in SEQ ID NOs 94 or 98 and 95 or 99, respectively, including post-translational modifications of those sequences.
In some embodiments, antibodies are provided that compete for binding to CCR8 with anti-CCR 8 antibodies provided herein. In some embodiments, the antibodies compete for binding to an epitope on CCR8 with anti-CCR 8 antibodies provided herein.
In some embodiments, competition assays can be used to identify monoclonal antibodies that compete with anti-CCR 8 antibodies described herein (such as 1-K16, 1-K17, 6-B09, 7-B16, 13-E16, and/or 19-O07) for binding to CCR 8. Competition assays can be used to determine whether two antibodies bind to the same epitope by recognizing the same or spatially overlapping epitopes, or one antibody competitively inhibits the binding of the other antibody to the antigen. In some embodiments, such a competing antibody binds to the same epitope as the epitope bound by the antibody described herein. Exemplary competition assays include, but are not limited to, conventional assays, such as those provided in the following references: harlow and Lane (1988) Antibodies Chapter 14 of A Laboratory Manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.). Detailed exemplary Methods for Mapping the Epitope bound by an antibody are provided in Morris (1996) "Epitope Mapping Protocols," Methods in Molecular Biology Vol.66 (Humana Press, Totowa, N.J.). In some embodiments, two antibodies are said to bind the same epitope if each blocks 50% or more of the binding of the other antibody. In some embodiments, the antibody that competes with the anti-CCR 8 antibodies described herein is a chimeric, humanized, or human antibody. In some embodiments, antibodies are provided that compete with chimeric, humanized, or human anti-CCR 8 antibodies as described herein.
In addition, the present disclosure also provides variants of the above disclosed antibodies, such as variants of the 7-B16 antibody. For example, in some embodiments, the disclosure provides an isolated antibody that binds human CCR8, wherein the antibody comprises HCDR3, the HCDR3 comprises SEQ ID NO:86 or a variant of SEQ ID NO:86 containing 1, 2, or 3 mutations, and wherein the antibody binds human CCR8 and has ADCC activity. In some embodiments, the mutation is a substitution (e.g., a conservative or non-conservative substitution), a deletion, or an insertion. In some embodiments, the 1, 2, or 3 mutations are at least one of amino acid positions 1-4, 6, 7, or 12 of SEQ ID No. 86. In some embodiments, the substitution is a conservative substitution. In some embodiments, the conservative substitution is at amino acid position 1, 4, or 12 of SEQ ID No. 86. In some embodiments, the substitution is a non-conservative substitution. In some embodiments, the non-conservative substitution is at amino acid position 7 of SEQ ID NO 86. In some embodiments, the antibody comprises at least 2 substitutions in HCDR 3. In some embodiments, the at least 2 substitutions are at least one of amino acid positions 1-4, 6, 7, or 12 of SEQ ID No. 86. In some embodiments, the at least 2 substitutions are conservative substitutions. In some embodiments, at least one conservative substitution is at SEQ ID NO 86 Amino acid position 1, 4 or 12. In some embodiments, the at least 2 substitutions are non-conservative substitutions. In some embodiments, the at least one non-conservative substitution is at amino acid position 7 of SEQ ID No. 86. In some embodiments, when more than one substitution mutation is present, the mutation comprises a conservative substitution and a non-conservative substitution. In some embodiments, the present disclosure provides an isolated antibody that binds human CCR8, wherein the antibody comprises HCDR3 that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:86, and wherein the antibody binds human CCR8 and has ADCC activity. In some embodiments, HCDR3 comprises an amino acid sequence selected from any one of SEQ ID NO 86 and SEQ ID NO 104-119. In some embodiments, the antibody comprises HCDR1 comprising SEQ ID NO 84 or SEQ ID NO 123. In some embodiments, the antibody comprises HCDR2 comprising SEQ ID NO:85 or SEQ ID NO: 124. In some embodiments, the antibody comprises LCDR1 comprising SEQ ID No. 87 or SEQ ID No. 120. In some embodiments, the antibody comprises LCDR2 comprising SEQ ID No. 88 or SEQ ID No. 121. In some embodiments, the antibody comprises LCDR3 comprising SEQ ID No. 89 or SEQ ID No. 122. In some embodiments, the ADCC activity comprises EC50 values of less than 200ng/ml, 175ng/ml, 150ng/ml, 125ng/ml, 100ng/ml, 75ng/ml, 50ng/ml, 25ng/ml, 20ng/ml, 15ng/ml, 10ng/ml, 9ng/ml, 8ng/ml, 7ng/ml, 6ng/ml, 5ng/ml, 4ng/ml, 3ng/ml, 2ng/ml, or 1ng/ml, as measured by an ADCC reporter mechanism of action (MOA) based bioassay, such as the Promega reporter assay disclosed in example 7 below. In some embodiments, the ADCC activity is more potent than the 7-B16 antibody. In some embodiments, the ADCC activity is at least as effective as the 7-B16 antibody. In some embodiments, the antibody is to K of human CCR8 D Equal to or lower than the 7-B16 antibody (e.g., as determined by kinetic exclusion assay (i.e., KinExA)). In some embodiments, the antibody is K on a cell of human CCR8 D Equal to or lower than 7-B16 antibody (e.g., as determined by kinetic exclusion assay (i.e., KinExA)). In some embodiments, the antibody comprises at least one that enhances cell killingAnd (5) modifying. In some embodiments, the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC). In some embodiments, the at least one modification is afucosylation. In some embodiments, the at least one modification is one or more heavy chain constant region mutations at one or more positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334 and P396. In some embodiments, the one or more heavy chain constant region mutations are one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L. In some embodiments, the one or more heavy chain constant region mutations are selected from the group consisting of: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E. In some embodiments, the at least one modification is galactosylation. In some embodiments, the antibody has an affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75pM, or less than 50pM, or less than 25pM D ) (as determined by kinetic exclusion assay (i.e., KinExA)) binds to human CCR 8. In some embodiments, the antibody has an on-cell affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75, or less than 50pM, or less than 25pM D ) Binds to human CCR8 as determined by, for example, a kinetic exclusion assay (i.e., KinExA). In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a human or humanized antibody. In some embodiments, the antibody is a full-length antibody. In some embodiments, the antibody is an IgG1 or IgG3 antibody. Such variants may be used in methods of treating cancer (including both hematologic cancers and solid tumors).
In some embodiments, antibodies that bind to any one or more epitopes bound by antibodies provided herein are provided. In some embodiments, antibodies are provided that bind to and overlap the epitope bound by the antibodies of the invention. In some embodiments, an antibody that competes with at least one antibody provided herein is provided. In some embodiments, an antibody that competes with at least two of the antibodies provided herein is provided. In some embodiments, an antibody that competes with at least three of the antibodies provided herein is provided. In some embodiments, the epitope bound by the antibody overlaps with the epitope bound by the antibody described in the examples herein. In some embodiments, the entire epitope is bound and/or blocked by the competing antibody. In some embodiments, a portion of the epitope is bound and/or blocked by the competing antibody. In some embodiments, the paratope of the competing antibody binds to at least a portion of an epitope of an antibody provided herein. In some embodiments, the paratope of the competing antibody binds to the target, and different portions of the structure of the competing antibody block at least a portion of an epitope of an antibody provided herein.
Exemplary chimeric antibodies
In some embodiments, the antibodies provided herein are chimeric antibodies. Certain chimeric antibodies are described, for example, in U.S. Pat. nos. 4,816,567; and Morrison et al, (1984) Proc. Natl. Acad. Sci. USA,81:6851-6855 (1984)). In one example, a chimeric antibody comprises a non-human variable region (e.g., a variable region derived from a mouse, rat, hamster, rabbit, or non-human primate, such as a monkey) and a human constant region. In another example, a chimeric antibody is a "class switch" antibody in which the class or subclass has been changed from that of the parent antibody. Chimeric antibodies include antigen-binding fragments thereof.
Non-limiting exemplary chimeric antibodies include chimeric antibodies comprising the heavy chain variable region and/or the light chain variable region of an antibody selected from, for example, antibodies 1-K16, 1-K17, 6-B09, 7-B16, 13-E16, and 19-O07 disclosed herein. Additional non-limiting exemplary chimeric antibodies include chimeric antibodies comprising heavy chain CDRs 1, CDRs 2 and CDRs 3 and/or light chain CDRs 1, CDRs 2 and CDRs 3 of an antibody selected from the antibodies 1-K16, 1-K17, 6-B09, 7-B16, 13-E16 and 19-O07 as disclosed herein. In some embodiments, the chimeric anti-CCR 8 antibody comprises the variable regions described above and binds to CCR 8. In some embodiments, the chimeric anti-CCR 8 antibody comprises the variable regions described above, binds CCR8 and inhibits binding of CCR8 to CCL 1. In some embodiments, the anti-CCR 8 antibody comprises a variable region as described above, binds to CCR8 and enhances the immune response in the subject, and/or increases the activation of T cells in the subject upon administration of the antibody to the subject. In some embodiments, administration of an anti-CCR 8 antibody described herein stimulates the activity of immune cells, reduces down-regulation of immune cells, or increases T cell responses in a subject.
In some embodiments, a chimeric antibody described herein comprises one or more human constant regions. In some embodiments, the human heavy chain constant region has an isotype selected from IgA, IgG, IgD, and IgE. In some embodiments, the human light chain constant region has an isotype selected from κ and λ. In some embodiments, a chimeric antibody described herein comprises a human IgG constant region. In some embodiments, the chimeric antibodies described herein comprise a human IgG4 heavy chain constant region. In some embodiments, the chimeric antibodies described herein comprise a human IgG4 constant region and a human kappa light chain.
As noted above, whether effector function is desirable may depend on the particular therapeutic approach intended for the antibody. Thus, in some embodiments, when effector function is desired, a chimeric anti-CCR 8 antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is not required, a chimeric anti-CCR 8 antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected. In some embodiments, enhanced effector function is desired.
Exemplary humanized antibodies
In some embodiments, humanized antibodies that bind CCR8 are provided. Humanized antibodies are useful as therapeutic molecules because humanized antibodies reduce or eliminate human immune responses as compared to non-human antibodies, which may result in an immune response to an antibody therapeutic, such as a human anti-mouse antibody (HAMA) response, and reduced efficacy of the therapeutic.
In some embodiments, the chimeric antibody is a humanized antibody. Typically, non-human antibodies are humanized to reduce immunogenicity to humans, while retaining the specificity and affinity of the parent non-human antibody. Typically, a humanized antibody comprises one or more variable domains in which the CDRs (or portions thereof) are derived from a non-human antibody and the FRs (or portions thereof) are derived from a human antibody sequence. The humanized antibody optionally will also comprise at least a portion of a human constant region. In some embodiments, some FR residues in the humanized antibody are substituted with corresponding residues of a non-human antibody (e.g., the antibody from which the CDR residues are derived), e.g., to restore or improve antibody specificity or affinity.
Humanized antibodies and methods for their preparation are reviewed, for example, in Almagro and Fransson (2008) front.biosci.13:1619-1633, and further described, for example, in Riechmann et al (1988) Nature 332: 323-329; queen et al, (1989) Proc. Natl Acad. Sci. USA 86: 10029-10033; U.S. Pat. nos. 5,821,337, 7,527,791, 6,982,321, and 7,087,409; kashmiri et al, (2005) Methods 36: 25-34; padlan (1991) mol.Immunol.28:489-498 (describing "resurfacing"); dall' Acqua et al, (2005) Methods 36:43-60 (describing "FR shuffling"); and Osbourn et al, (2005) Methods 36:61-68 and Klimka et al, (2000) Br.J. cancer,83: 252-.
Human framework regions that may be used for humanization include, but are not limited to: framework regions selected using the "best fit" approach (see, e.g., Sims et al (1993) J.Immunol.151: 2296); framework regions derived from human antibody consensus sequences of a particular subset of light chain variable regions or heavy chain variable regions (see, e.g., Carter et al (1992) Proc. Natl. Acad. Sci. USA,89: 4285; and Presta et al (1993) J.Immunol,151: 2623); human mature (somatic mutation) framework regions or human germline framework regions (see, e.g., Almagro and Fransson, (2008) front. biosci.13: 1619-1633); and the framework regions derived from screening FR libraries (see, e.g., Baca et al, (1997) J.biol. chem.272:10678-10684 and Rosok et al, (1996) J.biol. chem.271: 22611-22618).
In some embodiments, the humanized anti-CCR 8 antibody comprises at least 90% of the amino acid sequence of SEQ ID NO 68 or 74,A heavy chain variable domain (VH) sequence of 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity and a light chain variable domain (VL) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:69 or 75. In some embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, and a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, but an anti-CCR 8 antibody comprising the sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:68 or 74. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:69 or 75. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). In some embodiments, the anti-CCR 8 antibody comprises: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO. 60; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 63; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO. 64; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 65. In some embodiments, the antibody has ADCC activity. In some embodiments, the ADCC activity comprises an EC50 value of less than 200ng/ml, 175ng/ml, 150ng/ml, 125ng/ml, 100ng/ml, 75ng/ml, 50ng/ml, 25ng/ml, 20ng/ml, 15ng/ml, 10ng/ml, 9ng/ml, 8ng/ml, 7ng/ml, 6ng/ml, 5ng/ml, 4ng/ml, 3ng/ml, 2ng/ml, or 1ng/ml, as measured by an ADCC reporter mechanism of action (MOA) based bioassay. In some cases In embodiments, the ADCC activity is more potent than the 7-B16 antibody. In some embodiments, the ADCC activity is at least as effective as the 7-B16 antibody. In some embodiments, the antibody is to K of human CCR8 D Equal to or lower than the 7-B16 antibody (e.g., as determined by kinetic exclusion assay (i.e., KinExA)). In some embodiments, the antibody is K on a cell of human CCR8 D Equal to or lower than the 7-B16 antibody (e.g., as determined by kinetic exclusion assay (i.e., KinExA)).
In some embodiments, the humanized anti-CCR 8 antibody comprises a VH sequence in SEQ ID NO:68 or 74, including post-translational modifications of one or both sequences, and comprises a VL sequence in SEQ ID NO:69 or 75, including post-translational modifications of one or both sequences.
In some embodiments, a humanized anti-CCR 8 antibody comprises a heavy chain variable domain (VH) sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:92 or 96 and a light chain variable domain (VL) having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% sequence identity to the amino acid sequence of SEQ ID NO:93 or 97. In some embodiments, a VH sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, and a VL sequence having at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity contains a substitution (e.g., a conservative substitution), insertion, or deletion relative to a reference sequence, but an anti-CCR 8 antibody comprising the sequence retains the ability to bind CCR 8. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:92 or 96. In some embodiments, a total of 1 to 10 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acids) have been substituted, inserted, and/or deleted in SEQ ID NO:93 or 97. In some embodiments, the substitution, insertion, or deletion occurs in a region outside of the CDRs (i.e., in the FRs). In some embodiments, the anti-CCR 8 antibody comprises: (a) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100; (b) HCDR2 comprising the amino acid sequence of SEQ ID NO. 85; (c) HCDR3 comprising the amino acid sequence of SEQ ID NO 86; (d) LCDR1 comprising the amino acid sequence of SEQ ID NO: 87; (e) LCDR2 comprising the amino acid sequence of SEQ ID NO: 88; and (f) LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
In some embodiments, the humanized anti-CCR 8 antibody comprises the VH sequence in SEQ ID NO:92 or 96, including post-translational modifications of one or both sequences, and comprises the VL sequence in SEQ ID NO:93 or 97, including post-translational modifications of one or both sequences.
Exemplary humanized anti-CCR 8 antibodies include antibodies that compete for binding to CCR8 with the antibodies or fragments thereof described herein. Thus, in some embodiments, humanized anti-CCR 8 antibodies are provided that compete for binding to CCR8 with an antibody or fragment thereof selected from the group consisting of antibodies 1-K16, 1-K17, 6-B09, 7-B16, 13-E16, and 19-O07. In some embodiments, the humanized anti-CCR 8 antibody competes for binding to CCR8 with an antibody described herein and inhibits binding of CCR8 to CCL 1. In some embodiments, the humanized anti-CCR 8 antibody competes for binding to CCR8 with an antibody described herein.
Exemplary human antibodies
In some embodiments, the anti-CCR 8 antibodies provided herein are human antibodies. Human antibodies can be produced using various techniques known in the art. Human antibodies are generally described in van Dijk and van de Winkel, (2001) curr. opin. pharmacol.5:368-374 and Lonberg, (2008) curr. opin. immunol.20: 450-459. In some embodiments, the human antibody is not a naturally occurring antibody. In some embodiments, the human antibody is a monoclonal antibody; thus, in some embodiments, each human antibody in a set can bind to the same epitope on an antigen.
Human antibodies can be made by administering an immunogen to a transgenic animal that has been modified to produce fully human antibodies or to have human variable properties in response to antigen challengeIntact antibodies of the region. Such animals typically contain all or part of a human immunoglobulin locus that replaces an endogenous immunoglobulin locus or that is present extrachromosomally or randomly integrated into the animal chromosome. In such transgenic mice, the endogenous immunoglobulin loci have typically been inactivated. For an overview of the methods for obtaining human antibodies from transgenic animals, see Lonberg, (2005) nat. Biotech.23: 1117-1125. See also, e.g., the description XENOMOUSE TM U.S. Pat. nos. 6,075,181 and 6,150,584 to the art; description of the preferred embodimentU.S. patent numbers 5,770,429 for technology; description of K-MU.S. Pat. No. 7,041,870 and description of the technologyU.S. patent application publication No. US 2007/0061900 to the art. The human variable regions from intact antibodies produced by such animals may be further modified, for example by combination with different human constant regions.
Human antibodies can also be made by hybridoma-based methods. Human myeloma and mouse-human heteromyeloma cell lines have been described for the production of human monoclonal antibodies. (see, e.g., Kozbor (1984) J.Immunol,133: 3001; Brodeur et al, Monoclonal Antibody Production Techniques and Applications, pp.51-63 (Marcel Dekker, Inc., New York, 1987); and Boerner et al (1991) J.Immunol, 147: 86). Human antibodies produced by human B-cell hybridoma technology are also described in Li et al, (2006) proc.natl.acad.sci.usa,103: 3557-3562. Additional methods include, for example, those described in U.S. Pat. No. 7,189,826 (describing the production of human IgM monoclonal antibodies from hybridoma cell lines) and Ni, (2006) Xiaoandai Mianyixue,26(4):265-268 (describing human-human hybridomas). The human hybridoma technique (Trioma (Trioma) technique) is also described in Vollmers and Brandlein, (2005) history and Histopathology,20(3): 927-.
Human antibodies can also be produced by isolating Fv clone variable domain sequences selected from phage display libraries of human origin. Such variable domain sequences can then be combined with the desired human constant domains. Techniques for selecting human antibodies from antibody libraries are described below.
Antibodies can be isolated by screening combinatorial libraries for antibodies having one or more desired activities. For example, various methods are known in the art for generating phage display libraries and screening such libraries for antibodies having desired binding characteristics. Such Methods are reviewed, for example, in Hoogenboom et al, in Methods in Molecular Biology 178:1-37(O' Brien et al, eds., Human Press, Totowa, NJ,2001), and are further described, for example, in McCafferty et al, (1990) Nature 348: 552-; clackson et al, (1991) Nature 352: 624-; marks et al (1992) J.mol.biol 222: 581-597; marks and Bradbury in Methods in Molecular Biology 248:161-175(Lo eds., Human Press, Totowa, NJ, 2003); sidhu et al, (2004) J.mol.biol.338(2): 299-; lee et al, (2004) J.mol.biol.340(5): 1073-1093; fellouse, (2004) Proc.Natl.Acad.Sci.USA101(34): 12467-12472; and Lee et al (2004) J.Immunol.Methods284(1-2):119-132 and PCT publication WO 99/10494.
In certain phage display methods, V H And V L Genomic libraries were individually cloned by Polymerase Chain Reaction (PCR) and randomly recombined in phage libraries, which can then be screened for antigen-binding phage as described by Winter et al, (1994) Ann.Rev.Immunol.,12: 433-455. Phage typically display antibody fragments, either as single chain fv (scfv) fragments or as Fab fragments. Libraries from immune sources provide high affinity antibodies to the immunogen without the need to construct hybridomas. Alternatively, the primary repertoire can be cloned (e.g., from humans) to provide a single source of antibodies against a wide range of non-self and self-antigens without any immunization, as described by Griffiths et al, (1993) EMBO J12: 725-. Finally, it is also possible to clone unrearranged V gene segments from stem cells andnatural libraries were prepared synthetically using PCR primers containing random sequences to encode the highly variable CDR3 regions and to effect in vitro rearrangement, as described by Hoogenboom and Winter (1992), J.mol.biol., 227: 381-388. Patent publications describing human antibody phage libraries include, for example: U.S. Pat. No. 5,750,373 and U.S. patent publication nos. 2005/0079574, 2005/0119455, 2005/0266000, 2007/0117126, 2007/0160598, 2007/0237764, 2007/0292936 and 2009/0002360.
In some embodiments, the human anti-CCR 8 antibody binds to CCR8 and inhibits the binding of CCR8 to CCL 1.
Exemplary human anti-CCR 8 antibodies also include antibodies that compete for binding to CCR8 with the human antibodies or fragments thereof described herein. Thus, in some embodiments, there is provided a human anti-CCR 8 antibody that competes for binding to CCR8 with an antibody or fragment thereof selected from the group consisting of antibodies 1-K16, 1-K17, 6-B09, 7-B16, 13-E16, and 19-O07. In some embodiments, a human anti-CCR 8 antibody competes for binding to CCR8 with the antibodies described herein and inhibits binding of CCR8 to CCL 1.
In some embodiments, chimeric human anti-CCR 8 antibodies are provided, wherein the antibodies comprise variable regions from a human antibody that binds CCR8 and constant regions from a different human antibody. In some embodiments, chimeric human anti-CCR 8 antibodies are provided, wherein the antibodies comprise CDRs from a human antibody that binds CCR8 and framework regions from a different human antibody. In some embodiments, the antibody is not a naturally occurring human antibody.
In some embodiments, a human anti-CCR 8 antibody comprises one or more human constant regions. In some embodiments, the human heavy chain constant region has an isotype selected from IgA, IgG, IgD, and IgE. In some embodiments, the human light chain constant region has an isotype selected from κ and λ. In some embodiments, the human antibodies described herein comprise a human IgG constant region. In some embodiments, the human antibodies described herein comprise a human IgG4 heavy chain constant region. In some embodiments, the human antibodies described herein comprise a human IgG4 constant region and a human kappa light chain.
In some embodiments, when effector function is desired, a human anti-CCR 8 antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is not required, a human anti-CCR 8 antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected.
As used herein, the term "human antibody" refers to the genus of possible sequences of an antibody construct, rather than the source of the antibody.
Fusion proteins
In some embodiments, a fusion protein described herein comprises CCL1 or an active fragment of CCL 1. As used herein, an "active fragment" of CCL1 refers to a CCL1 fragment that binds CCR 8. One of ordinary skill in the art can identify CCL1 active fragments that bind CCR 8. In some embodiments, an active fragment of CCL1 binds CCR8 with an affinity that is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% as strong as the affinity of native CCL1 for CCR 8. For the avoidance of doubt, at least 50% of 1nM affinities are strong affinities of 2nM or less. One of ordinary skill in the art can determine the affinity of an active fragment of CCL1 for CCR8 and compare it to the affinity of native CCL1 for CCR 8. In some embodiments, the fusion protein described herein comprises SEQ ID NO. 2 or amino acids 24-96 of SEQ ID NO. 2.
In some embodiments, a fusion protein comprising CCL1, or an active fragment thereof, comprises an Fc region, such as an Fc region provided herein. In some embodiments, the Fc region comprises at least one modification that enhances cell killing. In some such embodiments, the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC). Non-limiting exemplary modifications that enhance cell killing are provided herein and include, for example, afucosylation.
In some embodiments, a fusion protein described herein comprises MC148 or an active fragment of MC 148. As used herein, an "active fragment" of MC148 refers to a fragment of MC148 that binds CCR 8. One of ordinary skill in the art can identify MC148 active fragments that bind CCR 8. In some embodiments, an active fragment of MC148 binds CCR8 with an affinity that is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95% stronger than the affinity of native MC148 for CCR 8. For the avoidance of doubt, at least 50% of 1nM affinities are strong affinities of 2nM or less. One of ordinary skill in the art can determine the affinity of an active fragment of MC148 for CCR8 and compare it to the affinity of native MC148 for CCR 8. In some embodiments, the fusion protein described herein comprises SEQ ID NO. 2 or amino acids 24-96 of SEQ ID NO. 2.
In some embodiments, a fusion protein comprising MC148 or an active fragment thereof comprises an Fc region, such as an Fc region provided herein. In some embodiments, the Fc region comprises at least one modification that enhances cell killing. In some such embodiments, the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC). Non-limiting exemplary modifications that enhance cell killing are provided herein and include, for example, afucosylation.
Exemplary antibody constant regions and Fc regions
In some embodiments, an antibody described herein comprises one or more human constant regions. In some embodiments, the human heavy chain constant region has an isotype selected from IgA, IgG, IgD, and IgE. In some embodiments, the antibodies described herein comprise a human IgG constant region. In some embodiments, when effector function is desired, an anti-CCR 8 antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, an anti-CCR 8 antibody comprising a human IgG4 or IgG2 heavy chain constant region is selected when effector function is not required. In some embodiments, the human light chain constant region has an isotype selected from κ and λ. In some embodiments, the antibodies described herein comprise a human IgG1 heavy chain constant region. In some embodiments, the antibodies described herein comprise a human IgG1 constant region and a human kappa light chain.
In some embodiments, a fusion protein described herein comprises one or more human Fc regions. In some embodiments, the Fc region has an isotype selected from IgA, IgG, IgD, and IgE. In some embodiments, a fusion protein described herein comprises a human Fc region. In some embodiments, when effector function is desired, a fusion protein comprising a human IgG1Fc region or a human IgG3Fc region is selected. In some embodiments, when effector function is not required, a fusion protein comprising a human IgG4 or IgG2Fc region is selected.
Throughout this specification and claims, unless explicitly stated or known to those skilled in the art, the numbering of residues in an immunoglobulin heavy chain is that of the EU index as in Kabat et al, Sequences of Proteins of Immunological Interest, published Health Service 5 th edition, National Institutes of Health, Bethesda, Md. (1991), which is expressly incorporated herein by reference. The "EU index in Kabat" refers to the residue numbering of the human IgG1EU antibody.
As noted above, whether effector function is desirable may depend on the particular therapeutic approach intended for the antibody. Thus, in some embodiments, when effector function is desired, an anti-CCR 8 antibody comprising a human IgG1 heavy chain constant region or a human IgG3 heavy chain constant region is selected. In some embodiments, when effector function is desired, a fusion protein comprising a human IgG1Fc region or a human IgG3Fc region is selected.
In some embodiments, the antibody or fusion protein comprises a variant Fc region having at least one amino acid substitution compared to the Fc region of a wild-type IgG Fc region. In some embodiments, the variant Fc region has two or more amino acid substitutions as compared to a wild-type Fc region. In some embodiments, the variant Fc region has three or more amino acid substitutions as compared to a wild-type Fc region. In some embodiments, the variant Fc region has at least one, two, or three or more amino acid substitutions in the Fc region described herein. In some embodiments, the variant Fc regions herein will have at least about 80% homology with a native sequence Fc region and/or with an Fc region of a parent polypeptide. In some embodiments, the variant Fc regions herein will have at least about 90% homology to a native sequence Fc region and/or to an Fc region of a parent polypeptide. In some embodiments, the variant Fc regions herein will have at least about 95% homology with a native sequence Fc region and/or with an Fc region of a parent polypeptide. In some embodiments, the heavy chain constant region or Fc region lacks the C-terminal lysine (K) residue. In some such embodiments, the heavy chain constant region or Fc region may be referred to as "desK". In some embodiments, the heavy chain constant region or Fc region lacking the C-terminal lysine is an IgG, such as IgG1, IgG2, IgG3, or IgG 4.
In some embodiments, the antibodies or fusion proteins provided herein are altered to increase or decrease the extent to which the antibody is glycosylated. Addition or deletion of antibody glycosylation sites can be conveniently accomplished by altering the amino acid sequence so as to create or remove one or more glycosylation sites.
The carbohydrate attached to the Fc region may be altered. Natural antibodies produced by mammalian cells typically comprise branched bi-antennary oligosaccharides, which are typically linked by Asn 297N-linked to the CH2 domain of the Fc region. See, e.g., Wright et al TIBTECH 15:26-32 (1997). Oligosaccharides may include various carbohydrates such as mannose, N-acetylglucosamine (GlcNAc), galactose, and sialic acid, as well as fucose attached to GlcNAc in the "stem" of the biantennary oligosaccharide structure. In some embodiments, the oligosaccharides in an antibody or fusion protein may be modified in order to produce antibody variants with certain improved properties.
In some embodiments, antibody or fusion protein variants are provided that have a carbohydrate structure that lacks (directly or indirectly) fucose attached to an Fc region (i.e., afucosylated). For example, the amount of fucose in such variants may be 1% to 80%, 1% to 65%, 5% to 65%, or 20% to 40%. The amount of fucose is determined by calculating the average amount of fucose in the sugar chain at Asn297 relative to the sum of all sugar structures (e.g. complex, hybrid and high mannose structures) attached to Asn297, as measured by MALDI-TOF mass spectrometry, as described e.g. in WO 2008/077546. Asn297 refers to the asparagine residue at about position 297 in the Fc region (EU numbering of Fc region residues); however, due to minor sequence variations in the antibody, Asn297 can also be located about ± 3 amino acids upstream or downstream of position 297, i.e., between positions 294 and 300. Such fucosylated variants may have improved ADCC function. See, e.g., U.S. patent publication No. US 2003/0157108(Presta, L.); US 2004/0093621(Kyowa Hakko Kogyo co., Ltd). Examples of publications related to "defucosylated" or "fucose-deficient" antibody variants include: US 2003/0157108; WO 2000/61739; WO 2001/29246; US 2003/0115614; US 2002/0164328; US 2004/0093621; US 2004/0132140; US 2004/0110704; US 2004/0110282; US 2004/0109865; WO 2003/085119; WO 2003/084570; WO 2005/035586; WO 2005/035778; WO 2005/053742; WO 2002/031140; okazaki et al, J.mol.biol.336:1239-1249 (2004); Yamane-Ohnuki et al, Biotech.Bioeng.87:614 (2004). Examples of cell lines capable of producing defucosylated antibodies include Lec13CHO cells lacking protein fucosylation (Ripka et al, Arch. biochem. Biophys.249: 533. 545 (1986); U.S. patent application No. US 2003/0157108A 1, Presta, L; and WO 2004/056312A 1, Adams et al, particularly in example 11), and knock-out cell lines (such as alpha-1, 6-fucosyltransferase gene FUT8 knock-out CHO cells) (see, e.g., Yamane-Ohnuki et al, Biotech. Bioeng.87:614 (2004); Kanda, Y. et al, Biotechnol. Bioeng.,94(4): 680. 688 (2006); and WO 2003/085107).
Antibodies and fusion protein variants also have bisected oligosaccharides, for example where the biantennary oligosaccharide attached to the Fc region of the antibody is bisected by GlcNAc. Such antibody or fusion protein variants may have reduced fucosylation and/or improved ADCC function. Examples of such variants are described, for example, in WO 2003/011878(Jean-Mairet et al), U.S. Pat. No. 6,602,684(Umana et al), and U.S. Pat. No. 2005/0123546(Umana et al). Also provided are variants having at least one galactose residue in an oligosaccharide linked to an Fc region. Such variants may have improved CDC function. Such variants are described, for example, in WO 1997/30087(Patel et al), WO 1998/58964(Raju, S.) and WO 1999/22764(Raju, S.).
The antibody or variant Fc region has an Fc mutation that increases ADCC activity. In some embodiments, the antibody or Fc region variant comprises one or more mutations that enhance Fc γ RIIIa binding and/or reduce Fc γ RIIIb binding. Non-limiting exemplary such mutations may be made at one or more amino acid positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334 and P396. Non-limiting exemplary mutations include L234Y, L235Q, G236W, S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L. In some embodiments, the antibody or Fc region variant comprises the mutations F243L/R292P/Y300L/V305I/P396L. See, e.g., Stavenhagen et al, 2007, Cancer Res.67: 8882-. In some embodiments, the antibody or Fc region variant comprises the mutations S239D/I332E or S239D/I332E/a 330L. See, e.g., Lazar et al, 2006, PNAS USA,103: 4005-. In some embodiments, the antibody or Fc region variant comprises the mutations S298A/E333A/K334A. See, e.g., shiplds et al, 2001, j.biol.chem.,276: 6591-. In some embodiments, the antibody or Fc region variant comprises the mutations L234Y/L235Q/G236W/S239M/H268D/D270E/S298A or the mutations D270E/K326D/a330M/K334E, or one heavy chain constant region or Fc comprises the mutations L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and the other heavy chain constant region or Fc comprises the mutations D270E/K326D/a 330M/K334E. See, e.g., Mimoto et al, 2013, MAbs,5: 229-.
Antibodies and Fc region variants also have an amino-terminal leader extension. For example, one or more amino acid residues of the amino-terminal leader sequence are present at the amino terminus of any one or more heavy or light chains of the antibody. An exemplary amino-terminal leader extension comprises or consists of the three amino acid residues VHS present on one or both light chains of an antibody variant.
The in vivo or serum half-life of a human FcRn high affinity binding polypeptide can be determined, for example, in a transgenic mouse, human or non-human primate to which is administered a polypeptide having a variant Fc region. See also, e.g., Petkova et al International Immunology 18(12): 1759-.
In some embodiments, the antibody or Fc region variant mediates ADCC in the presence of human effector cells more effectively than the parent antibody. In some embodiments, the antibody or variant Fc region is substantially more effective in mediating ADCC in vitro when the amounts of polypeptide variant and parent antibody or Fc region used in the assay are substantially the same. In some embodiments, the antibody or variant Fc region is substantially more effective in mediating ADCC in vivo when the amounts of polypeptide variant and parent antibody or Fc region used in the assay are substantially the same. Typically, such variants will be identified using an in vitro ADCC assay as disclosed herein, although other assays or methods for determining ADCC activity, e.g. in animal models and the like, are also contemplated.
Exemplary conjugates
In some embodiments, an antibody or fusion protein provided herein is conjugated to another molecule. In some embodiments, the additional molecule may be a detectable marker, such as a label. In some embodiments, the additional molecule may be a therapeutic molecule, such as a cytotoxic agent. In some embodiments, the label and/or cytotoxic agent may be conjugated to an antibody or fusion protein provided herein. As used herein, a label is a moiety that facilitates detection of an antibody or fusion protein and/or facilitates detection of a molecule to which the antibody or fusion protein binds. Non-limiting exemplary labels include, but are not limited to, radioisotopes, fluorescent groups, enzyme groups, chemiluminescent groups, biotin, epitope tags, metal binding tags, and the like. One skilled in the art can select the appropriate label depending on the particular application.
As used herein, a cytotoxic agent is a moiety that reduces the proliferative capacity of one or more cells. A cell has a reduced proliferative capacity when the cell becomes less capable of proliferation, e.g., because the cell undergoes apoptosis or otherwise dies, the cell fails to pass through the cell cycle and/or fails to divide, the cell differentiates, etc. Non-limiting exemplary cytotoxic agents include, but are not limited to, radioisotopes, toxins, and chemotherapeutic agents. One skilled in the art can select an appropriate cytotoxic agent depending on the intended application. In some embodiments, the cytotoxic agent is at least one of an antimetabolite, an alkylating agent, an antibiotic, a growth factor, a cytokine, an antiangiogenic agent, an antimitotic agent, an anthracycline, a toxin, or an apoptotic agent.
In some embodiments, the label and/or cytotoxic agent is conjugated to an antibody or fusion protein provided herein using in vitro chemistry methods. Non-limiting exemplary chemical conjugation methods are known in the art and include services, methods and/or reagents commercially available from the following suppliers: such as Thermo Scientific Life Science Research products (formerly Pierce; Rockford, Ill.), Prozyme (Hayward, Calif.), SACRI Antibody Services (Calgary, Canada), AbD Serotec (Raleigh, N.C.), etc. In some embodiments, when the marker and/or cytotoxic agent is a polypeptide, the marker and/or cytotoxic agent may be expressed from the same expression vector with at least one antibody chain or fusion protein to produce a polypeptide comprising the marker and/or cytotoxic agent fused to the antibody or fusion protein. One skilled in the art can select an appropriate method of conjugating the label and/or cytotoxic agent to the antibody or fusion protein depending on the intended application.
In some embodiments, conjugation may be covalent. In some embodiments, conjugation may be non-covalent. In some embodiments, conjugation may be via a specific binding interaction, for example by binding a second antibody.
Exemplary embodiments of conjugates comprising a drug (or drug derivative) and an antibody or fusion protein as disclosed herein may have general formula 1:
mab- [ linker ] -drug (formula 1)
Wherein the linker is a cleavable or non-cleavable linker, the Mab is an antibody or fusion protein disclosed herein, and the drug is any drug or cytotoxic agent.
In some embodiments, the conjugate may comprise general formula 2:
MAb-[L2]-[L1]-[AA] m -[A′]-medicine (formula 2)
Wherein the MAb is an antibody or fusion protein disclosed herein; l2 is a component of a cross-linker comprising an antibody-coupling moiety and one or more acetylene (or azide) groups; l1 contains a defined PEG having an azide (or acetylene) complementary to the acetylene (or azide) moiety in L2 at one terminus and a reactive group such as a carboxylic acid or hydroxyl group at the other terminus; AA is an L-amino acid; m is an integer having a value of 0, 1, 2, 3 or 4; and a' is an additional spacer selected from the group consisting of: ethanolamine, 4-hydroxybenzyl alcohol, 4-aminobenzyl alcohol or substituted or unsubstituted ethylenediamine. The L amino acid of 'AA' is selected from alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine. If the A' group contains a hydroxyl group, it is attached to the hydroxyl group or amino group of the drug in the form of a carbonate or carbamate, respectively.
In some embodiments of formula 2, A' is a substituted ethanolamine derived from an L-amino acid, wherein the carboxylic acid group of the amino acid is replaced with a hydroxymethyl moiety. A' may be derived from any of the following L-amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine, and valine.
In an example of such a conjugate of the embodiment of formula 2, m is 0, A' is L-valinol, and the drug is exemplified by SN-38. The resulting structure is shown in formula 3.
In another example of the conjugate of this embodiment of formula 2, m is 1 and is represented by derivatized L-lysine, A' is L-valinol, and the drug is exemplified by SN-38. The structure is shown in formula 4.
In this embodiment, an amide bond is first formed between a carboxylic acid of an amino acid such as lysine and an amino group of valinol using an orthogonal protecting group for the lysine amino group. The protecting group on the N-terminus of lysine was removed, leaving the protecting group on the lysine side chain intact, and the N-terminus was coupled to the carboxyl group on the defined PEG with azide (or acetylene) at the other end. The hydroxyl group of valinol is then attached to the 20-chloroformate derivative of 10-hydroxy-protected SN-38 and this intermediate is coupled to an L2 component carrying a targeting carrier binding moiety and a complementary acetylene (or azide) group participating in click cycloaddition chemistry. Finally, removal of the protecting groups on both the lysine side chain and SN-38 gives the product of this example, as shown in formula 3.
While not wishing to be bound by theory, the small MW SN-38 product produced after intracellular proteolysis (i.e., valinol-SN-38 carbonate) has an additional pathway to release intact SN-38 through intramolecular cyclization involving the amino group of valinol and the carbonyl group of the carbonate.
In another embodiment, A' of formula 2 is A-OH, wherein A-OH is a foldable moiety, such as 4-aminobenzyl alcohol or substituted with C at the benzyl position 1 -C 10 Substituted 4-aminobenzyl alcohol substituted with an alkyl group and the latter is linked via its amino group to an L-amino acid or a polypeptide comprising up to four L-amino acid moieties; wherein the N-terminus is linked to a cross-linking agent that terminates in a binding group for the targeting moiety.
An example of an embodiment is given below, wherein the a-OH embodiment of a ' of formula (2) is derived from a substituted 4-aminobenzyl alcohol and ' AA ' consists of a single L-amino acid with m ═ 1 in formula (2), and the drug is exemplified by SN-38. This structure is represented by (formula 5, referred to as MAb-CLX-SN-38). The single amino acid of AA is selected from any one of the following L-amino acids: alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tryptophan, tyrosine and valine. The substituent R on the 4-aminobenzyl alcohol moiety (A' in the A-OH embodiment) is hydrogen or an alkyl group selected from C1-C10 alkyl groups.
An embodiment of MAb-CLX-SN-38 of formula 5, wherein the single amino acid AA is L-lysine and R ═ H, and the drug is exemplified by SN-38 (formula 6; referred to as MAb-CL 2A-SN-38).
Other embodiments are possible in the context of 10-hydroxy-containing camptothecins such as SN-38. In the example of SN-38 as a drug, the more reactive 10-hydroxy group of the drug is derivatized while the 20-hydroxy group is unaffected. In formula 2, a' is a substituted ethylenediamine. An example of this embodiment is represented by the following formula '7', where the phenolic hydroxyl group of SN-38 is derivatized with a substituted ethylenediamine to a carbamate, the other amine of the diamine is derivatized with 4-aminobenzyl alcohol to a carbamate, and the amino group of the latter is attached to a Phe-Lys dipeptide. In this structure (formula 7), R and R' are independently hydrogen or methyl. When R ═ R' ═ methyl, it is known as MAb-CL17-SN-38 or MAb-CL 2E-SN-38.
In some embodiments, the AA comprises a polypeptide moiety that is cleavable by an intracellular peptidase, such as a dipeptide, tripeptide, or tetrapeptide. Examples are: Ala-Leu, Leu-Ala-Leu and Ala-Leu-Ala-Leu (SEQ ID NO: 129; Trouet et al, 1982).
In some embodiments, the L1 component of the conjugate contains a defined polyethylene glycol (PEG) spacer having 1-30 repeating monomer units. In another embodiment, the PEG is a defined PEG having 1-12 repeating monomer units. The introduction of PEG may involve the use of commercially available heterobifunctional PEG derivatives. The heterobifunctional PEG may contain an azide or acetylene group. An example of a heterobifunctional defined PEG containing 8 repeating monomer units (where "NHS" is succinimide) is given in formula 8 below:
In some embodiments, L2 has a plurality of acetylene (or azide) groups and a single targeting vector binding moiety in the range of 2 to 40, but preferably 2 to 20 and more preferably 2 to 5.
Representative SN-38 conjugates of antibodies containing multiple drug molecules and a single targeting vector binding moiety are shown below. The 'L2' component of the structure is attached to 2 acetylene groups, resulting in the attachment of two azide-attached SN-38 molecules. The linkage to the MAb is denoted as succinimide.
Wherein the R residues are:
in some embodiments, when the bifunctional drug contains a thiol-reactive moiety as the antibody binding group, a thiolating reagent is used to generate a thiol on the antibody on a lysine group of the antibody. Methods for introducing thiol groups onto antibodies by modifying lysine groups of MAbs are well known in the art (Wong, Chemistry of protein conjugation and cross-linking, CRC Press, Inc., Boca Raton, Fla. (1991), pages 20-22). Alternatively, mild reduction of the interchain disulfide bond on an antibody using a reducing agent such as Dithiothreitol (DTT) (Willner et al, Bioconjugate chem. 4: 521-527(1993)) can produce 7 to 10 thiols on the antibody; this has the advantage of incorporating multiple drug moieties in the inter-chain regions of the MAb remote from the antigen binding region.
In some embodiments, the chemotherapeutic moiety is selected from the group consisting of: doxorubicin (DOX), epirubicin, morpholinodoxorubicin (morpholino-DOX), cyanomorpholino-doxorubicin (cyanomorpholino-DOX), 2-pyrrolinyl-doxorubicin (2-PDOX), CPT, 10-hydroxycamptothecin, SN-38, topotecan (topotecan), lurtotecan (lurtotcan), 9-aminocamptothecin, 9-nitrocamptothecin, taxane, geldanamycin, ansamycin, and epothilone. In another embodiment, the chemotherapeutic moiety is SN-38. Preferably, in the conjugate of some embodiments, the targeting moiety is conjugated to at least one chemotherapeutic moiety; preferably 1 to about 12 chemotherapeutic moieties; most preferably from about 6 to about 12 chemotherapeutic moieties are attached.
Furthermore, in some embodiments, linker component 'L2' comprises a thiol group that reacts with a thiol-reactive residue introduced at one or more lysine side chain amino groups of the targeting moiety. In such cases, the antibody may be pre-derivatized with a thiol-reactive group such as maleimide, vinyl sulfone, bromoacetamide, or iodoacetamide by methods well described in the art.
Exemplary preamble sequences
In order for some secreted proteins to be expressed and secreted in large quantities, a leader sequence from a heterologous protein may be required. In some embodiments, it may be advantageous to use a heterologous leader sequence, as the resulting mature polypeptide may remain unchanged when the leader sequence is removed in the ER during the secretion process. The addition of heterologous leader sequences can be used for expression and secretion of some proteins.
The sequences of certain exemplary Leader sequences are described, for example, in an online Leader Sequence Database (Leader Sequence Database) maintained by the National University of Biochemistry, National University of Singapore. See Choo et al, BMC Bioinformatics,6:249 (2005); and PCT publication No. WO 2006/081430.
Polypeptide expression and production
Nucleic acid molecules encoding antibodies or fusion proteins
Provided herein are nucleic acid molecules comprising polynucleotides encoding one or more chains of an anti-CCR 8 antibody. In some embodiments, the nucleic acid molecule comprises a polynucleotide encoding a heavy chain or a light chain of an anti-CCR 8 antibody. In some embodiments, the nucleic acid molecule comprises a polynucleotide encoding a heavy chain and a polynucleotide encoding a light chain of an anti-CCR 8 antibody. In some embodiments, the first nucleic acid molecule comprises a first polynucleotide encoding a heavy chain and the second nucleic acid molecule comprises a second polynucleotide encoding a light chain.
In some embodiments, the heavy and light chains are expressed from one nucleic acid molecule or from two separate nucleic acid molecules as two separate polypeptides. In some embodiments, such as when the antibody is an scFv, a single polynucleotide encodes a single polypeptide comprising both a heavy chain and a light chain linked together.
In some embodiments, the polynucleotide encoding the heavy or light chain of an anti-CCR 8 antibody comprises a nucleotide sequence encoding at least one CDR provided herein. In some embodiments, the polynucleotide encoding the heavy or light chain of an anti-CCR 8 antibody comprises a nucleotide sequence encoding at least 3 CDRs provided herein. In some embodiments, the polynucleotide encoding the heavy or light chain of an anti-CCR 8 antibody comprises a nucleotide sequence encoding at least 6 CDRs provided herein. In some embodiments, the polynucleotide encoding the heavy or light chain of the anti-CCR 8 antibody comprises a nucleotide sequence encoding a leader sequence that is translationally N-terminal to the heavy or light chain. As discussed above, the leader sequence may be the native heavy or light chain leader sequence, or may be another heterologous leader sequence.
Provided herein are nucleic acid molecules comprising polynucleotides encoding fusion proteins. In some embodiments, the polynucleotide encoding the fusion protein comprises a nucleotide sequence encoding a leader sequence that is positioned N-terminally of the fusion protein upon translation. The leader sequence may be the native leader sequence or may be another heterologous leader sequence.
In some embodiments, the nucleic acid is a nucleic acid encoding any of the amino acid sequences of the antibodies and fusion proteins in the sequence listing herein. In some embodiments, the nucleic acid is a nucleic acid that is at least 80% identical (e.g., at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical) to a nucleic acid encoding any of the amino acid sequences of the antibodies and fusion proteins in the sequence listing herein. In some embodiments, a nucleic acid is a nucleic acid that hybridizes to any one or more of the nucleic acid sequences provided herein. In some embodiments, hybridization is performed under mild conditions. In some embodiments, hybridization is performed under highly stringent conditions, such as: at least about 6 XSSC and 1% SDS at 65 ℃, a first wash with about 20% (v/v) formamide in 0.1 XSSC at about 42 ℃ for 10 minutes, followed by a wash with 0.2 XSSC and 0.1% SDS at 65 ℃.
Nucleic acid molecules can be constructed using recombinant DNA techniques conventional in the art. In some embodiments, the nucleic acid molecule is an expression vector suitable for expression in a selected host cell.
Vectors comprising polynucleotides encoding anti-CCR 8 heavy chain and/or anti-CCR 8 light chain are provided. Vectors comprising polynucleotides encoding the heavy chain of anti-CCR 8 and/or the light chain of anti-CCR 8 are also provided. Such vectors include, but are not limited to, DNA vectors, phage vectors, viral vectors, retroviral vectors, and the like. In some embodiments, the vector comprises a first polynucleotide sequence encoding a heavy chain and a second polynucleotide sequence encoding a light chain. In some embodiments, the heavy and light chains are expressed from the vector as two separate polypeptides. In some embodiments, the heavy and light chains are expressed as part of a single polypeptide, such as when the antibody is a scFv.
In some embodiments, the first vector comprises a polynucleotide encoding a heavy chain and the second vector comprises a polynucleotide encoding a light chain. In some embodiments, the first vector and the second vector are transfected into the host cell in similar amounts (such as similar molar amounts or similar mass). In some embodiments, the first vector and the second vector are transfected into the host cell at a molar or mass ratio of between 5:1 and 1: 5. In some embodiments, a vector encoding a heavy chain and a vector encoding a light chain are used in a mass ratio of between 1:1 and 1: 5. In some embodiments, a 1:2 mass ratio of vector encoding the heavy chain and vector encoding the light chain is used.
Vectors comprising polynucleotides encoding fusion proteins are provided. Such vectors include, but are not limited to, DNA vectors, phage vectors, viral vectors, retroviral vectors, and the like.
In some embodiments, a vector optimized for expression of the polypeptide in CHO or CHO-derived cells or in NSO cells is selected. Exemplary such antibodies are described, for example, in Running Deer et al, Biotechnol. Prog.20:880-889 (2004).
Host cell
In some embodiments, the antibodies or fusion proteins provided herein can be expressed in prokaryotic cells, such as bacterial cells; or in eukaryotic cells such as fungal cells (such as yeast), plant cells, insect cells and mammalian cells. Such expression can be performed, for example, according to methods known in the art. Exemplary eukaryotic cells that may be used to express the polypeptide include, but are not limited to, COS cells, including COS 7 cells; 293 cells, including 293-6E cells; CHO cells, including CHO-S, DG 44; lec13 CHO cells and FUT8CHO cells; Cells (Crucell); and NSO cells. In some embodiments, the antibodies or fusion proteins provided herein can be expressed in yeast. See, e.g., U.S. publication No. US 2006/0270045 a 1. In some embodiments, a particular eukaryotic host cell is selected based on its ability to perform the desired post-translational modifications to the antibodies or fusion proteins provided herein. For example, in some embodiments, a polypeptide produced by a CHO cell has a higher level of sialylation than the same polypeptide produced in a 293 cell.
Introduction of one or more nucleic acids into a desired host cell can be accomplished by any method, including but not limited to calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid mediated transfection, electroporation, transduction, infection, and the like. Non-limiting exemplary methods are described, for example, in Sambrook et al, Molecular Cloning, A Laboratory Manual, 3 rd edition, Cold Spring Harbor Laboratory Press (2001). The nucleic acid may be transiently or stably transfected into the desired host cell according to any suitable method.
Also provided are host cells comprising any of the polynucleotides or vectors described herein. In some embodiments, host cells comprising the antibodies or fusion proteins are provided. Any host cell capable of overexpressing heterologous DNA can be used to isolate the gene encoding the antibody, polypeptide or protein of interest. Non-limiting examples of mammalian host cells include, but are not limited to, COS, HeLa, and CHO cells. See also PCT publication No. WO 87/04462. Suitable non-mammalian host cells include prokaryotes such as E.coli (E.coli) or Bacillus subtilis (B.subtilis) and yeasts such as Saccharomyces cerevisiae (S.cerevisae), Schizosaccharomyces pombe (S.pombe); or Kluyveromyces lactis (K.lactis).
Purification of polypeptides
The antibodies and fusion proteins provided herein can be purified by any suitable method. Such methods include, but are not limited to, the use of affinity matrices or hydrophobic interaction chromatography. Suitable affinity ligands include ROR1ECD and ligands that bind to the constant region of an antibody. For example, protein a, protein G, protein a/G or antibody affinity columns can be used to bind a constant region or Fc region and purify the antibody or fusion protein. Hydrophobic interaction chromatography (e.g., butyl or phenyl columns) may also be suitable for purifying some polypeptides. Ion exchange chromatography (e.g., anion exchange chromatography and/or cation exchange chromatography) may also be suitable for purifying some polypeptides. Mixed mode chromatography (e.g., reverse phase/anion exchange, reverse phase/cation exchange, hydrophilic interaction/anion exchange, hydrophilic interaction/cation exchange, etc.) may also be suitable for purifying some polypeptides. Many methods of purifying polypeptides are known in the art.
Cell-free production of polypeptides
In some embodiments, the antibody or fusion protein is produced in a cell-free system. Non-limiting exemplary cell-free systems are described, for example, in Sitaraman et al, Methods mol.biol.498:229-44 (2009); spirin, Trends Biotechnol.22:538-45 (2004); endo et al, Biotechnol. adv.21: 695-.
Composition comprising a metal oxide and a metal oxide
In some embodiments, an antibody or fusion protein prepared by the method described above is provided. In some embodiments, the antibody or fusion protein is produced in a host cell. In some embodiments, the antibody or fusion protein is produced in a cell-free system. In some embodiments, the antibody or fusion protein is purified. In some embodiments, a cell culture medium comprising an antibody or fusion protein is provided. In some embodiments, a host cell culture fluid comprising an antibody or fusion protein is provided.
In some embodiments, compositions comprising an antibody or fusion protein prepared by the methods described above are provided. In some embodiments, the composition comprises an antibody or fusion protein produced in a host cell. In some embodiments, the composition comprises an antibody or fusion protein produced in a cell-free system. In some embodiments, the composition comprises a purified antibody or fusion protein.
In some embodiments, compositions are provided comprising an antibody or fusion protein at a concentration greater than about any one of: 10mg/mL, 20mg/mL, 30mg/mL, 40mg/mL, 50mg/mL, 60mg/mL, 70mg/mL, 80mg/mL, 90mg/mL, 100mg/mL, 125mg/mL, 150mg/mL, 175mg/mL, 200mg/mL, 225mg/mL, or 250 mg/mL.
Therapeutic compositions and methods
Methods of treating diseases using antibodies or fusion proteins
Antibodies (e.g., 7-B16, 1-K17, etc.) and fusion proteins and compositions comprising the antibodies and fusion proteins are provided for use in methods of treatment of humans or animals. Pharmaceutical and therapeutic uses of the disclosed antibodies and fusion proteins are also provided herein. Also provided are methods and pharmaceutical/therapeutic uses for treating diseases, comprising administering the antibodies and fusion proteins provided herein. Non-limiting exemplary diseases that can be treated with the antibodies and fusion proteins provided herein include, but are not limited to, cancer.
In some embodiments, a method of treating cancer (or therapeutic/pharmaceutical use of the disclosed antibodies or fusion proteins) comprises administering to a subject having cancer an effective amount of an antibody (e.g., 7-B16, 1-K17, etc.) or fusion protein provided herein. In some embodiments, the cancer is a solid cancer. In some embodiments, the cancer is a hematologic (i.e., liquid) cancer. In some embodiments, the cancer comprises tumor-infiltrating Treg cells. In some embodiments, the subject has received prior checkpoint inhibitor therapy (CPI) or has CPI-resistant or refractory cancer, as many subjects with CPI-resistant or refractory cancer will exhibit elevated levels of intratumoral tregs. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprizumab, and cepacilinumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab. In some embodiments, the cancer comprises cells that express CCR8 (e.g., tregs that express CCR 8). Expression of CCR8 can be determined by, for example, immunohistochemistry, Fluorescence Activated Cell Sorting (FACS), gene expression analysis (such as Q-PCR or RT-PCR), Western blotting, ELISA, or other known methods of assessing expression at the gene or protein level. In some embodiments, the CCR 8-expressing cells are Treg cells (e.g., intratumoral tregs), and in some embodiments, the expression levels of CCR8 of such intratumoral tregs are elevated relative to peripheral tregs. In some embodiments, CCR8 is expressed on the surface of Treg cells at less than 10,000 copies per cell, as can be determined by, for example, Fluorescence Activated Cell Sorting (FACS) and flow cytometry. In some embodiments, the cancer comprises tumor cells expressing CCR 8. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs express less than 10,000 copies of CCR 8/cell on the cell surface.
In some embodiments, the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the cancer may be metastatic. In some embodiments, when the cancer is breast cancer, the breast cancer is triple negative breast cancer (i.e., TNBC). In some embodiments, when the cancer is breast cancer, the breast cancer is metastatic TNBC.
In some embodiments, a method of treating a solid cancer having tumor-infiltrating tregs that express CCR8 comprises administering to a subject having a cancer an effective amount of an antibody (e.g., 7-B16, 1-K17, etc.) or fusion protein provided herein. In some embodiments, the tumor-infiltrating tregs express CCR8 at an elevated level relative to the level of CCR8 expression of peripheral tregs. Expression of CCR8 can be determined by, for example, immunohistochemistry, Fluorescence Activated Cell Sorting (FACS), gene expression analysis (such as Q-PCR or RT-PCR), Western blot, ELISA, or other known methods of assessing expression at the gene or protein level. In some embodiments, the solid cancer may be selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma.
In some embodiments, a method (or treatment/pharmaceutical use) of treating a hematologic cancer comprises administering to a subject having a hematologic cancer an effective amount of an antibody that binds human CCR8 (e.g., 7-B16, 1-K17, etc.). In some embodiments, the antibody inhibits the binding of CCL1 to CCR 8. In some embodiments, the hematologic cancer expresses CCR 8. In some embodiments, the subject has received a previous checkpoint inhibitor therapy (CPI) or has CPI-resistant or refractory cancer, as many subjects with CPI-resistant or refractory cancer will exhibit elevated levels of tregs expressing CCR 8. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, durolizumab, cimiciprizumab, and saprolizumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of tegralitumumab, viburnumab, donaldumab, AB308, BMS-986207, and covalendronumab. In some embodiments, the hematologic cancer comprises cells expressing CCR8 (e.g., tregs expressing CCR 8). In some embodiments, the cell expressing CCR8 is a Treg cell. In some embodiments, the hematologic cancer can be metastatic.
In some embodiments, a method or use of selecting a subject with a hematologic cancer for treatment with an antibody that binds human CCR8 comprises detecting CCR8 expression in a sample from the subject. In some embodiments, the methods or uses further comprise administering an effective amount of an antibody that binds human CCR 8.
In some embodiments, a method (or treatment/pharmaceutical use) of treating a hematologic cancer comprises administering to a subject having a hematologic cancer an effective amount of a fusion protein. In some embodiments, a method of selecting a subject with a hematologic cancer for treatment with a fusion protein comprising (a) CCL1 or an active fragment thereof, or MC148 or an active fragment thereof, and (b) an Fc region, comprises detecting CCR8 expression in a sample from the subject. In some embodiments, the method further comprises administering an effective amount of a fusion protein comprising (a) CCL1, or an active fragment thereof, or MC148, or an active fragment thereof, and (b) an Fc region. In some embodiments, the hematologic cancer expresses CCR 8. In some embodiments, the subject has received prior checkpoint inhibitor therapy (CPI) or has CPI-resistant or refractory cancer, as many subjects with CPI-resistant or refractory cancer will exhibit elevated levels of tregs expressing CCR 8. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprizumab, and cepacilinumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab. In some embodiments, the hematologic cancer comprises cells expressing CCR8 (e.g., tregs expressing CCR 8). In some embodiments, the cell expressing CCR8 is a Treg cell. In some embodiments, the tregs express CCR8 in less than 10,000 copies per cell, as can be determined by, for example, Fluorescence Activated Cell Sorting (FACS) and flow cytometry. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs express less than 10,000 copies of CCR 8/cell on the cell surface. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs in the sample obtained from the tumor express less than 10,000 copies of CCR 8/cell. In some embodiments, the hematologic cancer can be metastatic.
In some embodiments, the hematologic cancer is a B-cell and T-cell mixed leukemia, B-cell lymphoma, Chronic Myeloid Leukemia (CML), chronic myelomonocytic leukemia, diffuse large B-cell lymphoma (DLBC), lymphoma, Mantle Cell Lymphoma (MCL), multiple myeloma, myelodysplastic syndrome (MDS), myeloproliferative disorders, peripheral T-cell lymphoma, T-cell leukemia, Acute Myeloid Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic Lymphoma (SLL), CLL/SLL, mature T-cell and NK-cell lymphoma, follicular lymphoma, Acute Lymphocytic Leukemia (ALL), T-cell acute lymphocytic leukemia (TALL), T-cell adult acute lymphocytic leukemia, T-cell childhood acute lymphocytic leukemia, lymphoblastic lymphoma, lymphoblastic leukemia, and lymphoblastic leukemia, Cutaneous T-cell lymphoma (CTCL), adult T-cell leukemia/lymphoma (ATLL), T-cell lymphoblastic leukemia/lymphoma (TLLL), angioimmunoblastic T-cell lymphoma (ATCL), hepatosplenic T-cell lymphoma (htll), peripheral T-cell lymphoma not otherwise specified (PTCL NOS), Burkitt Lymphoma (BL), chronic myelomonocytic leukemia (CMML), extranodal NK/T-cell lymphoma (NKTCL), Primary Effusion Lymphoma (PEL), acute lymphocytic leukemia/acute myeloid leukemia (ALL, AML), Histiocytic Lymphoma (HL), Marginal Zone Lymphoma (MZL), B-cell acute lymphocytic leukemia, or Anaplastic Large Cell Lymphoma (ALCL).
In some embodiments, the hematologic cancer is T cell adult acute lymphoblastic leukemia, T cell childhood acute lymphoblastic leukemia, lymphoblastic lymphoma, acute lymphoblastic leukemia, cutaneous T cell lymphoma, T cell acute lymphoblastic leukemia, adult T cell leukemia/lymphoma, T cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
In some embodiments, a method (or treatment/pharmaceutical use) of treating a solid cancer comprises administering to a subject having a solid cancer an effective amount of an antibody that binds human CCR8 (e.g., 7-B16, 1-K17, etc.). In some embodiments, the antibody inhibits the binding of CCL1 to CCR 8. In some embodiments, the solid cancer (or tumor-infiltrating Treg) expresses CCR 8. In some embodiments, the solid cancer comprises intratumoral tregs. In some embodiments, the intratumoral tregs have elevated expression levels of CCR8 relative to peripheral tregs. In some embodiments, CCR8 is expressed on the surface of intratumoral Treg cells in less than 10,000 copies per cell (e.g., if the entire Treg population has low Treg expression), as can be determined by, for example, Fluorescence Activated Cell Sorting (FACS) and flow cytometry. In some embodiments, the intratumoral tregs comprise tregs that express CCR8 in less than 10,000 copies per cell (referred to as a subset of tregs with low CCR8 expression). In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs express less than 10,000 copies of CCR 8/cell on the cell surface. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of tumor-infiltrating tregs in a sample obtained from a tumor express less than 10,000 copies of CCR 8/cell. In some embodiments, the subject has received a previous checkpoint inhibitor therapy (CPI) or has CPI-resistant or refractory cancer, as many subjects with CPI-resistant or refractory cancer will exhibit elevated levels of tregs within the tumor. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, durolizumab, cimiciprizumab, and saprolizumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of tegralitumumab, viburnumab, donaldumab, AB308, BMS-986207, and covalendronumab. In some embodiments, the intratumoral tregs are depleted. Depletion of tregs may be a result of ADCC and/or CDC activity possessed by the disclosed antibodies.
In some embodiments, a method or use of selecting a subject with a solid cancer for treatment with an antibody that binds human CCR8 comprises detecting CCR8 expression in a sample from the subject. In some embodiments, CCR8 expression is detected in a population of tumor-infiltrating tregs in a sample. In some embodiments, tumor-infiltrating tregs express CCR8 in less than 10,000 copies per cell, as can be determined by, for example, Fluorescence Activated Cell Sorting (FACS) and flow cytometry. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs express less than 10,000 copies of CCR 8/cell on the cell surface. In some embodiments, the methods or uses further comprise administering an effective amount of an antibody that binds human CCR 8.
In some embodiments, a method (or therapeutic/pharmaceutical use) of treating a solid cancer comprises administering to a subject having a solid cancer an effective amount of a fusion protein. In some embodiments, a method of selecting a subject with a solid cancer for treatment with a fusion protein comprising (a) CCL1 or an active fragment thereof, or MC148 or an active fragment thereof, and (b) an Fc region, comprises detecting CCR8 expression in a sample from the subject. In some embodiments, the method further comprises administering an effective amount of a fusion protein comprising (a) CCL1, or an active fragment thereof, or MC148, or an active fragment thereof, and (b) an Fc region. In some embodiments, the solid cancer expresses CCR 8. In some embodiments, the subject has received a previous checkpoint inhibitor therapy (CPI) or has CPI-resistant or refractory cancer, as many subjects with CPI-resistant or refractory cancer will exhibit elevated levels of tregs expressing CCR 8. In some embodiments, the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody. In some embodiments, the anti-PDL 1 antibody is selected from the group consisting of palivizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprizumab, and cepacilinumab; the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab. In some embodiments, the solid cancer comprises cells expressing CCR8 (e.g., tregs expressing CCR 8). In some embodiments, the CCR 8-expressing cell is a Treg cell. In some embodiments, the tregs express CCR8 in less than 10,000 copies per cell (as determined by, e.g., FACS and flow cytometry), and may be tumor-infiltrating tregs. In some embodiments, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 99%, or 100% of the tumor-infiltrating tregs express less than 10,000 copies of CCR 8/cell on the cell surface. In some embodiments, the hematologic cancer can be metastatic.
In some embodiments, the solid cancer may be selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma. In some embodiments, the solid cancer may be metastatic. In some embodiments, when the solid cancer is breast cancer, the breast cancer is triple negative breast cancer (i.e., TNBC). In some embodiments, when the solid cancer is breast cancer, the breast cancer is metastatic TNBC.
In some embodiments of any of the foregoing methods or uses, the administered antibody (e.g., 7-B16, 1-K17, etc.) or fusion protein may deplete tregs in a subject due to ADCC and/or CDC properties of the disclosed antibody and fusion protein.
The antibody or fusion protein can be administered to the subject as needed. The determination of the frequency of administration can be made by a person skilled in the art, such as an attending physician, based on considerations of the condition being treated, the age of the subject being treated, the severity of the condition being treated, the general health of the subject being treated, and the like. In some embodiments, an effective dose of the antibody or fusion protein is administered to the subject one or more times. In some embodiments, an effective dose of the antibody or fusion protein is administered to the subject once a month, less than once a month (e.g., once every two months or once every three months). In some embodiments, an effective dose of the antibody or fusion protein is administered less than once a month (e.g., once every three weeks, once every two weeks, or once a week). An effective dose of the antibody or fusion protein is administered to the subject at least once. In some embodiments, an effective dose of the antibody or fusion protein can be administered multiple times, including for a period of at least one month, at least six months, or at least one year.
In some embodiments, the pharmaceutical composition is administered in an amount effective to treat (including prevent) cancer. The therapeutically effective amount will generally depend upon the weight of the subject being treated, his or her physical or health condition, the scope of the condition being treated, or the age of the subject being treated. Generally, the antibody and fusion protein can be administered in an amount ranging from about 10 μ g/kg body weight to about 100mg/kg body weight per dose. In some embodiments, the antibody and fusion protein can be administered in an amount ranging from about 50 μ g/kg body weight to about 5mg/kg body weight per dose. In some embodiments, the antibody and fusion protein can be administered in an amount ranging from about 100 μ g/kg body weight to about 10mg/kg body weight per dose. In some embodiments, the antibody and fusion protein can be administered in an amount ranging from about 100 μ g/kg body weight to about 20mg/kg body weight per dose. In some embodiments, the antibody and fusion protein may be administered in an amount ranging from about 0.5mg/kg body weight to about 20mg/kg body weight per dose.
Pharmaceutical composition
In some embodiments, compositions comprising The antibodies and fusion proteins provided herein are provided in formulations with a variety of pharmaceutically acceptable carriers (see, e.g., Gennaro, Remington: The Science and Practice of Pharmacy with products and companies: drugs Plus, 20 th edition (2003); Ansel et al, Pharmaceutical DosaForms and Drug Delivery Systems, 7 th edition, Lippencott Williams and Wilkins (2004); Kibbe et al, Handbook of Pharmaceutical Excipients, 3 rd edition, Pharmaceutical Press (2000)). A variety of pharmaceutically acceptable carriers are available, including vehicles, adjuvants, and diluents. In addition, various pharmaceutically acceptable auxiliary substances are also available, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizing agents, wetting agents and the like. Non-limiting exemplary carriers include saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.
In some embodiments, pharmaceutical compositions comprising the antibodies or fusion proteins are provided. In some embodiments, the pharmaceutical composition comprises a humanized antibody. In some embodiments, the pharmaceutical composition comprises an antibody or fusion protein prepared in a host cell or cell-free system as described herein. In some embodiments, the pharmaceutical composition comprises a pharmaceutically acceptable carrier.
In some embodiments, the pharmaceutical composition is administered in an amount effective to treat (including prevent) cancer. The therapeutically effective amount will generally depend upon the weight of the subject being treated, his or her physical or health condition, the scope of the condition being treated, or the age of the subject being treated.
Route of administration
In some embodiments, the antibodies and/or fusion proteins provided herein can be administered in vivo by various routes including, but not limited to, intravenous, intra-arterial, parenteral, intratumoral, intraperitoneal, or subcutaneous. The formulation and route of administration may be selected as appropriate for the intended application.
Combination therapy
The antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) can be administered alone or with other modes of treatment. They may be provided prior to, substantially simultaneously with and/or after other modes of treatment, e.g., surgery, chemotherapy, radiotherapy or administration of a biological agent, such as another therapeutic antibody. In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) are administered in combination with another anti-cancer agent. In some embodiments, the antibodies and/or fusion proteins provided herein are administered in combination with radiotherapy, such as ablative or non-ablative radiotherapy.
In some embodiments, the antibody and/or fusion protein (e.g., 7-B16, 1-K17, etc.) is administered concurrently with the second therapeutic agent. For example, the two or more therapeutic agents are administered at intervals of no more than about 60 minutes, such as no more than about any of 30 minutes, 15 minutes, 10 minutes, 5 minutes, or 1 minute. In some embodiments, the antibody and/or fusion protein and the second therapeutic agent are administered sequentially. For example, administration of the two or more therapeutic agents is administered at intervals of greater than about 15 minutes, such as about 20 minutes, 30 minutes, 40 minutes, 50 minutes, or 60 minutes, 1 day, 2 days, 3 days, 1 week, 2 weeks, or 1 month or more.
In some embodiments, the antibody and/or fusion protein (e.g., 7-B16, 1-K17, etc.) is administered for treatment with a second method of treatment. Thus, administration of the antibodies provided herein can be combined with another therapeutic system.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) are administered with anti-PD-1 or anti-PD-L1 therapy. In some embodiments, the antibodies and/or fusion proteins provided herein are administered with an anti-PD-1 antibody or an anti-PD-L1 antibody (e.g., acilizumab).
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) are administered with anti-ICOS therapy. In some embodiments, the antibodies and/or fusion proteins provided herein are administered with an antibody that binds inducible T cell costimulatory factor (ICOS). In some embodiments, the antibodies and/or fusion proteins provided herein are administered with an isolated antibody that binds ICOS, wherein the anti-ICOS antibody is an agonist of CD4+ T cells, such as CD4+ T effector (Teff) cells. In some embodiments, the antibodies that bind ICOS are agonists of CD4+ T cells (such as CD4+ Teff cells) and deplete T regulatory (Treg) cells.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) are administered with an agonist anti-OX 40 antibody (such as Medi6469, mediimmune; MOXR0916/RG7888, Roche). In some embodiments, the antibodies and/or fusion proteins provided herein are combined with an anti-CTLA 4 antibody (such as ipilimumab,BMS) are administered together.
In some embodiments, the additional therapeutic agent is a chemotherapeutic agent. Exemplary chemotherapeutic agents that may be combined with the antibodies and/or fusion proteins provided herein include, but are not limited to, capecitabine, cyclophosphamide, dacarbazine, temozolomide, cyclophosphamide, docetaxel, doxorubicin, daunorubicin, cisplatin, carboplatin, epirubicin, eribulin, 5-FU, gemcitabine, irinotecan, ixabepilone, methotrexate, mitoxantrone, oxaliplatin, paclitaxel, nabu-paclitaxel, and the like, (protein-bound paclitaxel), pemetrexed, vinorelbine, and vincristine. In some embodiments, the antibodies and/or fusion proteins provided herein are administered with at least one kinase inhibitor. Non-limiting exemplary kinase inhibitionAgents include erlotinib (erlotinib), afatinib (afatinib), gefitinib (gefitinib), crizotinib (crizotinib), dabrafenib (dabrafenib), trametinib (trametinib), vemurafenib (vemurafenib) and cobimetinib (cobimetinib).
In some embodiments, the additional therapeutic agent is an IDO inhibitor. Non-limiting exemplary IDO inhibitors are described in, for example, US 2016/0060237; and US 2015/0352206. Non-limiting exemplary IDO inhibitors include indoximod (New Link Genetics), INCB024360(Incyte Corp), 1-methyl-D-tryptophan (New Link Genetics), and GDC-0919 (Genentech).
In some embodiments, the antibodies and/or fusion proteins provided herein are administered in combination with an immune modifying drug (IMiD). Non-limiting exemplary imids include thalidomide, lenalidomide, and pomalidomide.
In some embodiments, the additional therapeutic agent is a cancer vaccine. Cancer vaccines have been investigated as a potential method of antigen transfer and dendritic cell activation. In particular, vaccination in combination with agonists of immune checkpoints or co-stimulatory pathways has shown evidence of overcoming tolerance and producing increased anti-tumor responses. A series of cancer vaccines have been tested that use different approaches to promote an immune response against a tumor (see, e.g., Emens LA, Expert Opin Emerg Drugs 13(2): 295-308 (2008)). Methods have been devised to enhance the response of B cells, T cells or professional antigen presenting cells to tumors. Exemplary cancer vaccine types include, but are not limited to, the use of peptide-based vaccines targeting different tumor antigens, which can be delivered as peptides/proteins or as genetically engineered DNA vectors, viruses, bacteria, etc.; and cell biology methods, such as for the development of cancer vaccines against less well-defined targets, including but not limited to vaccines developed from patient-derived dendritic cells, autologous tumor cells or tumor cell lysates, allogeneic tumor cells, and the like.
Thus, in certain embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) can be used in combination with a cancer vaccine. Exemplary cancer vaccines include, but are not limited to, dendritic cell vaccines, oncolytic viruses, tumor cell vaccines, and the like. In some embodiments, such vaccines enhance anti-tumor responses. Examples of cancer vaccines that can be used in combination with the antibodies and/or fusion proteins provided herein include, but are not limited to, MAGE3 vaccine (e.g., for melanoma and bladder cancer), MUC1 vaccine (e.g., for breast cancer), EGFRv3 (such as Rindopepimut, e.g., for brain cancer, including glioblastoma multiforme), or ALVAC-CEA (e.g., for CEA + cancer).
Non-limiting exemplary cancer vaccines also include Sipuleucel-T derived from autologous Peripheral Blood Mononuclear Cells (PBMCs) including antigen presenting cells (see, e.g., Kantoff PW et al, N Engl J Med 363:411-22 (2010)). In Sipuleucel-T production, PBMCs of patients were activated ex vivo with PA2024, a recombinant fusion protein of prostatic acid phosphatase (prostate antigen) and granulocyte-macrophage colony stimulating factor (immune cell activator). Another approach to candidate cancer vaccines is to generate an immune response against specific peptides mutated in tumor tissue such as melanoma (see, e.g., Carreno BM et al, Science 348:6236 (2015)). In some embodiments, such mutant peptides may be referred to as neoantigens. As a non-limiting example of the use of neoantigens in tumor vaccines, neoantigens predicted to bind major histocompatibility complex protein HLA-a 02:01 in tumors are identified for individual patients with cancer such as melanoma. Dendritic cells from patients are matured ex vivo and then incubated with neoantigens. The activated dendritic cells are then administered to the patient. In some embodiments, robust T cell immunity to neoantigens can be detected following administration of cancer vaccines.
In some such embodiments, a cancer vaccine is developed using the neoantigen. In some embodiments, the cancer vaccine is a DNA vaccine. In some embodiments, the cancer vaccine is an engineered virus comprising a cancer antigen, such as PROSTVAC (rivomogene galvano)/grifformyl). In some embodiments, the cancer vaccine comprises engineered tumor cells, such as GVAX, which is a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine (see, e.g., Nemunaitis,2005, Expert Rev Vaccines,4: 259-74).
In some embodiments, the antibodies and/or fusion proteins described herein are administered prior to, concurrently with, and/or after a cancer vaccine. In some embodiments, cancer vaccines developed using the neoantigens are used in combination with the antibodies and/or fusion proteins described herein. In some such embodiments, the combination is used to treat a cancer with a high mutational load, such as melanoma, lung, bladder or colorectal cancer.
In some embodiments, the antibodies and/or fusion proteins provided herein are administered in combination with chimeric antigen receptor T cell therapy (CAR-T therapy).
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16, 1-K17, etc.) are administered with one or more (e.g., one, two, three, or four) additional therapeutic agents. In some embodiments, the additional therapeutic agent includes, for example, an inhibitory immune checkpoint blocker or inhibitor, a stimulatory immune checkpoint stimulator, an agonist or activator, a chemotherapeutic agent, an anti-cancer agent, a radiotherapeutic agent, an antineoplastic agent, an antiproliferative agent, an anti-angiogenic agent, an anti-inflammatory agent, an immunotherapeutic agent, a therapeutic antigen binding molecule (e.g., monospecific and multispecific antibodies or fragments thereof in any form, such asBiKE、TriKE、scFv, Fab derivatives), bispecific antibodies, non-immunoglobulin antibody mimetics (including, for example, adnectin, affibody, affilin, affimer, affitin, alphabody, anticalin, peptide aptamers, armadillo-repeat protein (ARM), atrimer, avimer, designed ankyrin-repeat proteinfynomer, knottin, Kunitz domain peptide, mono-antibody and nanocomplex), antibody-drug conjugates (ADC),Antibody-peptide conjugates), oncolytic viruses, gene modifiers or gene editors, cells comprising a Chimeric Antigen Receptor (CAR) (e.g., including T cell immunotherapeutic agents, NK cell immunotherapeutic agents, or macrophage immunotherapeutic agents), cells comprising an engineered T cell receptor (TCR-T), or combinations thereof.
Exemplary targets
In some embodiments, the one or more additional therapeutic agents include, for example, inhibitors (inhibitors), agonists, antagonists, ligands, modulators, stimulators, blockers, activators, or inhibitors (supressors) of a target (e.g., a polypeptide or polynucleotide), such as: 2'-5' -oligoadenylate synthetase (OAS 1; NCBI gene ID: 4938); 5'-3' exoribonuclease 1(XRN 1; NCBI gene ID: 54464); 5' -exonucleotidase (NT5E, CD 73; NCBI gene ID: 4907); ABL protooncogene 1, non-receptor tyrosine kinase (ABL1, BCR-ABL, c-ABL, v-ABL; NCBI gene ID: 25); melanoma-deficient factor 2(AIM 2; NCBI gene ID: 9447); acetyl-CoA acyltransferase 2(ACAA 2; NCBI gene ID: 10499); acid phosphatase 3(ACP 3; NCBI gene ID: 55); adenosine deaminase (ADA, ADA 1; NCBI gene ID: 100); adenosine receptors (e.g., ADORA1(A1), ADORA2A (A2a, A2AR), ADORA2B (A2b, A2BR), ADORA3 (A3); NCBI gene IDs: 134, 135, 136, 137); AKT serine/threonine kinase 1(AKT1, AKT, PKB; NCBI gene ID: 207); alanyl aminopeptidase (membrane) (ANPEP, CD 13; NCBI gene ID: 290); ALK receptor tyrosine kinase (ALK, CD 242; NCBI gene ID: 238); alpha-fetoprotein (AFP; NCBI gene ID: 174); copper-containing amine oxidases (e.g., AOC1(DAO1), AOC2, AOC3(VAP 1); NCBI genes ID: 26, 314, 8639); androgen receptor (AR; NCBI gene ID: 367); angiogenin (ANGPT1, ANGPT 2; NCBI gene ID: 284, 285); angiotensin II receptor type 1 (AGTR 1; NCBI gene ID: 185); angiotensinogen (AGT; NCBI Gene ID: 183); apolipoprotein A1(APOA 1; NCBI Gene ID: 335); mitochondrial associated apoptosis-inducing factor 1(AIFM1, AIF; NCBI gene ID: 9131); arachidonic acid 5-lipoxygenase (ALOX 5; NCBI gene ID: 240); asparaginase (ASPG; NCBI gene ID: 374569); asteroid homolog 1(ASTE 1; NCBI gene ID: 28990); ATM serine/threonine kinase (ATM; NCBI gene ID: 472); ATP-binding cassette subfamily B member 1(ABCB1, CD243, GP 170; NCBI gene ID: 5243); ATP-dependent Clp protease (CLPP; NCBI gene ID: 8192); ATR serine/threonine kinase (ATR; NCBI gene ID: 545); AXL receptor tyrosine kinase (AXL; NCBI gene ID: 558); b and T lymphocyte-related (BTLA, CD 272; NCBI gene ID: 151888); proteins containing baculovirus IAP repeats (BIRC2(cIAP1), BIRC3(cIAP2), XIAP (BIRC4, IAP3), BIRC5 (survivin); NCBI gene ID: 329, 330, 331, 332); baigin (Ok blood group) (BSG, CD 147; NCBI gene ID: 682); b cell lymphoma 2(BCL 2; NCBI gene ID: 596); BCL2 binding component 3(BBC3, PUMA; NCBI gene ID: 27113); BCL 2-like (e.g., BCL2L1(Bcl-x), BCL2L2 (BIM); Bcl-x; NCBI gene ID: 598, 10018); beta 3-adrenergic receptor (ADRB 3; NCBI gene ID: 155); bone gamma-carboxyglutamic acid protein (BGLAP; NCBI gene ID: 632); bone morphogenetic protein-10 ligand (BMP 10; NCBI gene ID: 27302); bradykinin receptors (e.g., BDKRB1, BDKRB 2; NCBI gene ID: 623, 624); B-RAF (BRAF; NCBI gene ID: 273); a breakpoint cluster region (BCR; NCBI gene ID: 613); bromodomain and outer domain (BET) bromodomain-containing proteins (e.g., BRD2, BRD3, BRD4, BRDT; NCBI gene IDs: 6046, 8019, 23476, 676); bruton's tyrosine kinase (BTK; NCBI gene ID: 695); cadherins (e.g., CDH3 (p-cadherin), CDH6 (k-cadherin); NCBI gene ID: 1001, 1004); cancer/testis antigens (e.g., CTAG1A, CTAG1B, CTAG 2; NCBI gene ID: 1485, 30848, 246100); cannabinoid receptors (e.g., CNR1(CB1), CNR2(CB 2); NCBI gene ID: 1268, 1269); carbohydrate sulfotransferase 15(CHST 15; NCBI gene ID: 51363); carbonic anhydrases (e.g., CA1, CA2, CA3, CA4, CA5A, CA5B, CA6, CA7, CA8, CA9, CA10, CA11, CA12, CA13, CA 14; NCBI gene IDs: 759, 760, 761, 762, 763, 765, 766, 767, 768, 770, 771, 11238, 23632, 56934, 377677); carcinoembryonic antigen-related cell adhesion molecules (e.g., CEACAM3(CD66d), CEACAM5(CD66e), CEACAM6(CD66 c); NCBI gene IDs 1048, 1084, 4680); casein kinases (e.g., CSNK1A1(CK1), CSNK2A1(CK 2); NCBI gene ID: 1452, 1457); caspases (e.g., CASP3, CASP7, CASP 8; NCBI gene IDs: 836, 840, 841, 864); catenin beta 1(CTNNB 1; NCBI gene ID: 1499); cathepsin G (CTSG; NCBI gene ID: 1511); cbl proto-oncogene B (CBLB, Cbl-B; NCBI gene ID: 868); C-C motif chemokine ligand 21(CCL 21; NCBI Gene ID: 6366); C-C motif chemokine receptor 2(CCR 2; NCBI gene ID: 729230); C-C motif chemokine receptors (e.g., CCR3(CD193), CCR4(CD194), CCR5(CD195), CCR8(CDw 198); NCBI genes ID: 1232, 1233, 1234, 1237); the CCAAT enhancer binding protein alpha (CEBPA, CEBP; NCBI gene ID: 1050); cell adhesion molecule 1(CADM 1; NCBI gene ID: 23705); cell division cycle 7(CDC 7; NCBI gene ID: 8317); cell communication network factor 2(CCN 2; NCBI Gene ID: 1490); cereblon (CRBN; NCBI gene ID: 51185); checkpoint kinases (e.g., CHEK1(CHK1), CHEK2(CHK 2); NCBI gene ID: 1111, 11200); cholecystokinin B receptor (CCKBR;, NCBI gene ID: 887); chorionic lactogen 1(CSH 1; NCBI gene ID: 1442); blocking proteins (e.g., CLDN6, CLDN 18; NCBI gene ID: 9074, 51208); cluster of differentiation markers (e.g., CD1, CD α (TRA), CD β (TRB), CD γ (TRG), CD δ (TRD), CD8, CD (MS 4A), CD (IL2, TCGFR), CD (SIGLEC), CD (ENTPD), CD (TNFRSF), CD (MIC, PGP), CD (IAP), CD (BLAST), CD (DAF), CD (LFA), CD79, CD (B-1), CD (B-2), CD (tacile), CD (MIC), CD115(CSF 1), CD116(GMCSFR, CSF 2), CD122(IL 2), CD123(IL 3), CD128 IL 8R), CD132(IL 2), CD135 (FLT), CD (tnff, 4-BB), CD152 (193-1), CD (tff), CD142 (IGF), CD194), CD (IGF), CD (tff) 207, CD (tff), CD (IGF) 207, CD194), CD (IGF), CD (tff) and CD (tff), CD (IGF) 207, CD (tff) and CD (tff), CD) 21 (IGF) and CD (142, CD) and CD (IGF) 21 (tff), CD) and CD (tff), CD (IGF) 21 (CD) and CD123 (CD) and CD 190, CD (IL 8), CD) and CD (IGF), CD) and CD (CD) and CD 190 CD244, CD247, CD248, CD276(B7-H3), CD331(FGFR1), CD332(FGFR2), CD333(FGFR3), CD334(FGFR 4); NCBI Gene ID: 909. 911, 912, 913, 914, 919, 920, 923, 925, 926, 930, 931, 933, 940, 941, 942, 945, 951, 952, 953, 958, 960, 961, 962, 965, 972, 973, 974, 1043, 1232, 1233, 1234, 1237, 1436, 1438, 1493, 1604, 2152, 2260, 2261, 2263, 2322, 3480, 3482, 3559, 3560, 3561, 3563, 3577, 3579, 3604, 3902, 4267, 6955, 6957, 696, 8832, 10666, 11126, 50489, 51744, 80381, 100133941); clusterin (CLU; NCBI Gene ID: 1191); blood coagulation factors (e.g., F7, FXA; NCBI gene ID: 2155, 2159); collagen type IV alpha chains (e.g., COL4A1, COL4A2, COL4A3, COL4A4, COL4A 5; NCBI gene IDs: 1282, 1284, 1285, 1286, 1287); collectin subfamily member 10(COLEC 10; NCBI Gene ID: 10584); colony stimulating factors (e.g., CSF1(MCSF), CSF2(GMCSF), CSF3 (GCSF); NCBI gene IDs: 1435, 1437, 1440); complement factors (e.g., C3, C5; NCBI gene ID: 718, 727); COP9 semaphore subunit 5(COPS 5; NCBI gene ID: 10987); c-type lectin domain family members (e.g., CLEC4C (CD303), CLEC9A (CD370), CLEC12A (CD 371); CD 371; NCBI gene IDs: 160364, 170482, 283420); C-X-C motif chemokine ligand 12(CXCL 12; NCBI gene ID: 6387); C-X-C motif chemokine receptors (CXCR1(IL8R1, CD128), CXCR2(IL8R2, CD182), CXCR3(CD182, CD183, IP-10R), CXCR4(CD 184); NCBI gene IDs: 2833, 3577, 3579, 7852); cyclin D1(CCND1, BCL 1; NCBI gene ID: 595); cyclin-dependent kinases (e.g., CDK1, CDK2, CDK3, CDK4, CDK5, CDK6, CDK7, CDK8, CDK9, CDK10, CDK 12; NCBI gene IDs: 983, 1017, 1018, 1019, 1020, 1021, 1022, 1024, 1025, 8558, 51755; cyclin G1(CCNG 1; NCBI gene ID: 900), cytochrome P450 family members (e.g., CYP2D 1, CYP3A 1, CYP11B 1, CYP17A1, CYP19A1, CYP51A 1; NCBI gene ID: 1565, 1583, 1585, 1586, 1588, 1595; cytochrome P450 oxidoreductase (Ab; NCBI gene ID: 5447), cytokine-inducible SH-containing proteins (CISH 1; NCBI gene 1151654; CTLA toxic proteins (DDBI genes: 1656; DDBI gene ID; DDBI gene binding proteins) (e.g., DDDLL 72; DDBI gene ID: 1; CTLA 1; DDBI gene ID; CTLA 72; CTLA 1; CTLA receptor binding protein; 1; DDBI gene; 1; DDBI gene 1; CTLA receptor binding protein; 1; DDBI gene; 1; CTLA receptor 1; DDBI gene; CTLA receptor 1; CTLA receptor binding protein; 1; CTLA receptor 1; DDBI gene; CTLA receptor) (e.g. 1; DDBI gene; CTLA; CTD; 1; CTLA receptor 1; CTLA; CTD; 1; CTLA receptor 1; CTLA; 1; CTLA receptor 1; BIX; see: 1; see, SMAC; NCBI Gene ID: 56616) (ii) a Diacylglycerol kinases (e.g., DGKA, DGKZ; NCBI gene ID: 1606, 8525); dickkopf WNT signaling pathway inhibitors (e.g., DKK1, DKK 3; NCBI gene ID: 22943, 27122); dihydrofolate reductase (DHFR; NCBI gene ID: 1719); dihydropyrimidine dehydrogenase (DPYD; NCBI gene ID: 1806); dipeptidyl peptidase 4(DPP 4; NCBI gene ID: 1803); discoidin domain receptor tyrosine kinases (e.g., DDR1(CD167), DDR 2; CD 167; NCBI gene ID: 780, 4921); DNA-dependent protein kinase (PRKDC; NCBI gene ID: 5591); DNA topoisomerases (e.g., TOP1, TOP2A, TOP2B, TOP3A, TOP 3B; NCBI gene IDs 7150, 7153, 7155, 7156, 8940); dopachrome tautomerase (DCT; NCBI gene ID: 1638); dopamine receptor D2(DRD 2; NCBI Gene ID: 1318); DOT 1-like histone lysine methyltransferase (DOT 1L; NCBI gene ID: 84444); ectonucleotide pyrophosphatase/phosphodiesterase 3(ENPP3, CD203 c; NCBI gene ID: 5169); EMAP-like 4(EML 4; NCBI gene ID: 27436); endoglin (ENG; NCBI gene ID: 2022); endoplasmic reticulum aminopeptidases (e.g., ERAP1, ERAP 2; NCBI gene ID: 51752, 64167); enhancer of zeste homolog 2 multicomb inhibitory complex 2 subunit (EZH 2; NCBI Gene ID: 2146); ephrin (ephrin) receptors (e.g., EPHA1, EPHA2EPHA3, EPHA4, EPHA5, EPHA7, EPHB 4; NCBI gene IDs: 1969, 2041, 2042, 2043, 2044, 2045, 2050); ephrin (e.g., EFNA1, EFNA4, EFNB 2; NCBI Gene ID: 1942, 1945, 1948); epidermal growth factor receptor (e.g., ERBB1(HER1, EGFR), ERBB1 variant III (EGFRvIII), ERBB2(HER2, NEU, CD340), ERBB3(HER3), ERBB4(HER 4); NCBI gene ID: 1956, 2064, 2065, 2066); epithelial cell adhesion molecule (EPCAM; NCBI gene ID: 4072); epithelial mitogen (EPGN; NCBI gene ID: 255324); eukaryotic translation elongation factors (e.g., EEF1A2, EEF 2; NCBI gene ID: 1917, 1938); eukaryotic translation initiation factors (e.g., EIF4A1, EIF 5A; NCBI Gene ID: 1973, 1984); exportin-1 (XPO 1; NCBI gene ID: 7514); farnesoid X receptor (NR1H4, FXR; NCBI Gene ID: 9971); fas ligand (FASLG, FASL, CD95L, CD178, TNFSF 6; NCBI gene ID: 356); fatty acid amide hydrolase (FAAH; NCBI gene ID: 2166); fatty acid synthase (FASN; FAS; NCBI gene ID: 2194); fc fragments of Ig receptors (e.g., FCER1A, FCGRT, FCGR3A (CD 16); NCBI gene IDs: 2205, 2214, 2217); fc receptor-like 5(FCRL5, CD 307; NCBI gene ID: 83416); fibroblast activation protein alpha (FAP; NCBI gene ID: 2191); fibroblast growth factor receptors (e.g., FGFR1(CD331), FGFR2(CD332), FGFR3(CD333), FGFR4(CD 334); NCBI gene IDs: 2260, 2261, 2263, 2264); fibroblast growth factors (e.g., FGF1 (FGF. alpha.), FGF2 (FGF. beta.), FGF4, FGF 5; NCBI gene IDs: 2246, 2247, 2249, 2250); fibronectin 1(FN1, MSF; NCBI gene ID: 2335); fms-related receptor tyrosine kinases (e.g., FLT1(VEGFR1), FLT3(STK1, CD135), FLT4(VEGFR 2); NCBI gene IDs: 2321, 2322, 2324); fms-related receptor tyrosine kinase 3 ligand (FLT3 LG; NCBI gene ID: 2323); focal adhesion kinase 2(PTK2, FAK 1; NCBI gene ID: 5747); folate hydrolase 1(FOLH1, PSMA; NCBI gene ID: 2346); folate receptor 1(FOLR 1; NCBI gene ID: 2348); forkhead box protein M1(FOXM 1; NCBI Gene ID: 2305); FURIN (FURIN, PACE; NCBI gene ID: 5045); FYN tyrosine kinase (FYN, SYN; NCBI gene ID: 2534); galectins (e.g., LGALS3, LGALS8(PCTA1), LGALS 9; NCBI gene IDs: 3958, 3964, 3965); glucocorticoid receptor (NR3C1, GR; NCBI gene ID: 2908); glucuronidase beta (GUSB; NCBI gene ID: 2990); glutamate metabotropic receptor 1(GRM 1; NCBI gene ID: 2911); glutaminase (GLS; NCBI gene ID: 2744); glutathione S-transferase Pi (GSTP 1; NCBI gene ID: 2950); glycogen synthase kinase 3 β (GSK 3B; NCBI gene ID: 2932); glypican 3(GPC 3; NCBI gene ID: 2719); gonadotropin releasing hormone 1(GNRH 1; NCBI gene ID: 2796); gonadotropin releasing hormone receptor (GNRHR; NCBI gene ID: 2798); GPNMB glycoprotein nmb (GPNMB, osteoactivin; NCBI gene ID: 10457); growth differentiation factor 2(GDF2, BMP 9; NCBI gene ID: 2658); growth factor receptor binding protein 2(GRB2, ASH; NCBI gene ID: 2885); guanylate cyclase 2C (GUCY2C, STAR, MECIL, MUCIL, NCBI Gene ID: 2984); h19 imprinting of gene parental expression transcripts (H19; NCBI Gene ID: 283120); HCK proto-oncogene Src family tyrosine kinase (HCK; NCBI gene ID: 3055); heat shock proteins (e.g., HSPA5(HSP70, BIP, GRP78), HSPB1(HSP27), HSP90B1(GP 96); NCBI gene IDs: 3309, 3315, 7184); heme oxygenases (e.g., HMOX1(HO1), HMOX2(HO 1); NCBI gene ID: 3162, 3163); heparanase (HPSE; NCBI gene ID: 10855); hepatitis A virus cell receptor 2(HAVCR2, TIM3, CD 366; NCBI gene ID: 84868); hepatocyte growth factor (HGF; NCBI gene ID: 3082); HERV-H LTR-related 2(HHLA2, B7-H7; NCBI gene ID: 11148); histamine receptor H2(HRH 2; NCBI gene ID: 3274); histone deacetylases (e.g., HDAC1, HDAC7, HDAC 9; NCBI gene ID: 3065, 9734, 51564); HRas proto-oncogene GTP enzyme (HRAS; NCBI gene ID: 3265); hypoxia inducible factors (e.g., HIF1A, HIF2A (EPAS 1); NCBI gene ID: 2034, 3091); I-kappa-B kinase (IKK beta; NCBI gene ID: 3551, 3553); IKAROS family zinc fingers (IKKF 1(LYF1), IKKF 3; NCBI gene IDs: 10320, 22806); immunoglobulin superfamily member 11(IGSF 11; NCBI gene ID: 152404); indoleamine 2, 3-dioxygenase (e.g., IDO1, IDO 2; NCBI gene ID: 3620, 169355); inducible T cell costimulators (ICOS, CD 278; NCBI gene ID: 29851); inducible T cell costimulator ligands (ICOSLG, B7-H2; NCBI gene ID: 23308); insulin-like growth factor receptors (e.g., IGF1R, IGF 2R; NCBI gene ID: 3480, 3482); insulin-like growth factors (e.g., IGF1, IGF 2; NCBI gene ID: 3479, 3481); insulin receptor (INSR, CD 220; NCBI gene ID: 3643); integrin subunits (e.g., ITGA5(CD49e), ITGAV (CD51), ITGB1(CD29), ITGB2(CD18, LFA1, MAC1), ITGB 7; NCBI gene IDs: 3678, 3685, 3688, 3695, 3698); intercellular adhesion molecule 1(ICAM1, CD 54; NCBI gene ID: 3383); interleukin 1 receptor-associated kinase 4(IRAK 4; NCBI gene ID: 51135); interleukin receptors (e.g., IL2RA (TCGFR, CD25), IL2RB (CD122), IL2RG (CD132), IL3RA, IL6R, IL13RA2(CD213A2), IL22RA 1; NCBI gene IDs: 3598, 3559, 3560, 3561, 3563, 3570, 58985); interleukins (e.g., IL1A, IL1B, IL2, IL3, IL6(HGF), IL7, IL8(CXCL8), IL10(TGIF), IL12A, IL12B, IL15, IL17A (CTLA8), IL23A, IL24, IL-29(IFNL 1); NCBI gene IDs 3552, 3553, 3558, 3562, 3565, 3569, 3574, 3586, 3592, 3593, 3600, 3605, 11009, 51561, 282618); isocitrate dehydrogenase (NADP (+)1) (e.g., IDH1, IDH 2; NCBI gene ID: 3417, 3418); janus kinases (e.g., JAK1, JAK2, JAK 3; NCBI gene ID: 3716, 3717, 3718); kallikrein related peptidase 3(KLK 3; NCBI gene ID: 354); killer cell immunoglobulin-like receptor Ig domains and long cytoplasmic tails (e.g., KIR2DL1(CD158A), KIR2DL2(CD158B1), KIR2DL3(CD158B), KIR2DL4(CD158D), KIR2DL5A (CD158F), KIR2DL5B, KIR3DL1(CD158E1), KIR3DL2(CD158K), KIR3DP1(CD158c), KIR2DS2(CD 158J); NCBI gene IDs 3802, 3803, 3804, 3805, 3811, 3812, 57292, 553128, 548594, 100132285); killer lectin-like receptors (e.g., KLRC1(CD159A), KLRC2(CD159c), KLRC3, KLRRC4, KLRD1(CD94), KLRG1, KLRK1(NKG2D, CD 314); NCBI gene IDs 3821, 3822, 3823, 3824, 8302, 10219, 22914); kinase insert domain receptors (KDR, CD309, VEGFR 2; NCBI gene ID: 3791); kinesin family member 11(KIF 11; NCBI gene ID: 3832); KiSS-1 transfer inhibitory factor (KISS 1; NCBI gene ID: 3814); KIT proto-oncogene receptor tyrosine kinase (KIT, C-KIT, CD 117; NCBI gene ID: 3815); KRAS proto-oncogene GTP enzyme (KRAS; NCBI gene ID: 3845); lactotransferrin (LTF; NCBI gene ID: 4057); LCK proto-oncogene Src family tyrosine kinase (LCK; NCBI gene ID: 3932); LDL receptor-related protein 1(LRP1, CD91, IGFBP 3R; NCBI gene ID: 4035); leucine-rich repeat containing protein 15(LRRC 15; NCBI Gene ID: 131578); leukocyte immunoglobulin-like receptors (e.g., LILRB1(ILT2, CD85J), LILRB2(ILT4, CD 85D); NCBI gene IDs: 10288, 10859); leukotriene A4 hydrolase (LTA 4H; NCBI gene ID: 4048); a linker for activating T cells (LAT; NCBI gene ID: 27040); luteinizing hormone/chorionic gonadotropin receptor (LHCGR; NCBI gene ID: 3973); LY6/PLAUR domain-containing protein 3(LYPD 3; NCBI gene ID: 27076); lymphocyte activation gene 3(LAG 3; CD 223; NCBI gene ID: 3902); lymphocyte antigens (e.g., LY9(CD229), LY75(CD 205); NCBI gene ID: 4063, 17076); LYN proto-oncogene Src family tyrosine kinase (LYN; NCBI gene ID: 4067); lymphocyte cytoplasmic protein 2(LCP 2; NCBI gene ID: 3937); lysine demethylase 1A (KDM 1A; NCBI Gene ID: 23028); lysophosphatidic acid receptor 1(LPAR1, EDG2, LPA1, GPR 26; NCBI Gene ID: 1902); lysyl oxidase (LOX; NCBI gene ID: 4015); lysyl oxidase-like protein 2(LOXL 2; NCBI gene ID: 4017); macrophage migration inhibitory factor (MIF, GIF; NCBI gene ID: 4282); macrophage stimulating factor 1 receptor (MST1R, CD 136; NCBI gene ID: 4486); MAGE family members (e.g., MAGEA1, MAGEA2, MAGEA2B, MAGEA3, MAGEA4, MAGEA5, MAGEA6, MAGEA10, MAGEA11, MAGEC1, MAGEC2, MAGED1, MAGED 2; NCBI gene IDs: 4100, 4101, 4102, 4103, 4104, 4105, 4109, 4110, 9500, 9947, 10916, 51438, 266740); major histocompatibility complexes (e.g., HLA-A, HLA-E, HLA-F, HLA-G; NCBI gene ID: 3105, 3133, 3134, 3135); major VAULT protein (MVP, VAULT 1; NCBI gene ID: 9961); MALT1 caspase (paracasease) (MALT 1; NCBI gene ID: 10892); MAPK activates protein kinase 2(MAPKAPK 2; NCBI gene ID: 9261); MAPK-interacting serine/threonine kinases (e.g., MKNK1, MKNK 2; NCBI gene IDs: 2872, 8569); matrix metallopeptidases (e.g., MMP1, MMP2, MMP3, MMP7, MMP8, MMP9, MMP10, MMP11, MMP12, MMP13, MMP14, MMP15, MMP16, MMP17, MMP19, MMP20, MMP21, MMP24, MMP25, MMP26, MMP27, MMP 28; NCBI gene ID: 4312, 4313, 4314, 4316, 4317, 4318, 4319, 4320, 4321, 4322, 4323, 4324, 4325, 4326, 4327, 9313, 10893, 56547, 64066, 64386, 79148, 118856); MCL1 apoptosis regulator, BCL2 family member (MCL 1; NCBI gene ID: 4170); the MDM2 protooncogene (MDM 2; NCBI gene ID: 4193); MDM4 regulator of p53 (MDM 4; BMFS 6; NCBI gene ID: 4194); mechanistic targets of rapamycin kinase (MTOR, FRAP 1; NCBI gene ID: 2475); melanin-A (MLANA; NCBI Gene ID: 2315); melanocortin receptors (MC1R, MC 2R; NCBI gene ID: 4157, 4148); MER proto oncogene tyrosine kinase (MERRTK; NCBI gene ID: 10461); mesothelin (MSLN; NCBI gene ID: 10232); MET proto-oncogene receptor tyrosine kinases (MET, c-Met, HGFR; NCBI gene ID: 4233); methionyl aminopeptidase 2(METAP2, MAP 2; NCBI gene ID: 10988); MHC class I polypeptide-related sequences (e.g., MICA, MICB; NCBI gene ID: 4277, 100507436); mitogen-activated protein kinases (e.g., MAPK1(ERK2), MAPK3(ERK1), MAPK8(JNK1), MAPK9(JNK2), MAPK10(JNK3), MAPK11(p38 β), MAPK 12; NCBI gene IDs: 5594, 5595, 5599, 5600, 5601, 5602, 819251); mitogen-activated protein kinase kinases (e.g., MAP3K5(ASK1), MAP3K8(TPL2, AURA 2); NCBI gene IDs: 4217, 1326); mitogen-activated protein kinase 1(MAP4K1, HPK 1; NCBI gene ID: 11184); mitogen-activated protein kinase kinases (e.g., MAP2K1(MEK1), MAP2K2(MEK2), MAP2K7(MEK 7); NCBI genes ID: 5604, 5605, 5609); the MPL proto-oncogene, thrombopoietin receptor (MPL; NCBI gene ID: 4352); mucins (e.g., MUC1 (including splice variants thereof (e.g., including MUC1/A, C, D, X, Y, Z and REP)), MUC5AC, MUC16(CA 125); NCBI gene IDs: 4582, 4586, 94025); MYC proto-oncogene bHLH transcription factor (MYC; NCBI gene ID: 4609); myostatin (MSTN, GDF 8; NCBI gene ID: 2660); myristoylated alanine-rich protein kinase C substrate (MARCKS; NCBI Gene ID: 4082); natriuretic peptide receptor 3(NPR 3; NCBI gene ID: 4883); ligand 1 of natural killer cytotoxic receptor 3 (NCR3LG1, B7-H6; NCBI gene ID: 374383); necdin, a member of the MAGE family (NDN; NCBI gene ID: 4692); NECTIN cell adhesion molecules (e.g., NECTN 2(CD112, PVRL2), NECTN 4(PVRL 4); NCBI gene ID: 5819, 81607); neural cell adhesion molecule 1(NCAM1, CD 56; NCBI gene ID: 4684); neuropilins (e.g., NRP1(CD304, VEGF165R), NRP2(VEGF165R 2); NCBI gene ID: 8828, 8829); neurotrophic receptor tyrosine kinases (e.g., NTRK1(TRKA), NTRK2(TRKB), NTRK3 (TRKC); NCBI gene IDs: 4914, 4915, 4916); NFKB activating protein (NKAP; NCBI gene ID: 79576); NIMA-associated kinase 9(NEK 9; NCBI gene ID: 91754); NLR family heat protein domain containing protein 3(NLRP3, NALP 3; NCBI gene ID: 114548); NOTCH receptors (e.g., NOTCH1, NOTCH2, NOTCH3, NOTCH 4; NCBI gene IDs: 4851, 4853, 4854, 4855); NRAS protooncogene GTP enzyme (NRAS; NCBI Gene ID: 4893); nuclear factor κ B (NFKB1, NFKB 2; NCBI gene IDs: 4790, 4791); erythroid-derived nuclear factor 2-like protein 2(NFE2L 2; NRF 2; NCBI gene ID: 4780); nuclear receptor subfamily 4 group a member 1(NR4a 1; NCBI gene ID: 3164); nucleolin (NCL; NCBI gene ID: 4691); nucleophosmin 1(NPM 1; NCBI gene ID: 4869); nucleotide binding oligomerization domain containing protein 2(NOD 2; NCBI gene ID: 64127); nudix hydrolase 1(NUDT 1; NCBI gene ID: 4521); O-6-methylguanine-DNA methyltransferase (MGMT; NCBI gene ID: 4255); opioid receptor delta 1(OPRD 1; NCBI gene ID: 4985); ornithine decarboxylase 1(ODC 1; NCBI gene ID: 4953); ketoglutarate dehydrogenase (OGDH; NCBI gene ID: 4967); parathyroid hormone (PTH; NCBI gene ID: 5741); PD-L1(CD 274; NCBI gene ID: 29126); periostin (POSTN; NCBI gene ID: 10631); peroxisome proliferator-activated receptors (e.g., PPARA (PPAR α), PPARD (PPAR δ), PPARG (PPAR γ); NCBI gene IDs: 5465, 5467, 5468); phosphatase and tensin homologs (PTEN; NCBI Gene ID: 5728); phosphatidylinositol-4, 5-bisphosphate 3-kinase (PIK3CA (PI 3K. alpha.), PIK3B (PI 3K. beta.), PIK3D (PI 3K. delta.), PIK3CG (PI 3K. gamma.); NCBI gene ID: 5290, 5291, 5293, 5294); phospholipases (e.g., PLA2G1B, PLA2G2A, PLA2G2D, PLA2G3, PLA2G4A, PLA2G5, PLA2G7, PLA2G10, PLA2G12A, PLA2G12B, PLA2G 15; NCBI gene IDs 5319, 5320, 5321, 5322, 7941, 8399, 50487, 23659, 26279, 81579, 84647); pim proto-oncogene serine/threonine kinases (e.g., PIM1, PIM2, PIM 3; NCBI gene IDs: 5292, 11040, 415116); placental growth factor (PGF; NCBI gene ID: 5228); plasminogen activator, urokinase (PLAU, u-PA, ATF; NCBI gene ID: 5328); platelet-derived growth factor receptors (e.g., PDGFRA (CD140A, PDGFR2), FDGFRB (CD140B, PDGFR 1); NCBI gene ID: 5156, 5159); plexin B1(PLXNB 1; NCBI Gene ID: 5364); poliovirus receptor (PVR) cell adhesion molecules (PVR, CD 155; NCBI gene ID: 5817); polo-like kinase 1(PLK 1; NCBI gene ID: 5347); poly (ADP-ribose) polymerase (e.g., PARP1, PARP2, PARP 3; NCBI gene ID: 142, 10038, 10039); polycombin EED (EED; NCBI gene ID: 8726); porcupine O-acyltransferase (PORCN; NCBI gene ID: 64840); PRAME nuclear receptor transcription regulator (PRAME; NCBI gene ID: 23532); prepro-melanosome protein (PMEL; NCBI gene ID: 6490); progesterone receptor (PGR; NCBI gene ID: 5241); programmed cell death 1(PDCD1, PD-1, CD 279; NCBI gene ID: 5133); programmed cell death protein 1 ligand 2(PDCD1LG2, CD273, PD-L2; NCBI gene ID: 80380); prominin 1(PROM1, CD 133; NCBI gene ID: 8842); promyelocytic leukemia (PML; NCBI gene ID: 5371); sphingolipid activating Protein (prosaposin) (PSAP; NCBI Gene ID: 5660); prostaglandin E receptor 4(PTGER 4; NCBI gene ID: 5734); prostaglandin E synthase (PTGES; NCBI gene ID: 9536); prostaglandin-endoperoxide synthase (PTGS1(COX1), PTGS2(COX 2); NCBI gene ID: 5742, 5743); proteasome 20S subunit beta 9(PSMB 9; NCBI gene ID: 5698); protein arginine methyltransferases (e.g., PRMT1, PRMT 5; NCBI gene ID: 3276, 10419); protein kinase N3(PKN 3; NCBI gene ID: 29941); protein phosphatase 2A (PPP2 CA; NCBI gene ID: 5515); protein tyrosine kinase 7 (inactive) (PTK 7; NCBI gene ID: 5754); protein tyrosine phosphatase receptor (PTPRB (PTPB), PTPRC (CD 45R); NCBI gene ID: 5787, 5788); prothymosin alpha (PTMA; NCBI gene ID: 5757); purine nucleoside phosphorylase (PNP; NCBI Gene ID: 4860); purinergic receptor P2X7(P2RX 7; NCBI gene ID: 5027); contains PVR-associated immunoglobulin domains (PVRIG, CD 112R; NCBI gene ID: 79037); raf-1 proto-oncogene serine/threonine kinase (RAF1, c-Raf; NCBI Gene ID: 5894); RAR-associated orphan receptor gamma (RORC; NCBI gene ID: 6097); ras homolog family member c (rhoc); NCBI Gene ID: 389) (ii) a Ras homolog to which mTORC1 binds (RHEB; NCBI gene ID: 6009); RB transcriptional co-repressor 1(RB 1; NCBI gene ID: 5925); receptor-interacting serine/threonine protein kinase 1(RIPK 1; NCBI gene ID: 8737); RET protooncogene (RET; NCBI gene ID: 5979); retinoic acid early transcripts (e.g., RAET1E, RAET1G, RAET 1L; NCBI gene ID: 135250, 154064, 353091); retinoic acid receptor alpha (e.g., RARA, RARG; NCBI gene ID: 5914, 5916); retinoid X receptors (e.g., RXRA, RXRB, RXRG; NCBI gene ID: 6256, 6257, 6258); rho-associated coiled coil-containing protein kinases (e.g., ROCK1, ROCK 2; NCBI Gene ID: 6093, 9475); ribosomal protein S6 kinase B1(RPS6KB1, S6K-. beta.1; NCBI gene ID: 6198); ring finger protein 128(RNF128, GRAIL; NCBI gene ID: 79589); ROS proto-oncogene 1 receptor tyrosine kinase (ROS 1; NCBI gene ID: 6098); circumcision-directed receptor 4(ROBO 4; NCBI gene ID: 54538); RUNX family transcription factor 3(RUNX 3; NCBI Gene ID: 864); s100 calbindin A9(S100A 9; NCBI gene ID: 6280); secreted frizzled-related protein 2(SFRP 2; NCBI gene ID: 6423); secreted phosphoprotein 1(SPP 1; NCBI gene ID: 6696); secretory globin family 1A member 1(SCGB1A 1; NCBI gene ID: 7356); selectins (e.g., SELE, SELL (CD62L), SELP (CD 62); NCBI gene ID: 6401, 6402, 6403); semaphorin 4D (SEMA 4D; CD 100; NCBI gene ID: 10507); sialic acid binds to Ig-like lectins (SIGLEC9(CD329), SIGLEC 10; NCBI gene ID: 27180, 89790); signal regulatory protein alpha (SIRPA, CD 172A; NCBI gene ID: 140885); signal transducers and transcriptional activators (e.g., STAT1, STAT3, STAT5A, STAT 5B; NCBI gene IDs: 6772, 6774, 6776, 6777); sirtuin-3(SIRT 3; NCBI gene ID: 23410); signaling Lymphocyte Activation Molecule (SLAM) family members (e.g., SLAMF1(CD150), SLAMF6(CD352), SLAMF7(CD319), SLAMF8(CD353), SLAMF 9; NCBI gene IDs: 56833, 57823, 89886, 114836); SLIT and NTRK-like family member 6(SLITRK 6; NCBI Gene ID: 84189); smooth coil class receptors (SMO; NCBI gene ID: 6608); soluble epoxide hydrolase 2(EPHX 2; NCBI gene ID: 2053); solute vector family members (e.g., SLC3A2(CD98), SLC5A5, SLC6A2, SLC10A3, SLC34A2, SLC39A6, SLC43A2(LAT4), SLC44A 4; NCBI gene IDs: 6520, 6528, 6530, 8273, 10568, 25800, 80736, 124935); somatostatin receptors (e.g., SSTR1, SSTR2, SSTR3, SSTR4, SSTR 5; NCBI gene IDs: 6751, 6752, 6753, 6754, 6755); sonic hedgehog signal molecule (SHH; NCBI gene ID: 6469); sp1 transcription factor (SP 1; NCBI gene ID: 6667); sphingosine kinases (e.g., SPHK1, SPHK 2; NCBI gene ID: 8877, 56848); sphingosine-1-phosphate receptor 1(S1PR1, CD 363; NCBI gene ID: 1901); spleen-related tyrosine kinase (SYK; NCBI gene ID: 6850); splicing factor 3B factor 1(SF3B 1; NCBI gene ID: 23451); SRC proto-oncogene non-receptor tyrosine kinase (SRC; NCBI gene ID: 6714); stabilin 1(STAB1, CLEVER-1; NCBI gene ID: 23166); STEAP family member 1(STEAP 1; NCBI gene ID: 26872); steroid sulfatase (STS; NCBI gene ID: 412); interferon response stimulator cGAMP interactor 1(STING 1; NCBI gene ID: 340061); superoxide dismutase 1(SOD1, ALS 1; NCBI gene ID: 6647); cytokine signaling inhibitors (SOCS1(CISH1), SOCS3(CISH 3); NCBI gene ID: 8651, 9021); synapsin 3(SYN 3; NCBI gene ID: 8224); syndecan 1(SDC1, CD138, syndecan; NCBI gene ID: 6382); synuclein alpha (SNCA, PARK 1; NCBI gene ID: 6622); t cell immunoglobulin and mucin domain containing protein 4(TIMD4, SMUCKLER; NCBI gene ID: 91937); t cell immunoreceptors with Ig and ITIM domains (TIGIT; NCBI gene ID: 201633); tachykinin receptors (e.g., TACR1, TACR 3; NCBI gene ID: 6869, 6870); TANK binding kinase 1(TBK 1; NCBI gene ID: 29110); tankyrase (TNKS; NCBI gene ID: 8658); TATA-box binding protein-associated factor, RNA polymerase I subunit B (TAF 1B; NCBI gene ID: 9014); t-box transcription factor T (TBXT; NCBI gene ID: 6862); TCDD inducible poly (ADP-ribose) polymerase (TIPARP, PAPR 7; NCBI gene ID: 25976); TEC protein tyrosine kinase (TEC; NCBI gene ID: 7006); TEK receptor tyrosine kinases (TEK, CD202B, TIE 2; NCBI Gene ID: 7010); telomerase reverse transcriptase (TERT; NCBI gene ID: 7015); tenascin-C (TNC; NCBI gene ID: 3371); three major repair exonucleases (e.g., TREX1, TREX 2; NCBI Gene ID: 11277, 11219); thrombomodulin (THBD, CD 141; NCBI gene ID: 7056); thymidine kinases (e.g., TK1, TK 2; NCBI gene IDs: 7083, 7084); thymidine phosphorylase (TYMP; NCBI gene ID: 1890); thymidylate synthase (TYMS; NCBI Gene ID: 7298); thyroid hormone receptors (THRA, THRB; NCBI gene ID: 7606, 7608); thyroid stimulating hormone receptor (TSHR; NCBI gene ID: 7253); TNF superfamily members (e.g., TNFSF4(OX40L, CD252), TNFSF5(CD40L), TNFSF7(CD70), TNFSF8(CD153, CD30L), TNFSF9(4-1BB-L, CD137L), TNFSF10(TRAIL, CD253, APO2L), TNFSF11(CD254, RANKL2, TRANCE), TNFSF13(APRIL, CD256, TRAIL2), TNFSF13b (BAFF, BLYS, CD257), TNFSF14(CD258, LIGHT), TNFSF18 (GITRL); NCBI gene IDs: 944, 959, 970, 7292, 8600, 8740, 8741, 8743, 8744, 8995); toll-like receptors (e.g., TLR1(CD281), TLR2(CD282), TLR3(CD283), TLR4(CD284), TLR5, TLR6(CD286), TLR7, TLR8(CD288), TLR9(CD289), TLR10(CD 290); NCBI gene IDs: 7096, 7097, 7098, 7099, 10333, 51284, 51311, 54106, 81793); transferrin (TF; NCBI gene ID: 7018); transferrin receptor (TFRC, CD 71; NCBI gene ID: 7037); transforming growth factors (e.g., TGFA, TGFB 1; NCBI gene ID: 7039, 7040); transforming growth factor receptors (e.g., TGFBR1, TGFBR2, TGFBR 3; NCBI gene IDs: 7046, 7048, 7049); the transforming protein E7 (E7; NCBI gene ID: 1489079); transglutaminase 5(TGM 5; NCBI gene ID: 9333); transient receptor potential cation channel subfamily V member 1(TRPV1, VR 1; NCBI gene ID: 7442); transmembrane and immunoglobulin domain containing protein 2(TMIGD2, CD28H, IGPR 1; NCBI gene ID: 126259); trigger receptors expressed on bone marrow cells (e.g., TREM1(CD354), TREM 2; NCBI gene ID: 54209, 54210); tropinin (TRO, MAGED 3; NCBI Gene ID: 7216); trophoblast glycoprotein (TPBG; NCBI gene ID: 7162); tryptophan 2, 3-dioxygenase (TDO 2; NCBI gene ID: 6999); tryptophan hydroxylase (e.g., TPH1, TPH 2; NCBI gene ID: 7166, 121278); tumor associated calcium signal transducer 2 (TACTD 2, TROP 2; NCBI gene ID: 4070); tumor necrosis factor (TNF; NCBI gene ID: 7124); tumor Necrosis Factor (TNF) receptor superfamily members (e.g., TNFRSF 1(CD 120), TNFRSF (OX), TNFRSF (CD, FAS receptor), TNFRSF (CD137, 4-1BB), TNFRSF10 (CD261), TNFRSF10 (TRAIL, DR, CD262), TNFRSF10, TNFRSF11 (OPG), TNFRSF12, TNFRSF13, TNFRR 13 (CD268, BAFFR), TNFRSF (CD270, LIGHT, TNFRSF (CD269, BCMA), TNFRSF (GITR, CD357), TNFRSF 871BI gene IDs 355, 608, 9, 93943, 958, 4804, 7182, 7132, 7133, 498792, 8793, 878, 8793, 8795); tumor protein p53(TP 53; NCBI gene ID: 7157); tumor suppressor factor 2, mitochondrial calcium regulatory factor (TUSC 2; NCBI Gene ID: 11334); TYRO3 protein tyrosine kinase (TYRO 3; BYK; NCBI gene ID: 7301); tyrosinase (TYR; NCBI Gene ID: 7299); tyrosine hydroxylase (TH; NCBI gene ID: 7054); tyrosine kinase 1 with immunoglobulin-like and EGF-like domains (e.g., TIE1, TIE 1; NCBI Gene ID: 7075); tyrosine-protein phosphatase non-receptor type 11(PTPN11, SHP 2; NCBI gene ID: 5781); ubiquitin conjugating enzyme E2I (UBE2I, UBC 9; NCBI gene ID: 7329); ubiquitin C-terminal hydrolase L5(UCHL 5; NCBI Gene ID: 51377); ubiquitin-specific peptidase 7(USP 7; NCBI gene ID: 7874); ubiquitin-like modifier activating enzyme 1(UBA 1; NCBI gene ID: 7317); UL16 binding proteins (e.g., ULBP1, ULBP2, ULBP 3; NCBI gene ID: 79465, 80328, 80328); a valosin-containing protein (VCP, CDC 48; NCBI gene ID: 7415); vascular cell adhesion molecule 1(VCAM1, CD 106; NCBI gene ID: 7412); vascular endothelial growth factors (e.g., VEGFA, VEGFB; NCBI gene ID: 7422, 7423); vimentin (VIM; NCBI gene ID: 7431); vitamin D receptor (VDR; NCBI gene ID: 7421); v-set domain-containing T cell activation inhibitor 1(VTCN1, B7-H4; NCBI gene ID: 79679); v-set immunoregulatory receptor (VSIR, VISTA, B7-H5; NCBI Gene ID: 64115); WEE1G2 checkpoint kinase (WEE 1; NCBI Gene ID: 7465); WRN RecQ-like helicases (WRN; RECQ 3; NCBI gene ID: 7486); WT1 transcription factor (WT 1; NCBI gene ID: 7490); WW domain-containing transcription regulator 1(WWTR 1; TAZ; NCBI gene ID: 25937); X-C motif chemokine ligand 1(XCL1, ATAC; NCBI gene ID: 6375); X-C motif chemokine receptor 1(XCR1, GPR5, CCXCR 1; NCBI gene ID: 2829); yes 1-related transcriptional regulator (YAP 1; NCBI gene ID: 10413); zeta-chain-associated protein kinase 70(ZAP 70; NCBI gene ID: 7535).
In some embodiments, the one or more additional therapeutic agents include, for example, inhibitors or antagonists of: protein tyrosine phosphatase non-receptor type 11(PTPN11 or SHP 2; NCBI Gene ID: 5781); myeloid leukemia sequence 1(MCL1) apoptosis regulator (NCBI gene ID: 4170); mitogen-activated protein kinase 1(MAP4K1) (also known as hematopoietic progenitor kinase 1(HPK1), NCBI gene ID: 11184); phosphatidylinositol-4, 5-bisphosphate 3-kinase including catalytic subunit alpha (PIK3 CA; NCBI gene ID: 5290), catalytic subunit beta (PIK3 CB; NCBI gene ID: 5291), catalytic subunit gamma (PIK3 CG; NCBI gene ID: 5294) and catalytic subunit delta (PIK3 CD; NCBI gene ID: 5293), diacylglycerol kinase alpha ((DGKA, DACK 1 or DGK-alpha; NCBI gene ID: 1606), 5' -extracellular nucleotide enzyme (NT5E or CD 73; NCBI gene ID: 4907), extracellular nucleotide triphosphate bisphosphate hydrolase 1 (TPEND 1 or CD 39; NCBI gene ID: 593), transforming growth factor beta 1(TGFB1 or TGF beta; NCBI gene ID: 7040), heme oxygenase 1(HMOX1, HO-1 or 1; NCBI gene ID: 3162; NCBI gene ID: 312; VEGF 33: 312; VEGF 33: 33; VEGF 33: 7433; VEGF 33: IVBI gene ID: 7425; VEGF receptor II: 7425; VEGF receptor II: 3125; VEGF receptor II: 7425; VEGF receptor II: 3125; VEGF receptor II; VEGF receptor III; VEGF receptor II: 7425; VEGF receptor III; VEGF receptor II; VEII; VEI; VEII; VEI; VEII; VEI; VEII; VEI; VEII; VEI; VEII; VEIII; VEI; VEIII; VEI; VEIII; VEI; VEIII; VEI; VEIII; VEI; VEIII; VE, HER2, HER2/neu or CD 340; NCBI Gene ID: 2064) epidermal growth factor receptor (EGFR, ERBB1, or HER 1; NCBI Gene ID: 1956) (ii) a ALK receptor tyrosine kinase (ALK, CD 246; NCBI gene ID: 238); poly (ADP-ribose) polymerase 1(PARP 1; NCBI gene ID: 142); poly (ADP-ribose) polymerase 2(PARP 2; NCBI gene ID: 10038); TCDD inducible poly (ADP-ribose) polymerase (TIPARP, PARP 7; NCBI gene ID: 25976); cyclin-dependent kinase 4(CDK 4; NCBI gene ID: 1019); cyclin-dependent kinase 6(CDK 6; NCBI gene ID: 1021); TNF receptor superfamily member 14(TNFRSF14, HVEM, CD 270; NCBI gene ID: 8764); t cell immunoreceptors with Ig and ITIM domains (TIGIT; NCBI Gene ID: 201633); x-linked apoptosis inhibitors (XIAP, BIRC4, IAP-3; NCBI gene ID: 331); 2 containing baculovirus IAP repeats (BIRC2, cIAP 1; NCBI gene ID: 329); baculovirus IAP repeat containing 3(BIRC3, cIAP 2; NCBI gene ID: 330); 5 containing baculovirus IAP repeats (BIRC5, survivin; NCBI gene ID: 332); C-C motif chemokine receptor 2(CCR2, CD 192; NCBI gene ID: 729230); C-C motif chemokine receptor 5(CCR5, CD 195; NCBI gene ID: 1234); C-C motif chemokine receptor 8(CCR8, CDw 198; NCBI gene ID: 1237); C-X-C motif chemokine receptor 2((CXCR2, CD 182; NCBI gene ID: 3579); C-X-C motif chemokine receptor 3(CXCR3, CD182, CD 183; NCBI gene ID: 2833); C-X-C motif chemokine receptor 4((CXCR4, CD 184; NCBI gene ID: 7852); cytokine-inducible SH 2-containing protein (CISH; NCBI gene ID: 1154); arginase (ARG1(NCBI gene ID: 383), ARG2(NCBI gene ID: 384)), carbonic anhydrase (CA1(NCBI gene ID: 759), CA2(NCBI gene ID: 760), CA3(NCBI gene ID: 761), CA4(NCBI gene ID: 762), CA 967637 (NCBI gene ID: 3), CA5 CA B (NCBI gene ID: 11238), NCBI gene 6(NCBI gene ID: 768), NCBI gene ID 39768 (NCBI gene ID: 7638), NCBI gene ID 397626 (NCBI gene ID: 7626), CA 967626) (NCBI gene ID: 397626), CA 58ID: 3978: 596), and CA 967637 (NCBI gene ID: 596), CA11(NCBI gene ID: 770), CA12(NCBI gene ID: 771), CA13(NCBI gene ID: 377677), CA14(NCBI gene ID: 23632)), prostaglandin-endoperoxide synthase 1(PTGS1, COX-1; NCBI Gene ID: 5742) prostaglandin-endoperoxide synthase 2(PTGS2, COX-2; NCBI Gene ID: 5743) secreted phospholipase a2, prostaglandin E synthase (PTGES, PGES; gene ID: 9536) arachidonic acid 5-lipoxygenase (ALOX5, 5-LOX; NCBI Gene ID: 240) and/or soluble epoxide hydrolase 2(EPHX2, SEH; NCBI Gene ID: 2053) (ii) a Secreted phospholipase A2 (e.g., PLA2G1B (NCBI gene ID: 5319); PLA2G7(NCBI gene ID: 7941), PLA2G3(NCBI gene ID: 50487), PLA2G2A (NCBI gene ID: 5320); PLA2G4A (NCBI gene ID: 5321); PLA2G12A (NCBI gene ID: 81579); PLA2G12B (NCBI gene ID: 84647); PLA2G10(NCBI gene ID: 8399); PLA2G5(NCBI gene ID: 5322); PLA2G2D (NCBI gene ID: 26279); PLA2G15(NCBI gene ID: 23659)); indoleamine 2, 3-dioxygenase 1(IDO 1; NCBI gene ID: 3620); indoleamine 2, 3-dioxygenase 2(IDO 2; NCBI gene ID: 169355); hypoxia inducible factor 1 subunit alpha (HIF 1A; NCBI gene ID: 3091); angiopoietin 1(ANGPT 1; NCBI gene ID: 284); endothelial TEK tyrosine kinase (TIE-2, TEK, CD 202B; NCBI gene ID: 7010); janus kinase 1(JAK 1; NCBI gene ID: 3716); catenin beta 1(CTNNB 1; NCBI gene ID: 1499); histone deacetylase 9(HDAC 9; NCBI gene ID: 9734), 5'-3' exoribonuclease 1(XRN 1; NCBI gene ID: 54464), or WRN RecQ-like helicase (WRN; NCBI gene ID: 7486).
Exemplary mechanisms of action
Immune checkpoint modulators
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with one or more blockers or inhibitors of inhibitory immune checkpoint proteins or receptors and/or with one or more stimulators, activators or agonists of one or more stimulatory immune checkpoint proteins or receptors. Blockade or inhibition of inhibitory immune checkpoints can positively regulate T cell or NK cell activation and prevent immune escape of cancer cells within the tumor microenvironment. Activation or stimulation of a stimulatory immune checkpoint may enhance the effect of immune checkpoint inhibitors in the treatment of cancer. In some embodiments, the immune checkpoint protein or receptor modulates a T cell response (e.g., reviewed in Xu et al, J Exp Clin Cancer Res. (2018)37: 110). In some embodiments, the immune checkpoint protein or receptor modulates NK cell responses (e.g., reviewed in Davis et al, seminin Immunol. (2017)31: 64-75 and Chiossone et al, Nat Rev Immunol. (2018)18(11): 671-.
Examples of immune checkpoint proteins or receptors include CD27(NCBI gene ID: 939), CD70(NCBI gene ID: 970); CD40(NCBI gene ID: 958), CD40LG (NCBI gene ID: 959); CD47(NCBI gene ID: 961), SIRPA (NCBI gene ID: 140885); CD48(SLAMF 2; NCBI gene ID: 962), transmembrane and immunoglobulin domain containing 2(TMIGD2, CD 28H; NCBI gene ID: 126259), CD84(LY9B, SLAMF 5; NCBI gene ID: 8832), CD96(NCBI gene ID: 10225), CD160(NCBI gene ID: 11126), MS4A1(CD 20; NCBI gene ID: 931), CD244(SLAMF 4; NCBI gene ID: 51744); CD276(B7H 3; NCBI gene ID: 80381); v-set domain containing inhibitor of T cell activation 1(VTCN1, B7H 4); v-set immunoregulatory receptors (VSIR, B7H5, VISTA; NCBI gene ID: 64115); immunoglobulin superfamily member 11(IGSF11, VSIG 3; NCBI gene ID: 152404); natural killer cytotoxic receptor 3 ligand 1(NCR3LG1, B7H 6; NCBI gene ID: 374383); HERV-H LTR-related 2(HHLA2, B7H 7; NCBI gene ID: 11148); inducible T cell costimulators (ICOS, CD 278; NCBI gene ID: 29851); inducible T cell costimulator ligand (ICOSLG, B7H 2; NCBI gene ID: 23308); TNF receptor superfamily member 4(TNFRSF4, OX 40; NCBI gene ID: 7293); TNF superfamily member 4(TNFSF4, OX 40L; NCBI gene ID: 7292); TNFRSF8(CD 30; NCBI gene ID: 943), TNFSF8(CD 30L; NCBI gene ID: 944); TNFRSF10A (CD261, DR4, TRAILR 1; NCBI gene ID: 8797), TNFRSF9(CD 137; NCBI gene ID: 3604), TNFRSF9(CD 137L; NCBI gene ID: 8744); TNFRSF10B (CD262, DR5, TRAILR 2; NCBI gene ID: 8795), TNFRSF10 (TRAIL; NCBI gene ID: 8743); TNFRSF14(HVEM, CD 270; NCBI Gene ID: 8764), TNFSF14 (HVEML; NCBI Gene ID: 8740); CD272(B & T lymphocyte-associated ((BTLA); NCBI gene ID: 151888); TNFRSF17(BCMA, CD 269; NCBI gene ID: 608), TNFRSF 13B (BAFF; NCBI gene ID: 10673); TNFRSF18 (GITR; NCBI gene ID: 8784), TNFRSF18 (GITRL; NCBI gene ID: 8995); MHC class I polypeptide-related sequence A (MICA; NCBI gene ID: 100507436); MHC class I polypeptide-related sequence B (MICB; NCBI gene ID: 4277); CD274(CD 274), PDL1, PD-L1; NCBI gene ID: 29126); programmed cell death 1(PDCD1, PD1, PD-1; NCBI gene ID: 5133); cytotoxic T lymphocyte-related protein 4(CTLA 2, CD 152; PVR gene ID: 2913); CD80 (CD 3-1; NCBI gene ID: 941), NCBI gene ID: 73742; NCBI receptor adhesion molecule CD 226: 62; CD adhesional receptor CD 240; CD adhesional gene ID: 10642; CD 15; CD) and CD adhesional receptor molecule II: 226: 68519; CD adhesional receptor II CD 155; NCBI Gene ID: 5817) (ii) a Contains PVR-associated immunoglobulin domains (PVRIG, CD 112R; NCBI gene ID: 79037); t cell immunoreceptors with Ig and ITIM domains (TIGIT; NCBI Gene ID: 201633); t-cell immunoglobulin and mucin domain 4(TIMD 4; TIM 4; NCBI gene ID: 91937); hepatitis A virus cell receptor 2(HAVCR2, TIMD3, TIM 3; NCBI gene ID: 84868); galectin 9(LGALS 9; NCBI Gene ID: 3965); lymphocyte activation gene 3(LAG3, CD 223; NCBI gene ID: 3902); signaling lymphocyte activation molecule family member 1(SLAMF1, SLAM, CD 150; NCBI gene ID: 6504); lymphocyte antigen 9(LY9, CD229, SLAMF 3; NCBI gene ID: 4063); SLAM family member 6(SLAMF6, CD 352; NCBI gene ID: 114836); SLAM family member 7(SLAMF7, CD 319; NCBI gene ID: 57823); UL16 binding protein 1(ULBP 1; NCBI gene ID: 80329); UL16 binding protein 2(ULBP 2; NCBI gene ID: 80328); UL16 binding protein 3(ULBP 3; NCBI gene ID: 79465); retinoic acid early transcript 1E (RAET 1E; ULBP 4; NCBI gene ID: 135250); retinoic acid early transcript 1G (RAET 1G; ULBP 5; NCBI gene ID: 353091); retinoic acid early transcript 1L (RAET 1L; ULBP 6; NCBI gene ID: 154064); killer cell immunoglobulin-like receptor, three Ig domains and long cytoplasmic tail 1(KIR, CD158E 1; NCBI gene ID: 3811, e.g., Rireluzumab (IPH-2102, IPH-4102)); killer lectin-like receptor C1(KLRC1, NKG2A, CD 159A; NCBI gene ID: 3821); killer lectin-like receptor K1(KLRK1, NKG2D, CD 314; NCBI gene ID: 22914); killer lectin-like receptor C2(KLRC2, CD159C, NKG 2C; NCBI gene ID: 3822); killer lectin-like receptor C3(KLRC3, NKG 2E; NCBI gene ID: 3823); killer lectin-like receptor C4(KLRC4, NKG 2F; NCBI gene ID: 8302); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 1(KIR2DL 1; NCBI gene ID: 3802); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 2(KIR2DL 2; NCBI gene ID: 3803); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 3(KIR2DL 3; NCBI gene ID: 3804); killer cell immunoglobulin-like receptor, three Ig domains and long cytoplasmic tail 1(KIR3DL 1); killer lectin-like receptor D1(KLRD 1; NCBI gene ID: 3824); killer lectin-like receptor G1(KLRG 1; CLEC15A, MAFA, 2F 1; NCBI gene ID: 10219); sialic acid binds to Ig-like lectin 7(SIGLEC 7; NCBI gene ID: 27036); and sialic acid binds to Ig-like lectin 9(SIGLEC 9; NCBI gene ID: 27180).
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with one or more blockers or inhibitors of one or more T cell inhibitory immune checkpoint proteins or receptors. Exemplary T cell inhibitory immune checkpoint proteins or receptors include CD274(CD274, PDL1, PD-L1); programmed cell death 1 ligand 2(PDCD1LG2, PD-L2, CD 273); programmed cell death 1(PDCD1, PD1, PD-1); cytotoxic T lymphocyte-associated protein 4(CTLA4, CD 152); CD276(B7H 3); v-set domain containing inhibitor of T cell activation 1(VTCN1, B7H 4); v-set immunoregulatory receptor (VSIR, B7H5, VISTA); immunoglobulin superfamily member 11(IGSF11, VSIG 3); TNFRSF14(HVEM, CD270), TNFSF14 (HVEML); CD272(B and T lymphocyte-associated (BTLA)); contains a PVR-associated immunoglobulin domain (PVRIG, CD 112R); t cell immunoreceptors with Ig and ITIM domains (TIGIT); lymphocyte activation 3(LAG3, CD 223); hepatitis a virus cell receptor 2(HAVCR2, TIMD3, TIM 3); galectin 9(LGALS 9); killer cell immunoglobulin-like receptor, three Ig domains and long cytoplasmic tail 1(KIR, CD158E 1); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 1(KIR2DL 1); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 2(KIR2DL 2); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 3(KIR2DL 3); and killer cell immunoglobulin-like receptor, three Ig domains and long cytoplasmic tail 1(KIR3DL 1). In some embodiments, the antibodies and/or fusion proteins provided herein are administered with one or more agonists or activators of one or more T cell stimulatory immune checkpoint proteins or receptors. Exemplary T cell stimulatory immune checkpoint proteins or receptors include, but are not limited to, CD27, CD 70; CD40, CD40 LG; inducible T cell co-stimulatory factors (ICOS, CD 278); inducible T cell costimulator ligand (ICOSLG, B7H 2); TNF receptor superfamily member 4(TNFRSF4, OX 40); TNF superfamily member 4(TNFSF4, OX 40L); TNFRSF9(CD137), TNFSF9(CD 137L); TNFRSF18(GITR), TNFSF18 (GITRL); CD80(B7-1), CD 28; NECTIN cell adhesion molecule 2(NECTIN2, CD 112); CD226 (DNAM-1); CD244(2B4, SLAMF4), poliovirus receptor (PVR) cell adhesion molecules (PVR, CD 155). See, e.g., Xu et al, J Exp Clin Cancer Res. (2018)37: 110.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with one or more blockers or inhibitors of one or more NK cell inhibitory immune checkpoint proteins or receptors. Exemplary NK cell inhibitory immune checkpoint proteins or receptors include killer cell immunoglobulin-like receptor, three Ig domains and long cytoplasmic tail 1(KIR, CD158E 1); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 1(KIR2DL 1); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 2(KIR2DL 2); killer cell immunoglobulin-like receptor, two Ig domains and long cytoplasmic tail 3(KIR2DL 3); killer cell immunoglobulin-like receptor, three Ig domains and long cytoplasmic tail 1(KIR3DL 1); killer lectin-like receptor C1(KLRC1, NKG2A, CD 159A); killer cell lectin-like receptor D1(KLRD1, CD 94); killer cell lectin-like receptor G1(KLRG 1; CLEC15A, MAFA, 2F 1); sialic acid binds to Ig-like lectin 7(SIGLEC 7); and sialic acid binds to Ig-like lectin 9(SIGLEC 9). In some embodiments, the antibodies and/or fusion proteins provided herein are administered with one or more agonists or activators of one or more NK cell-stimulating immune checkpoint proteins or receptors. Exemplary NK cell-stimulating immune checkpoint proteins or receptors include CD16, CD226 (DNAM-1); CD244(2B4, SLAMF 4); killer lectin-like receptor K1(KLRK1, NKG2D, CD 314); SLAM family member 7(SLAMF 7). See, e.g., Davis et al, Semin Immunol. (2017)31: 64-75; fang et al, Semin Immunol, (2017)31: 37-54; and Chiossone et al, Nat Rev Immunol (2018)18(11) 671-688.
In some embodiments, the one or more immune checkpoint inhibitors comprise a protein (e.g., an antibody or fragment thereof or antibody mimetic) inhibitor of PD-L1(CD274), PD-1(PDCD1), CTLA4, or TIGIT. In some embodiments, the one or more immune checkpoint inhibitors comprise small organic molecule inhibitors of PD-L1(CD274), PD-1(PDCD1), CTLA4, or TIGIT.
Examples of CTLA4 inhibitors that may be co-administered include ipilimumab, tremelimumab, BMS-986218, AGEN1181, AGEN1884 (zeolizumab), BMS-986249, MK-1308, REGN-4659, ADU-1604, CS-1002, BCD-145, APL-509, JS-007, BA-3071, ONC-392, AGEN-2041, JHL-1155, KN-044, CG-0161, ATOR-1144, PBI-5D3H5, BPI-002, and the multispecific inhibitor FPT-155(CTLA4/PD-L1/CD28), PF-06936308(PD-1/CTLA4), MGD-019(PD-1/CTLA4), KN-046(PD-1/CTLA4), MEDI-5752(CTLA4/PD-1), XmAb-20717(PD-1/CTLA4) and AK-104(CTLA 4/PD-1).
Examples of inhibitors of PD-L1(CD274) or PD-1(PDCD1) that may be co-administered include palivizumab, nivolumab, cimaprimab, pidilizumab, AMP-224, MEDI0680(AMP-514), sibatuzumab, atizumab tiuxelizumab, avilumab, Duvaliuzumab, BMS-936559, CK-301, PF-06801591, BGB-A317 (tirezumab), GLS-010(WBP-3055), JNS-103 (HX-008), AK-105, CS-1003, HLX-10, MGA-012, BI-754091, AGEN-2034, JS-001 (terlizumab), JNP-63723283, Jennuzumab (CBT-501), LZM-009, BCD-100, Rayley-3300054, SHR-1201, SHR-1210, Rayleigh-1210, Sym-021, ABBV-181, PD1-PIK, BAT-1306(MSB0010718C), CX-072, CBT-502, TSR-042 (Dutalimumab), MSB-2311, JTX-4014, BGB-A333, SHR-1316, CS-1001(WBP-3155, KN-035, IBI-308 (Cedilitumumab), HLX-20, KL-A167, STI-A1014, STI-A1015(IMC-001), BCD-135, FAZ-053, TQB-2450, MDX1105-01, GS-4224, GS-4416, INCB086550, 101MAX 81, AGEN2034 (Batulimumab), Sepalivizumab, and bispecific inhibitor T-155(CTLA 4/35L 1/CD28), PF-06936308 (MGB-1/4), PD (CTLA-1-3/013), FS-118(LAG-3/PD-L1) MGD-019(PD-1/CTLA4), KN-046(PD-1/CTLA4), MEDI-5752(CTLA4/PD-1), RO-7121661(PD-1/TIM-3), XmAb-20717(PD-1/CTLA4), AK-104(CTLA4/PD-1), M7824(PD-L1/TGF β -EC domain), CA-170(PD-L1/VISTA), CDX-527(CD27/PD-L1), LY-3415244 (PDL 3/PDL1) and INBRX-105(4-1BB/PDL 1).
Examples of TIGIT inhibitors that may be co-administered include tegralitumumab (RG-6058), Venbrizumab, Dunalizumab, AB308, Dunalizumab (AB154), AB308, BMS-986207, AGEN-1307, COM-902, or eptilibab (etilizab).
TNF receptor superfamily (TNFRSF) member agonists or activators
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an agonist of one or more TNF receptor superfamily (TNFRSF) members, such as an agonist of one or more of: TNFRSF1A (NCBI gene ID: 7132), TNFRSF1 (NCBI gene ID: 7133), TNFRSF4(OX40, CD 134; NCBI gene ID: 7293), TNFRSF 40 (CD 40; NCBI gene ID: 958), TNFRSF 40 (FAS, NCBI gene ID: 355), TNFRSF 40 (CD40, NCBI gene ID: 939), TNFRSF 40 (CD40, NCBI gene ID: 943), TNFRSF 40 (4-1BB, CD137, NCBI gene ID: 3604), TNFRSF10 40 (CD261, DR 40, TRAILR 40, NCBI gene ID: 8797), TNFRSF10 40 (CD262, DR 40, TRAILR 40, NCBI gene ID: 8795), TNFRSF10 40 (CD263, TRAILR 36264, NCBI gene ID: 8794), TNFRSF 8772 (CD 8772, NCFRSF 8772, NCFR 8772, TNFRSF 8772, TNFRID 40, TNFRID 3673, TNFRID 3678, TNFRID 3673, TNFRSF 8795, TNFRSF 8772, TNFRID 40, TNFRID 3678, TNFRID 8795, TNFRID 3678, TNFRID 8795, TNFRID 8772, TNFRID 3678, TNFRID 8795, TNFRID 8772, TNFRID 3678, TNFRID 3678, TNFRID 8772, TNFRID 8772, TNFRID 3678, TNFRID 8772, TNFRID 3678, TNFRID 8772, TNFRID 3678, TNFRID 8772, TNFRID 40, TNFRID 8772, TNFRID 40, TNFRID 8772, TNFRID 3677, TNFRID 8772, TNFRID 3677, TNFRID 8772, TNFRID 8772, TNFRID 8772, TNFRSF19(NCBI gene ID: 55504), TNFRSF21(CD358, DR6, NCBI gene ID: 27242) and TNFRSF25(DR3, NCBI gene ID: 8718).
Exemplary anti-TNFRSF 4(OX40) antibodies that may be co-administered include MEDI6469, MEDI6383, MEDI0562 (tavorlizumab), MOXR0916, PF-04518600, RG-7888, GSK-3174998, incag 1949, BMS-986178, GBR-8383, ABBV-368, and those described in WO2016179517, WO2017096179, WO2017096182, WO2017096281, and WO 2018089628.
Exemplary anti-TNFRSF 5(CD40) antibodies that may be co-administered include RG7876, SEA-CD40, APX-005M, and ABBV-428.
In some embodiments, the anti-TNFRSF 7(CD27) antibody, valrubizumab (CDX-1127), is co-administered.
Exemplary anti-TNFRSF 9(4-1BB, CD137) antibodies that may be co-administered include Uluzumab, Utuzumab (PF-05082566), AGEN2373, and ADG-106.
In some embodiments, an anti-TNFRSF 17(BCMA) antibody GSK-2857916 is co-administered.
Exemplary anti-TNFRSF 18(GITR) antibodies that can be co-administered include MEDI1873, FPA-154, incag-1876, TRX-518, BMS-986156, MK-1248, GWN-323, and those described in WO2017096179, WO2017096276, WO2017096189, and WO 2018089628. In some embodiments, an antibody or fragment thereof that co-targets TNFRSF4(OX40) and TNFRSF18(GITR) is co-administered. Such antibodies are described, for example, in WO2017096179 and WO 2018089628.
Bispecific antibodies targeting TNFRSF family members that may be co-administered include PRS-343(CD-137/HER2), AFM26(BCMA/CD16A), AFM-13(CD16/CD30), REGN-1979(CD20/CD3), AMG-420(BCMA/CD3), INHIBRX-105(4-1BB/PDL1), FAP-4-IBBL (4-1BB/FAP), XBAM-13676 (CD3/CD20), RG-7828(CD20/CD3), CC-93269(CD3/BCMA), REGN-5458(CD3/BCMA), and IMM-0306(CD47/CD 20).
Bispecific T cell engagers
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with a bispecific T cell engager (e.g., without Fc) or an anti-CD 3 bispecific antibody (e.g., with Fc). Exemplary anti-CD 3 bispecific antibodies or BiTEs that may be co-administered include JNJ-64052781(CD19/CD3), AMG-211(CEA/CD3), RG7802(CEA/CD3), ERY-974(CD3/GPC3), PF-3 (cadherin/CD 3), APVO436(CD123/CD3), fluorotuzumab (CD123/CD3), REGN-1979(CD 3/CD3), MCLA-117(CD3/CLEC12 3), JNJ-0819, JNJ-7564(CD3/heme), AMG-757(DLL3-CD3), AMG-330(CD 3/CD3), AMG-420(BCMA/CD3), JNJ-3 (CD123/CD3), MGD-36007 (CD 3/gpA/g3672), MGCD 14072/GmG-3 (CD3/CD 3), CG3672/CD 3), CGI 3/3, CD 3(CD 3/CGI 3/3), CGI 3, CD3/CD3, and CD 3/CGI-3 (CD 3/CGI-3/mGlu-3, CD3/CD3, and mAb-3, etc, XmAb-18087(SSTR2/CD3), Katuzumab (CD3/EpCAM), REGN-4018(MUC16/CD3), RG-7828(CD20/CD3), CC-93269(CD3/BCMA), REGN-5458(CD3/BCMA), GRB-1302(CD3/Erbb2), GRB-1342(CD38/CD3), GEM-333(CD3/CD 33). The anti-CD 3 binding bispecific molecule may or may not have an Fc as appropriate. Exemplary bispecific T cell adaptors that can be co-administered target CD3 and tumor associated antigens as described herein, including, for example, CD19 (e.g., bornauzumab); CD33 (e.g., AMG 330); CEA (e.g., MEDI-565); receptor tyrosine kinase-like orphan receptor 1(ROR1) (Gohil et al, Oncoimmunology.2017, 5 months and 17 days; 6(7): e 1326437); PD-L1(Horn et al, Oncotarget.2017, 8/3; 8(35): 57964-; and EGFRvIII (Yang et al, Cancer Lett.2017, 9, 10; 403: 224-.
Bi-and tri-specific Natural Killer (NK) cell adapters
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with a bispecific NK cell engager (BiKE) or trispecific NK cell engager (TriKE) (e.g., without Fc) or a bispecific antibody (e.g., with Fc) against: NK cell activating receptors such as CD16A, C-type lectin receptors (CD94/NKG2C, NKG2D, NKG2E/H and NKG2F), natural cytotoxic receptors (NKp30, NKp44 and NKp46), killer cell C-type lectin-like receptors (NKp65, NKp80), Fc receptor Fc γ R (which mediates antibody-dependent cytotoxicity), SLAM family receptors (e.g., 2B4, SLAM6 and SLAM7), killer cell immunoglobulin-like receptors (KIR) (KIR-2DS and KIR-3DS), DNAM-1 and CD137(41 BB). Exemplary anti-CD 16 bispecific antibodies, BiKE or TriKE that can be co-administered include AFM26(BCMA/CD16A) and AFM-13(CD16/CD 30). As the case may be, the anti-CD 16 binding bispecific molecule may or may not have an Fc. Exemplary bispecific NK cell adaptors that may be co-administered target CD16 and one or more tumor associated antigens as described herein, including, for example, CD19, CD20, CD22, CD30, CD33, CD123, EGFR, EpCAM, ganglioside GD2, HER2/neu, HLA class II, and FOLR 1. BiKE and TriKE are described, for example, in the following documents: felices et al, Methods Mol Biol. (2016)1441: 333-; fang et al, Semin Immunol, (2017)31: 37-54.
MCL1 apoptosis regulator, BCL2 family member (MCL1) inhibitor
In some embodiments, the antibodies and/or fusion proteins provided herein are administered with an inhibitor of: MCL1 apoptosis regulator, BCL2 family member (MCL1, TM; EAT; MCL 1L; MCL 1S; Mcl-1; BCL2L 3; MCL 1-ES; BCL 2-L-3; MCL 1/EAT; NCBI gene ID: 4170). Examples of MCL1 inhibitors include AMG-176, AMG-397, S-64315, AZD-5991, 483-LM, A-1210477, UMI-77, JKY-5-037, GS-9716, and those described in WO2018183418, WO2016033486, and WO 2017147410.
SHP2 inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an inhibitor of: protein tyrosine phosphatase non-receptor type 11(PTPN 11; BPTP3, CFC, JMML, METCDS, NS1, PTP-1D, PTP2C, SH-PTP2, SH-PTP3, SHP 2; NCBI gene ID: 5781). Examples of SHP2 inhibitors include TNO155(SHP-099), RMC-4550, JAB-3068, RMC-4630, and those described in WO2018172984 and WO 2017211303.
Inhibitors of hematopoietic progenitor kinase 1(HPK1)
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: mitogen-activated protein kinase 1(MAP4K1, HPK 1; NCBI gene ID: 11184). Examples of hematopoietic progenitor kinase 1(HPK1) inhibitors include, but are not limited to, those described in WO2020092621, WO2018183956, WO2018183964, WO2018167147, WO2018183964, WO2016205942, WO2018049214, WO2018049200, WO2018049191, WO2018102366, WO2018049152, and WO 2016090300.
Apoptosis signal-regulating kinase (ASK) inhibitors
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: ASK inhibitors (e.g., mitogen-activated protein kinase 5(MAP3K 5; ASK1, MAPKKKK 5, MEKK 5; NCBI gene ID: 4217.) examples of ASK1 inhibitors include those described in WO2011008709(Gilead Sciences) and WO 2013112741(Gilead Sciences).
Bruton's Tyrosine Kinase (BTK) inhibitors
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: bruton's tyrosine kinase (BTK, AGMX1, AT, ATK, BPK, IGHD3, IMD1, PSCTK1, XLA; NCBI Gene ID: 695). Examples of BTK inhibitors include (S) -6-amino-9- (1- (but-2-ynoyl) pyrrolidin-3-yl) -7- (4-phenoxyphenyl) -7H-purin-8 (9H) -one, acartib (acallabrutinib) (ACP-196), BGB-3111, CB988, HM71224, ibrutinib (ibrutinib), M-2951 (ibrutinib), M7583, tirarotinib (tirarobrutinib) (ONO-4059), PRN-1008, serpentinib (spertitinib) (CC-292), TAK-020, vecarbutinib (vecabrutinib), ARQ-531, SHR-1459, DTMWXHS-12, and TAS-5315.
Cluster of differentiation 47(CD47) inhibitors
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: CD47(IAP, MER6, OA 3; NCBI gene ID: 961). Examples of CD47 inhibitors include anti-CD 47mAb (Vx-1004), anti-human CD47mAb (CNTO-7108), CC-90002-ST-001, humanized anti-CD 47 antibody or CD47 blocker (Hu5F9-G4), NI-1701, NI-1801, RCT-1938 and TTI-621. In some embodiments, the CD47 inhibitor is molorezumab.
SIRP alpha targeting agents
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with: SIRPαTargeting agent (NCBI Gene ID: 140885; UniProt P78324) combinations. SIRPαExamples of targeting agents include SIRPαInhibitors (such as AL-008, RRx-001, and CTX-5861) and anti-SIRPαAntibodies (such as FSI-189(GS-0189), ES-004, BI765063, ADU1805 and CC-95251). Additional sirpa targeting agents used are described, for example, in the following documents: WO200140307, WO2002092784, WO2007133811, WO2009046541, WO2010083253, WO2011076781, WO2013056352, WO2015138600, WO2016179399, WO2016205042, WO2017178653, WO2018026600, WO2018057669, WO2018107058, WO2018190719, WO2018210793, WO2019023347, WO2019042470, WO2019175218, WO2019183266, WO2020013170 and WO 2020068752.
Cyclin Dependent Kinase (CDK) inhibitors
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: cyclin-dependent kinase 1(CDK1, CDC 2; CDC 28A; P34CDC 2; NCBI gene ID: 983); cyclin-dependent kinase 2(CDK2, CDKN 2; p33(CDK 2); NCBI gene ID: 1017); cyclin-dependent kinase 3(CDK 3; NCBI gene ID: 1018); cyclin-dependent kinase 4(CDK4, CMM 3; PSK-J3; NCBI gene ID: 1019); cyclin-dependent kinase 6(CDK6, MCPH 12; PLSTIRE; NCBI gene ID: 1021); cyclin-dependent kinase 7(CDK7, CAK; CAK 1; HCAK; MO 15; STK 1; CDKN 7; p39MO 15; NCBI gene ID: 1022) or cyclin-dependent kinase 9(CDK9, TAK; C-2 k; CTK 1; CDC2L 4; PITALRE; NCBI gene ID: 1025). Inhibitors of CDK1, 2, 3, 4, 6, 7 and/or 9 include Abelici (abemacicib), Avocidib (alvocidib) (HMR-1275, flaperot (flavopiridol)), AT-7519, dinaciclib (dinaciclib), Ebosebin (ibance), FLX-925, LEE001, Palbociclib (palbociclib), Ribosicib (ribociclib), Rigortib (rigosenib), selinex, UCN-01, SY1365, CT-7001, SY-1365, G1T38, Millicib (milciib), Trilaciclib (trilicib) and TG-02.
Discotic Domain Receptor (DDR) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are combined with inhibitors of the following kinases: discotic domain receptor tyrosine kinase 1(DDR1, CAK, CD167, DDR, EDDR1, HGK2, MCK10, NEP, NTRK4, PTK3, PTK3A, RTK6, TRKE; NCBI gene ID: 780); and/or the discotic domain receptor tyrosine kinase 2(DDR2, MIG20a, NTRKR3, TKT, TYRO10, WRCN; NCBI Gene ID: 4921). Examples of DDR inhibitors include dasatinib (dasatinib) and those disclosed in WO2014/047624(Gilead Sciences), US 2009-0142345(Takeda Pharmaceutical), US 2011-0287011 (oncommune Pharmaceuticals), WO2013/027802(Chugai Pharmaceutical) and WO2013/034933(Imperial Innovations).
Targeting E3 ligase ligand conjugates
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with a targeting E3 ligase ligand conjugate. Such conjugates have a target protein binding moiety and an E3 ligase binding moiety (e.g., Inhibitors of Apoptosis Proteins (IAPs) (e.g., XIAP, c-IAP1, c-IAP2, NIL-IAP, Bruce, and survivin) E3 ubiquitin ligase binding moiety, Von Hippel-Lindau E3 ubiquitin ligase (VHL) binding moiety, cereblon E3 ubiquitin ligase binding moiety, mouse two minute 2 homolog (MDM2) E3 ubiquitin ligase binding moiety), and can be used to promote or increase degradation of targeted proteins, e.g., via the ubiquitin pathway. In some embodiments, the targeted E3 ligase ligand conjugate comprises: a targeting or binding moiety that targets or binds to a protein described herein, and an E3 ligase ligand or binding moiety. In some embodiments, the targeted E3 ligase ligand conjugate comprises a targeting or binding moiety that targets or binds a protein selected from the group consisting of: cbl protooncogene B (CBLB; Cbl-B, Nbl 00127, RNF 56; NCBI gene ID: 868) and hypoxia inducible factor 1 subunit alpha (HIF 1A; NCBI gene ID: 3091). In some embodiments, the targeted E3 ligase ligand conjugate comprises a kinase inhibitor (e.g., a small molecule kinase inhibitor, such as a small molecule kinase inhibitor of BTK and E3 ligase ligands or binding moieties). See, for example, WO 2018098280. In some embodiments, the targeted E3 ligase ligand conjugate comprises: targeting or binding to interleukin-1 (IL-1) receptor associated kinase-4 (IRAK-4); a binding moiety of rapid-accelerating fibrosarcoma (RAF, such as c-RAF, A-RAF and/or B-RAF), c-Met/p38 or BRD protein; and an E3 ligase ligand or binding moiety. See, e.g., WO2019099926, WO2018226542, WO2018119448, WO2018223909, WO 2019079701. Further targeting E3 ligase ligand conjugates that may be co-administered are described for example in WO2018237026, WO2019084026, WO2019084030, WO2019067733, WO2019043217, WO2019043208 and WO 2018144649.
Histone Deacetylase (HDAC) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an inhibitor of a histone deacetylase, such as histone deacetylase 9(HDAC9, HD7, HD7B, HD9, HDAC7, HDAC7B, HDAC9B, HDAC9FL, HDRP, MITR; gene ID: 9734). Examples of HDAC inhibitors include abebestat (abexinostat), ACY-241, AR-42, BEBT-908, belinostat (belinostat), CKD-581, CS-055(HBI-8000), CUDC-907 (non-minostat), entinostat (entinostat), gibvista (givinostat), moxystat (mocetinostat), panobinostat (panobinostat), pruinostat (pracinostat), quisinostat (JNJ-26481585), remimitat, ricolinostat (ricolinostat), SHP-141, valproic acid (VAL-001), vorinostat, moxystine (ostastin), rementintine (rementostatin), rementinostat (rementinostat), and rementinostat (rementinostat).
Indoleamine-pyrrole-2, 3-dioxygenase (IDO1) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein are administered with an inhibitor of: indoleamine 2, 3-dioxygenase 1(IDO 1; NCBI gene ID: 3620). Examples of IDO1 inhibitors include BLV-0801, indole stat (epacadostat), F-001287, GBV-1012, GBV-1028, GDC-0919, indomod (indoximod), NKTR-218, NLG-919 based vaccines, PF-06840003, naphthopyranone derivatives (SN-35837), reminostat (reminiostat), SBLK-200802, BMS-986205 and shIDO-ST, EOS-200271, KHK-2455 and LY-3381916.
Janus kinase (JAK) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an inhibitor of the following kinases: janus kinase 1(JAK1, JAK1A, JAK1B, JTK 3; NCBI gene ID: 3716); janus kinase 2(JAK2, JTK10, THCYT 3; NCBI gene ID: 3717); and/or Janus kinase 3(JAK3, JAK-3, JAK3_ HUMAN, JAKL, L-JAK, LJAK; NCBI gene ID: 3718). Examples of JAK inhibitors include AT9283, AZD1480, baricitinib (baricitinib), BMS-911543, phenanthratinib (fedratinib), nonglutinib (filgoninib) (GLPG0634), gancitinib (gandottinib) (LY2784544), INCB039110 (itatinib), lestatinib (lestaurtinib), mometinib (momelotinib) (CYT0387), NS-018, pactinib (pactinib) (SB1518), pefinitib (pefinitinib) (ASP015K), ruxotinib (ruxotinib), tofacitinib (tofacitinib) (formerly tasocitinib 052793 and XL 019.
Inhibitors of lysyl oxidase-like protein (LOXL)
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: LOXL proteins, for example LOXL1(NCBI gene ID: 4016), LOXL2(NCBI gene ID: 4017), LOXL3(NCBI gene ID: 84695), LOXL4(NCBI gene ID: 84171) and/or LOX (NCBI gene ID: 4015). Examples of LOXL2 inhibitors include antibodies described in WO 2009017833(Arresto Biosciences), WO 2009035791(Arresto Biosciences) and WO 2011097513(Gilead Biologics).
Matrix Metalloproteinase (MMP) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an inhibitor of Matrix Metalloproteases (MMPs), such as an inhibitor of: MMP1(NCBI gene ID: 4312), MMP2(NCBI gene ID: 4313), MMP3(NCBI gene ID: 4314), MMP7(NCBI gene ID: 4316), MMP8(NCBI gene ID: 4317), MMP9(NCBI gene ID: 4318), MMP10(NCBI gene ID: 4319), MMP11(NCBI gene ID: 4320); MMP12(NCBI gene ID: 4321), MMP13(NCBI gene ID: 4322), MMP14(NCBI gene ID: 4323), MMP15(NCBI gene ID: 4324), MMP16(NCBI gene ID: 4325), MMP17(NCBI gene ID: 4326), MMP19(NCBI gene ID: 4327), MMP20(NCBI gene ID: 9313), MMP21(NCBI gene ID: 118856), MMP24(NCBI gene ID: 10893), MMP25(NCBI gene ID: 64386), MMP26(NCBI gene ID: 56547), MMP27(NCBI gene ID: 64066), and/or MMP28(NCBI gene ID: 79148). Examples of MMP9 inhibitors include marimastat (BB-2516), cimastat (Ro 32-3555), GS-5745 (Andecaliximab), and those described in WO 2012027721(Gilead Biologics).
RAS and RAS pathway inhibitors
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: KRAS proto-oncogene GTPase (KRAS; also known as NS; NS 3; CFC 2; RALD; K-Ras; KRAS 1; KRAS 2; RASK 2; KI-RAS; C-K-RAS; K-RAS 2A; K-RAS 2B; K-RAS 4A; K-RAS 4B; C-Ki-Ras 2; NCBI gene ID: 3845); NRAS protooncogene GTPase (NRAS; also known as NS 6; CMNS; NCMS; ALPS 4; N-Ras; NRAS 1; NCBI gene ID: 4893) or HRAS protooncogene GTPase (HRAS; also known as CTLO; KRAS; HAMSV; HRAS 1; KRAS 2; RASH 1; RASK 2; Ki-Ras; p21 Ras; C-H-RAS; C-K-Ras; H-RASIDX; C-Ki-Ras; C-BAS/HAS; C-HA-RAS 1; NCBI gene ID: 3265). Ras inhibitors can inhibit Ras at the polynucleotide (e.g., transcription inhibitors) or polypeptide (e.g., gtpase inhibitors) level. In some embodiments, the inhibitor targets one or more proteins in the Ras pathway, e.g., inhibits one or more of EGFR, Ras, Raf (A-Raf, B-Raf, C-Raf), MEK (MEK1, MEK2), ERK, PI3K, AKT, and mTOR. Exemplary K-Ras inhibitors that may be co-administered include ARS-1620(G12C), SML-8-73-1(G12C), Compound 3144(G12D), Kobe0065/2602(Ras GTP), RT11, MRTX-849(G12C), and K-Ras (G12D) -Selective inhibitory peptides, including KRpep-2 (Ac-RRCPLYISYDPVCRR-NH) 2 ) (SEQ ID NO:126) and KRpep-2d (Ac-RRRRCPLYISYDPVCRRRR-NH) 2 ) (SEQ ID NO: 127). Exemplary KRAS mRNA inhibitors include anti-KRAS U1 aptamer, AZD-4785, siG12D-LODER TM And siG12D exosomes. Exemplary MEK inhibitors that may be co-administered include bimetinib (binimetinib), cobimetinib (cobimetinib), PD-0325901, pimatinib (pimasetib), RG-7304, semetinib (selumetinib), trimetinib, and those described below and herein. Exemplary Raf dimer inhibitors that can be co-administered include BGB-283, HM-95573, LXH-254, LY-3009120, RG7304, andTAK-580. Exemplary ERK inhibitors that may be co-administered include LTT-462, LY-3214996, MK-8353, lavendib (ravoxertinib), and ulitinib (ulixertiib). Exemplary Ras gtpase inhibitors that can be co-administered include agoutib. Exemplary PI3K inhibitors that can be co-administered include idelixibAlbuliform, bupiriform, and pirtilib (pictilisib). Exemplary PI3K/mTOR inhibitors that may be co-administered include daptomiflozin, omipacib, and voraxiflozin. In some embodiments, Ras-driven cancers (e.g., NSCLC) with a CDKN2A mutation can be inhibited by co-administration of the MEK inhibitor semetinib and the CDK4/6 inhibitor palbociclib. See, e.g., Zhou et al, Cancer lett.2017, 11/1; 408:130-137. Furthermore, K-RAS and mutant N-RAS can be reduced by the irreversible ERBB1/2/4 inhibitor neratinib. See, e.g., Booth et al, Cancer Biol ther.2018, 2 months and 1 day; 19(2):132-137.
Mitogen-activated protein kinase (MEK) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an inhibitor of: mitogen-activated protein kinase 7(MAP2K7, JNKK2, MAPKK7, MEK 7, MKK7, PRKMK7, SAPKK-4, SAPKK 4; NCBI gene ID: 5609). Examples of MEK inhibitors include antroquinanol (antroquinanol), bimetinib, cobicisib (GDC-0973, XL-518), MT-144, semetinib (AZD6244), sorafenib (sorafenib), trametinib (GSK 0211122), Yopopertib (uplasertib) + trametinib, PD-0325901, pimatinib, LTT462, AS703988, CC-90003, and refametinib (refametinib).
Phosphatidylinositol 3-kinase (PI3K) inhibitors
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an inhibitor of: phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit, exampleSuch as phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA, CLAPO, CLOVE, CWS5, MCAP, MCM, MCMTC, PI3K, PI 3K-alpha, p 110-alpha; NCBI gene ID: 5290); phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit BETA (PIK3CB, P110BETA, PI3K, PI3KBETA, PIK3C 1; NCBI Gene ID: 5291); phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit gamma (PIK3CG, PI3CG, PI3K, PI3K gamma, PIK3, p110 gamma, p120-PI 3K; gene ID 5494); and/or phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit DELTA (PIK3CD, APDS, IMD14, P110DELTA, PI3K, P110D, NCBI Gene ID: 5293). In some embodiments, the PI3K inhibitor is a pan PI3K inhibitor. Examples of PI3K inhibitors include ACP-319, AEZA-129, AMG-319, AS252424, AZD8186, BAY 10824391, BEZ235, bupalicet (BKM120), BYL719 (Abolisibu), CH5132799, copericide (BAY 80-6946), Duvirassib, GDC-0032, GDC-0077, GDC-0941, GDC-0980, GSK2636771, GSK2269557, Adelisibu INCB50465, IPI-145, IPI-443, IPI-549, KAR4141, LY294002, LY3023414, MLN1117, OXY111A, PA799, PX-866, RG7604, reglitinib, RP5090, RP6530, SRX3177, Tacelecoxib, TG100115, TGR-1202 (embralsib)), TGX221, WX-037, X-339, X-414, XL147(SAR 2454408), XL499, XL756, wortmannin, ZSTK 311474, and the compounds described in WO2005113556(ICOS), WO 2013/052699(Gilead Calistoga), WO 2016562 (Gilead Calistoga), WO 100765(Gilead Calistoga), WO 100767(Gilead Calistoga) and WO 100201409 (Gilecesa).
Spleen tyrosine kinase (SYK) inhibitors
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an inhibitor of: spleen-related tyrosine kinase (SYK, p72-Syk, Gene ID: 6850). Examples of SYK inhibitors include 6- (1H-indazol-6-yl) -N- (4-morpholinophenyl) imidazo [1,2-a ] pyrazin-8-amine, BAY-61-3606, cerdulatinib (PRT-062607), etostatinib (entospentinib), fotatatinib (foslatinib) (R788), HMPL-523, NVP-QAB 205 AA, R112, R343, tamatinib (R406), and those described in US 50350321 (Gilead Connecticut) and US 20150175616.
Toll-like receptor (TLR) agonists
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with agonists for toll-like receptors (TLRs), such as agonists for TLR1(NCBI gene ID: 7096), TLR2(NCBI gene ID: 7097), TLR3(NCBI gene ID: 7098), TLR4(NCBI gene ID: 7099), TLR5(NCBI gene ID: 7100), TLR6(NCBI gene ID: 10333), TLR7(NCBI gene ID: 51284), TLR8(NCBI gene ID: 51311), TLR9(NCBI gene ID: 54106), and/or TLR10(NCBI gene ID: 81793). Exemplary TLR7 agonists that may be co-administered include DS-0509, GS-9620 (visamott), visamott analogues, LHC-165, TMX-101 (imiquimod), GSK-2245035, resiquimod, DSR-6434, DSP-3025, IMO-4200, MCT-465, MEDI-647, 3M-051, SB-9922, 3M-052, Limtop, TMX-30X, TMX-202, RG-7863, RG-7795, and US20100143301(Gilead Sciences), US 0098248(Gilead Sciences) and US 2009004747sciences (jalead Sciences), US 20115849 (20140042014) 2014, US 033642 (Janssen 695), WO 2011695 (jansen 3), WO Pharma Pharma (Pharma 0721), WO 201162249 (ja × 2014), US 201120145820142014 2014, (WO 20000779) (2014), WO 0336699 (WO 0336695), WO 0320136695), WO 201695 (Janarx) 50779) (bioirus 920082050) (1969), bioirus 509231 (bioirus 92002014) (bioirus 920082050), WO 2005046) (19651485) (bioirus) Compounds disclosed in US20140088085(Ventirx Pharma), US20140275167(Novira Therapeutics) and US20130251673(Novira Therapeutics). The TLR7/TLR8 agonist that may be co-administered is NKTR-262. Exemplary TLR8 agonists that may be co-administered include E-6887, IMO-4200, IMO-8400, IMO-9200, MCT-465, MEDI-9197, motolimod, resiquimod, GS-9688, VTX-1463, VTX-763, 3M-051, 3M-052, and U S20140045849(Janssen), US20140073642(Janssen), WO2014/056953(Janssen), WO2014/076221(Janssen), WO2014/128189(Janssen), US20140350031(Janssen), WO2014/023813(Janssen), US20080234251(Array Biopharma), US 3062008020080050 (Array Biopharma), US20100029585(Ventirx Pharma), US20110092485(Ventirx Pharma), US 20118235 (Ventirx Pharma), US20120082658 658 (ventx Pharma), US20120219615(Ventirx Pharma), US 0066432(Ventirx Pharma), US 0085 (Ventirx Pharma), US Pharma 201467 (Pharma 513025130251673 (novirra) and novirra 6720146720143. Exemplary TLR9 agonists that may be co-administered include AST-008, CMP-001, IMO-2055, IMO-2125, ranimod (litenimod), MGN-1601, BB-001, BB-006, IMO-3100, IMO-8400, IR-103, IMO-9200, alogtomoid (agotolimod), DIMS-9054, DV-1079, DV-1179, AZD-1419, leftiliomod (MGN-1703), CYT-003-QbG10, and PUL-042. Examples of TLR3 agonists include letamimod, poly ICLC, b,Apoxxim、IPH-33, MCT-465, MCT-475 and ND-1.1.
Tyrosine Kinase Inhibitor (TKI)
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with a Tyrosine Kinase Inhibitor (TKI). TKIs can target Epidermal Growth Factor Receptor (EGFR) as well as receptors for Fibroblast Growth Factor (FGF), platelet-derived growth factor (PDGF), and Vascular Endothelial Growth Factor (VEGF). Examples of TKIs include, but are not limited to, Afatinib, ARQ-087 (Delazatinib), ASP5878, AZD3759, AZD4547, bosutinib, bugatitinib, cabozantinib, cedanib, kroneonib, dacomitinib, dasatinib, dorivitinib, E-6201, ervatinib, erlotinib, gefitinib, giritinib (ASP-2215), FP-1039, HM61713, icotinib, imatinib, KX2-391(Src), lapatinib, lestatinib, lenvatinib, midostaurin, nintedanib, ODM-203, Oxitinib (AZD-9291), panatinib, pozzatinib, quinitinib, ladotinib, rocitinib, solitinib (HMPL-012), sunitinib, L-malic acid famitinib (MAC-4), tivoranib, TH-4000 and MEDI-575 (anti-PDGFR antibody).
Chemotherapeutic agents
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with a chemotherapeutic or anti-neoplastic agent.
As used herein, the term "chemotherapeutic agent" or "chemotherapeutic agent" (or "chemotherapy" in the context of treatment with a chemotherapeutic agent) is intended to encompass any non-proteinaceous (e.g., non-peptidic) compound that can be used to treat cancer. Examples of chemotherapeutic agents include, but are not limited to: alkylating agents, such as thiotepa and cyclophosphamideAlkyl sulfonates such as busulfan, improsulfan, and piposulfan; aziridines such as benzotepa, carboquone, metotepipa, and uretepa; ethyleneimine and methylmelamine, including altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, and trimeylomelamine; lactones such as brazic acid and brazilin; camptothecin, including the synthetic analog topotecan; bryodin, callystatin (callystatin); CC-1065, including its aldorexin, kazelesxin and bizelesin synthetic analogs; nostoc, especially nostoc 1 and nostoc 8; dolastatin; duocarmycins, including the synthetic analogs KW-2189 and CBI-TMI; eiscosahol (eleutherobin); 5-azacytidine; coprinus atrata base (pancratistatin); sarcandra glabra alcohol (sarcodictyin); spongistatin (spongistatin); nitrogen mustards, such as chlorambucil, cyclophosphamide, glufosfamide, eflucfosfamide, bendamustine, estramustine, ifosfamide, dichloromethyl diethylamine, dichloromethyl diethylin hydrochloride Amines, melphalan, neoenbisine, benzene mustard cholesterol, prednimustine, trofosfamide, and uracil mustard; nitrosoureas such as carmustine, chlorouramicin, fotemustine, lomustine, nimustine and ranimustine; antibiotics, such as enediyne antibiotics (e.g., calicheamicin, particularly calicheamicin γ II and calicheamicin phiI1), danamycin (including daptomycin a), bisphosphonates (such as clodronate), espiramicin, neocancerin chromophores and related chromoproteins enediyne chromophores, aclacinomycin, actinomycin, anthracycline (auramycin), azaserine, bleomycin, actinomycin, calicheacin (caraicin), carnosinomycin (carzinomomycin), carcinomycin (carzinophilin), tryptomycin (chromomycin), dactinomycin, daunorubicin, ditorelbirubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin (including morpholino-doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolo-doxorubicin and deoxydoxorubicin); antibiotics, Epirubicin, esorubicin, idarubicin, sisomycin, mitomycins (such as mitomycin C), mycophenolic acid, nogaxomycin, olivomycin, pelomycin, pofiomycin, puromycin, quinomycin, roxobicin, streptomycin, streptozotocin, tubercidin, ubenimex, setastatin and zorubicin; antimetabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogs such as norpterin, methotrexate, pteropterin, and trimetrexate; purine analogs such as cladribine, pentostatin, fludarabine, 6-mercaptopurine, thioimisine, and thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, and floxuridine; androgens such as carpoterone, drostandrosterone propionate, epitioandrostanol, meindroxane, and testolactone; anti-adrenal species such as aminoglutethimide, mitotane and trostane; folic acid replenishers such as leucovorin; radiotherapeutic agents, such as radium-223; trichothecenes, especially T-2 toxin, verrucin (verrucin) A, tubercidin (roridin) A and serpentin (anguidine); taxanes, such as paclitaxel Albumin-bound paclitaxel (abraxane) and docetaxelCabazitaxel, BIND-014, tesetaxel; platinum analogs such as cisplatin and carboplatin, NC-6004 nanoplatinum; acetic acid glucurolactone; an aldehydic phosphoramide glycoside; (ii) aminolevulinic acid; eniluracil; amsacrine; hebrschil (hestrabucil); a bisantrene group; edatrexate; desphosphamide; colchicine; diazaquinone; eflornithine (elformmthine); ammonium etiolate; an epothilone; etoglut; gallium nitrate; a hydroxyurea; lentinan; leucovorin; lonidamine; maytansinoids such as maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidanol; diamine nitracridine; pentostatin; methionine; pirarubicin; losoxanthraquinone; fluoropyrimidines; folinic acid; podophyllinic acid; 2-ethyl hydrazide; procarbazine; polysaccharide-K (PSK); lezoxan; rhizomycin; a texaphyrin; a germanium spiroamine; alternarionic acid; trabectedin, triaminoquinone; 2,2',2 "-trichlorotrimethylamine; a carbamate; vindesine; dacarbazine; mannomustine; dibromomannitol; dibromodulcitol; pipobroman; gatifloxacin (gacytosine); arabinoside ("Ara-C"); cyclophosphamide; thiotepa; chlorambucil; gemcitabine 6-thioguanine; mercaptopurine; methotrexate; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitoxantrone; vincristine; vinorelbineNoxiaoling; (ii) teniposide; edatrexae; daunomycin; aminopterin; silloda (xeoloda); ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DFMO); retinoids, such as retinoic acid; capecitabine; NUC-1031; FOLFOX (folinic acid, 5-fluorouracil, oxa)Platinum (ll) salts; FOLFIRI (leucovorin, 5-fluorouracil, irinotecan); FOLFOXIRI (folinic acid, 5-fluorouracil, oxaliplatin, irinotecan), FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, oxaliplatin), and pharmaceutically acceptable salts, acids or derivatives of any of the foregoing. Such agents may be conjugated to an antibody or any of the targeting agents described herein to form an antibody-drug conjugate (ADC) or a targeted drug conjugate.
Anti-hormonal agents
Also included within the definition of "chemotherapeutic agent" are anti-hormonal agents such as anti-estrogens and Selective Estrogen Receptor Modulators (SERMs), aromatase inhibitors, anti-androgens, as well as pharmaceutically acceptable salts, acids, or derivatives of any of the foregoing, which are used to modulate or inhibit the action of hormones on tumors.
Examples of antiestrogens and SERMs include tamoxifen (including NOLVADEXTM), raloxifene, droloxifene, 4-hydroxyttamoxifen, troloxifene, raloxifene hydrochloride, LY117018, onapristone, and toremifene
Inhibitors of aromatase modulate the production of estrogen in the adrenal glands. Examples include 4(5) -imidazole, aminoglutethimide, megestrol acetateExemestane, formestane, fadrozole and vorozoleLetrozoleAnd anastrozole
Examples of antiandrogens include apalutamide, abiraterone, enzalutamide, flutamide, galatel, nilutamide, bicalutamide, leuprorelin, goserelin, ODM-201, APC-100, ODM-204.
Examples of progesterone receptor antagonists include onapristone.
Anti-angiogenic agents
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an anti-angiogenic agent. Anti-angiogenic agents that may be co-administered include retinoid acids and derivatives thereof, 2-methoxyestradiol, beta-glucosidase, and beta-glucosidase,Regorafenib, nigupanib, suramin, squalamine, tissue inhibitors of metalloproteinase 1, tissue inhibitors of metalloproteinase 2, plasminogen activator inhibitor 1, plasminogen activator inhibitor 2, cartilage derived inhibitors, paclitaxel (nabu-paclitaxel), platelet factor 4, protamine sulfate (herring protamine), sulfated chitin derivatives (prepared from queen shells), sulfated peptidoglycan complexes (sp-pgs), staurosporines, modulators of matrix metabolism (including proline analogs such as l-azetidine-2-carboxylic acid (LACA), cis-hydroxyproline, d, I-3, 4-dehydroproline, thioproline), α' -bipyridine, β -aminopropionitrile fumarate, beta-aminopropionitrile fumarate, 4-propyl-5- (4-pyridyl) -2(3h) -oxazolone, methotrexate, mitoxantrone, heparin, interferon, 2-macroglobulin-serum, chicken metalloproteinase 3 inhibitor (ChIMP-3), chymotrypsin inhibitor, beta-cyclodextrin tetradecyl sulfate, epothilones, fumagillins, aureothiomalate, d-penicillamine, beta-1-anticementarase-serum, alpha-2-antiplasmin, bisantrene, disodium clobenzaprine, disodium n-2-carboxyphenyl-4-chloroanthranilate or "CCA", thalidomide, an antiangiogenic steroid, a carboxyamidoimidazole, a metalloproteinase inhibitor such as BB-94, an S100a9 inhibitor such as taquinomod. Other anti-angiogenic agents include antibodies, preferably monoclonal antibodies directed against these angiogenic growth factors: beta-FGF, alpha-FGF, FGF- 5. VEGF isoforms, VEGF-C, HGF/SF and Ang-1/Ang-2.
Anti-fibrotic agents
In some embodiments, the antibodies and/or fusion proteins provided herein are administered with an anti-fibrotic agent. Anti-fibrotic agents that may be co-administered include (e.g., 7-B16) compounds such as Beta Aminopropionitrile (BAPN), as well as compounds disclosed in US4965288 relating to lysyl oxidase inhibitors and their use in treating diseases and disorders associated with abnormal deposition of collagen, and compounds disclosed in US4997854 relating to compounds that inhibit LOX to treat various pathological fibrotic states, which patents are incorporated herein by reference. Further exemplary inhibitors are described in US4943593 relating to compounds such as 2-isobutyl-3-fluoro-, chloro-or bromo-allylamine, US5021456 relating to 2- (1-naphthyloxymethyl) -3-fluoroallylamine, US5059714, US5120764, US5182297, US5252608, and US 20040248871, which are incorporated herein by reference.
Exemplary anti-fibrotic agents also include primary amines that react with the carbonyl group of the active site of lysyl oxidase, and more specifically, those that, upon binding to the carbonyl group, produce a product that is stabilized by resonance, such as the following primary amines: ethylenediamine, hydrazine, phenylhydrazine, and derivatives thereof; semicarbazides and urea derivatives; aminonitriles such as BAPN or 2-nitroacetamine; unsaturated or saturated haloamines such as 2-bromo-ethylamine, 2-chloroethylamine, 2-trifluoroethylamine, 3-bromopropylamine and p-halobenzylamine; and selenium homocysteine lactone.
Other anti-fibrotic agents are copper chelators that either penetrate or do not penetrate the cell. Exemplary compounds include indirect inhibitors that block aldehyde derivatives derived from the oxidative deamination of lysyl and hydroxyllysyl residues by lysyl oxidase. Examples include thiolamines (particularly D-penicillamine) and their analogs such as 2-amino-5-mercapto-5-methylhexanoic acid, D-2-amino-3-methyl-3- ((2-acetamidoethyl) dithio) butanoic acid, p-2-amino-3-methyl-3- ((2-aminoethyl) dithio) butanoic acid, sodium 4- ((p-1-dimethyl-2-amino-2-carboxyethyl) dithio) butanesulfate, 2-acetamidoethyl-2-acetamidoethylmercaptan sulfate, and sodium 4-mercaptobutanesulfinate trihydrate.
Anti-inflammatory agents
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an anti-inflammatory agent. Exemplary anti-inflammatory agents include, but are not limited to, inhibitors of one or more of the following: arginase (ARG1(NCBI gene ID: 383), ARG2(NCBI gene ID: 384)), carbonic anhydrase (CA1(NCBI gene ID: 759), CA2(NCBI gene ID: 760), CA3(NCBI gene ID: 761), CA4(NCBI gene ID: 762), CA5A (NCBI gene ID: 763), CA5B (NCBI gene ID: 11238), CA6(NCBI gene ID: 765), CA7(NCBI gene ID: 766), CA8(NCBI gene ID: 767), CA9(NCBI gene ID: 768), CA10(NCBI gene ID: 56934), CA 84 (NCBI gene ID: 58770), CA12(NCBI gene ID: 771), CA13(NCBI gene ID: 377677), CA14 (prostaglandin ID: GS synthase (NCBI gene ID: 23632), COX-peroxidase synthase (COX-375, COX gene ID: 58571), prostaglandin-PTBI gene ID 5724, PTBI gene ID 5743, PTBI gene ID-5723 (NCBI gene ID: 5724), prostaglandin I5743, PTBI gene ID-2000, PTBI gene synthase (NCBI gene ID: 5723), prostaglandin I5743, PTBI gene ID-type PTBI gene 5743, PTBI gene ID-S-2000, and prostaglandin E synthase (NCBI gene ID type PTBI gene I-S-763), and/or a, PGES; gene ID: 9536) arachidonic acid 5-lipoxygenase (ALOX5, 5-LOX; NCBI Gene ID: 240) soluble epoxide hydrolase 2(EPHX2, SEH; NCBI Gene ID: 2053) and/or mitogen-activated protein kinase 8(MAP3K8, TPL 2; NCBI Gene ID: 1326). In some embodiments, the inhibitor is a dual inhibitor, such as a dual inhibitor of COX-2/COX-1, COX-2/SEH, COX-2/CA, COX-2/5-LOX.
Examples of inhibitors of prostaglandin-endoperoxide synthase 1(PTGS1, COX-1; NCBI gene ID: 5742) that may be co-administered include moxidectin, GLY-230, and TRK-700.
Examples of prostaglandin-endoperoxide synthase 2(PTGS2, COX-2; NCBI gene ID: 5743) inhibitors that may be co-administered include diclofenac, meloxicam, parecoxib, etoricoxib, AP-101, celecoxib, AXS-06, diclofenac potassium, DRGT-46, AAT-076, meloxicam, lumiracoxib, meloxicam, valdecoxib, zatoprofen, nimesulide, anizafen, alicoxib, cimicib, deracoxib, fluroimidazole, nonoxib, mavaxib, NS-398, pamidrogrel, parecoxib, robesib, rofecoxib, rutaecarpine, temoxib, and zatoprofen. Examples of inhibitors of COX1/COX2 that may be co-administered include HP-5000, lornoxicam, ketorolac tromethamine, bromfenac sodium, ATB-346, HP-5000. Examples of bis-COX-2/Carbonic Anhydrase (CA) inhibitors that may be co-administered include Bomexicam and Eloxib.
Examples of inhibitors of secreted phospholipase A2, prostaglandin E synthase (PTGES, PGES; gene ID: 9536) which may be co-administered include LY3023703, GRC 27864, and the compounds described in WO2015158204, WO2013024898, WO2006063466, WO2007059610, WO 20071245889, WO2010100249, WO2010034796, WO2010034797, WO2012022793, WO2012076673, WO2012076672, WO2010034798, WO 20100300300399, WO2012022792, WO2009103778, WO2011048004, WO 2012082087771, WO 302161965, WO 2013113113113113118031171, WO 20172825, WO 16716716791376, WO 2009138812, WO 2010860, WO 2012012012013153535, WO 2009130230230242, WO 20046913026986, WO 2012013186692, WO 201606905690518, WO 2016069056905690569056978, WO 20020020020064919898989898989820091374, WO 20020064989898989898989898987, WO 20071989898989898989898989898915 and WO 2015989. Metformin has also been found to inhibit COX2/PGE2/STAT3 axis and metformin can be co-administered. See, e.g., Tong et al, Cancer Lett. (2017)389: 23-32; and Liu et al, Oncotarget, (2016)7(19) 28235-46.
Examples of inhibitors of carbonic anhydrase that may be co-administered (e.g., one or more of CA1(NCBI gene ID: 759), CA2(NCBI gene ID: 760), CA3(NCBI gene ID: 761), CA4(NCBI gene ID: 762), CA5A (NCBI gene ID: 763), CA5B (NCBI gene ID: 11238), CA6(NCBI gene ID: 765), CA7(NCBI gene ID: 766), CA8(NCBI gene ID: 767), CA9(NCBI gene ID: 768), CA10(NCBI gene ID: 56934), CA11(NCBI gene ID: 770), CA12(NCBI gene ID: 771), CA13(NCBI gene ID: 377677), CA14(NCBI gene ID: 23632)) include acetazolamide, methazolamide, dorzolamide, nizamide, clothianidin, zolamide, and dichlorobenzene. bis-COX-2/CA 1/CA2 inhibitors that may be co-administered include CG 100649.
Examples of inhibitors of arachidonic acid 5-lipoxygenase (ALOX5, 5-LOX; NCBI gene ID: 240) that may be co-administered include meclofenamate sodium, zileuton.
Double inhibitors of soluble epoxide hydrolase 2(EPHX2, SEH; NCBI gene ID: 2053) which may be co-administered include compounds described in WO 2015148954. Dual inhibitors of COX-2/SEH that may be co-administered include the compounds described in WO 2012082647. Dual inhibitors of SEH and fatty acid amide hydrolase (FAAH; NCBI gene ID: 2166) that may be co-administered include the compounds described in WO 2017160861.
Examples of inhibitors of mitogen-activated protein kinase 8(MAP3K8, tumor progression locus-2, TPL 2; NCBI gene ID: 1326) that may be co-administered include GS-4875, GS-5290, BHM-078, and those described in: WO2006124944, WO2006124692, WO2014064215, WO 2018005435; teli et al, J Enzyme Inhib Med Chem. (2012)27(4) 558-70; gangwall et al, Curr Top Med Chem. (2013)13(9): 1015-35; wu et al, Bioorg Med Chem Lett. (2009)19(13): 3485-8; kaila et al, Bioorg Med Chem. (2007)15(19): 6425-42; and Hu et al, Bioorg Med Chem Lett. (2011)21(16): 4758-61.
Tumor oxygen mixture
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with an agent that promotes or increases tumor oxygenation or reoxygenation or prevents or reduces tumor hypoxia. Exemplary agents that may be co-administered include, for example, hypoxia inducible factor 1 alpha (HIF-1 alpha) inhibitors, such as PT-2977, PT-2385; VEGF inhibitors such as bevacizumab (bevasizumab), IMC-3C5, GNR-011, tinibizumab, LYN-00101, ABT-165; and/or oxygen carrier proteins (e.g., heme nitric oxide and/or oxygen binding protein (HNOX)), such as OMX-302 and HNOX proteins described in WO2007137767, WO 20071397691, WO2014107171, and WO 2016149562.
Immunotherapeutic agent
In some embodiments, reference will be made herein toThe antibody and/or fusion protein (e.g., 7-B16) provided is administered with an immunotherapeutic agent. Exemplary immunotherapeutic agents that may be co-administered include abamectin (abagozumab), AB308, ABP-980, adalimumab (adelimumab), alfuzumab (afutuzumab), alemtuzumab (alemtuzumab), amatuzumab (amatuximab), analimumab (anatumumab), acipimox (aritemab), alemtuzumab (arituzumab), atilizumab, bavizumab (bavituximab), betuzumab (bectmomab), bevacizumab (bivatuzumab), bonatumumab (blinatumomab), bentuximab (brentuximab), kantuzumab (tuzumab), carbamtuzumab (catatumumab), 49, bevacizumab (blinatuzumab), netuzumab (brentuximab), brazetuzumab (brentuximab), bevacizumab (brazemazumab), bevacizumab (rituximab (brazimab), bevacizumab (rabuzumab), bevacizumab (catazetuzumab), bevacizumab (bevacizumab), bevacizumab (bevacizumab), bevaciz (bevacizumab), bevacizumab (bevaciz), bevaciz (bevaciz), bevaciz (e), bevaciz (bevaciz), bevaciz (e), bevaciz (e (bevaciz (e), bevaciz (bevacizb), bevaciz (e), bevaciz (b), bevaciz (e), bevaciz (e, Dextuximab (dinutuximab), donamizumab (domfanalimab), trastuzumab (drozitumab), durigotazumab (duligotumab), durigotamab (dumigiumab), ekitumumab (dummieximab), ekimuximab (ecromeximab), elotuzumab (elotuzumab), emituzumab (emituzumab), ensuximab (ensituximab), ertuzumab (ertuzomab), edazumab (etazemab), fartuzumab (fartuzumab), non-trastuzumab (firtatuzumab), fititumumab (figitumab), frantuzumab (fungitumumab), frantuzumab (fuflutuzumab), flutuximab (futuximab), gantuximab (gantuzumab), futuximab (gantuximab), gemtuximab), gantuximab (gemtuzumab), gemtuzumab (gimitumumab), immitumumab (immitumumab), immitumumab (rituximab (imob), rituximab (icotuzumab), rituximab (orizumab), orizumab (orituzumab), orituzumab (orimetuzumab), orimetuzumab (orimex), orimetub), orimejitub (orimetub), orimetub (orimetuzumab), orimetuzumab (orimuitux (orimetub), orimetuzumab (orimejituzumab (orimex (orimetub), ir MDX-010, BMS-734016, and MDX-101), itumumab (iratuzumab), labetazumab (labetuzumab), lexatuzumab (lexatuzumab), lintuzumab (l)intuzumab, lomuzumab (lorvotuzumab), lucatumab (lucatumumab), mapatumumab (mapatumumab), matuzumab (matuzumab), milatuzumab (matuzumab), mirtazumab (matuzumab), minritumumab (minretumumab), mitumumab (mitumumab), moguzumab (mogamuzumab), moxitumumab (moxitumumab), naptuzumab (naptumomab), natalizumab (nertuzumab), nimotuzumab (nimotuzumab), nofitumumab (nofetummomab), OBI-833, obinutuzumab (obiumutuzumab), austotuzumab (pactuzumab), pasurizumab (pantuzumab), trastuzumab (oteuzumab), obian obile (notuzumab), obile (otetuzumab), OBI-833, obium (obile), obium (pertuzumab), pertuzumab (paseuzumab), trastuzumab (abotuzumab), trastuzumab (abotuzumab), tuzumab (abotuzumab), trastuzumab (abotuzumab), and related (abotuzumab), or (adotuzumab (abotuzumab), or (abotuotuzumab), or (abotuzumab), or (abotuzumab (abotuvu (abotuzumab), or (abotuvu), or (abotuzumab), or, Ritumomab (pintumomab), primitumumab (primumumab), ranibizumab (ractumomab), ranituzumab (radretumab), ramucirumab (ramucirumab) Rituximab (rilotuzumab), rituximab (rituximab), rituximab (robitumumab), sibutrumab (robitumumab), semuximab (situximab), solituzumab (solitomab), sintuzumab (simtuzumab), tacatuzumab (tacatuzumab), tatupuzumab (tapetum umab), ternatalizumab (tenatumumab), tetuzumab (teprotuzumab), tegafuzumab (tigatuzumab), tositumomab (tositumomab), trastuzumab (tuzumab), tuzumab (tuzumab), ubuzumab (trastuzumab), tuzumab (trastuzumab), rituzumab (rituxuzumab), rituxuzumab (trastuzumab), rituxuzumab (8), trastuzumab (netuzumab), trastuzumab (trastuzumab), and trastuzumab (netuzumab). Rituximab is useful in the treatment of indolent B cell cancers, including marginal zone lymphoma, WM, CLL and small lymphocytic lymphomas. Combinations of rituximab and chemotherapeutic agents are particularly effective.
Exemplary therapeutic antibodies can be further labeled or combined with radioisotope particles such as indium-111, yttrium-90 (90Y-clerituzumab), or iodine-131.
In some embodiments, the immunotherapeutic agent is an antibody-drug conjugate (ADC). Exemplary ADCs that may be co-administered include, but are not limited to, drug-conjugated antibodies, fragments thereof, or antibody mimetics that target the proteins or antigens listed above and herein. Exemplary ADCs that may be co-administered include gemtuzumab ozogamicin, present trastuzumab, infliximab, granuabamab, alemtuzumab, mivirtuximab, dirtuzumab ozogamicin, rovacizumab, vadatuximab, labuzumab, saxituzumab (e.g., safituzumab gavatica), rituximab, inflituzumab ozogamicin, pertuzumab, Poluotuzumab, pinitumumab, Cotuximab, inflatauximab, Millatuzumab, Rovatuzumab, ABBV-399, AGS-16C3F, ASG-22ME, AGS67E, AMG172, AMG575, BAY1129980, Y1187982, BAY94-9343, GSK2857916, max-TF-ADC, IMTF 529, GN853, LOP 85628, LOP 06265547, MDS-3978 PF-5934 PF-7450, MDRG-7450, GEV-7450, and RG, RG7458, RG7598, SAR566658, SGN-CD19A, SGN-CD33A, SGN-CD70A, SGN-LIV1A and SYD 985. ADCs that can be co-applied are described, for example, in Lambert et al, Adv Ther (2017)34: 1015-. In some embodiments, the antibodies and/or fusion proteins provided herein are administered with safirtuzumab gavitegam.
Exemplary therapeutic agents (e.g., anti-cancer or anti-tumor agents) that can be conjugated to a drug-conjugated antibody, fragment thereof, or antibody mimetic include, but are not limited to, monomethyl reoxidine e (mmae), monomethyl reoxidine f (mmaf), calicheamicin, ansamitocins, maytansine or analogs thereof (e.g., maytansine (mertansine)/emtansine (DM1), ravitant (ravtanine)/soratane (soravtansine) (DM4)), anthracyclines (e.g., doxorubicin, daunorubicin, epirubicin, idarubicin), Pyrrolobenzodiazepine (PBD) DNA cross-linker SC-DR002(D6.5), duocarmycin, microtubule inhibitor (MTI) (e.g., taxane, vinca alkaloid, epothilone), Pyrrolobenzodiazepine (PBD) or dimers thereof, duocarmycin (A, B1, B2, C1, C2, D, SA, C D, SA, etc.) CC-1065) and other anti-cancer or anti-tumor agents described herein. In some embodiments, the therapeutic agent conjugated to the drug-conjugated antibody is a topoisomerase I inhibitor (e.g., a camptothecin analog, such as irinotecan or its active metabolite SN 38). In some embodiments, therapeutic agents (e.g., anti-cancer or anti-tumor agents) that can be conjugated to drug-conjugated antibodies, fragments thereof, or antibody mimetics include immune checkpoint inhibitors. In some embodiments, the conjugated immune checkpoint inhibitor is a conjugated small molecule inhibitor of CD274(PDL1, PD-L1), programmed cell death 1(PDCD1, PD1, PD-1), or CTLA 4. In some embodiments, the conjugated small molecule inhibitor of CD274 or PDCD1 is selected from the group consisting of GS-4224, GS-4416, INCB086550, and MAX 10181. In some embodiments, the conjugated small molecule inhibitor of CTLA4 comprises BPI-002.
Cancer gene therapy and cell therapy
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with cancer gene therapy and cell therapy. Cancer gene therapy and cell therapy include the insertion of normal genes into cancer cells to replace mutated or altered genes; a genetic modification of a silent mutant gene; genetic methods to kill cancer cells directly; including infusions of immune cells designed to replace most of the patient's own immune system to enhance an immune response to cancer cells, or to activate the patient's own immune system (T cells or natural killer cells) to kill cancer cells, or to find and kill cancer cells; genetic approaches to altering cellular activity to further alter the endogenous immune response against cancer.
Cell therapy
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with one or more cell therapiesAnd applying. Exemplary cell therapies include, but are not limited to, co-administration of one or more of a population of Natural Killer (NK) cells, NK-T cells, Cytokine Induced Killer (CIK) cells, Macrophage (MAC) cells, Tumor Infiltrating Lymphocytes (TILs), and/or Dendritic Cells (DCs). In some embodiments, the cell therapy requires T cell therapy, e.g., co-administration of α/β TCR T cells, γ/δ TCR T cells, regulatory T (treg) cells, and/or TRuC TM A population of T cells. In some embodiments, the cell therapy requires NK cell therapy, e.g., co-administration of NK-92 cells. As the case may be, cell therapy may require co-administration of cells that are autologous, syngeneic, or allogeneic to the subject.
In some embodiments, the cell therapy entails co-administration of cells comprising a Chimeric Antigen Receptor (CAR). In such therapies, the population of immune effector cells is engineered to express a CAR, wherein the CAR comprises a tumor antigen binding domain. In T cell therapy, T Cell Receptors (TCRs) are engineered to target tumor-derived peptides presented on the surface of tumor cells.
With respect to the structure of the CAR, in some embodiments, the CAR comprises an antigen binding domain, a transmembrane domain, and an intracellular signaling domain. In some embodiments, the intracellular domain comprises a primary signaling domain, a costimulatory domain, or both a primary signaling domain and a costimulatory domain. In some embodiments, the primary signal domain comprises a functional signal domain of one or more proteins selected from the group consisting of: CD3 ζ, CD3 γ, CD3 δ, CD3 ε, common FcR γ (FCERIG), FcR β (fcepsilon Rlb), CD79a, CD79b, fcyriia, DAP10, and DAP 12.
In some embodiments, the co-stimulatory domain comprises a functional domain of one or more proteins selected from the group consisting of: CD, 4-1BB (CD137), OX, CD, PD-1, ICOS, CD, LIGHT, NKG2, B-H, a ligand that specifically binds to CD, CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHT TR), SLAMF, NKp (KLRFI), CD160, CD α, CD β, IL2 γ, IL7 α, ITGA, VLA, CD49, ITGA, IA, CD49, ITGA, VLA-6, CD49, ITGAD, ITGAE, CD103, ITGAL, CD1 (NCBI gene ID: 909), CD1 (NCBI gene ID: 910), CD1 (NCBI gene ID: 911), CD1 (NCBI gene ID: 912), CD1 (NCBI gene ID: ITGAL), ITGAM, ITGAX, ITGB, CD, ITGB (ITGB, ACAA-160, ACAA-229), TNFAMGL (CD-100, CD-100, TNGL, CD-100, CD-CD, CD-CD, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, LAT, GADS, SLP-76, PAG/Cbp, NKp44, NKp30, NKp46 and NKG 2D.
In some embodiments, the transmembrane domain comprises a transmembrane domain of a protein selected from the group consisting of: the alpha, beta or zeta chain of the T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80(KLRF1), CD160, CD19, IL2R beta, IL2R gamma, IL7R, ITGA1, VLA1, CD49a, ITGA4, 695IA 4, CD49D, ITGA D, VLA-6, CD49D, ITGAD, CD1D, ITGAE, CD103, ITGAL, ITGAM, ITGAX, ITGB D, CD D, ITGB D (LFA-1, CD D), ITGB D, TNFR D, DNAM D (CD226), SLAMF D (CD244, 2B D), CD D (TACTILE), CEM ACAM D, CRTAM, Ly D (CD229), CD160 (D), PSGL D, CD100(SEMA4D), SLAMF D (NTB-D), SLAM (SLAMF D, CD150, IPO-3), SLAME (SLAMG D), PAGS (PAGS 36162, PAGS D), NKPLG D, NKG D, and NKG D.
In some embodiments, a TCR or CAR antigen binding domain or an immunotherapeutic agent described herein (e.g., a monospecific or multispecific antibody or antigen-binding fragment thereof or antibody mimetic) binds a tumor-associated antigen (TAA). In some embodiments, the tumor-associated antigen is selected from the group consisting of: CD 19; CD 123; CD 22; CD 30; CD 171; CS-1 (also known as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A 24); c-type lectin-like molecule-1 (CLL-1 or CLECLI); CD 33; epidermal growth factor receptor variant iii (egfrvlll); ganglioside G2(GD 2); ganglioside GD3 (alpha NeuSAc (2-8) alpha NeuSAc (2-3) beta DGaip (1-4) bDGIcp (1-1) Cer); ganglioside GM3 (alpha NeuSAc (2-3) beta DGalp (1-4) beta DGlcp (1-1) Cer); TNF receptor superfamily member 17(TNFRSF17, BCMA); tn antigen ((Tn Ag) or (GaINAcu-Ser/Thr)); prostate Specific Membrane Antigen (PSMA); receptor tyrosine kinase-like orphan receptor 1 (RORI); tumor associated glycoprotein 72(TAG 72); CD 38; CD44v 6; carcinoembryonic antigen (CEA); epithelial cell adhesion molecule (EPCAM); B7H3(CD 276); KIT (CD 117); interleukin-13 receptor subunit alpha-2 (IL-13Ra2 or CD213a 2); mesothelin; interleukin 11 receptor alpha (IL-11 Ra); prostate Stem Cell Antigen (PSCA); protease serine 21(Testisin or PRSS 21); vascular endothelial growth factor receptor 2(VEGFR 2); lewis (Y) antigen; CD 24; platelet-derived growth factor receptor beta (PDGFR-beta); stage-specific embryonic antigen-4 (SSEA-4); CD 20; δ -like 3(DLL 3); a folate receptor alpha; receptor tyrosine protein kinase, ERBB2(Her 2/neu); cell surface associated mucin 1(MUC 1); epidermal Growth Factor Receptor (EGFR); neural Cell Adhesion Molecule (NCAM); prostasin; prostatic Acid Phosphatase (PAP); mutant elongation factor 2(ELF 2M); ephrin B2; fibroblast activation protein alpha (FAP); insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase ix (caix); proteasome (Prosome, Macropain) subunit beta type 9(LMP 2); glycoprotein 100(gp 100); an oncogene fusion protein consisting of the region of the Breakcluster (BCR) and Abelson murine leukemia virus oncogene homolog 1(Abl) (BCR-Abl); a tyrosinase enzyme; ephrin type a receptor 2(EphA 2); fucosyl GM 1; sialylated Lewis adhesion molecules (sLe); transglutaminase 5(TGS 5); high Molecular Weight Melanoma Associated Antigen (HMWMAA); o-acetyl-GD 2 ganglioside (OAcGD 2); folate receptor β; tumor endothelial marker 1(TEM1/CD 248); tumor endothelial marker 7 related (TEM 7R); prostate six transmembrane epithelial antigen I (STEAP 1); blocking protein 6(CLDN 6); thyroid Stimulating Hormone Receptor (TSHR); class 5 members of the G protein-coupled receptor C (gprcsd); chromosome X open reading frame 61(CXORF 61); CD 97; CD179 a; anaplastic Lymphoma Kinase (ALK); polysialic acid; placenta-specific 1(PLAC 1); the hexasaccharide moiety of the globoH glycoceramide (globoH); mammary differentiation antigen (NY-BR-1); uroblast protein (uroplakin)2(UPK 2); hepatitis a virus cell receptor 1(HAVCR 1); adrenoceptor β 3(ADRB 3); ubiquitin 3(PANX 3); g protein-coupled receptor 20(GPR 20); lymphocyte antigen 6 complex, locus K9 (LY 6K); olfactory receptor 51E2(ORS IE 2); TCR γ alternate reading frame protein (TARP); wilms tumor protein (WT 1); cancer/testis antigen 1 (NY-ESO-1); cancer/testis antigen 2 (LAGE-la); melanoma associated antigen 1 (MAGE-A1); ETS translocation variant gene 6 located on chromosome 12p (ETV 6-AML); sperm protein 17(SPA 17); x antigen family member 1A (XAGE 1); angiopoietin binds to cell surface receptor 2(Tie 2); melanoma cancer testis antigen-1 (MADCT-1); melanoma cancer testis antigen-2 (MAD-CT-2); fos-related antigen 1; tumor protein p53(p 53); a p53 mutant; a prostein; a survivin; a telomerase; prostate cancer tumor antigen-1 (PCTA-1 or galectin 8), melanoma antigen recognized by T cell 1 (MelanA or MARTI); rat sarcoma (Ras) mutant; human telomerase reverse transcriptase (hTERT); a sarcoma translocation breakpoint; an inhibitor of melanoma apoptosis (ML-IAP); ERG (transmembrane protease, serine 2(TMPRSS2) ETS fusion gene); n-acetylglucosaminyltransferase V (NA 17); paired box protein Pax-3(PAX 3); an androgen receptor; cyclin B1; a v-myc avian myelocytoma virus oncogene neuroblastoma-derived homolog (MYCN); ras homolog family member c (rhoc); tyrosinase-related protein 2 (TRP-2); cytochrome P4501B 1(CYP IBI); CCCTC binding factor (zinc finger protein) like (BORIS or brother of imprinted site regulator), squamous cell carcinoma antigen recognized by T cells 3(SART 3); paired box protein Pax-5(PAX 5); the anterior vertex voxel binding protein sp32(OY-TES I); lymphocyte-specific protein tyrosine kinase (LCK); a kinase anchoring protein 4 (AKAP-4); synovial sarcoma, X breakpoint 2(SSX 2); receptor for advanced glycation end products (RAGE-I); renal ubiquitin 1 (RUI); renal ubiquitin 2(RU 2); legumain; human papilloma virus E6(HPV E6); human papilloma virus E7(HPV E7); intestinal carboxylesterase; mutated heat shock protein 70-2(mut hsp 70-2); CD79 a; CD79 b; CD 72; leukocyte-associated immunoglobulin-like receptor 1 (LAIRI); an Fc fragment of IgA receptor (FCAR or CD 89); leukocyte immunoglobulin-like receptor subfamily a member 2(LILRA 2); CD300 molecular-like family member f (CD300 LF); c-type lectin domain family 12 member a (CLEC 12A); bone marrow stromal cell antigen 2(BST 2); EGF-like module 2(EMR2) containing mucin-like hormone receptor-like; lymphocyte antigen 75(LY 75); glypican-3 (GPC 3); fc receptor like 5(FCRL 5); and immunoglobulin lambda-like polypeptide 1(IGLL 1). In some embodiments, the target is an epitope of a tumor associated antigen presented in the MHC.
In some embodiments, the tumor antigen is selected from the group consisting of CD150, 5T4, ActRIIA, B7, TNF receptor superfamily member 17(TNFRSF17, BCMA), CA-125, CCNA1, CD123, CD126, CD138, CD1, CD148, CD1, CD200, CD1, CD261, CD262, CD1, CD362, CD1, CD66 1-d, CD1, CE 1, CS-1 glycoprotein, CSPG 1, EGFP 36III, fibronectin, EGFRP 2-EGFP-72, EGFP-7, EGFP-7-EGFP-7, EGFP-72, EGFP-7, EGFP-3-7-EGFP-7, EGFP-3-EGFP-EGB-3, EGB-3-EGB-3, EGB-3-EGB-3, EGB-3-EGB-3-EGB-3, EGB-3-EGB-3-E-3-E-3-E, EGB-E, EGB-E-3-E-3, EGB-E-3, EGB-E-3, EGB-E-, IL-2, IL-22R-alpha, IL-6R, Ia, Ii, L1-CAM, L1-cell adhesion molecule, Lewis Y, Ll-CAM, MAGE A3, MAGE-A1, MART-1, MUC1, NKG2C ligand, NKG2D ligand, NYESO-1, OEPHa2, PIGF, PSCA, PSMA, ROR1, T101, TAC, TAG72, TIM-3, TRAIL-R1, TRAIL-R1(DR4), TRAIL-R2(DR5), VEGF, VEGFR2, WT-I, G protein coupled receptor, alpha-fetoprotein (AFP), angiogenic factor, exogenous cognate binding molecule (ExoCBM), oncogene product, anti-folate receptor, c-estrogen, carcinoembryonic antigen (CEA), cyclin (1), tumor ligand B2, epithelial receptor antigen, acetylcholine receptor binding protein, and binding protein, gp100, hepatitis b surface antigen, k chain, k light chain, kdr, λ chain, activin, melanoma-associated antigen, mesothelin, mouse double minute 2 homolog (MDM2), mucin 16(MUC16), mutant P53, mutant ras, necrotic antigen, oncofetal antigen, ROR2, progesterone receptor, prostate specific antigen, tfegfr, tenascin, P2-microgiobiuin, Fc receptor-like 5(FcRL 5).
In some embodiments, the antigen binding domain binds to an epitope of a target or Tumor Associated Antigen (TAA) that is presented in a Major Histocompatibility Complex (MHC) molecule. In some embodiments, the TAA is a cancer testis antigen. In some embodiments, the cancer testis antigen is selected from the group consisting of: sperm noggin binding protein (ACRBP; CT23, OY-TES-1, SP 32; NCBI gene ID: 84519), alpha fetoprotein (AFP; AFPD, FETA, HPAFP; NCBI gene ID: 174); a kinase anchoring protein 4(AKAP 4; AKAP82, AKAP-4, AKAP82, CT99, FSC1, HI, PRKA4, hAKAP82, p 82; NCBI gene ID: 8852), ATPase family AAA domain-containing 2(ATAD 82; ANCCA, CT137, PRO 2000; NCBI gene ID: 29028), centromere scaffold 1(KNL 82; AF15Q 82, CASC 82, CT 82, D82, MCPH 82, PPP1R 82, Spc 82, hKNL-1, hSpc 105; NCBI gene ID: 82), protein 55(CEP 82; C10orf 82, CT111, MARCH, URCC 82; CTABI gene ID: 55165), cancer/testis antigen 1A (CTAG1 ESG 72; CTAO 366.1; CT 2-2, CT 2-2, CTALAG 72; CTALAG 82, CTAB 2-82; CTABI gene 82, CTABI 2-82, CTABI gene 82, CTAB 2 LAG 1-82, CTAB 2-1-82, CTAB 2, CTAB 82, CTA 82, CTAB 2, CTA 82, CTAB 2, CTAB 82, CTAB 2, CTAB 82, CTA 82, CTAB 2, CTAB 82, CTA 82, CTAB 2, CTAB 82, CTAB 2, CTA 82, CTAB 3, CTA 82, and CTA 82, CTAB 2, CTA 82, CTAB 2, CTAB, LAGE 2B; NCBI Gene ID: 30848) CCCTC-binding factor-like (CTCFL; BORIS, CT27, CTCF-T, HMGB1L1, dJ579F20.2; NCBI Gene ID: 140690), catenin α 2(CTNNA 2; CAPR, CDCBM9, CT114, CTNR; NCBI Gene ID: 1496) cancer/testis antigen 83(CT 83; CXorf61, KK-LC-1, KKLC 1; NCBI Gene ID: 203413), cyclin a1(CCNA 1; CT 146; NCBI Gene ID: 8900) DEAD-box helicase 43(DDX 43; CT13, HAGE; NCBI Gene ID: 55510) Developmental pluripotency-related 2(DPPA 2; CT100, ECAT15-2, PESCRG 1; NCBI Gene ID: 151871), fetal and adult testis expressed 1(FATE 1; CT43, FATE; NCBI Gene ID: 89885) FMR1 neighbors (FMR1 NB; CT37, NY-SAR-35, NYSAR 35; NCBI Gene ID: 158521), HORMA domain containing 1(HORMAD 1; CT46, NOHMA; NCBI Gene ID: 84072) Insulin-like growth factor 2mRNA binding protein 3(IGF2BP 3; CT98, IMP-3, IMP3, KOC1, VICKZ 3; NCBI Gene ID: 10643) Leucine zipper protein 4(LUZP 4; CT-28, CT-8, CT28, HOM-TES-85; NCBI Gene ID: 51213) Lymphocyte antigen 6 family member K (LY 6K; CT97, HSJ001348, URLC10, ly-6K; NCBI Gene ID: 54742) Vortex spermatogenesis transposon silencer (MAEL; CT128, SPATA 35; NCBI Gene ID: 84944) MAGE family member a1(MAGEA 1; CT1.1, MAGE 1; NCBI Gene ID: 4100) (ii) a MAGE family member A3(MAGEA 3; CT1.3, HIP8, HYPD, MAGE3, MAGEA 6; NCBI gene ID: 4102); MAGE family member A4(MAGEA 4; CT1.4, MAGE-41, MAGE-X2, MAGE4, MAGE4A, MAGE 4B; NCBI gene ID: 4103); MAGE family member A11(MAGEA 11; CT1.11, MAGE-11, MAGE11, MAGEA-11; NCBI gene ID: 4110); MAGE family member C1(MAGEC 1; CT7, CT 7.1; NCBI gene ID: 9947); MAGE family member C2(MAGEC 2; CT10, HCA587, MAGE 1; NCBI gene ID: 51438); MAGE family member D1(MAGED 1; DLXIN-1, NRAGE; NCBI gene ID: 9500); MAGE family member D (MAGED; 11B, BARTS, BCG-1, BCG, HCA, MAGE-D; NCBI gene ID: 10916), kinesin family member 20B (KIF 20; CT, KRMP, MPHOSPH, MPP-1, MPP; NCBI gene ID: 9585), NUF component of NDC centromere complex ((NUF; CDCA, CT106, NUF 2; NCBI gene ID:)), nuclear RNA export factor 2 (; CT, TAPL-2, TCP 11X; NCBI gene ID: 56001), repressor protein 1 containing PAS domain (PASD; CT, OXTES; NCBI gene ID:), PDZ binding kinase (PBK; CT, HEL164, Nori-3, SPK, TOPK; NCBI gene ID:), piwi-like RNA mediated gene 2 (PIWIL; CT, HILI, PIWIL1, NCBI:, MABI gene:, MAPI-related antigen in SPAI:, MPBI gene (PRIMI-4; PRIME-7, PRIME, PRI, MAR-4, MAR-II, MAR gene, MAR-1, MAR-II, MAR-1, MAR, HLC-6, HLC4, HLC6, JIP-4, JIP4, JLP, PHET, PIG 6; NCBI Gene ID: 9043) nuclear-associated sperm protein X linked family member a1(SPANXA 1; CT11.1, CT11.3, NAP-X, SPAN-X, SPAN-Xa, SPAN-Xb, SPANX-A; NCBI Gene ID: 30014) SPANX family member a2(SPANXA 2; CT11.1, CT11.3, SPANX-A, SPANX-C, SPANXA, SPANXC; NCBI Gene ID: 728712), SPANX family member C (SPANXC; CT11.3, CTp11, SPANX-C, SPANX-E, SPANXE; NCBI Gene ID: 64663) SPANX family member D (SPANXD; CT11.3, CT11.4, SPANX-C, SPANX-D, SPANX-E, SPANXC, SPANXE, dJ171K16.1; NCBI Gene ID: 64648) SSX family member 1(SSX 1; CT5.1, SSRC; NCBI Gene ID: 6756) SSX family member 2(SSX 2; CT5.2, CT5.2A, HD21, HOM-MEL-40, SSX; NCBI Gene ID: 6757) synaptophysin complex protein 3(SYCP 3; COR1, RPRGL4, SCP3, SPGF 4; NCBI Gene ID: 50511) Testis-expressed 14-cell bridge-forming factor (TEX 14; CT113, SPGF 23; NCBI Gene ID: 56155) Transcription factor Dp family member 3(TFDP 3; CT30, DP4, HCA 661; NCBI Gene ID: 51270) Serine protease 50(PRSS 50; CT20, TSP 50; NCBI Gene ID: 29122) TTK protein kinase (TTK; CT96, ESK, MPH1, MPS1, MPS1L1, PYT; NCBI Gene ID: 7272) and zinc finger proteins 165(ZNF 165; CT53, LD65, ZSCAN 7; NCBI Gene ID: 7718). T Cell Receptors (TCRs) and TCR-like antibodies that bind to epitopes of cancer testis antigens presented in Major Histocompatibility Complex (MHC) molecules are known in the art and can be used in the heterodimers described herein. Cancer testis antigens associated with neoplasia are described, for example, in Gibbs et al, Trends Cancer; year 2018, month 10; 4(10) 701-712 and CT database website cta. lncc. br/index. php. Exemplary TCR and TCR-like antibodies that bind to an epitope of NY-ESO-1 presented in the MHC are described, for example, in: Stewart-Jones et al, Proc Natl Acad Sci USA, 2009, 4 months and 7 days; 106(14) 5784-8; WO2005113595, WO2006031221, WO2010106431, WO2016177339, WO2016210365, WO2017044661, WO2017076308, WO2017109496, WO2018132739, WO2019084538, WO2019162043, WO2020086158 and WO 2020086647. Exemplary TCR and TCR-like antibodies that bind to an epitope of PRAME presented in the MHC are described, for example, in WO2011062634, WO2016142783, WO2016191246, WO2018172533, WO2018234319, and WO 2019109821. Exemplary TCRs and TCR-like antibodies that bind to MAGE epitopes presented in MHC are described, for example, in WO2007032255, WO2012054825, WO2013039889, WO2013041865, WO2014118236, WO2016055785, WO2017174822, WO2017174823, WO2017174824, WO2017175006, WO2018097951, WO2018170338, WO2018225732, and WO 2019204683. Exemplary TCRs and TCR-like antibodies that bind to an epitope of Alpha Fetoprotein (AFP) presented in the MHC are described, for example, in WO 2015011450. Exemplary TCRs and TCR-like antibodies that bind to an epitope of SSX2 presented in the MHC are described, for example, in WO 2020063488. Exemplary TCRs and TCR-like antibodies that bind to an epitope of KK-LC-1(CT83) presented in the MHC are described, for example, in WO 2017189254.
Examples of cell therapy include Algenpancuce-L, Sipuleucel-T, Rivogenlecucel (BPX-501) US9089520, WO2016100236, AU-105, ACTR-087, activated allogeneic Natural killer cell CNDO-109-AANK, MG-4101, AU-101, BPX-601, FATE-NK100, LFU-835 hematopoietic Stem cells, Imileckexecel-T, baltaleucel-T, PNK-007, UCARTCS1, ET-1504, ET-1501, ET-1502, ET-190, CD19-ARTEMIS, ProHema, FT 1050-treated bone marrow Stem cell therapy, CD4 NK-92 cells, CryoStim, AlloStimCAR, lentivirus transduced Hu T-meso cells, CART-22 cells, EGFP/19-28-BBT/4-1L BBT cells, GREFT-4628/5912/GREFT-25/5912 cells, GREFT-22 cells, GREFT-5912, and GREFT-190 cells, CCR5-SBC-728-HSPC, CAR4-1BBZ, CH-296, dnTGFbRII-NY-ESOc259T, Ad-RTS-IL-12, IMA-101, IMA-201, CARMA-0508, TT-18, CMD-501, CMD-503, CMD-504, CMD-502, CMD-601, CMD-602, CSG-005.
In some embodiments, one or more additional coadministered therapeutic agents may be classified according to their mechanism of action, for example, into the following groups:
agents targeting adenosine deaminase, such as pentostatin or cladribine;
agents targeting AKT1, such as GSK 2141795;
ATM-targeting agents such as AZD 1390;
MET-targeting agents, such as savotinib (savolitinib), carbamatinib (caplatib), terpatinib (tepotinib), ABT-700, AG213, JNJ-38877618(OMO-1), meletinib (merstinib), HQP-8361, BMS-817378, or TAS-115;
agents targeting mitogen-activated protein kinases, such as, e.g., quinuclidine, bimetinib, cobitinib, semetinib, trametinib, europlastinib (mirdometinib) (PD-0325901), pimatinib, rafatinib, or compounds disclosed in: WO2011008709, WO2013112741, WO2006124944, WO2006124692, WO2014064215, WO2018005435, Zhou et al, Cancer Lett.2017, 11/1/month, 408: 130-; teli et al, J Enzyme Inhib Med Chem. (2012)27(4): 558-70; gangwall et al, Curr Top Med Chem. (2013)13(9): 1015-35; wu et al, Bioorg Med Chem Lett. (2009)19(13): 3485-8; kaila et al, Bioorg Med Chem. (2007)15(19): 6425-42; or Hu et al, Bioorg Med Chem Lett. (2011)21(16) 4758-61;
agents targeting MAP4K1, such as compounds disclosed in WO2018183956, WO2018183964, WO2018167147, WO2018183964, WO2016205942, WO2018049214, WO2018049200, WO2018049191, WO2018102366, WO2018049152, WO2020092528, WO2020092621 or WO 2016090300;
Agents targeting thymidine kinase, such as beamakino (protepicture besadenovec (promatak, PancAtak, GliAtak, GMCI or AdV-tk);
agents targeting androgen receptors, such as enoboxate (enobosarm) (GTX-024) or daloluamide (daroluamide);
agents targeting CD47, such as RRx-001, DSP-107, VT-1021, IMM-02, SGN-CD47M, SIRPa-Fc-CD40L (SL-172154), or molorezumab;
agents targeting the interleukin pathway, such as pergiline interleukin (pegilenecokin) (AM-0010) (pegylated IL10), CA-4948(IRAK 4);
agents targeting markers of cluster of differentiation, such as A6, AD-IL24, neratinib, Tokatinib (ONT 380), mobocitinib (TAK-788), tervatinib (tesevatinib), trastuzumabTrastuzumab biosimilar (HLX-02), Margeritumumab (margetuximab), BAT-8001, pertuzumab (Perjeta), pefilgrastim (pegfilgrastim), RG6264, Zernida tuzumab (zanidamab) (ZW25), cavatak, AIC-100, talafurt (tagaxofusp) (SL-401), HLA-A2402/HLA-A0201 restricted epitope peptide vaccine, dasatinib, imatinib, nilotinibNib, sorafenib, lenvatinib mesylate (lentivatinib mesylate), phydroxynil (ofanergent obedienotec), cabozantinib malate (cabozantinib malate), AL-8326, ZLJ-33, KBP-7018, sunitinib malate (sunitinib malate), pazopanib derivatives (pazopanib derivatives), AGX-73, rebastinib (rebastinib), NMS-088, dritinib hydrochloride (drochloride), midostaurin, cediranib (cediranib), dolastatin, stritinib (cerivanib), trovatinib (sidetinib), tizanib (tivanib), masitinib (masisinib), regorafenib (regorafenib), mellitinib (scba besylate), valtinib (scytinib mesylate), valcanib), valcaninib (nesinib), valcaninib (359-170-Cbx-359), CDMC-2023-170, CDK-2021, CDF-170, CDK-170, BCA-31, SCB, GS-3583, asunercept (APG-101), APO-010, or a compound disclosed in: WO2016033570, WO2006009755, WO2007078034, WO2007092403, WO2007127317, WO2008005877, WO2012154480, WO 2012012154480, WO2008005877, WO 2012154414, WO2014039714, WO 2012015671536, WO 2018112112136, WO 201811218112140, WO 2012015509167, WO 2020076106106105, PCT/US2019/063091, WO19173692, WO 179517, WO2017096179, WO 201201201201709696182, WO2017096281, WO2018089628, 2018195321, WO 2020014646882, WO 2016091882, WO 201818191321, WO 2012012012012012012012012012012012012019091201201201201201819191919191307, WO 200200333390273333333333332012012012012012012012012012012012012012012012012012012012012012018167332012012012012012012012012018167332012002012002012002012002012002012002012002013391712012012012012012012012012012012012012012002012002012002012002012002012012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002019, WO 2002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002012002019, WO 3,979, WO 3,201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200201200; or gaierpy j. et al, 106 th american society for immunologists (AAI) (2019, 5 months, 9 days to 13 days, San Diego, abstract 71.5);
Agents targeting members of the cytochrome P450 family, such as letrozole, anastrozole, aminoglutethimide, megestrol acetateExemestane, formestane, fadrozole and vorozoleLetrozoleOr anastrozole
Agents targeting DKK3, such as MTG-201;
agents targeting EEF1a2, such as prilin;
agents targeting EIF4a1, such as loratinib (rohinitib);
agents targeting endoglin, such as TRC105 (carotuximab);
EGFR-targeting agents such as neratinib, tocatinib (ONT-380), tervatinib, mobotinib (TAK-788), DZD-9008, varlitinib, Avertinib (abortinib) (ACEA-0010), EGF816 (azartinib), ormotinib (olmutinib) (BI-1482694), Oxicitinib (AZD-9291), AMG-596, Lifeinib (lifrafenib) (BGB-283), panitumumab injection one (vectibix), or compounds disclosed in: booth et al, Cancer Biol ther.2018, 2.1.month; 19(2) 132 and 137;
agents targeting exporter 1, such as eltamisol (eltanexor);
agents targeting fatty acid amide hydrolases, such as the compounds disclosed in WO 2017160861;
Agents targeting heat shock protein 90 β family member 1, such as angutinib (anlotinib);
agents targeting interleukins, such as SAR 441000;
agents targeting lactoferrin, such as rutemide (ruxotemitide) (LTX-315);
agents targeting lysyl oxidase such as the compounds disclosed in US4965288, US4997854, US4943593, US5021456, US5059714, US5120764, US5182297, US5252608 or US 20040248871;
agents targeting members of the MAGE family, such as KITE-718, MAGE-A10C796T or MAGE-A10 TCR;
agents targeting matrix metallopeptidase 9, such as the compounds disclosed in WO 2012027721;
an agent targeting MCL1, such as tapotoclax (AMG-176), AMG-397, S-64315, AZD-5991, 483-LM, a-1210477, UMI-77, or a compound disclosed in WO2018183418, WO2016033486, or WO 2017147410;
agents targeting MDM2, such as ALRN-6924, CMG-097, miladeptan monothiosulfonate monohydrate (DS-3032b) or AMG-232;
agents targeting MDM4, such as ALRN-6924;
agents targeting melano a, such as MART-1F5 TCR-engineered PBMC;
An agent targeting mesothelin, such as CSG-MESO or TC-210;
agents targeting METAP2, such as M8891 or APL-1202;
agents targeting NLRP3, such as BMS-986299;
agents targeting ketoglutarate dehydrogenase such as tevimistat (CPI-613);
agents targeting placental growth factor, such as aflibercept;
agents targeting progesterone receptors, such as TRI-CYCLEN LO (norethindrone + ethinyl estradiol);
agents targeting prostaglandin E synthase, such as GRC 27864 or compounds disclosed in: WO2015158204, WO2013024898, WO2006063466, WO2007059610, WO 20071245889, WO2010100249, WO2010034796, WO2010034797, WO2012022793, WO2012076673, WO2012076672, WO2010034798, WO2010034799, WO2012022792, WO2009103778, WO2011048004, WO2012087771, WO2012161965, WO2013118071, WO2013072825, WO 1672014444, WO2009138376, WO2011023812, WO2012110860, WO2013153535, WO 2009130230242, WO2009146696, WO2013186692, WO 059618, WO 696969374, WO2009117985, WO 20090646464251, WO 2009182347, WO 2009198347, WO 200907117987, WO 2002016063376, WO 20020120160201389, WO 20090173, WO 2009098173, WO 20090173, WO 20090987, WO 20090389, WO 2009032173, WO 20090329, WO 20090567, and so on (200809032); liu et al, Oncotarget, (2016)7(19) 28235-46;
Agents targeting signal-regulatory protein α, such as CC-95251, GS-0189(FSI-189), ES-004, BI765063, ADU1805, AL-008, BAT-6030, APX-700, CTX-5861, or RRx-001;
an agent targeting SLC10a3, such as a compound disclosed in WO2015148954, WO2012082647 or WO 2017160861;
agents targeting transforming growth factor alpha, such as the compounds disclosed in WO 2019103203;
agents targeting the tumor protein p53, such as kevetrin (stimulating agent);
agents targeting vascular endothelial growth factor a, such as aflibercept;
agents targeting vascular endothelial growth factor receptors, such as furoquintinib (fruquintinib) or MP 0250;
agents targeting VISTA, such as CA-170 or HMBD-002;
agents targeting WEE1, such as adatanserin (avasertib) (AZD-1775);
small molecule inhibitors targeting ABL1, such as imatinib, rebatinib, asciminib or ponatinib
Small molecule antagonists targeting adenosine receptors such as CPI-444, AZD-4635, prokineticide or PBF-509;
small molecule antagonists targeting the androgen receptor, such as apalutamide, enzalutamide, flutamide, galatel, nilutamide, bicalutamide, leuprorelin, goserelin, ODM-201, APC-100, or ODM-204;
Small molecule inhibitors targeting arachidonic acid 5-lipoxygenase, such as meclofenamate sodium or zileuton;
small molecule inhibitors targeting ATR serine/threonine kinases such as BAY-937, ceralasertib (AZD6738), AZD6783, VX-803, or VX-970 (bezoar tryptib);
small molecule inhibitors targeting AXL receptor tyrosine kinase, such as bemcentinib (BGB-324), SLC-0211 or giritinib (AXL/Flt 3);
small molecule inhibitors targeting bruton' S tyrosine kinase (BTK), such as (S) -6-amino-9- (1- (but-2-alkynoyl) pyrrolidin-3-yl) -7- (4-phenoxyphenyl) -7H-purin-8 (9H) -one, acatinib (ACP-196), zetinib (BGB-3111), CB988, poisetinib (HM71224), ibrutinib (ibruvica), M-2951 (ibrutinib), tiraglutinib (ONO-4059), rizatritinib (PRN-1008), spetinib (CC-292), vicarib, ARQ-531(MK-1026), SHR-1459, dtrmxhws S-12, or TAS-5315;
small molecule inhibitors targeting neurotrophic receptor tyrosine kinases, such as larotinib, emtrictinib or selitestrinib (LOXO-195);
small molecule inhibitors targeting ROS protooncogene 1 receptor tyrosine kinase, such as enrotinib, reboectininib (TPX-0005), or loratinib;
Small molecule inhibitors targeting spleen-related tyrosine kinases, such as, for example, foxitinib (ASN-002), 6- (1H-indazol-6-yl) -N- (4-morpholinophenyl) imidazo [1,2-a ] pyrazin-8-amine, cerdulatinib, entotinib (entospletinib), fotatatinib disodium (R788, R406), HMPL-523, NVP-QAB 205AA, R112, or compounds disclosed in US8450321 or US 20150175616;
small molecule inhibitors targeting the SRC protooncogene non-receptor tyrosine kinase, such as VAL-201, terbinafine (KX2-391), or ibrutinib maleate (NS-018);
small molecule inhibitors targeting B-cell lymphoma 2, such as quintoclat (navitoclax) (ABT-263), venectal (venetaclax) (ABT-199, RG-7601), or AT-101 (gossypol);
small molecule inhibitors targeting the breakpoint cluster region, such as RG-7304;
small molecule inhibitors targeting bromodomain-and outer domain (BET) bromodomain-containing proteins such as ABBV-744, INCB-054329, INCB057643, AZD-5153, ABT-767, BMS-986158, CC-90010, NHWD-870, ODM-207, ZBC246, ZEN3694, CC-95775(FT-1101), mivebresib (mivebresib), BI-894999, PLX-2853, PLX-51107, CPI-0610, or GS-5829;
Small molecule inhibitors targeting carbohydrate sulfotransferase 15, such as STNM-01;
small molecule inhibitors targeting carbonic anhydrase, such as pamacoxib (polmacoxib), acetazolamide or methazolamide;
small molecule inhibitors targeting catenin β 1, such as CWP-291 or PRI-724;
small molecule antagonists targeting C-C motif chemokine receptors, such as CCX-872, BMS-813160(CCR2/CCR5) or MK-7690 (viriviroc);
small molecule inhibitors targeting cell division cycle 7, such as simuloresertib hydrate (TAK 931);
small molecule inhibitors targeting cereblon, such as atorvastatin (avadomide) (CC-122), CC-92480, CC-90009, or ibberdomide (iberdomide);
small molecule inhibitors targeting checkpoint kinase 1, such as SRA 737;
small molecule inhibitors targeting cluster of differentiation markers such as PBF-1662, BLZ-945, pemitinib (pemigatinib) (INCB-054828), BAY-1163877 (rogatinib)), AZD4547, FGF-401 (robitinib), quinitinib (quizartinib) dihydrochloride, SX-682, AZD-5069, PLX-9486, avatinib (avaritinib) (BLU-285), Ripidinib (DCC-2618), imatinib mesylate, JSP-191, BLU-263, CD117-ADC, AZD 9, tiratinib, volvulib (vorolinib), GO-203-2C, AB-680, PSB-1233979, PSB-12441, PSB-12425, HM-30181, HM-30181A, motixafortide, TG-24-25125, TG 24-25138, TG-362514 (TG-094), TG-2514-4P, TG 4-362518, and TG, Mavoroxaford (X4P-001-IO), plerixafor, CTX-5861 or REGN-5678(PSMA/CD 28);
Small molecule inhibitors targeting complement components, such as Imprime PGG (Biothera Pharmaceuticals);
small molecule inhibitors targeting C-X-C motif chemokine ligands (e.g., CXCL12), such as perortella (olapted pegol) (NOX-a 12);
small molecule inhibitors targeting cyclin-dependent kinases, such as palbociclib;
small molecule inhibitors targeting the cytochrome P450 family, such as ODM-209, LAE-201, sevieronel (VT-464), CFG920, abiraterone or abiraterone acetate;
small molecule inhibitors targeting DEAD box helicase 5, such as supinoxin (RX-5902);
small molecule inhibitors targeting the binding of diablo IAPs to mitochondrial proteins, such as BI-891065;
small molecule inhibitors targeting dihydrofolate reductase, such as pralatrexate or pemetrexed disodium;
small molecule inhibitors targeting DNA-dependent protein kinases, such as MSC2490484A (nedistertib), VX-984, AsiDNA (DT-01), LXS-196 or sotroparin;
small molecule inhibitors targeting mitogen-activated protein kinase such as rilitinib (ralimetinib), RG-7304, GS-4875 or GS-5290;
small molecule inhibitors targeting MARCKS, such as BIO-11006;
Small molecule inhibitors targeting RIPK1, such as GSK-3145094;
small molecule inhibitors targeting Rho-associated coiled-coil containing protein kinases, such as AT13148 or KD 025;
small molecule inhibitors targeting DNA topoisomerase such as irinotecan, pegafenac (fistecan pegol) or amrubicin;
small molecule inhibitors targeting dopamine receptor D2, such as ONC-201;
small molecule inhibitors targeting DOT 1-like histone lysine methyltransferases, such as pinometstat (EPZ-5676);
small molecule inhibitors targeting EZH2, such as tasetastat, CPI-1205 or PF-06821497;
small molecule inhibitors targeting fatty acid synthase, such as TVB-2640(Sagimet Biosciences);
small molecule inhibitors targeting fibroblast growth factor, such as bematuzumab (bemartuzumab) (FPA 144);
small molecule inhibitors targeting focal adhesion kinase 2, such as VS-4718, efatinib (deffectib), or GSK 2256098;
small molecule inhibitors targeting folate receptor 1, such as pralatrexate;
small molecule inhibitors targeting FOXM1, such as thiostrepton;
small molecule inhibitors targeting galectin 3, such as bepiastin (GR-MD-02);
Small molecule antagonists targeting glucocorticoid receptors, such as relacolan (relasporiant) (CORT-125134);
small molecule inhibitors targeting glutaminase include, but are not limited to CB-839 (telaglenastat) or bis-2- (5-phenylacetamido-1, 3, 4-thiadiazol-2-yl) ethyl sulfide (BPTES);
small molecule inhibitors targeting GNRHR, such as elargol, regorax (relugolix) or degarelix;
small molecule inhibitors targeting EPAS1, such as befetifan (belzutifan) (PT-2977(Merck & Co.));
small molecule inhibitors targeting IDO, such as alcazastat, linodostat (F-001287), PF-06840003, reminostat, linospestat (BMS-986205), EOS-200271 or KHK-2455;
small molecule inhibitors targeting isocitrate dehydrogenase (NADP (+)) such as Avenib (ivosidenb) (AG-120), Vorrinib (vorasidenb) (AG-881) (IDH1 and IDH2), IDH-305 or enzidipine (enasidenb) (AG-221);
small molecule inhibitors targeting Janus kinases such as ZD-1480, barrecinib (baricitinib), non-golitinib (filicinib) (GLPG0634), itacinib (itacinib) (INCB039110), molutetinib (momelotinib) (CYT0387), pefinitib (pefinitinib) (ASP015K), ruxotinib (ruxolitinib), tofacitinib (tofacitinib), INCB052793, ZD-1480, barrecinib (baritinib), phenanthroitinib (fedratinib), ganfinib (gandottinib) (LY2784544), letitinib (lertinib) (FLT 2/JAK 2/trtrt), molutetinib (CYT0387), monopatitinib (NS 018-018), cinia malactib (NS 15182/2);
KRAS-targeted small molecule inhibitors such as Sotolira cloth (sotoranib) (AMG-510), COTI-219, ARS-3248, WDB-178, BI-3406, BI-1701963, SML-8-73-1(G12C), Compound 3144(G12D), Kobe0065, Kobe/2602, (Ras GTP), RT11, or adaras raschib (adagrib) (MRTX-849);
small molecule inhibitors targeting lysine demethylase 1A, such as CC-90011;
small molecule inhibitors targeting MAPK-interacting serine/threonine kinases, such as tomivotettib (eFT-508);
small molecule inhibitors targeting the mechanistic target of rapamycin kinase (mTOR), such as daptomisib (dactolisib), omissib (omissib), voxilisib (voxtalisib), gedatolisib (gedatolisib), GSK2141795, avalisib (RG6114), saparsertib (sapanisibib), ME-344, sirolimus (oral nano-amorphous formulation, cancer), racemic tyrosine (TYME-88 (mTOR/cytochrome P4503A 4)), sirolimus, temsirolimus (mTOR/cytochrome P4503A 4)), (Tasirolimus)CCI-779), CC-115, onadotti (onatastib) (CC-223), SF-1126, PQR-309 (bimiralisib), Votaliseil or GSK-2126458;
small molecule inhibitors targeting the notch receptor, such as AL-101 (BMS-906024);
Small molecule inhibitors targeting the catalytic subunit of phosphatidylinositol-4, 5-bisphosphate 3-kinase, such as ASN-003, inacolisib (RG6114), Abelisib, and pentixib (pi)ctilisib) or idelixib
Small molecule inhibitors targeting polo-like kinase 1, such as volasertib (volasertib) or onvansertib (onvansertib);
small molecule inhibitors targeting poly (ADP-ribose) polymerase such as olaparib (MK7339), lucapanib (rucapanib), veliparib (veliparib), talapanib (talazoparib), ABT-767, pamiparib (BGB-290), fluazolarib (fluzolepali) (SHR-3162), nilapanib (JNJ-64091742), stenoparib (2X-121(e-7499)), cimiparib (simiparib), IMP-4297, SC-10914, IDX-1197, HWH-340, or CK 102;
small molecule inhibitors targeting polycombin EED, such as MAK 683;
small molecule inhibitors targeting porcupine O-acyltransferase, such as WNT-974;
small molecule antagonists targeting the progesterone receptor, such as onapristone;
small molecule inhibitors targeting prostaglandin-endoperoxide synthase such as HP-5000, lornoxicam, ketorolac tromethamine, bromfenac sodium, oxprenoxsule (ATB-346), moxifloxacin (mofezalac), GLY-230, TRK-700, diclofenac, meloxicam, parecoxib, etoricoxib, celecoxib, AXS-06, diclofenac potassium, reformulated celecoxib (DRGT-46), AAT-076, mexosulide (meisuoshuli), lumiracoxib, meloxicam, valdecoxib, zaltoprofen, nimesulide, anizafene, alicoxib, cimicib, deracoxib, flumizole, feloxicib, marvacoxib, pamigrel, parecoxib, robecoxib, rofecoxib, rutaecarpine, temacoxib, zaltoprofen or erexib (imreoxib);
Small molecule inhibitors targeting the protein arginine N methyltransferase, such as MS203, PF-06939999, GSK3368715 or GSK 3326595;
small molecule inhibitors targeting PTPN11, such as TNO155(SHP-099), RMC-4550, JAB-3068, RMC-4630(SAR442720), or the compounds disclosed in WO2018172984 or WO 2017211303;
small molecule inhibitors targeting Raf-1 protooncogenes, serine/threonine kinases, such as RG-7304;
small molecule antagonists targeting retinoic acid receptors, such as tamibarotene (SY-1425);
small molecule inhibitors targeting ribosomal protein S6 kinase B1, such as MSC 2363318A;
small molecule inhibitors targeting S100 calbindin a9, such as taquinomod;
small molecule inhibitors targeting selectin E, such as uproreselan sodium (GMI-1271);
small molecule inhibitors targeting SF3B1, such as H3B-8800;
small molecule inhibitors targeting Sirtuin-3, such as YC 8-02;
SMO-targeted small molecule inhibitors such as Sonidegib (sonidegib) ((R))Formerly known as LDE-225), vismodegib (vismodegib) (GDC-0449), glasdegib (PF-04449913), itraconazole, patridegib (patridegib), or taladegib (taladegib);
Small molecule antagonists targeting somatostatin receptors, such as OPS-201;
small molecule inhibitors targeting sphingosine kinase 2, such as opaganib (opaganib) ((ii))ABC294640);
Small molecule inhibitors targeting STAT3, such as napacacine (napabucain) (BBI-608);
small molecule inhibitors targeting tankyrase, such as G007-LK or stenoparib (2X-121 (e-7499));
small molecule inhibitors targeting TFGBR1, such as galinisertib, PF-06952229;
small molecule inhibitors targeting thymidylate synthase, such as idetrexed (ONX-0801);
small molecule inhibitors targeting tumor protein p53, such as CMG-097;
small molecule inhibitors targeting valosin containing proteins such as CB-5083;
small molecule inhibitors targeting WT1, such as ombipepimut-S (DSP-7888);
small molecule agonists that target adenosine receptors, such as AB928 or naminoson (namodenoson) (CF 102);
small molecule agonists targeting asparaginase, such as creitase (crisantapase)GRASPA (ERY-001, ERY-ASP), pergola-aspartase (calasapagase pegol) or permendiol;
small molecule agonists targeting CCAAT enhancer binding protein alpha, such as MTL-501;
Small molecule agonists targeting cluster of differentiation markers, such as Interleukin 2 receptor subunit γ, Eltepopapa, Retimod, Poly ICLC (NSC-301463), Riboxxon, Apoxxim, Abraxin, and,MCT-465, MCT-475, G100, PEPA-10, etonestrel alpha (eftozaneremin alfa) (ABBV-621), E-6887, mototimod (motolimod), resiquimod, selgantimod (selgantliomod) (GS-9688), VTX-1463, NKTR-262, AST-008, CMP-001, cototimod (cobiolimod), tisoimod (tilsotolomod), linimod (litemimod), MGN-1601, BB-006, IMO-8400, IMO-9200, atrophimod (agatolimod), DIMS-9054, DV-1079, lefitolimod (MGN-1703), CYT-003, or PUL-042;
small molecule agonists targeting the cytochrome P450 family, such as mitotane;
small molecule agonists targeting the DExD/H box helicase 58, such as RGT-100;
small molecule agonists targeting the GNRHR, such as leuprolide acetate, leuprolide acetate sustained release depot (ATRIGEL), triptorelin pamoate or goserelin acetate;
small molecule agonists targeting GRB2, such as progrebersen (BP 1001);
small molecule agonists targeting NFE2L2, such as omasolone (RTA-408);
Small molecule agonists targeting NOD2, such as mifamutide (liposomes);
small molecule agonists targeting PD-L1, such as CA-170, GS-4224, GS-4416, Lazetinib (GNS-1480; PD-L1/EGFR);
small molecule agonists targeting the progesterone receptor, such as norgestimate + ethinylestradiol (Tri-Cyclen) or levonorgestrel;
small molecule agonists targeting RAR-associated orphan receptor gamma, such as sintroger (LYC-55716);
small molecule agonists targeting retinoic acid receptors, such as tretinoin;
small molecule agonists targeting STING1, such as ADU-S100(MIW-815), SB-11285, MK-1454, SR-8291, AdVCA0848, GSK-532, SYN-STING, MSA-1, SR-8291, cyclic-gamp (cgamp), or cyclic-di-AMP;
small molecule agonists targeting thyroid hormone receptor beta, such as levothyroxine sodium;
a small molecule agonist targeting TLR7, such as BDB-001, DSP-0509, visatimod (versatolimod) (GS-9620), LHC-165, imiqimod (TMX-101), resiquimod, MCT-465, RG-7854, NKTR-262, or a compound disclosed in US20100143301, US20110098248, US20090047249, US20140045849, US20140073642, WO2014056953, WO2014076221, WO2014128189, US 20140350088031, WO2014023813, US20080234251, US 20080306306050 306050, US20100029585, US 201192485, US 01120112011201135, US20120082658 658, US20120219615, US 00620146432, US 00201485, US 0275167, or US 20130251673;
Small molecule agonists targeting tumor necrosis factor, such as tasolinamine;
KRAS-targeting inhibitory peptides such as KRpep-2(Ac-RRCPLYISYDPVCRR-NH2) or KRpep-2d (Ac-RRRRCPLYISYDPVCRRRR-NH 2);
antisense agents targeting AKT1, such as RX-0201;
antisense agents targeting the androgen receptor, such as IONIS-AR-2.5 Rx;
antisense agents targeting protein 5 containing baculovirus IAP repeats, such as EZN-3042;
antisense agents targeting GRB2, such as propyberson (presenigebersen);
antisense agents targeting heat shock protein 27, such as apatossen;
KRAS-targeting antisense agents, such as anti-KRAS U1 linker, siG12D-LODER TM Or siG12D exosomes;
antisense agents targeting STAT3, such as danvatissen (danvatissen) (IONIS-STAT3-2.5 Rx);
gene therapy targeting C-C motif chemokine receptors, such as SB-728-T;
interleukin-targeted gene therapy, such as EGENE-001, tavokinese telseplasmid, nogendehkin alpha (ALT-803), NKTR-255, NIZ-985(hetIL-15) or MDNA-55;
antibodies targeting CD47, such as CC-90002, Morelizumab (magrolimab) (Hu5F9-G4), AO-176(Vx-1004), IBI-188, Lyzorelizumab (lemzoprilumab) (TJC-4), SHR-1603, HLX-24, LQ-001, IMC-002, ZL-1201, IMM-01, B6H12, GenSci-059, TAY-018, TAY-240, 1F8-GMCSF, SY-102, KD-015, ALX-148, AK-117, TTI-621, TTI-622, or in WO199727873, WO 1999409940940, WO 20020120984, WO2005044857, WO2009046541, WO 201007919191624, WO 201201915702624, WO 20114391570250, WO 2013119752, WO 201311919191915720120120120120120120120120120172975778, WO 201201201201722017297423, WO 20120120120120120120172427242779, WO 20172975778, WO 20172979, WO 20120120172979, WO 20120120120120172979, WO 20120120020172979, WO 20120020172979, WO 20120020120020120020120020172979, WO 20120020120020120020120020120020120020120020120020120091579, WO 20120020120020120020120072979, WO 20120020120020120020120020120072979, WO 20120072979, WO 20120020120020120072979, WO 20120072979, WO 20120020120020120020120020120020120072979, WO 20120020120020120020120072979, WO 20120020120072979, WO 20120072979, WO 20120020120020120072979, WO 2012009, WO 2012002012002012002012002012002012002012002012009, WO 2012002012002012002012002012009, WO 2012009, WO 2012002012002012002012002002009, WO 2012002002002012002012002012002002002012002012002002009, WO 2012009, WO 2012002012009, WO 2012009, WO 2012002009, WO 2012002002002012002009, WO 2012002012002002009, WO 2012009, WO 2012002012002012009, WO 2012002012009, WO 2012009, WO 2012002012002012002012002012002012002012002012009, WO 2012009, WO 2012002012002012002012002012009,9750, WO 2012009,9750, WO 2012002012002012002012002012002012009, WO 2012002012002012009,9750, WO 2012009,9750, WO 2012002012009,9750, WO 2012009,9750, WO 2012009,9750,9750, WO 2012009,9750, WO 2012002012009,9750,9750, WO 2012009,9750, WO 2012002012009,9750, WO 2012009,9750, WO 2012002012002012009,9750, WO 2012009,, Compounds disclosed in WO2019201236, WO2019238012, WO2019241732, WO2020019135, WO2020036977, WO2020043188, WO 2020009725;
Antibodies targeting claudin 18, such as claudiximab;
antibodies targeting cluster of differentiation markers, such as temoxizumab (tafasitamab) (MOR 208; MorphoSys AG), einzelizumab (Inebilizumab) (MEDI-551), obinutuzumab (obinutuzumab), IGN-002, rituximab biosimilar (PF-05280586), vallizumab (varliumab) (CDX-1127), AFM-13(CD16/CD30), AMG330, ottotuzumab (otlertuzumab) (TRU-016), iximab (isatuximab), filzezumab (felferzamab) (MOR-202), TAK-079, TAK573, daratumumab (daratumumab)TTX-030, Seluzumab (selicrelumab) (RG7876), APX-005M, ABBV-428, ABBV-927, mizolizumab (mitazalimab) (JNJ-64457107), Ranjuzumab (lenzilucma), alemtuzumab (alemtuzumab), Emituzumab (emactuzumab), AMG-820, FPA-008 (cabiliazumab), PRS-343(CD-137/Her2), AFM-13(CD16/CD30), Belatumab molfopristine (Belatamab madoda madatin) (GSK-2857916), AFM 7 (BCMA/CD A), Xin Lonqu Ifupra alpha (56lukas alfa) (7461), Uluzumab (ureluuzumab), Utuzumab (Utuumu), Utuumu Luuku (AFE-05082566), HER-3573, FAP-7426-FAP-BEFAP-3673, FAP-744/FAP), FAP-3673, FAP-3/FAP-36343, FAP-343, FAP-3, FAP-BENb (FAP-3673, FAP-BENb) and FAP-3/FAP-E, CDX-0158, CDX-0159 and FSI-174, Riralizumab (relimab) (ONO-4482), LAG-525, MK-4280, fralimab (fianlimab) (REGN-3767), INCAN 2385, Ansarizumab (enceimab) (TSR-033), atituzumab, Brevivex (Mab-AR-20.5), MEDI-9447 (olelumab), CPX-006, IPH-53, BMS-986179, NZV-aRamI, CPI-006, PAT-SC1, liragumab (lirilumab) (IPH-2102), lacunab (lacuramab) (IPH-4102), Monralizumab (monelizumab), BAY-1834942, NEO-201 (ACAM 5/6), iodine (ACAMII) 131 (Aitustaitumumab) (IPH-4102), Monelizumab (monelizumab), and others b) (131I-BC8(lomab-B)), MEDI0562(tavolixizumab), GSK-3174998, INCAN 1949, BMS-986178, GBR-8383, ABBV-368, denosumab (denosumab), BION-1301, MK-4166, INCAN-1876, TRX-518, BMS-986156, MK-1248, GWN-323, CTB-006, INBRX-109, GEN-1029, Pepinemab (pepinemab) (VX-15), Vopril (vopraatelimiab) (JTX-2011), GSK3359609, cobellimab (cobilimab) (TSR-022), MBG-453, INCAN-2390, or a compound disclosed in WO 2017096179, WO2017096189 or WO 2018089276;
antibodies targeting clusterin, such as AB-16B 5;
antibodies targeting complement components, such as Rayleigh mab (ravulizumab) (ALXN-1210);
antibodies targeting CTLA4, such as ipilimumab, tremelimumab, BMS-986218, AGEN1181, zetilizumab (AGEN1884), BMS-986249, MK-1308, REGN-4659, ADU-1604, ipilimumab biosimilar (CS-1002), BCD-145, APL-509, JS-007, BA-3071, ONC-392, KN-044, CG-0161, BPI-002 or HBM-4003;
antibodies targeting C-X-C motif chemokine ligands, such as BMS-986253 (HuMax-Inflam);
antibodies targeting delta-like classical Notch ligand 4, such as dexecazumab (demcizumab);
Antibodies targeting the EPH receptor A3, such as fibatuzumab (KB-004);
antibodies targeting epidermal growth factor receptors, such as motuximab (modotuximab), cetuximab (cetuximab sarotalocan) (RM-1929), serurituzumab (seribant), neuximab, cetuximab mofetil (depatuzumab) (ABT-414), tolitumumab (tomotuximab), dextuximab (ABT-806), or cetuximab;
antibodies targeting epithelial cell adhesion molecules, such as mototuzumab (oportuzumab monatx) (VB 4-845);
antibodies targeting fibroblast growth factors, such as GAL-F2, B-701 (voravamab);
antibodies targeting hepatocyte growth factor, such as MP-0250;
antibodies targeting interleukins, such as canakinumab (ACZ885), gemuzumab (gevokizumab) (VPM087), CJM-112, gucekumab (gusekumab), taltuzumab (talacotuzumab) (JNJ-56022473), cetuximab, or toluzumab;
antibodies targeting LRRC15, such as ABBV-085 or trastuzumab (ARGX-110);
antibodies targeting mesothelin, such as BMS-986148, SEL-403 or anti-MSLN-MMAE;
Antibodies targeting MET, such as vildagliptin-tertuzumab (teliostuzumab vedotin) (ABBV-399);
antibodies targeting myostatin, such as dolizumab (landogrozumab);
antibodies targeting notch receptors, such as tarrituximab (tarextumab);
antibodies targeting PD-1, such as Pabollizumab, nivolumab, cimeprinizumab, pidilizumab, AMG-404, MEDI0680(AMP-514), atilizumab, Duvaliuzumab, Coxilizumab (cosibelizumab) (CK-301), Saxalizumab (sasanlizumab) (PF-06801591), BGB-A317 (tirelelizumab), Serratizumab (WBP-3055), AK-103(HX-008), AK-105, CS-10, HLX-10, Rifflumab (retifanlizumab) (MGA-012), BI-754091, Batlizumab (balstinilimab) (AG2034), JS-001 (terilizumab), Celizumab (cetriluzumab) (cetrij 63723283), Germin-39501), and Jiejimab (LZJ-009-501), Palivizumab (prolelimab) (BCD-100), lodalizumab (lodapolimab) (LY-3300054), SHR-1201, SHR-1210 (camerelizumab), Sym-021, buglimab (budigallimab) (ABBV-181), BAT-1306, Avluzumab (MSB0010718C), CX-072, CBT-502, TSR-042 (dolastalizab)), MSB-2311, JTX-4014, BGB-A333, SHR-1316, CS-1001(WBP-3155), enwoolimab (enfolimab) (KN-035), IBIDlizumab I-308 (IBindilizumab (sintilimab)), HLX-20, KL-A167, STI-1014, STI-A1015(IMC-001), FALgolizmab (TQD-135, TQTQZ-135, TQARX-050, MDTArX-0881, MDX-1105 (ABBV-181), and SALvX-3155), Or a compound disclosed in WO2018195321, WO2020014643, WO2019160882 or WO 2018195321;
An antibody targeting TGFB1, such as SAR439459, ABBV-151, NIS793, XOMA 089, or a compound disclosed in WO 2019103203;
antibodies targeting vascular endothelial growth factor a, such as bevacizumab, valnoulli-mab (vanucizumab) or faricimab (faricimab);
bispecific antibodies targeting CD47, such as IBI-322(CD47/PD-L1), IMM-0306(CD47/CD20), TJ-L1C4(CD47/PD-L1), HX-009(CD47/PD-1), PMC-122(CD47/PD-L1), PT-217, (CD 47/3), IMM-26011(CD47/FLT3), IMM-0207(CD47/VEGF), IMM-2902(CD47/HER2), BH29xx (CD47/PD-L1), IMM-03(CD47/CD20), IMM-2502(CD47/PD-L1), HMBD-004B (CD47/BCMA), HMBD-NI-004A (CD47/CD33), TG-1801 (TG-1801) or TG-1801;
bispecific antibodies targeting PD-1 such as PF-06936308(PD-1/CTLA4), MGD-013(PD-1/LAG-3), RO-7247669(PD-1/LAG-3), MGD-019(PD-1/CTLA4), KN-046(PD-1/CTLA4), MEDI-5752(CTLA4/PD-1), RO-7121661(PD-1/TIM-3), XmAb-20717(PD-1/CTLA4), CTLA-104 (CTLA4/PD-1), RG7769(PD-1/TIM-3) or TAK-252(PD-1/OX 40L);
bispecific antibodies targeting PD-L1, such as FS-118(LAG-3/PD-L1), FPT-155(CTLA4/PD-L1/CD28), GEN-1046(PD-L1/4-1BB), bintrafusisp alpha (M7824(PD-L1/TGF β -EC domain)), CDX-527(CD27/PD-L1) or INBRX-105(4-1BB/PDL 1);
T-cell adapters (targeting CD3), such as AMG-160(PSMA/CD3), AMG-212(PSMA/CD3), AMG-330(CD33/CD3), AMG-420(BCMA/CD3), AMG-427(FLT3/CD3), AMG-562(CD19/CD3), AMG-596(EGFRvIII/CD3), AMG-673(CD33/CD3), AMG-701(BCMA/CD3), AMG-757(DLL3/CD3), AMG-211(CEA/CD3),(CD19/CD3), RG7802(CEA/CD3), ERY-974(CD3/GPC3), huGD2-BsAb (CD3/GD2), PF-06671008 (cadherins)APVO436(CD123/CD3), ERY974, trastuzumab (flotetuzumab) (CD123/CD3), GEM333(CD3/CD 3), GEMoab (CD3/PSCA), REGN-1979(CD 3/CD3), MCLA-117(CD3/CLEC12 3), JNJ-0819, JNJ-7564(CD 3/heme), JNJ-3 (CD123/CD3), MGD-007(CD3/gpA 3), MGD-009(CD3/B7H3), IMCgp100(CD3/gp100), XmAb-14045(CD123/CD3), XmAb-13676(CD3/CD 3), Katuotuzumab (CD 3/CAM), CD 4018 (CD3/CD 40172), MUGN/361346, CD 611346, CD 60RG-3, GRRG-3/3660272), GRRG 3/3, GRRG 3/3660272, GRRG 3/3, GRRG 3/3660272, CD3, GRRG 3, CD3, GRRG 3660272, CD3, GRRG 3, GRC 3, GRRG 3, CD 3660272, CD3, GRRG 3, CD3, GRRG 3660272, GRRG 3, GRC 3, GRRG 3, GRC 3, GRRG 3, GRC 3660272, CD3, GRRG 3, GRC 3, GRRG 3, CD 3660272, GRRG 3, CD3, GRRG 3, CD 3660272, CD3, CD 3660272, CD3, GRRG 3, CD3, GRRG 3660272, CD3, GRRG 3, GEM-333(CD3/CD33), PF-06863135(BCMA/CD3), SAR440234(CD3/CDw123), JNJ-9383(MGD-015), bornauzumab (blinatumomab) (CD19/CD3), obinutuzumab (odronextamab) (REGN-1979(CD20xCD3)), palisadumab (PLAMOTAMAb) (XmAb-13676(CD3/CD20)), moseutuzumab (RG-7828(CD20/CD3)), AMG-424(CD38/CD3), tituzumab (tidutamab) (XmAb-18087(SSTR2/CD3)) or tuotuzumab (duvoruzumab) (JNJ-64052781) (CD19/CD 3);
Vaccines targeting fms-related receptor tyrosine kinases, such as HLA-a2402/HLA-a0201 restricted epitope peptide vaccines;
vaccines targeting heat shock protein 27, such as PSV-aml (phosphosynvax);
vaccines targeting PD-L1, such as IO-120+ IO-103(PD-L1/PD-L2 vaccine) or IO-103;
vaccines targeting the tumor protein p53, such as MVA-p 53;
a vaccine targeting WT1, such as a WT-1 analog peptide vaccine (WT 1-CTL);
cell therapy targeting protein 5 containing baculovirus IAP repeats such as tumor lysate/MUC 1/survivin PepTivator loaded dendritic cell vaccine;
cell therapies targeting carbonic anhydrase, such as DC-Ad-GMCAIX;
cell therapies targeting C-C motif chemokine receptors, such as CCR 5-SBC-728-HSPC;
cell therapies targeting cluster of differentiation markers include, but are not limited to, CD19-ARTEMIS, TBI-1501, CTL-119huCART-19T cells, liso-cel, Likimersen (lisocabtagen)e maraaleucel) (JCAR-017), Alferulacee ciloleucel (kTE-C19,) Ackeratex (KTE-X19), US7741465, US6319494, UCART-19, Taberencel (EBV-CTL), T esceracil (tisagenleceucel) -T (CTL019), T cells expressing CD19CAR-CD28-CD3 ζ -EGFRT, CAR T cell therapy of CD19/4-1BBL armoring, C-CAR-011, CIK-CAR.CD19, CD19 CAR-28-zeta T cells, PCAR-019, MatchT, DSCAR-01, IM 19-T, TC-110, anti-CD 19CAR T cell therapy (B cell acute lymphoblastic leukemia, Malaysia national University (Univasular University)), anti-CD 19T cell therapy (acute lymphoblastic leukemia/non-German Hodgmental CAR lymphoma, Hodgkin's Hocquification-II-Hodgrain), anti-CD 19, cancer, Shanghai Ukadi biomedical science and Technology Limited (Shanghai Unicar-Therapy Bio-medicine Technology)), MB-CART2019.1(CD19/CD20), GC-197(CD19/CD7), CLIC-1901, ET-019003, anti-CD 19-STAR-T cells, AVA-001, BCMA-CD19cCAR (CD19/APRIL), ICG-134, ICG-132(CD19/CD20), CTA-101, WZTL-002, dual anti-CD 19/anti-CD 20CAR T cells (chronic lymphocytic leukemia/B cell lymphoma), HY-001, ET-019002, YTB-323, GC-012(CD19/APRIL), GC-36ICT (CD19/CD22), zeta Tn/Tn expressing CD 19-CD 28-CD3 zeta, RtR-9, RtR-014, RtC-014, GC-022, GC-014, GC-19/BCG-9, and GC-01410, PTG-01, CC-97540, GC-007G, TC-310, GC-197, tesalasin-T, CART-19, tesalasin (CTL-019)), anti-CD 20CAR T cell therapy (non-Hodgkin's lymphoma), MB-CART2019.1(CD19/CD20), WZTL-002 dual anti-CD 19/anti-CD 20CAR-T cells, ICG-132(CD19/CD20), ACTR707ATTCK-20, PBCA-20A, LB-1905, CIK-CAR.CD33, CD33CART, dual anti-BCMA/anti-CD 38CAR T cell therapy, CART-ddBCMA, MB-102, IM-23, JEZ-567, UCART-123, PD-1 knock-out T cell therapy (esophageal cancer/NSCLC), ICTAT-052, Tn-1-PD-T, ICTCAR-053, CAR-1 knock out T cell therapy (NSCLC/NSCLC), AUTO-2, anti-BCMA CAR T cell therapy, Descarats-011, anti-BCMA/anti-CD 38CAR T cell therapy, CART-ddBCMA, BCMA-CS1cCAR, CYAD-01(NKG2D LIGAN) D MODULATOR), KD-045, PD-L1t-haNK, BCMA-CS1cCAR, MEDI5083, anti-CD 276CART or a therapy disclosed in WO2012079000 or WO 2017049166;
(ii) a cell therapy targeting folate hydrolase 1, such as CIK-car. PSMA or CART-PSMA-TGF β RDN;
cell therapy targeting GSTP1, such as CPG3-car (glycar);
cell therapy targeting HLA-a, such as FH-MCVA2TCR or NeoTCR-P1;
interleukin-targeted cell therapies, such as CST-101;
KRAS-targeted cell therapies, such as anti-KRAS G12D mTCR PBL;
cell therapies targeting MET, such as anti-cMet RNA CAR T;
cell therapies targeting MUC16, such as JCAR-020;
cell therapy targeting PD-1, such as PD-1 knock-out T cell therapy (esophageal cancer/NSCLC);
cell therapies targeting PRAME, such as BPX-701;
cell therapy targeting the transforming protein E7, such as kit-439;
a cell therapy targeting WT1, such as WT1-CTL, ASP-7517, or JTCR-016;
exemplary combination therapies
Combination therapy for lymphoma or leukemia
Some chemotherapeutic agents are suitable for treating lymphoma or leukemia. These agents include aldesleukin, avocadi (alvocidib), amifostine trihydrate, aminocamptothecin, antineodone A10, antineodone AS2-1, antithymocyte globulin, arsenic trioxide, Bcl-2 family protein inhibitor ABT-263, beta-arrhine (beta-alethine), BMS-345541, bortezomib Bortezomib (b)PS-341), MossBryoid 1, Bulsulfan, Campats-1H, carboplatin, and carfilzomibCarmustine, caspofungin acetate, CC-5103, chlorambucil, CHOP (cyclophosphamide, doxorubicin, vincristine and prednisone), cisplatin, cladribine, clofarabine, curcumin, CVP (cyclophosphamide, vincristine and prednisone), cyclophosphamide, cyclosporine, cytarabine, dinil interleukin (denileukin difitox), dexamethasone, docetaxel, urodoline 10, doxorubicin hydrochloride, DT-PACE (dexamethasone, thalidomide, cisplatin, doxorubicin, cyclophosphamide and etoposide), enzal, alfacastin, etoposide, everolimus (RAD001), FCM (fludarabine, cyclophosphamide and mitoxantrone), FCR (fludarabine, cyclophosphamide and rituximab), fenvirginine, filgrastim, lattuzumab (flavopirol), fludarabine, ritabine (fludarabine), and rituximab (fludarabine), Geldanamycin (17AAG), HyperCVAD (super-fractionated cyclophosphamide, vincristine, doxorubicin, dexamethasone, methotrexate and cytarabine), ICE (ifosfamide, carboplatin and etoposide), ifosfamide, irinotecan hydrochloride, interferon alpha-2 b, ixabepilone, lenalidomide (R) ((R)) CC-5013), lymphokine-activated killer cells, MCP (mitoxantrone, chlorambucil, and prednisolone), melphalan, mesna, methotrexate, mitoxantrone hydrochloride, motoxafen gadolinium, mycophenolate mofetil, nelarabine, obacara (obatoclatax) (GX15-070), oblimersen (oblimersen), octreotide acetate, omega-3 fatty acids, Omr-IgG-am (WNIG, Omrix), oxaliplatin, paclitaxel, pabuxib (PD 2903391), pegylated filgrastim, pegylated liposomal doxorubicin hydrochloride, perifosine, prednisolone, prednisone, recombinant flt3 ligand, recombinant human thrombopoietin, recombinant interferon alpha, recombinant interleukin-11, recombinant interleukin-12, rituximab, R-CHOP (rituximab and CHOP),R-CVP (Rituximab and CVP), R-FCM (Rituximab and FCM), R-ICE (Rituximab and ICE) and R-MCP (Rituximab and MCP), Roscovitine (R-roscovitine) (Selixivib), CYC202), sargrastim, sildenafil citrate, simvastatin, sirolimus, styryl sulfone, tacrolimus, Tacrolimus (tanespimacin), temsirolimus (CCl-779), thalidomide, therapeutic allogenic lymphocytes, Titipiper, tipifarnib, vincristine sulfate, vinorelbine ditartrate, SAHA (suberoylanilide hydroxamic acid or suberoyl, aniline and hydroxamic acid), Vemurafenib Venetork (ABT-199).
An improved method is radioimmunotherapy, in which monoclonal antibodies are combined with radioisotope particles (such as indium-111, yttrium-90, and iodine-131). Examples of combination therapies include, but are not limited to, iodine-131 tositumomabYttrium-90 ibritumomab tiuxetanAndand CHOP.
The above-described therapies may be supplemented with or combined with stem cell transplantation or therapy. Therapeutic procedures include peripheral blood stem cell transplantation, autologous hematopoietic stem cell transplantation, autologous bone marrow transplantation, antibody therapy, biological therapy, enzyme inhibitor therapy, systemic irradiation, stem cell infusion, bone marrow ablation with stem cell support, peripheral blood stem cell transplantation with extracorporeal treatment, cord blood transplantation, immunoenzymatic techniques, low LET cobalt-60 gamma ray therapy, bleomycin, routine surgery, radiotherapy and non-myeloablative allogeneic hematopoietic stem cell transplantation.
Combination therapy for non-hodgkin's lymphoma
Treatment of non-hodgkin's lymphoma (NHL), especially those of B-cell origin, includes the use of monoclonal antibodies, standard chemotherapy approaches (e.g., CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone), CVP (cyclophosphamide, vincristine, and prednisone), FCM (fludarabine, cyclophosphamide, and mitoxantrone), MCP (mitoxantrone, chlorambucil, prednisolone), all optionally including rituximab (R), etc.), radioimmunotherapy, and combinations thereof, especially integration of antibody therapy with chemotherapy.
Examples of unconjugated monoclonal antibodies for treating NHL/B cell cancers include rituximab, alemtuzumab, human or humanized anti-CD 20 antibody, lumiximab (lumiximab), anti-TNF related apoptosis inducing ligand (anti-TRAIL), bevacizumab (bevacizumab), galiximab (galiximab), epratuzumab (epratuzumab), SGN-40, and anti-CD 74.
Examples of experimental antibody agents for treating NHL/B cell cancers include ofatumumab (ofatumumab), ha20, PRO131921, alemtuzumab, galiximab, SGN-40, CHIR-12.12, epratuzumab, lumiximab, aprezumab (apolizumab), milatuzumab (milatuzumab), and bevacizumab.
Examples of standard regimens for chemotherapy of NHL/B cell cancers include CHOP, FCM, CVP, MCP, R-CHOP (Rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone), R-FCM, R-CVP, and R MCP.
Examples of radioimmunotherapy for NHL/B cell cancers include yttrium-90 ibritumomab tiuxetanAnd iodine-131 tositumomab
Combined therapy for mantle cell lymphoma
Therapeutic treatment of Mantle Cell Lymphoma (MCL) includes combination chemotherapy, such as CHOP, hyperCVAD, and FCM. These regimens may also be supplemented with the monoclonal antibody rituximab to form combination therapies R-CHOP, hypercVAD-R and R-FCM. Any of the above therapies can be combined with stem cell transplantation or ICE to treat MCL.
An alternative method of treating MCL is immunotherapy. One immunotherapy uses monoclonal antibodies such as rituximab. Another use is for cancer vaccines, such as GTOP-99, which are based on the genetic makeup of individual patient tumors.
An improved method of treating MCL is radioimmunotherapy, in which monoclonal antibodies are combined with radioisotope particles (such as iodine-131 tositumomab)And yttrium-90 ibritumomab tiuxetanAnd (4) combining. In a further example of the use of the present invention,for sequential treatment with CHOP.
Other methods of treating MCL include autologous stem cell transplantation in combination with high dose chemotherapy, administration of proteasome inhibitors such as bortezomib (r) ((r))Or PS-341) or administering an anti-angiogenic agent such as thalidomide, particularly in combination with rituximab.
Another therapeutic approach is the administration of drugs that cause degradation of Bcl-2 protein and increase the sensitivity of cancer cells to chemotherapy (such as olymerson) in combination with other chemotherapeutic agents.
Another method of treatment involves administration of an mTOR inhibitor that can result in inhibition of cell growth and even cell death. Non-limiting examples are sirolimus, temsirolimus (CCI-779), CC-115, CC-223, SF-1126, PQR-309 (bimiralisib), Wotassib (voxtalisib), GSK-2126458 and Or other chemotherapeutic agent combinations of temsirolimus.
Other recent MCL therapies have been disclosed. Examples of this include frataxin, palbociclib (PD0332991), R-roscovitine (Celisib, CYC202), styrylsulfone, olbara (GX15-070), TRAIL, anti-TRAIL death receptor DR4 and DR5 antibodies, temsirolimus (Tryprolimus: (R))CCl-779), everolimus (RAD001), BMS-345541, curcumin, SAHA, thalidomide, lenalidomide (R) ((R)CC-5013) and geldanamycin (17 AAG).
Combination therapy for Fahrenheit macroglobulinemia
Therapeutic agents for the treatment of Fahrenheit macroglobulinemia (WM) include aldesleukin, alemtuzumab, avoxidil, amifostine trihydrate, aminocamptothecin, antineoplastic A10, antineoplastic AS2-1, antithymocyte globulin, arsenic trioxide, HSPPC-96 from autologous human tumor sources, Bcl-2 family protein inhibitor ABT-263, beta-irclein, bortezomibBryodin 1, busufen, caparse-1H, carboplatin, carmustine, caspofungin acetate, CC-5103, cisplatin, clofarabine, cyclophosphamide, cyclosporine, cytarabine, dinil interleukin, dexamethasone, docetaxel, urodolastatin 10, doxorubicin hydrochloride, DT-PACE, enzastalin, alfa epoetin, epratuzumab (hLL 2-anti-CD 22 humanized antibody), etoposide, everolimus, fenretinide, filgrastim, fludarabine, ibrutinib, and isophosphoric acid Amines, indium-111 monoclonal antibody MN-14, iodine-131 tositumomab, irinotecan hydrochloride, ixabepilone, lymphokine-activated killer cells, melphalan, mesna, methotrexate, mitoxantrone hydrochloride, monoclonal antibody CD19 (such as tisagenleceucel-T, CART-19, CTL-019), monoclonal antibody CD20, motoxafen gadolinium, mycophenolate mofetil, nelarabine, oblimerson, octreotide acetate, omega-3 fatty acids, oxaliplatin, paclitaxel, pefilgrastim, PEGylated liposomal doxorubicin hydrochloride, pentostatin, parylene, prednisone, recombinant flt3 ligand, recombinant human thrombopoietin, recombinant interferon alpha, recombinant interleukin-11, recombinant interleukin-12, rituximab, sargrastim, sildenafil citrateSimvastatin, sirolimus, tacrolimus, temspiramycin, thalidomide, therapeutic allogenic lymphocytes, thiotepa, tipifarnib, tositumomab, eculumab (ulocuplumab), veltuzumab (veltuzumab), vincristine sulfate, vinorelbine ditartrate, vorinostat, WT 1126-134 peptide vaccine, WT-1 analog peptide vaccine, yttrium-90 ibritumomab, yttrium-90 humanized epratuzumab, and any combination thereof.
Examples of therapeutic procedures for treating WM include peripheral blood stem cell transplantation, autologous hematopoietic stem cell transplantation, autologous bone marrow transplantation, antibody therapy, biological therapy, enzyme inhibitor therapy, systemic irradiation, stem cell infusion, bone marrow ablation with stem cell support, ex vivo treated peripheral blood stem cell transplantation, umbilical cord blood transplantation, immunoenzymatic techniques, low LET cobalt-60 gamma ray therapy, bleomycin, general surgery, radiation therapy, and non-bone marrow ablative allogeneic hematopoietic stem cell transplantation.
Combination therapy for diffuse large B-cell lymphoma
Therapeutic agents for the treatment of diffuse large B-cell lymphoma (DLBCL) include cyclophosphamide, doxorubicin, vincristine, prednisone, an anti-CD 20 monoclonal antibody, etoposide, bleomycin, many of the agents listed for WM, and any combination thereof, such as ICE and RICE.
Combination therapy for chronic lymphocytic leukemia
Examples of therapeutic agents for the treatment of Chronic Lymphocytic Leukemia (CLL) include chlorambucil, cyclophosphamide, fludarabine, pentostatin, cladribine, doxorubicin, vincristine, prednisone, prednisolone, alemtuzumab, many of the agents listed for WM, as well as combination chemotherapies and chemoimmunotherapies, including the following common combination regimens: CVP, R-CVP, ICE, R-ICE, FCR, and FR.
Combination therapy for breast cancer
Therapeutic agents for treating breast cancer include albumin-bound paclitaxel, anastrozole, amitrazumab, capecitabine, carboplatin, cisplatin, cyclophosphamide, docetaxel, doxorubicin, epirubicin, everolimus, exemestane, fluorouracil, fulvestrant, gemcitabine, ixabepilone, lapatinib, letrozole, methotrexate, mitoxantrone, paclitaxel, pegylated liposomal doxorubicin, pertuzumab, tamoxifen, toremifene, trastuzumab, vinorelbine, and any combination thereof.
Triple negative breast cancer combination therapy
Therapeutic agents useful for treating triple negative breast cancer include amitrazumab, cyclophosphamide, docetaxel, doxorubicin, epirubicin, fluorouracil, paclitaxel, and combinations thereof.
Combined therapy for colorectal cancer
Therapeutic agents for treating colorectal cancer include bevacizumab, capecitabine, cetuximab (cetuximab), fluorouracil, irinotecan, leucovorin, oxaliplatin, panitumumab (panitumumab), aflibercept, and any combination thereof.
Combined therapy for esophageal and esophagogastric junction cancer
Therapeutic agents useful for treating esophageal and esophageal-gastric junction cancer include capecitabine, carboplatin, cisplatin, docetaxel, epirubicin, fluoropyrimidine, fluorouracil, irinotecan, leucovorin, oxaliplatin, paclitaxel, ramucirumab (ramucirumab), trastuzumab, and any combination thereof.
Combination therapy for gastric cancer
Therapeutic agents for the treatment of gastric cancer include capecitabine, carboplatin, cisplatin, docetaxel, epirubicin, fluoropyrimidine, fluorouracil, irinotecan, leucovorin, mitomycin, oxaliplatin, paclitaxel, ramucirumab, trastuzumab, and any combination thereof.
Combination therapy for head and neck cancer
Therapeutic agents for the treatment of head and neck cancer include afatinib, bleomycin, capecitabine, carboplatin, cetuximab, cisplatin, docetaxel, fluorouracil, gemcitabine, hydroxyurea, methotrexate, nivolumab, paclitaxel, palbociclumab, vinorelbine, and any combination thereof.
Combination therapy for non-small cell lung cancer
Therapeutic agents for treating non-small cell lung cancer (NSCLC) include afatinib, albumin-bound paclitaxel, eritinib, atilizumab, bevacizumab, cabozantinib, carboplatin, cisplatin, crizotinib, dabrafenib, docetaxel, erlotinib, etoposide, gemcitabine, nivolumab, paclitaxel, palbociclib, pemetrexed, ramucirumab, tremelimumab, trastuzumab, vandetanib, vemurafenib, vinblastine, vinorelbine, and any combination thereof.
Combination therapy for small cell lung cancer
Therapeutic agents useful for treating Small Cell Lung Cancer (SCLC) include amitrazumab, bendamustine, carboplatin, cisplatin, cyclophosphamide, docetaxel, doxorubicin, etoposide, gemcitabine, ipilimumab (ipilimumab), irinotecan, nivolumab, paclitaxel, temozolomide, topotecan, vincristine, vinorelbine, and any combination thereof.
Combination therapy for ovarian cancer
Therapeutic agents useful for treating ovarian cancer include 5-fluorouracil, albumin-bound paclitaxel, altretamine, anastrozole, bevacizumab, capecitabine, carboplatin, cisplatin, cyclophosphamide, docetaxel, doxorubicin, etoposide, exemestane, gemcitabine, ifosfamide, irinotecan, letrozole, leuprolide acetate, liposomal doxorubicin, megestrol acetate, melphalan, olaparib, oxaliplatin, paclitaxel, pazopanib, pemetrexed, tamoxifen, topotecan, vinorelbine, and any combination thereof.
Additional exemplary combination therapies
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with one or more therapeutic agents selected from Trop-2 binding agents, CD47 antagonists, sirpa antagonists, FLT3R agonists, PD-1 antagonists, PD-L1 antagonists, MCL1 inhibitors, CCR8 binding agents, HPK1 antagonists, DGKa inhibitors, CD73 inhibitors, adenosine receptor antagonists, TIGIT antagonists, TREM1 binding agents, TREM2 binding agents, TGF β (e.g., TGF β 1 or TGF β 3) binding agents, and CAR-T cell therapy.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with one or more therapeutic agents selected from an anti-Trop-2 antibody (e.g., sarcetuzumab gavitin, SKB-264, JS-108(DAC-002), dapotuzumab deluten, BAT-8003), an anti-CD 47 antibody or CD47 blocker (e.g., moloprizumab, DSP-107, AO-176, ALX-148, IBI-188, letuzumab, TTI-621, TTI-622), an anti-sirpa antibody (e.g., GS-0189), an FLT3L-Fc fusion protein (e.g., GS-3583), an anti-PD-1 antibody (pabulizumab, nivolumab, nigerumab), a small molecule-L1 inhibitor (e.g., PD, GS-4224), anti-PD-L1 antibodies (e.g., acilizumab), small molecule MCL1 inhibitors (e.g., GS-9716), small molecule HPK1 inhibitors (e.g., GS-6451), HPK1 degraders (PROTAC; e.g., ARV-766), small molecule DGKa inhibitors, small molecule CD73 inhibitors (e.g., AB680), anti-CD 73 antibodies (e.g., olaratumab), Dual A 2a /A 2b Adenosine receptor antagonists (e.g., itraconazole (AB928)), anti-TIGIT antibodies (e.g., tegraleigh mab, vibralizumab, donralizumab, AB308), anti-TREM 1 antibodies (e.g., PY159), anti-TREM 2 antibodies (e.g., PY314), TGF β -capturing agents (e.g., bintrafusip alpha, age-1423), anti-TGF β 1 antibodies (e.g., SRK-181), and CAR-T cell therapy (e.g., aliskiren, brij oritem, tesseram).
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with an anti-PD-1 antibody and a small molecule PD-L1 inhibitor or an anti-PD-L1 antibody (e.g., avilizumab) and an anti-TIGIT antibody.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are combined with an anti-PD-1 antibody and a dual a 2a /A 2b The adenosine receptor antagonist is administered together. In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are combined with an anti-PD-1 antibody, a small molecule CD73 inhibitor, and a dual a 2a /A 2b The adenosine receptor antagonist is administered together.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with one or more therapeutic agents selected from the group consisting of safituotuzumab gavatikukan-hziy, mololizumab, GS-0189, GS-3583, seraprimab, GS-4224, GS-9716, GS-6451, AB680, eltrina (AB928), donalizumab, AB308, PY159, PY314, SRK-181, argenzasirox, and briguerite.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with saritumumab gavatinib-hziy.
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with mololizumab.
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with the serpalizumab.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with serolizumab and dinolizumab.
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with serolizumab and eltromazine (AB 928).
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with one or more therapeutic agents selected from the group consisting of serolizumab, AB680, and eltromazine (AB 928).
In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with aliskiren and brij.
In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with a FLT3R agonist. In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with FLT3 ligand. In some embodiments, an antibody and/or fusion protein provided herein (e.g., 7-B16) is administered with FLT3L-Fc fusion protein (e.g., as described in WO 2020263830). In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with GS-3583 or CDX-301. In some embodiments, the antibodies and/or fusion proteins provided herein (e.g., 7-B16) are administered with GS-3583.
Diagnostic use
Provided herein are methods of using antibodies and/or fusion proteins (e.g., 7-B16, 1-K17, etc.), polypeptides, and polynucleotides to detect, diagnose, and monitor diseases, disorders, or conditions associated with CCR8 expression (increased or decreased relative to normal samples, and/or inappropriate expression, such as expression present in tissues and/or cells that normally lack epitope expression). Provided herein are methods of determining whether a patient will respond to treatment with an antibody and/or fusion protein provided herein.
In some embodiments, the method comprises detecting whether the patient has cells expressing CCR8 using an anti-CCR 8 antibody. In some embodiments, the detection method comprises contacting the sample with an antibody, polypeptide, or polynucleotide and determining whether the level of binding is different from the level of binding of a reference or comparative sample (such as a control). In some embodiments, the methods can be used to determine whether an antibody or polypeptide described herein is an appropriate treatment for a subject.
In some embodiments, the cells or cell/tissue lysates are contacted with an anti-CCR 8 antibody and binding between the antibody and the cells is determined. When a test cell exhibits binding activity as compared to a reference cell of the same tissue type, it can indicate that the subject will benefit from treatment with the antibodies and/or fusion proteins provided herein. In some embodiments, the test cells are from human tissue. In some embodiments, the test cells are from human blood.
Various methods known in the art for detecting specific antibody-antigen binding can be used. Exemplary immunoassays that can be performed include Fluorescence Polarization Immunoassay (FPIA), Fluorescence Immunoassay (FIA), Enzyme Immunoassay (EIA), turbidity inhibition immunoassay (NIA), enzyme-linked immunosorbent assay (ELISA), and Radioimmunoassay (RIA). Indicator moieties or labeling groups can be attached to the subject antibodies and selected to meet the needs of various uses of the method, which are typically dictated by the availability of assay equipment and compatible immunoassay procedures. Suitable labels include, but are not limited to, radionuclides (e.g., radionuclides) 125 I、 131 I、 35 S、 3 H or 32 P), an enzyme (e.g., alkaline phosphatase, horseradish peroxidase, luciferase, or beta-galactosidase), a fluorescent moiety or protein (e.g., fluorescein, rhodamine, phycoerythrin, GFP, or BFP), or a luminescent moiety (e.g., Qdot supplied by Quantum Dot Corporation (Palo Alto, Calif.) TM Nanoparticles). General techniques for performing the various immunoassays described above are known to those of ordinary skill in the art.
For diagnostic purposes, polypeptides comprising antibodies may be labeled with detectable moieties including, but not limited to, radioisotopes, fluorescent labels, and various enzyme-substrate labels known in the art. Methods of conjugating labels to antibodies are known in the art.
In some embodiments, the anti-CCR 8 antibody need not be labeled, and its presence can be detected using a second labeled antibody that binds to the first anti-CCR 8 antibody.
In some embodiments, the anti-CCR 8 antibodies can be used in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays. Zola, Monoclonal Antibodies A Manual of Techniques, pp.147-.
anti-CCR 8 antibodies and polypeptides may also be used in vivo diagnostic assays, such as in vivo imaging. Typically, the antibody or polypeptide is administered with a radionuclide (such as 111 In、 99 Tc、 14 C、 131 I、 125 I、 3 H or any other radionuclide label, including those outlined herein) so that the cells or tissues of interest can be located using immunoscintigraphy.
Antibodies may also be used as staining reagents in pathology using techniques well known in the art.
In some embodiments, the first antibody is used for diagnosis, and the second antibody is used as a therapeutic agent. In some embodiments, the first antibody and the second antibody are different. In some embodiments, the first antibody is from a non-human and the therapeutic agent is from a human. In some embodiments, the first antibody and the second antibody can both bind to the antigen simultaneously by binding to separate epitopes.
Kit/preparation
Also provided herein are kits, medicaments, compositions, and unit dosage forms for use in any of the methods described herein.
Kits can include one or more containers comprising an antibody and/or fusion protein provided herein (e.g., 7-B16, 1-K17, etc.) or unit dosage forms and/or articles of manufacture. In some embodiments, a unit dose is provided, wherein the unit dose contains a predetermined amount of a composition comprising an antibody and/or fusion protein provided herein, with or without one or more additional pharmaceutical agents. In some embodiments, such unit doses are provided in a disposable pre-filled syringe for injection. In some embodiments, the composition contained in a unit dose may comprise saline, sucrose, or the like; buffers such as phosphates and the like; and/or within a stable and effective pH range. In some embodiments, the composition can be provided as a lyophilized powder that can be reconstituted upon addition of an appropriate liquid (e.g., sterile water). In some embodiments, the composition comprises one or more substances that inhibit protein aggregation, including but not limited to sucrose and arginine. In some embodiments, the composition comprises heparin and/or proteoglycans.
In some embodiments, the amount of antibody and/or fusion protein used in a unit dose can be any amount provided herein for use in the various methods and/or compositions described.
In some embodiments, the kit further comprises instructions for treating cancer according to any of the methods described herein. The kit may further comprise instructions for selecting an individual suitable for treatment. The instructions provided in the kit are typically written instructions on a label or package insert (e.g., a paper sheet contained in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable. In some embodiments, the kit further comprises another therapeutic agent.
The kit is suitably packaged. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed mylar or plastic bags), and the like. The kit may optionally provide additional components, such as buffers and explanatory information. Accordingly, the present application also provides articles of manufacture including vials (such as sealed vials), bottles, cans, flexible packaging, and the like.
Examples
The embodiments discussed below are intended to be purely exemplary of the invention and should not be considered as limiting the invention in any way. The examples are not intended to represent that the following experiments are all or the only experiments performed. Although efforts have been made to ensure accuracy with respect to numbers used (e.g., amounts, temperature, etc.), some experimental errors and deviations should be accounted for. Unless otherwise indicated, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees celsius, and pressure is at or near atmospheric.
Example 1: assessment of CCR8mRNA expression in human tumors
The Expression of CCR8mRNA in a variety of human tumors was determined using data publicly available in the Gene Expression Omnibus database under serial accession number GSE13204 and accessed by Bloodspot (see Bagger et al, Nucleic Acids Research 44(D1): D917-924 (2016; Haferlach, Torsten et al, Journal of Clinical Oncology 28:2529-37 (2010); and Kohlmann, A. et al, British Journal of Haematology 142,802-807 (2008)). CCR8mRNA levels were found to be higher in 10% of T cell acute lymphoblastic leukemia (T-ALL) patients than the maximum observed in healthy bone marrow (fig. 1).
CCR8mRNA Expression levels were also determined from RNA sequencing data obtained from OmicSoft OncoLand Hematology (a collection of datasets of blood-related cancers from Multiple studies, including the Gene Expression Omnibus database and Multiple Myoloma Genomics Portal (MMGP)). Data were generated from primary patient samples, xenografts, or cell lines. Expression of CCR8 in these samples indicated that certain hematologic malignancies in this database had elevated expression levels compared to normal controls, including childhood T-cell acute lymphoblastic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, and the like (fig. 2).
Example 2: CCR8 expresses tregs specific for solid tumors.
Expression of CCR8 on tumor-infiltrating tregs and conventional T cells (Tconv) was determined by flow cytometry as described below. Human tumor biopsies were mechanically isolated with a cell filter to obtain single cell suspensions. Anti-human CCR8 antibody (BD Biosciences, clone #433H) or isotype matched controls were used for flow cytometry analysis. The density of CCR8 expression on the cell surface of tumor infiltrating Treg and Tconv cells was determined by quantitative flow cytometry using beads from bangs Laboratories, Inc (Quantum simple Cellular anti IgG) and following the manufacturer's instructions.
As shown in fig. 3A, CCR8 is preferentially expressed on tumor-infiltrating Treg cells in tumors of breast, colon, head and neck, lung and ovarian cancers. As shown in figure 3B, the density of CCR8 on the cell surface was higher in tumor infiltrating Treg cells compared to Tconv cells, ranging from 2,000-8,000 CCR8 receptors per cell, even though low.
Example 3: production of mouse anti-huCCR 8 antibodies
anti-CCR 8 monoclonal antibodies were generated by immunizing inbred mice BALB/C, CD1, B6/129, B6/SJL, NZB/NZW, JIL-E strains with one or more of the following antigens: 1) plasmid DNA encoding human CCR8, or 2) 3T12 cell line expressing human CCR 8.
Plasmid DNA immunization was performed by tail vein hydrodynamic injection. Cellular immunization was performed by intraperitoneal (i.p.) injection of mitomycin C-treated 3T12 cells overexpressing human CCR 8. Groups of animals were immunized with a cell line overexpressing CCR8, followed by DNA boosting, as described below. 200 μ l of the prepared antigen was injected into the peritoneal cavity or subcutaneous or footpad of the mice every 2-14 days. Intravenous injection of 10X 10 to animals producing anti-CCR 8 titers 6 3T12 cells overexpressing CCR 8. Lymph nodes and/or spleen were harvested and isolated lymph node cells and/or spleen cells were used for hybridoma preparation.
To select animals producing CCR8 binding antibodies, sera from the immunized animals were tested by flow cytometry to determine binding to the CHO-S cell line overexpressing human CCR8, but not to the parental CHO-S cell line. Briefly, binding of polyclonal sera was assessed by incubating CHO-S cells expressing human CCR8 with serial dilutions (1:4) of serum samples. The cells were then washed and binding was detected with Phycoerythrin (PE) -labeled species-specific anti-Fc IgG antibodies. Flow cytometry analysis was performed using a BD Fortessa flow cytometer (Becton Dickinson, San Jose, CA). Mice that produced the highest titers of anti-CCR 8 antibody were used for fusion. The fusion was performed as described below. Hybridoma supernatants were tested for anti-CCR 8 activity by flow cytometry.
Splenocytes and/or lymphocytes isolated from mice are fused by electrofusion with a mouse myeloma cell line. Briefly, a single cell suspension of splenocytes from immunized mice was fused with an equal number of IgG non-secreting mouse myeloma cells. The cells were cultured at approximately 2X 10 4 One/well plates were plated in 384-well flat-bottom microtiter plates, followed by incubation in selective medium containing l x hypoxanthine-aminopterin-thymidine (HAT) medium for about two weeks. One to two weeks later, cells were cultured in medium in which HAT was replaced with Hypoxanthine and Thymidine (HT) medium. Approximately 10-14 days after cell plating, supernatants from individual wells were first screened for mouse IgG. Supernatants scored positive for mouse IgG antibodies were then subsequently screened by flow cytometry for anti-CCR 8IgG antibodies. Hybridomas secreting anti-CCR 8 antibodies are then subjected to a subcloning process by limiting dilution. Stable subclones were cultured in vitro to determine binding to human CCR8 by flow cytometry. Confirmed positive clones were scaled up to produce small amounts of antibody in tissue culture medium for further characterization. Monoclonal antibodies from hybridoma supernatants were purified by protein a column chromatography.
Example 4: screening for anti-CCR 8 antibodies that bind to human CCR8 and human CCR4
Antibodies that specifically bind to human CCR8 were screened by flow cytometry using cell lines that overexpress human CCR8 and parental CHO-S cell lines. Antibodies that specifically bind human CCR8 were also tested for binding to CCR4 by flow cytometry. As shown in FIGS. 4A-4B, antibodies 1-K16, 1-K17, 6-B09, 7-B16, 13-E16, and 19-O07 bound to CHO-S cells expressing human CCR8 (4A), but not to CHO-S cells expressing human CCR4 (4B) or untransfected CHO-S cells.
Antibodies 1-K16, 1-K17, 6-B09, 7-B16, 13-E16 and 19-O07 were sequenced and reformatted into human IgG1 backbones for further characterization.
Binding of chimeric and humanized antibodies to CCR8, but not CCR4, was also demonstrated.
Example 5: screening for CCL1 blockade by anti-CCR 8 chimeric antibodies
Binding of CCL1 to surface-expressed CCR8 induced downstream signaling, and blocking CCR8 with anti-CCR 8 antibodies likely inhibited this signaling pathway. Thus, chimeric forms of anti-CCR 8 antibodies were screened for the ability to inhibit ligand-receptor interactions in a cell-based assay (discover x bioassays (Eurofins)) to identify antagonists of CCR8 signaling.
In short, will be derived fromBeta arrestin eXpress GPCR assay kit (Eurofins) 5000 PathHunter eXpress beta arrestin GPCR cells were plated in each well of a 96 well assay plate at 37 deg.C, 5% CO 2 Culturing in medium. After 48 hours of incubation, each chimeric antibody was serially diluted and added to the indicated row of the assay plate and incubated at 37 ℃ for 30 minutes. 13.7nM human CCL1 was then added and incubated for 90 min at 37 ℃. The working detection solution was then added to all wells of the assay plate and the assay plate was incubated for 1 hour at room temperature in the dark. Assay plates were read on a standard light plate reader at 0.1 to 1 second/well (for photomultiplier tube readers) or 5-10 seconds (for imagers). Data were plotted using GraphPad Prism software to obtain IC50 values and are summarized in fig. 5 and table 2.
Table 2: IC50 of chimeric anti-CCR 8 antibodies
As shown in fig. 5 and table 2, the antibodies showed different levels of antagonism. Antibodies 1-K17 and 7-B16 were the most effective antagonists in the discovery X assay.
Example 6: epitope binning (epitope) of anti-CCR 8 antibodies by flow cytometry and ELISA binning)
Antibody subsets directed against human CCR8 were epitope binned in tandem using flow cytometry methods. Briefly, a panel of antibodies was biotinylated. Cells overexpressing human CCR8 were incubated with 10 μ g/mL of non-biotinylated antibody for 1 hour at 4 ℃ and then biotinylated antibody was added at 10 μ g/mL without washing. After another 1 hour incubation, the cells were washed three times and then stained with a streptavidin-phycoerythrin (PE-strep) secondary antibody for 30 minutes. Binding competition was determined by flow cytometry detection of PE. Response values were normalized to% of maximal binding of a single antibody. The binning results are shown in table 3A and confirmed by a continuous format ELISA method using lipid particles overexpressing human CCR8 (table 3B).
Table 3A: epitope binning by FACS analysis
FACS 7-B16 1-K16 1-K17 19-O07 6-B09
7-B16 41 14 18 148 175
1-K16 88 31 32 149 167
1-K17 14 12 11 28 102
19-O07 108 109 91 54 39
6-B09 89 102 94 32 44
Table 3B: epitope binning by ELISA analysis
As shown in Table 3A and FIG. 3B, 7-B16, 1-K16, and 1-K17 are in the same epitope bin, and 19-O07 and 6-B09 are in the same epitope bin. 7-B16 and 1-K16 showed unidirectional binding.
Example 7: screening for ADCC function of anti-CCR 8 chimeric antibodies
Effector functions of anti-CCR 8 of human IgG1 chimeras and humanized versions were determined using an ADCC reporter bioassay kit from Promega. Briefly, CHO-S cells expressing human CCR8 were cultured at 10 6 The concentration of individual cells/mL was resuspended in pre-warmed assay buffer (37 ℃). 25,000 cells were mixed with serially diluted anti-CCR 8 antibodies in a 96-well flat clear bottom plate and then incubated at 37 ℃ with 5% CO 2 Incubate for 1 hour. Promega bioassay effector cells were added to each well at a different ratio to target cells and 5% CO at 37 ℃ 2 Incubate next for another 6 hours. After incubation, assay plates were equilibrated to room temperature for 15 minutes under foil on a bench top. Premixed Bio-Glo luciferase assay substrate was added to each well and incubated for 5 minutes at room temperature. Assay plates were read on a Bio-Tek plate reader within 30 minutes of substrate addition. Data were plotted using GraphPad Prism software to generate EC50 values for individual antibodies.
The results are shown in figure 6 and table 4, showing EC50 values for ADCC function for each test antibody. Human IgG1 chimeric 1-K17 and 7-B16 showed potent ADCC activity.
Table 4: EC50 values for ADCC function of chimeric anti-CCR 8 antibodies
mAb cloning EC50(ng/mL)
1-K17 6.182
7-B16 6.912
13-E16 54.880
Example 8: affinity characterization of antibodies 1-K17 and 7-B16
The affinity of the chimeric antibodies 1-K17 and 7-B16 was determined by kinetic exclusion assay (KinExA) at Sapidyne Instruments. To determine the equilibrium dissociation constant (Kd), CHO-S cells expressing human CCR8 were titrated in the background of 1-K17 or 7-B16 or negative control mAbs. The sample was gently shaken at 37 ℃ for 2.5 hours to reach equilibrium. After incubation, cells were centrifuged and the free portion of 1-K17 or 7-B16 or negative control mAb was removed without disturbing the cell pellet; PMMA beads coated with biotinylated goat anti-human IgG were used to capture a portion of free 1-K17 or 7-B16 or negative control mAb from an equilibrated sample of CHO-S cells expressing 1-K17 or 7-B16 or negative control mAb and human CCR 8. The captured 1-K17 or 7-B16 or negative control mAb was detected with a fluorescently labeled detection antibody. The fluorescent signal was converted to a voltage signal proportional to the amount of free 1-K17 or 7-B16 or negative control mAb in the equilibrated sample.
To determine the binding rate (kon), the same immobilized biotinylated goat anti-human IgG-PMMA column was used as capture reagent for the kinetic experiments. Measuring the amount of free 1-K17 or 7-B16 or negative control mAb in the sample prior to equilibration generates data points that monitor the decrease of free 1-K17 or 7-B16 or negative control mAb as the sample moves toward equilibration.
Data analysis was performed at the company Sapidyne Instruments using KinExA Pro software to generate Kd and kon. The dissociation rate (koff) was calculated based on the following equation.
koff=Kd×kon
As shown in Table 5, both antibodies showed high affinity in the range of 18-220 pM.
Table 5: on-cell affinity of anti-CCR 8 antibodies determined by KinExA
Antibody reference Ka[1/(M.s)] Kd[1/s] K D [M]
1-K17.hG1 chimeras 7.81×10 6 1.44×10 -4 0.18×10 -10
7-B16.hG1 chimera 6.08×10 6 1.34×10 -3 2.20×10 -10
Example 9: humanization and hotspot correction of antibodies 1-K17 and 7-B16
Antibodies 1-K17 and 7-B16 were humanized by grafting the CDRs of the lead antibody into a selected human IgG germline framework. Human germline IGHV, IGKV, IGHJ and IGKJ were selected based on sequence similarity within the two Frameworks (FR). The parent antibodies and selected germline are summarized in table 6. To maintain the classical loop structure and chain interface, some residues in the human germline framework were back-mutated to the corresponding mouse residues (bold in table 6).
Computer predictions suggest a high risk sequence bias in the CDRs of 1-K17 and 7-B16. For example, an NG motif is present in the CDR-H2 region of humanized 1-K17. Three preferential mutations at position N55 of the heavy chain were evaluated to see if potential deamidation sites in the VH could be removed without affecting activity. The residue substitution N55Q was selected to eliminate potential deamidation sites in the heavy chain (N55) and incorporated into the humanized sequence. In the CDR-L1 region of 1-K17, NG and NT motifs are present which may be susceptible to deamidation. Residue substitutions G34A and N35Q were selected to eliminate potential deamidation sites in the light chain (N33 and N35) and incorporated into the humanized sequence. The same mutant design as the humanized 1-K17 light chain was tested to remove potential deamidation tendencies in 7-B16CDR-L1, and the M56L mutation was also tested to remove oxidative tendencies in CDR-L2. The sequences are summarized in table 6.
All optimized antibodies were confirmed to bind to CHO-S expressing human CCR 8. The affinity constants for the humanized versions of antibodies 1-K17 and 7-B16 are shown in Table 7.
TABLE 7 byOn-cell affinity of anti-CCR 8 antibodies as determined by KinExA assay
Antibody reference Kon(M -1 s -1 ) Koff(s -1 ) K D [M]
Humanized 1-K17.015 9.78×10 6 1.42×10 -4 14.5×10 -12
Humanized 1-K17.044 8.29×10 6 6.67×10 -5 8.05×10 -12
Humanized 7-B16.001 4.99×10 6 7.93×10 -5 15.9×10 -12
Humanized 7-B16.033 5.25×10 6 3.91×10 -4 74.6×10 -12
Example 10: analysis of antibody afucosylation
ADCC reporter activity of afucosylated human IgG1 version of human IgG1 and anti-human CCR8 clone 1K17 was evaluated according to manufacturer recommendations using Promega ADCC reporter bioassay kit. Chinese Hamster Ovary (CHO) cells overexpressing 300,000 or 10,000 human CCR8 molecules on the cell surface were used as target cells at a ratio of effector cells to target cells of 3: 1.
The IK17 hIgG1 antibody did not trigger ADCC of cells expressing low density CCR8 (fig. 7A). In contrast, treatment with IK17 hIgG1 afucosylated antibody produced greater Fc-mediated ADCC in cells expressing both high and low levels of CCR8 (fig. 7B). Thus, afucosylation of anti-human CCR8 IgG1 antibodies increased ADCC activity against cells expressing low levels of CCR 8. As discussed herein, it was found that tumor-infiltrating tregs express low levels of CCR8, and therefore afucosylated anti-CCR 8 antibodies can provide effective ADCC activity against tumor-infiltrating tregs expressing CCR8, while non-fucosylated antibodies do not.
Example 11: in vivo analysis of antibodies
anti-CCR 8 mIgG2a and mIgG1 antibodies were evaluated in tumor models compared to mIgG2a isotype controls. 57Bl6 mice were inoculated with 2.5 × 10e5 MC38 cells. Each treatment group included 4 patients with established tumors (100 mm) on day 0 3 ) The animal of (1). Animals were injected intraperitoneally (i.p.) one 200 μ g of the above anti-mouse CCR8 or isotype control antibody (i.e., anti-mouse CCR8 mIgG2a or anti-mouse CCR8 mIgG1 or mouse IgG2a isotype control) on day 0 and three days later tumors were harvested for flow cytometry analysis.
As shown in fig. 8A-8C, the proportion of tumor-infiltrating tregs was reduced in the anti-CCR 8 mIgG2a group, but not in the other groups.
The therapeutic efficacy of the antibodies was also evaluated. To tumors with establishment on day 0 (100 mm) 3 ) Mice were intraperitoneally (i.p) injected twice weekly with 200 μ g of the above anti-mouse CCR8 or isotype control antibody (i.e., anti-mouse CCR8 mIgG2a or anti-mouse CCR8 mIgG1 or mouse IgG2a isotype control), once on each of days 0, 3, 7, 10, 14, and 17. Each treatment group included 10-15 animals and tumor growth was measured with a caliper instrument.
Fig. 9A-9B show Fc effector function driven therapeutic efficacy of the anti-mouse CCR8 monoclonal antibody. In this MC38 homology model, Fc activity (mouse IgG2a) but not Fc inactivity (mouse IgG1) anti-mouse CCR8 antibodies reduced tumor growth and promoted mouse survival.
Example 12: CCR8 mRNA expression in hematologic cancers
A database of whole transcriptome sequencing ("RNAseq") studies was constructed to explore CCR8 expression in multiple studies on leukemia and lymphoma. Datasets were selected for the keywords "leukamia" (leukemia) and "lymphoma" (lymphoma) and downloaded from record 2 (Collado-Torres et al, Nature Biotechnology 35.4(2017): 319-. The database included 2,679 samples from 98 studies spanning 22 cancer types. The data set includes samples from primary patient samples, cell lines and xenografts. Sources include bone marrow, peripheral blood and other tissue sources. mRNA expression levels were converted to number of Transcripts Per Million (TPM), quantile normalized between samples to adjust for total expression differences between study and sample and log2 transformation. Datasets with large numbers of non-expressed genes or datasets with incomplete metadata are omitted. CCR8 was assessed between cancer types, as well as log-fold change compared to normal healthy blood.
As shown in figure 10, the following indications have significant up-regulation of CCR8 compared to normal blood in the one-sided t-test when correcting the false discovery rate (adjusted p-value < 0.05): cutaneous T Cell Lymphoma (CTCL), adult T cell leukemia/lymphoma (ATLL), T cell lymphoblastic leukemia/lymphoma (TLLL), and T Acute Lymphocytic Leukemia (TALL). Anaplastic Large Cell Lymphoma (ALCL) was significant in nominal p-value, but not significant after FDR correction.
Example 13: screening for variants of 7-B16
Different classes of 7-B16 variants were generated and screened to establish binding affinity and activity. The first category included single substitution mutations in CDR H3 of 7-B16. The second category includes two or three substitution mutations in CDR H3 of 7-B16. The third class includes variants in which the CDRs of 7-B16, H1, H2, L1, L2, or L3, are exchanged with the corresponding CDRs from one of the other antibodies disclosed herein.
The first category of variants (with single substitution mutations in CDR H3) includes variants in which each amino acid residue of the heavy chain CDR3 (i.e., CDR H3) is iteratively substituted with alanine residues. These mutants were evaluated for ADCC activity (both EC50 and fold induction), octet response and CCR8 binding.
ADCC activity was evaluated according to the same method used in example 7 above. Briefly, the effector functions of the anti-CCR 8 of the chimeric and humanized versions of human IgG1 were determined using the ADCC reporter bioassay kit from Promega. CHO-S cells expressing human CCR8 were cultured at 10 6 The concentration of individual cells/mL was resuspended in pre-warmed assay buffer (37 ℃). 25,000 cells were mixed with serially diluted anti-CCR 8 antibodies in a 96-well flat clear bottom plate and then incubated at 37 ℃ with 5% CO 2 Incubate for 1 hour. Promega bioassay Effector cells were added to individual wells at different ratios to target cells and 5% CO at 37 ℃ 2 Followed by incubation for another 6 hours. After incubation, assay plates were equilibrated to room temperature for 15 minutes under foil on a bench top. Premixed Bio-Glo luciferase assay substrate was added to each well and incubated for 5 minutes at room temperature. Assay plates were read on a Bio-Tek plate reader within 30 minutes of substrate addition.
CCR8 binding was determined using flow cytometry according to the same method used in example 4. Briefly, cell lines overexpressing human CCR8 and parental CHO-S cell lines were used to screen for antibodies that specifically bind to human CCR8 by flow cytometry. Antibodies that specifically bind human CCR8 were also tested for binding to CCR4 by flow cytometry.
Finally, to understand the paratope of 7-B16, a CDR-H3 alanine scanning variant was generated. Binding of CDR-H3 alanine scanning variant of 7-B16 to CCR8 was determined by ForteBio Octet using an N-terminal peptide (aa 1-35) fused to maltose binding protein (CCR8-NT. MBP, the complete sequence of which is provided in SEQ ID NO: 128). Amino acids 406-440 of SEQ ID NO 128 (i.e. CCR8-NT. MBP) represent amino acids 1-35 of the CCR8 protein, and in this sequence all tyrosine residues shown in bold italics may be sulfated: MDYTLDLSVTTVTDYYYPDIFSSPCDAELIQTNGK (amino acids 406-440 of SEQ ID NO: 128). Amino acids 1-364 of SEQ ID NO 128 represent maltose binding protein.
For this evaluation, 10. mu.g/ml of the 7-B16 alanine scan variant was captured on an AHC biosensor. Mbp was used as an analyte to determine binding of the variant to CCR 8. Binding of the variant to CCR8-nt. mbp was recorded as Octet response (nM shift).
The results of these measurements are summarized in table 8 below and fig. 11.
Evaluation of CDR H3 variants of tables 8-7-B16
Variants 7B16.002, 7B16.008, and 7B16.013 were selected for the combination variants, which are discussed in more detail below.
The second and third class (two or three substitution mutations in CDR H3 or CDR-exchanged, respectively) of variants are shown in table 9 below.
TABLE 9 additional 7-B16 antibody variants
The octet response and CCR8 binding of the additional 7-B16 variants shown in Table 9 were evaluated according to the same method described for the CDR H3 single substitution variant disclosed in Table 8. The results are shown below. Specifically, table 10 shows the Octet response and binding to CHO CCR8 as determined by flow cytometry.
TABLE 10 evaluation of additional 7B16 variants
As shown in table 10, the Octet response was not significantly changed for any of the alanine combinatorial mutants. However, there was a loss of binding in the CDR-exchanged variants H1, H3, L1 and L3, and there was a modest reduction in binding for the CDR-exchanged variant L1.
The ADCC activity (both EC50 and fold induction) of the additional 7B16 variants shown in table 9 will also be evaluated according to the same method described for the CDR H3 single substitution variants disclosed in table 8. It is expected that these variants will also have similar ADCC activity.
*****
The present disclosure may be embodied in other specific forms without departing from its spirit or essential characteristics. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the disclosure. The scope of the disclosure is, therefore, indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.
Tables of some sequences
Sequence listing
<110> Shake therapy GmbH
<120> antibodies and fusion proteins binding to CCR8 and uses thereof
<130> 119835-0178
<140>
<141>
<150> 63/130,157
<151> 2020-12-23
<150> 62/976,869
<151> 2020-02-14
<160> 129
<170> PatentIn version 3.5
<210> 1
<211> 339
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 1
Met Gln Ile Ile Thr Thr Ala Leu Val Cys Leu Leu Leu Ala Gly Met
1 5 10 15
Trp Pro Glu Asp Val Asp Ser Lys Ser Met Gln Val Pro Phe Ser Arg
20 25 30
Cys Cys Phe Ser Phe Ala Glu Gln Glu Ile Pro Leu Arg Ala Ile Leu
35 40 45
Cys Tyr Arg Asn Thr Ser Ser Ile Cys Ser Asn Glu Gly Leu Ile Phe
50 55 60
Lys Leu Lys Arg Gly Lys Glu Ala Cys Ala Leu Asp Thr Val Gly Trp
65 70 75 80
Val Gln Arg His Arg Lys Met Leu Arg His Cys Pro Ser Lys Arg Lys
85 90 95
Ser Gly Gly Ser Gly Gly Gly Ser Gly Asp Lys Thr His Thr Cys Pro
100 105 110
Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe
115 120 125
Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val
130 135 140
Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe
145 150 155 160
Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro
165 170 175
Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr
180 185 190
Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val
195 200 205
Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala
210 215 220
Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg
225 230 235 240
Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly
245 250 255
Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro
260 265 270
Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser
275 280 285
Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln
290 295 300
Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His
305 310 315 320
Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys Gly Ser Gly Glu
325 330 335
Pro Glu Ala
<210> 2
<211> 96
<212> PRT
<213> Intelligent (Homo sapiens)
<400> 2
Met Gln Ile Ile Thr Thr Ala Leu Val Cys Leu Leu Leu Ala Gly Met
1 5 10 15
Trp Pro Glu Asp Val Asp Ser Lys Ser Met Gln Val Pro Phe Ser Arg
20 25 30
Cys Cys Phe Ser Phe Ala Glu Gln Glu Ile Pro Leu Arg Ala Ile Leu
35 40 45
Cys Tyr Arg Asn Thr Ser Ser Ile Cys Ser Asn Glu Gly Leu Ile Phe
50 55 60
Lys Leu Lys Arg Gly Lys Glu Ala Cys Ala Leu Asp Thr Val Gly Trp
65 70 75 80
Val Gln Arg His Arg Lys Met Leu Arg His Cys Pro Ser Lys Arg Lys
85 90 95
<210> 3
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 3
Ser Gly Gly Ser Gly Gly Gly Ser Gly
1 5
<210> 4
<211> 234
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 4
Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly
1 5 10 15
Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met
20 25 30
Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His
35 40 45
Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val
50 55 60
His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr
65 70 75 80
Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly
85 90 95
Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile
100 105 110
Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val
115 120 125
Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser
130 135 140
Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu
145 150 155 160
Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro
165 170 175
Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val
180 185 190
Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met
195 200 205
His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser
210 215 220
Pro Gly Lys Gly Ser Gly Glu Pro Glu Ala
225 230
<210> 5
<211> 322
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 5
Leu Ala Arg Arg Lys Cys Cys Leu Asn Pro Thr Asn Arg Pro Ile Pro
1 5 10 15
Asn Pro Leu Leu Gln Asp Leu Ser Arg Val Asp Tyr Gln Ala Ile Gly
20 25 30
His Asp Cys Gly Arg Glu Ala Phe Arg Val Thr Leu Gln Asp Gly Arg
35 40 45
Gln Gly Cys Val Ser Val Gly Asn Lys Ser Leu Leu Asp Trp Leu Arg
50 55 60
Gly His Lys Asp Leu Cys Pro Gln Ile Trp Ser Gly Cys Glu Ser Leu
65 70 75 80
Gly Gly Gly Ser Gly Gly Gly Ser Asp Lys Thr His Thr Cys Pro Pro
85 90 95
Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro
100 105 110
Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr
115 120 125
Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn
130 135 140
Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg
145 150 155 160
Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val
165 170 175
Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser
180 185 190
Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys
195 200 205
Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu
210 215 220
Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe
225 230 235 240
Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu
245 250 255
Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe
260 265 270
Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly
275 280 285
Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr
290 295 300
Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys Gly Ser Gly Glu Pro
305 310 315 320
Glu Ala
<210> 6
<211> 80
<212> PRT
<213> Molluscum contagiosum virus
<400> 6
Leu Ala Arg Arg Lys Cys Cys Leu Asn Pro Thr Asn Arg Pro Ile Pro
1 5 10 15
Asn Pro Leu Leu Gln Asp Leu Ser Arg Val Asp Tyr Gln Ala Ile Gly
20 25 30
His Asp Cys Gly Arg Glu Ala Phe Arg Val Thr Leu Gln Asp Gly Arg
35 40 45
Gln Gly Cys Val Ser Val Gly Asn Lys Ser Leu Leu Asp Trp Leu Arg
50 55 60
Gly His Lys Asp Leu Cys Pro Gln Ile Trp Ser Gly Cys Glu Ser Leu
65 70 75 80
<210> 7
<211> 8
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 7
Gly Gly Gly Ser Gly Gly Gly Ser
1 5
<210> 8
<211> 123
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 8
Glu Val Gln Leu Val Glu Thr Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Ile Asn
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Met
65 70 75 80
Leu Tyr Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Gly Gly Tyr Gly Ser Ser Pro Tyr Asp Met Asp Tyr
100 105 110
Trp Gly Gln Gly Thr Ser Val Thr Val Ser Ser
115 120
<210> 9
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 9
Asp Ile Val Met Thr Gln Ala Ala Pro Ser Val Pro Val Thr Pro Gly
1 5 10 15
Glu Ser Val Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Ala Phe Thr Leu Arg Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105 110
<210> 10
<211> 446
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 10
Glu Val Gln Leu Val Glu Thr Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Ile Asn
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Met
65 70 75 80
Leu Tyr Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Gly Gly Tyr Gly Ser Ser Pro Tyr Asp Met Asp Tyr
100 105 110
Trp Gly Gln Gly Thr Ser Val Thr Val Ser Ser Ala Lys Thr Thr Pro
115 120 125
Pro Ser Val Tyr Pro Leu Ala Pro Gly Ser Ala Ala Gln Thr Asn Ser
130 135 140
Met Val Thr Leu Gly Cys Leu Val Lys Gly Tyr Phe Pro Glu Pro Val
145 150 155 160
Thr Val Thr Trp Asn Ser Gly Ser Leu Ser Ser Gly Val His Thr Phe
165 170 175
Pro Ala Val Leu Glu Ser Asp Leu Tyr Thr Leu Ser Ser Ser Val Thr
180 185 190
Val Pro Ser Ser Pro Arg Pro Ser Glu Thr Val Thr Cys Asn Val Ala
195 200 205
His Pro Ala Ser Ser Thr Lys Val Asp Lys Lys Ile Val Pro Arg Asp
210 215 220
Cys Gly Cys Lys Pro Cys Ile Cys Thr Val Pro Glu Val Ser Ser Val
225 230 235 240
Phe Ile Phe Pro Pro Lys Pro Lys Asp Val Leu Thr Ile Thr Leu Thr
245 250 255
Pro Lys Val Thr Cys Val Val Val Asp Ile Ser Lys Asp Asp Pro Glu
260 265 270
Val Gln Phe Ser Trp Phe Val Asp Asp Val Glu Val His Thr Ala Gln
275 280 285
Thr Gln Pro Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg Ser Val Ser
290 295 300
Glu Leu Pro Ile Met His Gln Asp Trp Leu Asn Gly Lys Glu Phe Lys
305 310 315 320
Cys Arg Val Asn Ser Ala Ala Phe Pro Ala Pro Ile Glu Lys Thr Ile
325 330 335
Ser Lys Thr Lys Gly Arg Pro Lys Ala Pro Gln Val Tyr Thr Ile Pro
340 345 350
Pro Pro Lys Glu Gln Met Ala Lys Asp Lys Val Ser Leu Thr Cys Met
355 360 365
Ile Thr Asp Phe Phe Pro Glu Asp Ile Thr Val Glu Trp Gln Trp Asn
370 375 380
Gly Gln Pro Ala Glu Asn Tyr Lys Asn Thr Gln Pro Ile Met Asn Thr
385 390 395 400
Asn Gly Ser Tyr Phe Val Tyr Ser Lys Leu Asn Val Gln Lys Ser Asn
405 410 415
Trp Glu Ala Gly Asn Thr Phe Thr Cys Ser Val Leu His Glu Gly Leu
420 425 430
His Asn His His Thr Glu Lys Ser Leu Ser His Ser Pro Gly
435 440 445
<210> 11
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 11
Asp Ile Val Met Thr Gln Ala Ala Pro Ser Val Pro Val Thr Pro Gly
1 5 10 15
Glu Ser Val Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Ala Phe Thr Leu Arg Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105 110
Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu
115 120 125
Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe
130 135 140
Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg
145 150 155 160
Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu
180 185 190
Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser
195 200 205
Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys
210 215
<210> 12
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 12
Gly Phe Thr Phe Asn Ile Asn Ala Met Asn
1 5 10
<210> 13
<211> 17
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 13
Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp Ser
1 5 10 15
Val
<210> 14
<211> 14
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 14
Val Arg Gly Gly Tyr Gly Ser Ser Pro Tyr Asp Met Asp Tyr
1 5 10
<210> 15
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 15
Arg Ser Ser Lys Ser Leu Leu His Ser Asn Gly Asn Thr Tyr Leu Tyr
1 5 10 15
<210> 16
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 16
Arg Met Ser Asn Leu Ala Ser
1 5
<210> 17
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 17
Met Gln His Leu Glu Tyr Pro Phe Thr
1 5
<210> 18
<211> 452
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 18
Glu Val Gln Leu Val Glu Thr Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Ile Asn
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Met
65 70 75 80
Leu Tyr Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Gly Gly Tyr Gly Ser Ser Pro Tyr Asp Met Asp Tyr
100 105 110
Trp Gly Gln Gly Thr Ser Val Thr Val Ser Ser Ala Ser Thr Lys Gly
115 120 125
Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly
130 135 140
Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val
145 150 155 160
Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe
165 170 175
Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val
180 185 190
Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val
195 200 205
Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys
210 215 220
Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu
225 230 235 240
Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr
245 250 255
Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val
260 265 270
Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val
275 280 285
Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser
290 295 300
Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu
305 310 315 320
Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala
325 330 335
Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro
340 345 350
Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln
355 360 365
Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala
370 375 380
Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr
385 390 395 400
Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu
405 410 415
Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser
420 425 430
Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser
435 440 445
Leu Ser Pro Gly
450
<210> 19
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 19
Asp Ile Val Met Thr Gln Ala Ala Pro Ser Val Pro Val Thr Pro Gly
1 5 10 15
Glu Ser Val Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Ala Phe Thr Leu Arg Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 20
<211> 115
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 20
Gln Val Gln Leu Lys Glu Ser Gly Pro Gly Leu Val Ala Pro Ser Gln
1 5 10 15
Ser Leu Ser Ile Thr Cys Thr Val Ser Gly Phe Ser Leu Ala Arg Tyr
20 25 30
Asp Ile Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp Leu
35 40 45
Gly Val Ile Trp Thr Gly Gly Gly Thr Asn Tyr Asn Ser Ala Phe Met
50 55 60
Ser Arg Leu Ser Ile Ser Lys Asp Asn Ser Lys Ser Gln Val Phe Leu
65 70 75 80
Lys Met Asn Gly Leu Gln Thr Asp Asp Thr Ala Ile Tyr Tyr Cys Val
85 90 95
Ser Ile Arg Tyr Asp Glu Thr Tyr Trp Gly Gln Gly Thr Leu Val Thr
100 105 110
Val Ser Ala
115
<210> 21
<211> 106
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 21
Gln Ile Val Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ile Tyr Met
20 25 30
His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr
35 40 45
Ala Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Leu Thr
85 90 95
Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys
100 105
<210> 22
<211> 444
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 22
Gln Val Gln Leu Lys Glu Ser Gly Pro Gly Leu Val Ala Pro Ser Gln
1 5 10 15
Ser Leu Ser Ile Thr Cys Thr Val Ser Gly Phe Ser Leu Ala Arg Tyr
20 25 30
Asp Ile Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp Leu
35 40 45
Gly Val Ile Trp Thr Gly Gly Gly Thr Asn Tyr Asn Ser Ala Phe Met
50 55 60
Ser Arg Leu Ser Ile Ser Lys Asp Asn Ser Lys Ser Gln Val Phe Leu
65 70 75 80
Lys Met Asn Gly Leu Gln Thr Asp Asp Thr Ala Ile Tyr Tyr Cys Val
85 90 95
Ser Ile Arg Tyr Asp Glu Thr Tyr Trp Gly Gln Gly Thr Leu Val Thr
100 105 110
Val Ser Ala Ala Lys Thr Thr Ala Pro Ser Val Tyr Pro Leu Ala Pro
115 120 125
Val Cys Gly Asp Thr Thr Gly Ser Ser Val Thr Leu Gly Cys Leu Val
130 135 140
Lys Gly Tyr Phe Pro Glu Pro Val Thr Leu Thr Trp Asn Ser Gly Ser
145 150 155 160
Leu Ser Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Asp Leu
165 170 175
Tyr Thr Leu Ser Ser Ser Val Thr Val Thr Ser Ser Thr Trp Pro Ser
180 185 190
Gln Ser Ile Thr Cys Asn Val Ala His Pro Ala Ser Ser Thr Lys Val
195 200 205
Asp Lys Lys Ile Glu Pro Arg Gly Pro Thr Ile Lys Pro Cys Pro Pro
210 215 220
Cys Lys Cys Pro Ala Pro Asn Leu Leu Gly Gly Pro Ser Val Phe Ile
225 230 235 240
Phe Pro Pro Lys Ile Lys Asp Val Leu Met Ile Ser Leu Ser Pro Ile
245 250 255
Val Thr Cys Val Val Val Asp Val Ser Glu Asp Asp Pro Asp Val Gln
260 265 270
Ile Ser Trp Phe Val Asn Asn Val Glu Val His Thr Ala Gln Thr Gln
275 280 285
Thr His Arg Glu Asp Tyr Asn Ser Thr Leu Arg Val Val Ser Ala Leu
290 295 300
Pro Ile Gln His Gln Asp Trp Met Ser Gly Lys Glu Phe Lys Cys Lys
305 310 315 320
Val Asn Asn Lys Asp Leu Pro Ala Pro Ile Glu Arg Thr Ile Ser Lys
325 330 335
Pro Lys Gly Ser Val Arg Ala Pro Gln Val Tyr Val Leu Pro Pro Pro
340 345 350
Glu Glu Glu Met Thr Lys Lys Gln Val Thr Leu Thr Cys Met Val Thr
355 360 365
Asp Phe Met Pro Glu Asp Ile Tyr Val Glu Trp Thr Asn Asn Gly Lys
370 375 380
Thr Glu Leu Asn Tyr Lys Asn Thr Glu Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Tyr Phe Met Tyr Ser Lys Leu Arg Val Glu Lys Lys Asn Trp Val
405 410 415
Glu Arg Asn Ser Tyr Ser Cys Ser Val Val His Glu Gly Leu His Asn
420 425 430
His His Thr Thr Lys Ser Phe Ser Arg Thr Pro Gly
435 440
<210> 23
<211> 213
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 23
Gln Ile Val Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ile Tyr Met
20 25 30
His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr
35 40 45
Ala Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Leu Thr
85 90 95
Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys Arg Ala Asp Ala Ala Pro
100 105 110
Thr Val Ser Ile Phe Pro Pro Ser Ser Glu Gln Leu Thr Ser Gly Gly
115 120 125
Ala Ser Val Val Cys Phe Leu Asn Asn Phe Tyr Pro Lys Asp Ile Asn
130 135 140
Val Lys Trp Lys Ile Asp Gly Ser Glu Arg Gln Asn Gly Val Leu Asn
145 150 155 160
Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser Thr Tyr Ser Met Ser Ser
165 170 175
Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu Arg His Asn Ser Tyr Thr
180 185 190
Cys Glu Ala Thr His Lys Thr Ser Thr Ser Pro Ile Val Lys Ser Phe
195 200 205
Asn Arg Asn Glu Cys
210
<210> 24
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 24
Gly Phe Ser Leu Ala Arg Tyr Asp Ile Ser
1 5 10
<210> 25
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 25
Gly Gly Thr Asn Tyr Asn Ser Ala Phe Met Ser Arg Leu Ser Ile Ser
1 5 10 15
<210> 26
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 26
Val Ser Ile Arg Tyr Asp Glu Thr Tyr
1 5
<210> 27
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 27
Ser Ala Ser Ser Ser Val Ile Tyr Met His
1 5 10
<210> 28
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 28
Thr Ser Asn Leu Ala Ser Gly
1 5
<210> 29
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 29
Gln Gln Arg Ser Ser Tyr Pro Leu Thr
1 5
<210> 30
<211> 444
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 30
Gln Val Gln Leu Lys Glu Ser Gly Pro Gly Leu Val Ala Pro Ser Gln
1 5 10 15
Ser Leu Ser Ile Thr Cys Thr Val Ser Gly Phe Ser Leu Ala Arg Tyr
20 25 30
Asp Ile Ser Trp Ile Arg Gln Pro Pro Gly Lys Gly Leu Glu Trp Leu
35 40 45
Gly Val Ile Trp Thr Gly Gly Gly Thr Asn Tyr Asn Ser Ala Phe Met
50 55 60
Ser Arg Leu Ser Ile Ser Lys Asp Asn Ser Lys Ser Gln Val Phe Leu
65 70 75 80
Lys Met Asn Gly Leu Gln Thr Asp Asp Thr Ala Ile Tyr Tyr Cys Val
85 90 95
Ser Ile Arg Tyr Asp Glu Thr Tyr Trp Gly Gln Gly Thr Leu Val Thr
100 105 110
Val Ser Ala Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro
115 120 125
Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val
130 135 140
Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala
145 150 155 160
Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly
165 170 175
Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly
180 185 190
Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys
195 200 205
Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys
210 215 220
Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu
245 250 255
Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys
260 265 270
Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys
275 280 285
Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu
290 295 300
Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys
305 310 315 320
Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys
325 330 335
Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser
340 345 350
Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys
355 360 365
Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln
370 375 380
Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly
385 390 395 400
Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln
405 410 415
Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn
420 425 430
His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440
<210> 31
<211> 213
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 31
Gln Ile Val Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ile Tyr Met
20 25 30
His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr
35 40 45
Ala Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Ser Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Leu Thr
85 90 95
Phe Gly Ala Gly Thr Lys Leu Glu Leu Lys Arg Thr Val Ala Ala Pro
100 105 110
Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly Thr
115 120 125
Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala Lys
130 135 140
Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln Glu
145 150 155 160
Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser Ser
165 170 175
Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr Ala
180 185 190
Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser Phe
195 200 205
Asn Arg Gly Glu Cys
210
<210> 32
<211> 128
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 32
Glu Val Gln Leu Val Glu Thr Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Thr Asn
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Ile
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr His Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Ile
65 70 75 80
Leu His Leu Gln Met Asn Asn Leu Lys Asn Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Asp Ser His Tyr Tyr Val Ser Thr Tyr Val Gly Leu
100 105 110
Ala Trp Phe Ala Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ala
115 120 125
<210> 33
<211> 106
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 33
Gln Ile Val Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met
20 25 30
His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr
35 40 45
Ser Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Phe Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Tyr Thr
85 90 95
Phe Gly Gly Gly Thr Lys Leu Glu Arg Lys
100 105
<210> 34
<211> 457
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 34
Glu Val Gln Leu Val Glu Thr Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Thr Asn
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Ile
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr His Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Ile
65 70 75 80
Leu His Leu Gln Met Asn Asn Leu Lys Asn Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Asp Ser His Tyr Tyr Val Ser Thr Tyr Val Gly Leu
100 105 110
Ala Trp Phe Ala Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ala
115 120 125
Ala Lys Thr Thr Ala Pro Ser Val Tyr Pro Leu Ala Pro Val Cys Gly
130 135 140
Asp Thr Thr Gly Ser Ser Val Thr Leu Gly Cys Leu Val Lys Gly Tyr
145 150 155 160
Phe Pro Glu Pro Val Thr Leu Thr Trp Asn Ser Gly Ser Leu Ser Ser
165 170 175
Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Asp Leu Tyr Thr Leu
180 185 190
Ser Ser Ser Val Thr Val Thr Ser Ser Thr Trp Pro Ser Gln Ser Ile
195 200 205
Thr Cys Asn Val Ala His Pro Ala Ser Ser Thr Lys Val Asp Lys Lys
210 215 220
Ile Glu Pro Arg Gly Pro Thr Ile Lys Pro Cys Pro Pro Cys Lys Cys
225 230 235 240
Pro Ala Pro Asn Leu Leu Gly Gly Pro Ser Val Phe Ile Phe Pro Pro
245 250 255
Lys Ile Lys Asp Val Leu Met Ile Ser Leu Ser Pro Ile Val Thr Cys
260 265 270
Val Val Val Asp Val Ser Glu Asp Asp Pro Asp Val Gln Ile Ser Trp
275 280 285
Phe Val Asn Asn Val Glu Val His Thr Ala Gln Thr Gln Thr His Arg
290 295 300
Glu Asp Tyr Asn Ser Thr Leu Arg Val Val Ser Ala Leu Pro Ile Gln
305 310 315 320
His Gln Asp Trp Met Ser Gly Lys Glu Phe Lys Cys Lys Val Asn Asn
325 330 335
Lys Asp Leu Pro Ala Pro Ile Glu Arg Thr Ile Ser Lys Pro Lys Gly
340 345 350
Ser Val Arg Ala Pro Gln Val Tyr Val Leu Pro Pro Pro Glu Glu Glu
355 360 365
Met Thr Lys Lys Gln Val Thr Leu Thr Cys Met Val Thr Asp Phe Met
370 375 380
Pro Glu Asp Ile Tyr Val Glu Trp Thr Asn Asn Gly Lys Thr Glu Leu
385 390 395 400
Asn Tyr Lys Asn Thr Glu Pro Val Leu Asp Ser Asp Gly Ser Tyr Phe
405 410 415
Met Tyr Ser Lys Leu Arg Val Glu Lys Lys Asn Trp Val Glu Arg Asn
420 425 430
Ser Tyr Ser Cys Ser Val Val His Glu Gly Leu His Asn His His Thr
435 440 445
Thr Lys Ser Phe Ser Arg Thr Pro Gly
450 455
<210> 35
<211> 213
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 35
Gln Ile Val Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met
20 25 30
His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr
35 40 45
Ser Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Phe Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Tyr Thr
85 90 95
Phe Gly Gly Gly Thr Lys Leu Glu Arg Lys Arg Ala Asp Ala Ala Pro
100 105 110
Thr Val Ser Ile Phe Pro Pro Ser Ser Glu Gln Leu Thr Ser Gly Gly
115 120 125
Ala Ser Val Val Cys Phe Leu Asn Asn Phe Tyr Pro Lys Asp Ile Asn
130 135 140
Val Lys Trp Lys Ile Asp Gly Ser Glu Arg Gln Asn Gly Val Leu Asn
145 150 155 160
Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser Thr Tyr Ser Met Ser Ser
165 170 175
Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu Arg His Asn Ser Tyr Thr
180 185 190
Cys Glu Ala Thr His Lys Thr Ser Thr Ser Pro Ile Val Lys Ser Phe
195 200 205
Asn Arg Asn Glu Cys
210
<210> 36
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 36
Gly Phe Thr Phe Ser Thr Asn Ala Met Asn
1 5 10
<210> 37
<211> 17
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 37
Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr His Tyr Ala Asp Ser
1 5 10 15
Val
<210> 38
<211> 15
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 38
Val Arg Asp Ser His Tyr Tyr Val Ser Thr Tyr Val Gly Leu Ala
1 5 10 15
<210> 39
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 39
Ser Ala Ser Ser Ser Val Ser Tyr Met His
1 5 10
<210> 40
<211> 6
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 40
Thr Ser Asn Leu Ala Ser
1 5
<210> 41
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 41
Gln Gln Arg Ser Ser Tyr Pro Tyr Thr
1 5
<210> 42
<211> 457
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 42
Glu Val Gln Leu Val Glu Thr Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Ser Thr Asn
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Ile
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr His Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Ile
65 70 75 80
Leu His Leu Gln Met Asn Asn Leu Lys Asn Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Asp Ser His Tyr Tyr Val Ser Thr Tyr Val Gly Leu
100 105 110
Ala Trp Phe Ala Tyr Trp Gly Gln Gly Thr Leu Val Thr Val Ser Ala
115 120 125
Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys
130 135 140
Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr
145 150 155 160
Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser
165 170 175
Gly Val His Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser
180 185 190
Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr
195 200 205
Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys
210 215 220
Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys
225 230 235 240
Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro
245 250 255
Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys
260 265 270
Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp
275 280 285
Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu
290 295 300
Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu
305 310 315 320
His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn
325 330 335
Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly
340 345 350
Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu
355 360 365
Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr
370 375 380
Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn
385 390 395 400
Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe
405 410 415
Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn
420 425 430
Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr
435 440 445
Gln Lys Ser Leu Ser Leu Ser Pro Gly
450 455
<210> 43
<211> 213
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 43
Gln Ile Val Leu Thr Gln Ser Pro Ala Ile Met Ser Ala Ser Pro Gly
1 5 10 15
Glu Lys Val Thr Ile Thr Cys Ser Ala Ser Ser Ser Val Ser Tyr Met
20 25 30
His Trp Phe Gln Gln Lys Pro Gly Thr Ser Pro Lys Leu Trp Ile Tyr
35 40 45
Ser Thr Ser Asn Leu Ala Ser Gly Val Pro Ala Arg Phe Ser Gly Ser
50 55 60
Gly Ser Gly Thr Ser Tyr Phe Leu Thr Ile Ser Arg Met Glu Ala Glu
65 70 75 80
Asp Ala Ala Thr Tyr Tyr Cys Gln Gln Arg Ser Ser Tyr Pro Tyr Thr
85 90 95
Phe Gly Gly Gly Thr Lys Leu Glu Arg Lys Arg Thr Val Ala Ala Pro
100 105 110
Ser Val Phe Ile Phe Pro Pro Ser Asp Glu Gln Leu Lys Ser Gly Thr
115 120 125
Ala Ser Val Val Cys Leu Leu Asn Asn Phe Tyr Pro Arg Glu Ala Lys
130 135 140
Val Gln Trp Lys Val Asp Asn Ala Leu Gln Ser Gly Asn Ser Gln Glu
145 150 155 160
Ser Val Thr Glu Gln Asp Ser Lys Asp Ser Thr Tyr Ser Leu Ser Ser
165 170 175
Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu Lys His Lys Val Tyr Ala
180 185 190
Cys Glu Val Thr His Gln Gly Leu Ser Ser Pro Val Thr Lys Ser Phe
195 200 205
Asn Arg Gly Glu Cys
210
<210> 44
<211> 123
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 44
Glu Val Gln Leu Gln Gln Ser Val Ala Glu Leu Val Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Thr Ala Ser Gly Phe Asn Ile Lys Asn Thr
20 25 30
Gln Met His Trp Val Lys Gln Arg Pro Glu Gln Gly Leu Glu Trp Ile
35 40 45
Gly Arg Ile Asp Pro Ala Asn Gly Asn Thr Lys Tyr Ala Pro Lys Phe
50 55 60
Gln Gly Lys Ala Thr Ile Thr Gly Asp Thr Ser Ser Asn Thr Ala Tyr
65 70 75 80
Leu Gln Leu Ser Ser Leu Thr Ser Glu Asp Thr Ala Ile Tyr Tyr Cys
85 90 95
Ala Arg Phe Asp Tyr Tyr Tyr Gly Ser Gly Asp Tyr Ala Met Asp Tyr
100 105 110
Trp Gly Gln Gly Thr Ser Val Thr Val Ser Ser
115 120
<210> 45
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 45
Asp Val Val Met Thr Gln Thr Pro Leu Thr Leu Ser Val Thr Ile Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Tyr Ser
20 25 30
Asn Gly Lys Thr Tyr Leu Asn Trp Leu Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Lys Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro
50 55 60
Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Val Gln Gly
85 90 95
Thr Arg Phe Pro Trp Thr Phe Gly Gly Gly Thr Asn Leu Glu Ile Lys
100 105 110
<210> 46
<211> 521
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 46
Glu Val Gln Leu Gln Gln Ser Val Ala Glu Leu Val Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Thr Ala Ser Gly Phe Asn Ile Lys Asn Thr
20 25 30
Gln Met His Trp Val Lys Gln Arg Pro Glu Gln Gly Leu Glu Trp Ile
35 40 45
Gly Arg Ile Asp Pro Ala Asn Gly Asn Thr Lys Tyr Ala Pro Lys Phe
50 55 60
Gln Gly Lys Ala Thr Ile Thr Gly Asp Thr Ser Ser Asn Thr Ala Tyr
65 70 75 80
Leu Gln Leu Ser Ser Leu Thr Ser Glu Asp Thr Ala Ile Tyr Tyr Cys
85 90 95
Ala Arg Phe Asp Tyr Tyr Tyr Gly Ser Gly Asp Tyr Ala Met Asp Tyr
100 105 110
Trp Gly Gln Gly Thr Ser Val Thr Val Ser Ser Thr Thr Thr Ala Pro
115 120 125
Ser Val Tyr Pro Leu Val Pro Gly Cys Ser Asp Thr Ser Gly Ser Ser
130 135 140
Val Thr Leu Gly Cys Leu Val Lys Gly Tyr Phe Pro Glu Pro Val Thr
145 150 155 160
Val Lys Trp Asn Tyr Gly Ala Leu Ser Ser Gly Val Arg Thr Val Ser
165 170 175
Ser Val Leu Gln Ser Gly Phe Tyr Ser Leu Ser Ser Leu Val Thr Val
180 185 190
Pro Ser Ser Thr Trp Pro Ser Gln Thr Val Ile Cys Asn Val Ala His
195 200 205
Pro Ala Ser Lys Thr Glu Leu Ile Lys Arg Ile Glu Pro Arg Ile Pro
210 215 220
Lys Pro Ser Thr Pro Pro Gly Ser Ser Cys Pro Pro Gly Asn Ile Leu
225 230 235 240
Gly Gly Pro Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Ala Leu
245 250 255
Met Ile Ser Leu Thr Pro Lys Val Thr Cys Val Val Val Asp Val Ser
260 265 270
Glu Asp Asp Pro Asp Val His Val Ser Trp Phe Val Asp Asn Lys Glu
275 280 285
Val His Thr Ala Trp Thr Gln Pro Arg Glu Ala Gln Tyr Asn Ser Thr
290 295 300
Phe Arg Val Val Ser Ala Leu Pro Ile Gln His Gln Asp Trp Met Arg
305 310 315 320
Gly Lys Glu Phe Lys Cys Lys Val Asn Asn Lys Ala Leu Pro Ala Pro
325 330 335
Ile Glu Arg Thr Ile Ser Lys Pro Lys Gly Arg Ala Gln Thr Pro Gln
340 345 350
Val Tyr Thr Ile Pro Pro Pro Arg Glu Gln Met Ser Lys Lys Lys Val
355 360 365
Ser Leu Thr Cys Leu Val Thr Asn Phe Phe Ser Glu Ala Ile Ser Val
370 375 380
Glu Trp Glu Arg Asn Gly Glu Leu Glu Gln Asp Tyr Lys Asn Thr Pro
385 390 395 400
Pro Ile Leu Asp Ser Asp Gly Thr Tyr Phe Leu Tyr Ser Lys Leu Thr
405 410 415
Val Asp Thr Asp Ser Trp Leu Gln Gly Glu Ile Phe Thr Cys Ser Val
420 425 430
Val His Glu Ala Leu His Asn His His Thr Gln Lys Asn Leu Ser Arg
435 440 445
Ser Pro Glu Leu Glu Leu Asn Glu Thr Cys Ala Glu Ala Gln Asp Gly
450 455 460
Glu Leu Asp Gly Leu Trp Thr Thr Ile Thr Ile Phe Ile Ser Leu Phe
465 470 475 480
Leu Leu Ser Val Cys Tyr Ser Ala Ser Val Thr Leu Phe Lys Val Lys
485 490 495
Trp Ile Phe Ser Ser Val Val Gln Val Lys Gln Thr Ala Ile Pro Asp
500 505 510
Tyr Arg Asn Met Ile Gly Gln Gly Ala
515 520
<210> 47
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 47
Asp Val Val Met Thr Gln Thr Pro Leu Thr Leu Ser Val Thr Ile Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Tyr Ser
20 25 30
Asn Gly Lys Thr Tyr Leu Asn Trp Leu Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Lys Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro
50 55 60
Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Val Gln Gly
85 90 95
Thr Arg Phe Pro Trp Thr Phe Gly Gly Gly Thr Asn Leu Glu Ile Lys
100 105 110
Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu
115 120 125
Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe
130 135 140
Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg
145 150 155 160
Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu
180 185 190
Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser
195 200 205
Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys
210 215
<210> 48
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 48
Gly Phe Asn Ile Lys Asn Thr Gln Met His
1 5 10
<210> 49
<211> 13
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 49
Arg Ile Asp Pro Ala Asn Gly Asn Thr Lys Tyr Ala Pro
1 5 10
<210> 50
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 50
Ala Arg Phe Asp Tyr Tyr Tyr Gly Ser Gly Asp Tyr Ala Met Asp Tyr
1 5 10 15
<210> 51
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 51
Lys Ser Ser Gln Ser Leu Leu Tyr Ser Asn Gly Lys Thr Tyr Leu Asn
1 5 10 15
<210> 52
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 52
Leu Val Ser Lys Leu Asp Ser
1 5
<210> 53
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 53
Val Gln Gly Thr Arg Phe Pro Trp Thr
1 5
<210> 54
<211> 452
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 54
Glu Val Gln Leu Gln Gln Ser Val Ala Glu Leu Val Arg Pro Gly Ala
1 5 10 15
Ser Val Lys Leu Ser Cys Thr Ala Ser Gly Phe Asn Ile Lys Asn Thr
20 25 30
Gln Met His Trp Val Lys Gln Arg Pro Glu Gln Gly Leu Glu Trp Ile
35 40 45
Gly Arg Ile Asp Pro Ala Asn Gly Asn Thr Lys Tyr Ala Pro Lys Phe
50 55 60
Gln Gly Lys Ala Thr Ile Thr Gly Asp Thr Ser Ser Asn Thr Ala Tyr
65 70 75 80
Leu Gln Leu Ser Ser Leu Thr Ser Glu Asp Thr Ala Ile Tyr Tyr Cys
85 90 95
Ala Arg Phe Asp Tyr Tyr Tyr Gly Ser Gly Asp Tyr Ala Met Asp Tyr
100 105 110
Trp Gly Gln Gly Thr Ser Val Thr Val Ser Ser Ala Ser Thr Lys Gly
115 120 125
Pro Ser Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly
130 135 140
Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val
145 150 155 160
Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe
165 170 175
Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val
180 185 190
Thr Val Pro Ser Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val
195 200 205
Asn His Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys
210 215 220
Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu
225 230 235 240
Leu Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr
245 250 255
Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val
260 265 270
Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val
275 280 285
Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser
290 295 300
Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu
305 310 315 320
Asn Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala
325 330 335
Pro Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro
340 345 350
Gln Val Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln
355 360 365
Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala
370 375 380
Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr
385 390 395 400
Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu
405 410 415
Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser
420 425 430
Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser
435 440 445
Leu Ser Pro Gly
450
<210> 55
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 55
Asp Val Val Met Thr Gln Thr Pro Leu Thr Leu Ser Val Thr Ile Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Lys Ser Ser Gln Ser Leu Leu Tyr Ser
20 25 30
Asn Gly Lys Thr Tyr Leu Asn Trp Leu Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Lys Arg Leu Ile Tyr Leu Val Ser Lys Leu Asp Ser Gly Val Pro
50 55 60
Asp Arg Phe Thr Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Tyr Cys Val Gln Gly
85 90 95
Thr Arg Phe Pro Trp Thr Phe Gly Gly Gly Thr Asn Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 56
<211> 117
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 56
Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Met Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Lys Gln Thr Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Asn Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met His Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Leu Thr Val Ser Ala
115
<210> 57
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 57
Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly
1 5 10 15
Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 58
<211> 440
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 58
Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Met Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Lys Gln Thr Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Asn Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met His Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Leu Thr Val Ser Ala Ala Lys Thr Thr Pro Pro Ser Val Tyr Pro Leu
115 120 125
Ala Pro Gly Ser Ala Ala Gln Thr Asn Ser Met Val Thr Leu Gly Cys
130 135 140
Leu Val Lys Gly Tyr Phe Pro Glu Pro Val Thr Val Thr Trp Asn Ser
145 150 155 160
Gly Ser Leu Ser Ser Gly Val His Thr Phe Pro Ala Val Leu Glu Ser
165 170 175
Asp Leu Tyr Thr Leu Ser Ser Ser Val Thr Val Pro Ser Ser Pro Arg
180 185 190
Pro Ser Glu Thr Val Thr Cys Asn Val Ala His Pro Ala Ser Ser Thr
195 200 205
Lys Val Asp Lys Lys Ile Val Pro Arg Asp Cys Gly Cys Lys Pro Cys
210 215 220
Ile Cys Thr Val Pro Glu Val Ser Ser Val Phe Ile Phe Pro Pro Lys
225 230 235 240
Pro Lys Asp Val Leu Thr Ile Thr Leu Thr Pro Lys Val Thr Cys Val
245 250 255
Val Val Asp Ile Ser Lys Asp Asp Pro Glu Val Gln Phe Ser Trp Phe
260 265 270
Val Asp Asp Val Glu Val His Thr Ala Gln Thr Gln Pro Arg Glu Glu
275 280 285
Gln Phe Asn Ser Thr Phe Arg Ser Val Ser Glu Leu Pro Ile Met His
290 295 300
Gln Asp Trp Leu Asn Gly Lys Glu Phe Lys Cys Arg Val Asn Ser Ala
305 310 315 320
Ala Phe Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys Thr Lys Gly Arg
325 330 335
Pro Lys Ala Pro Gln Val Tyr Thr Ile Pro Pro Pro Lys Glu Gln Met
340 345 350
Ala Lys Asp Lys Val Ser Leu Thr Cys Met Ile Thr Asp Phe Phe Pro
355 360 365
Glu Asp Ile Thr Val Glu Trp Gln Trp Asn Gly Gln Pro Ala Glu Asn
370 375 380
Tyr Lys Asn Thr Gln Pro Ile Met Asn Thr Asn Gly Ser Tyr Phe Val
385 390 395 400
Tyr Ser Lys Leu Asn Val Gln Lys Ser Asn Trp Glu Ala Gly Asn Thr
405 410 415
Phe Thr Cys Ser Val Leu His Glu Gly Leu His Asn His His Thr Glu
420 425 430
Lys Ser Leu Ser His Ser Pro Gly
435 440
<210> 59
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 59
Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly
1 5 10 15
Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu
115 120 125
Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe
130 135 140
Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg
145 150 155 160
Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu
180 185 190
Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser
195 200 205
Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys
210 215
<210> 60
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 60
Gly Tyr Thr Phe Thr Ser Tyr Asn Met His
1 5 10
<210> 61
<211> 13
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 61
Ala Ile Tyr Pro Gly Asn Gly Asp Thr Ser Tyr Asn Gln
1 5 10
<210> 62
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 62
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr
1 5 10
<210> 63
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 63
Arg Ser Ser Gln Ser Leu Val His Ser Asn Gly Asn Thr Tyr Leu Tyr
1 5 10 15
<210> 64
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 64
Lys Val Ser Asn Arg Phe Ser
1 5
<210> 65
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 65
Ser Gln Ser Thr His Val Pro Tyr Thr
1 5
<210> 66
<211> 446
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 66
Gln Val Gln Leu Gln Gln Pro Gly Ala Glu Leu Val Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Met Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Lys Gln Thr Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Asn Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Lys Ala Thr Leu Thr Ala Asp Lys Ser Ser Ser Thr Ala Tyr
65 70 75 80
Met His Leu Ser Ser Leu Thr Ser Glu Asp Ser Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Leu Thr Val Ser Ala Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu
115 120 125
Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys
130 135 140
Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser
145 150 155 160
Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser
165 170 175
Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser
180 185 190
Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn
195 200 205
Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His
210 215 220
Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val
225 230 235 240
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
245 250 255
Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu
260 265 270
Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
275 280 285
Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser
290 295 300
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
305 310 315 320
Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile
325 330 335
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
340 345 350
Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
355 360 365
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
370 375 380
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
385 390 395 400
Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg
405 410 415
Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
420 425 430
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440 445
<210> 67
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 67
Asp Val Val Met Thr Gln Thr Pro Leu Ser Leu Pro Val Ser Leu Gly
1 5 10 15
Asp Gln Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Lys Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Leu Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 68
<211> 117
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 68
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Arg Gln Ala Pro Gly Gln Arg Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Gln Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Ala Thr Leu Thr Ala Asp Lys Ser Ala Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser
115
<210> 69
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 69
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Ala Gln Thr Tyr Leu Tyr Trp Tyr Gln Gln Arg Pro Gly Gln Ser
35 40 45
Pro Arg Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 70
<211> 446
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 70
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Arg Gln Ala Pro Gly Gln Arg Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Gln Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Ala Thr Leu Thr Ala Asp Lys Ser Ala Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu
115 120 125
Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys
130 135 140
Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser
145 150 155 160
Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser
165 170 175
Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser
180 185 190
Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn
195 200 205
Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His
210 215 220
Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val
225 230 235 240
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
245 250 255
Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu
260 265 270
Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
275 280 285
Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser
290 295 300
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
305 310 315 320
Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile
325 330 335
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
340 345 350
Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
355 360 365
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
370 375 380
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
385 390 395 400
Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg
405 410 415
Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
420 425 430
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440 445
<210> 71
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 71
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Ala Gln Thr Tyr Leu Tyr Trp Tyr Gln Gln Arg Pro Gly Gln Ser
35 40 45
Pro Arg Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 72
<211> 13
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 72
Ala Ile Tyr Pro Gly Gln Gly Asp Thr Ser Tyr Asn Gln
1 5 10
<210> 73
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 73
Arg Ser Ser Gln Ser Leu Val His Ser Asn Ala Gln Thr Tyr Leu Tyr
1 5 10 15
<210> 74
<211> 117
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 74
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Ala Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Ala Thr Leu Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser
115
<210> 75
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 75
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Ala Ala Thr Tyr Leu Tyr Trp Tyr Gln Gln Arg Pro Gly Gln Ser
35 40 45
Pro Arg Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 76
<211> 446
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 76
Glu Val Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Ser Tyr
20 25 30
Asn Met His Trp Val Arg Gln Ala Pro Gly Gln Gly Leu Glu Trp Ile
35 40 45
Gly Ala Ile Tyr Pro Gly Ala Gly Asp Thr Ser Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Ala Thr Leu Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Lys Gly Gly Thr Pro Phe Ala Tyr Trp Gly Gln Gly Thr Leu
100 105 110
Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu
115 120 125
Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys
130 135 140
Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser
145 150 155 160
Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gln Ser
165 170 175
Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser
180 185 190
Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His Lys Pro Ser Asn
195 200 205
Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys Asp Lys Thr His
210 215 220
Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val
225 230 235 240
Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr
245 250 255
Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu
260 265 270
Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys
275 280 285
Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser
290 295 300
Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys
305 310 315 320
Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile
325 330 335
Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro
340 345 350
Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu
355 360 365
Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn
370 375 380
Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser
385 390 395 400
Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg
405 410 415
Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu
420 425 430
His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly
435 440 445
<210> 77
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 77
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Leu Val His Ser
20 25 30
Asn Ala Ala Thr Tyr Leu Tyr Trp Tyr Gln Gln Arg Pro Gly Gln Ser
35 40 45
Pro Arg Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Phe Cys Ser Gln Ser
85 90 95
Thr His Val Pro Tyr Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 78
<211> 13
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 78
Ala Ile Tyr Pro Gly Ala Gly Asp Thr Ser Tyr Asn Gln
1 5 10
<210> 79
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 79
Arg Ser Ser Gln Ser Leu Val His Ser Asn Ala Ala Thr Tyr Leu Tyr
1 5 10 15
<210> 80
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 80
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Met
65 70 75 80
Leu Tyr Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Ala Gly Thr Thr Val Thr Val Ser Ser
115 120
<210> 81
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 81
Asp Ile Val Met Thr Gln Ala Glu Pro Ser Val Pro Val Thr Pro Gly
1 5 10 15
Glu Ser Ile Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Ala Phe Thr Leu Arg Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 82
<211> 444
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 82
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Met
65 70 75 80
Leu Tyr Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Ala Gly Thr Thr Val Thr Val Ser Ser Ala Lys Thr Thr Pro Pro Ser
115 120 125
Val Tyr Pro Leu Ala Pro Gly Ser Ala Ala Gln Thr Asn Ser Met Val
130 135 140
Thr Leu Gly Cys Leu Val Lys Gly Tyr Phe Pro Glu Pro Val Thr Val
145 150 155 160
Thr Trp Asn Ser Gly Ser Leu Ser Ser Gly Val His Thr Phe Pro Ala
165 170 175
Val Leu Glu Ser Asp Leu Tyr Thr Leu Ser Ser Ser Val Thr Val Pro
180 185 190
Ser Ser Pro Arg Pro Ser Glu Thr Val Thr Cys Asn Val Ala His Pro
195 200 205
Ala Ser Ser Thr Lys Val Asp Lys Lys Ile Val Pro Arg Asp Cys Gly
210 215 220
Cys Lys Pro Cys Ile Cys Thr Val Pro Glu Val Ser Ser Val Phe Ile
225 230 235 240
Phe Pro Pro Lys Pro Lys Asp Val Leu Thr Ile Thr Leu Thr Pro Lys
245 250 255
Val Thr Cys Val Val Val Asp Ile Ser Lys Asp Asp Pro Glu Val Gln
260 265 270
Phe Ser Trp Phe Val Asp Asp Val Glu Val His Thr Ala Gln Thr Gln
275 280 285
Pro Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg Ser Val Ser Glu Leu
290 295 300
Pro Ile Met His Gln Asp Trp Leu Asn Gly Lys Glu Phe Lys Cys Arg
305 310 315 320
Val Asn Ser Ala Ala Phe Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys
325 330 335
Thr Lys Gly Arg Pro Lys Ala Pro Gln Val Tyr Thr Ile Pro Pro Pro
340 345 350
Lys Glu Gln Met Ala Lys Asp Lys Val Ser Leu Thr Cys Met Ile Thr
355 360 365
Asp Phe Phe Pro Glu Asp Ile Thr Val Glu Trp Gln Trp Asn Gly Gln
370 375 380
Pro Ala Glu Asn Tyr Lys Asn Thr Gln Pro Ile Met Asn Thr Asn Gly
385 390 395 400
Ser Tyr Phe Val Tyr Ser Lys Leu Asn Val Gln Lys Ser Asn Trp Glu
405 410 415
Ala Gly Asn Thr Phe Thr Cys Ser Val Leu His Glu Gly Leu His Asn
420 425 430
His His Thr Glu Lys Ser Leu Ser His Ser Pro Gly
435 440
<210> 83
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 83
Asp Ile Val Met Thr Gln Ala Glu Pro Ser Val Pro Val Thr Pro Gly
1 5 10 15
Glu Ser Ile Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Ala Phe Thr Leu Arg Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Ala Asp Ala Ala Pro Thr Val Ser Ile Phe Pro Pro Ser Ser Glu
115 120 125
Gln Leu Thr Ser Gly Gly Ala Ser Val Val Cys Phe Leu Asn Asn Phe
130 135 140
Tyr Pro Lys Asp Ile Asn Val Lys Trp Lys Ile Asp Gly Ser Glu Arg
145 150 155 160
Gln Asn Gly Val Leu Asn Ser Trp Thr Asp Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Met Ser Ser Thr Leu Thr Leu Thr Lys Asp Glu Tyr Glu
180 185 190
Arg His Asn Ser Tyr Thr Cys Glu Ala Thr His Lys Thr Ser Thr Ser
195 200 205
Pro Ile Val Lys Ser Phe Asn Arg Asn Glu Cys
210 215
<210> 84
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 84
Gly Phe Thr Phe Asn Thr Tyr Ala Met Asn
1 5 10
<210> 85
<211> 19
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 85
Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp Ser
1 5 10 15
Val Lys Asp
<210> 86
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 86
Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val
1 5 10
<210> 87
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 87
Arg Ser Ser Lys Ser Leu Leu His Ser Asn Gly Asn Thr Tyr Leu Tyr
1 5 10 15
<210> 88
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 88
Arg Met Ser Asn Leu Ala Ser
1 5
<210> 89
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 89
Met Gln His Leu Glu Tyr Pro Phe Thr
1 5
<210> 90
<211> 450
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 90
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Lys Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Gln Ser Met
65 70 75 80
Leu Tyr Leu Gln Met Asn Asn Leu Lys Thr Glu Asp Thr Ala Met Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Ala Gly Thr Thr Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
115 120 125
Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala
130 135 140
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
145 150 155 160
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
165 170 175
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
180 185 190
Pro Ser Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His
195 200 205
Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys
210 215 220
Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly
225 230 235 240
Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met
245 250 255
Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His
260 265 270
Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val
275 280 285
His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr
290 295 300
Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly
305 310 315 320
Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile
325 330 335
Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val
340 345 350
Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser
355 360 365
Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu
370 375 380
Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro
385 390 395 400
Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val
405 410 415
Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met
420 425 430
His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser
435 440 445
Pro Gly
450
<210> 91
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 91
Asp Ile Val Met Thr Gln Ala Glu Pro Ser Val Pro Val Thr Pro Gly
1 5 10 15
Glu Ser Ile Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Arg Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Ala Phe Thr Leu Arg Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 92
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 92
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Ser Gly Lys Gly Leu Glu Trp Val
35 40 45
Gly Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Lys Asn Thr
65 70 75 80
Ala Tyr Leu Gln Met Asn Ser Leu Lys Thr Glu Asp Thr Ala Val Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Gln Gly Thr Thr Val Thr Val Ser Ser
115 120
<210> 93
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 93
Asp Ile Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Pro Gly
1 5 10 15
Glu Pro Ala Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
<210> 94
<211> 450
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 94
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Gly
1 5 10 15
Ser Leu Lys Leu Ser Cys Ala Ala Ser Gly Phe Thr Phe Asn Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Ser Gly Lys Gly Leu Glu Trp Val
35 40 45
Gly Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Lys Asn Thr
65 70 75 80
Ala Tyr Leu Gln Met Asn Ser Leu Lys Thr Glu Asp Thr Ala Val Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Gln Gly Thr Thr Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
115 120 125
Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala
130 135 140
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
145 150 155 160
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
165 170 175
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
180 185 190
Pro Ser Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His
195 200 205
Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys
210 215 220
Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly
225 230 235 240
Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met
245 250 255
Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His
260 265 270
Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val
275 280 285
His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr
290 295 300
Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly
305 310 315 320
Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile
325 330 335
Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val
340 345 350
Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser
355 360 365
Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu
370 375 380
Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro
385 390 395 400
Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val
405 410 415
Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met
420 425 430
His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser
435 440 445
Pro Gly
450
<210> 95
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 95
Asp Ile Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Pro Gly
1 5 10 15
Glu Pro Ala Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 96
<211> 121
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 96
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Thr Ala Ser Gly Phe Thr Phe Ala Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Lys Ser Ile
65 70 75 80
Leu Tyr Leu Gln Met Asn Ser Leu Lys Thr Glu Asp Thr Ala Val Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Gln Gly Thr Thr Val Thr Val Ser Ser
115 120
<210> 97
<211> 112
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 97
Asp Ile Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Pro Gly
1 5 10 15
Glu Pro Ala Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
<210> 98
<211> 450
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 98
Glu Val Gln Leu Val Glu Ser Gly Gly Gly Leu Val Gln Pro Gly Arg
1 5 10 15
Ser Leu Arg Leu Ser Cys Thr Ala Ser Gly Phe Thr Phe Ala Thr Tyr
20 25 30
Ala Met Asn Trp Val Arg Gln Ala Pro Gly Lys Gly Leu Glu Trp Val
35 40 45
Ala Arg Ile Arg Ser Lys Ser Asn Asn Tyr Ala Thr Tyr Tyr Ala Asp
50 55 60
Ser Val Lys Asp Arg Phe Thr Ile Ser Arg Asp Asp Ser Lys Ser Ile
65 70 75 80
Leu Tyr Leu Gln Met Asn Ser Leu Lys Thr Glu Asp Thr Ala Val Tyr
85 90 95
Tyr Cys Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val Trp Gly
100 105 110
Gln Gly Thr Thr Val Thr Val Ser Ser Ala Ser Thr Lys Gly Pro Ser
115 120 125
Val Phe Pro Leu Ala Pro Ser Ser Lys Ser Thr Ser Gly Gly Thr Ala
130 135 140
Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val
145 150 155 160
Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala
165 170 175
Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val
180 185 190
Pro Ser Ser Ser Leu Gly Thr Gln Thr Tyr Ile Cys Asn Val Asn His
195 200 205
Lys Pro Ser Asn Thr Lys Val Asp Lys Lys Val Glu Pro Lys Ser Cys
210 215 220
Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly
225 230 235 240
Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met
245 250 255
Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His
260 265 270
Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val
275 280 285
His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr
290 295 300
Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly
305 310 315 320
Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile
325 330 335
Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val
340 345 350
Tyr Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser
355 360 365
Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu
370 375 380
Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro
385 390 395 400
Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val
405 410 415
Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met
420 425 430
His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser
435 440 445
Pro Gly
450
<210> 99
<211> 219
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 99
Asp Ile Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Pro Gly
1 5 10 15
Glu Pro Ala Ser Ile Ser Cys Arg Ser Ser Lys Ser Leu Leu His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Tyr Trp Phe Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Arg Met Ser Asn Leu Ala Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Met Gln His
85 90 95
Leu Glu Tyr Pro Phe Thr Phe Gly Gly Gly Thr Lys Val Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 100
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 100
Gly Phe Thr Phe Ala Thr Tyr Ala Met Asn
1 5 10
<210> 101
<211> 355
<212> PRT
<213> Intelligent (Homo sapiens)
<400> 101
Met Asp Tyr Thr Leu Asp Leu Ser Val Thr Thr Val Thr Asp Tyr Tyr
1 5 10 15
Tyr Pro Asp Ile Phe Ser Ser Pro Cys Asp Ala Glu Leu Ile Gln Thr
20 25 30
Asn Gly Lys Leu Leu Leu Ala Val Phe Tyr Cys Leu Leu Phe Val Phe
35 40 45
Ser Leu Leu Gly Asn Ser Leu Val Ile Leu Val Leu Val Val Cys Lys
50 55 60
Lys Leu Arg Ser Ile Thr Asp Val Tyr Leu Leu Asn Leu Ala Leu Ser
65 70 75 80
Asp Leu Leu Phe Val Phe Ser Phe Pro Phe Gln Thr Tyr Tyr Leu Leu
85 90 95
Asp Gln Trp Val Phe Gly Thr Val Met Cys Lys Val Val Ser Gly Phe
100 105 110
Tyr Tyr Ile Gly Phe Tyr Ser Ser Met Phe Phe Ile Thr Leu Met Ser
115 120 125
Val Asp Arg Tyr Leu Ala Val Val His Ala Val Tyr Ala Leu Lys Val
130 135 140
Arg Thr Ile Arg Met Gly Thr Thr Leu Cys Leu Ala Val Trp Leu Thr
145 150 155 160
Ala Ile Met Ala Thr Ile Pro Leu Leu Val Phe Tyr Gln Val Ala Ser
165 170 175
Glu Asp Gly Val Leu Gln Cys Tyr Ser Phe Tyr Asn Gln Gln Thr Leu
180 185 190
Lys Trp Lys Ile Phe Thr Asn Phe Lys Met Asn Ile Leu Gly Leu Leu
195 200 205
Ile Pro Phe Thr Ile Phe Met Phe Cys Tyr Ile Lys Ile Leu His Gln
210 215 220
Leu Lys Arg Cys Gln Asn His Asn Lys Thr Lys Ala Ile Arg Leu Val
225 230 235 240
Leu Ile Val Val Ile Ala Ser Leu Leu Phe Trp Val Pro Phe Asn Val
245 250 255
Val Leu Phe Leu Thr Ser Leu His Ser Met His Ile Leu Asp Gly Cys
260 265 270
Ser Ile Ser Gln Gln Leu Thr Tyr Ala Thr His Val Thr Glu Ile Ile
275 280 285
Ser Phe Thr His Cys Cys Val Asn Pro Val Ile Tyr Ala Phe Val Gly
290 295 300
Glu Lys Phe Lys Lys His Leu Ser Glu Ile Phe Gln Lys Ser Cys Ser
305 310 315 320
Gln Ile Phe Asn Tyr Leu Gly Arg Gln Met Pro Arg Glu Ser Cys Glu
325 330 335
Lys Ser Ser Ser Cys Gln Gln His Ser Ser Arg Ser Ser Ser Val Asp
340 345 350
Tyr Ile Leu
355
<210> 102
<211> 353
<212> PRT
<213> little mouse (Mus musculus)
<400> 102
Met Asp Tyr Thr Met Glu Pro Asn Val Thr Met Thr Asp Tyr Tyr Pro
1 5 10 15
Asp Phe Phe Thr Ala Pro Cys Asp Ala Glu Phe Leu Leu Arg Gly Ser
20 25 30
Met Leu Tyr Leu Ala Ile Leu Tyr Cys Val Leu Phe Val Leu Gly Leu
35 40 45
Leu Gly Asn Ser Leu Val Ile Leu Val Leu Val Gly Cys Lys Lys Leu
50 55 60
Arg Ser Ile Thr Asp Ile Tyr Leu Leu Asn Leu Ala Ala Ser Asp Leu
65 70 75 80
Leu Phe Val Leu Ser Ile Pro Phe Gln Thr His Asn Leu Leu Asp Gln
85 90 95
Trp Val Phe Gly Thr Ala Met Cys Lys Val Val Ser Gly Leu Tyr Tyr
100 105 110
Ile Gly Phe Phe Ser Ser Met Phe Phe Ile Thr Leu Met Ser Val Asp
115 120 125
Arg Tyr Leu Ala Ile Val His Ala Val Tyr Ala Ile Lys Val Arg Thr
130 135 140
Ala Ser Val Gly Thr Ala Leu Ser Leu Thr Val Trp Leu Ala Ala Val
145 150 155 160
Thr Ala Thr Ile Pro Leu Met Val Phe Tyr Gln Val Ala Ser Glu Asp
165 170 175
Gly Met Leu Gln Cys Phe Gln Phe Tyr Glu Glu Gln Ser Leu Arg Trp
180 185 190
Lys Leu Phe Thr His Phe Glu Ile Asn Ala Leu Gly Leu Leu Leu Pro
195 200 205
Phe Ala Ile Leu Leu Phe Cys Tyr Val Arg Ile Leu Gln Gln Leu Arg
210 215 220
Gly Cys Leu Asn His Asn Arg Thr Arg Ala Ile Lys Leu Val Leu Thr
225 230 235 240
Val Val Ile Val Ser Leu Leu Phe Trp Val Pro Phe Asn Val Ala Leu
245 250 255
Phe Leu Thr Ser Leu His Asp Leu His Ile Leu Asp Gly Cys Ala Thr
260 265 270
Arg Gln Arg Leu Ala Leu Ala Ile His Val Thr Glu Val Ile Ser Phe
275 280 285
Thr His Cys Cys Val Asn Pro Val Ile Tyr Ala Phe Ile Gly Glu Lys
290 295 300
Phe Lys Lys His Leu Met Asp Val Phe Gln Lys Ser Cys Ser His Ile
305 310 315 320
Phe Leu Tyr Leu Gly Arg Gln Met Pro Val Gly Ala Leu Glu Arg Gln
325 330 335
Leu Ser Ser Asn Gln Arg Ser Ser His Ser Ser Thr Leu Asp Asp Ile
340 345 350
Leu
<210> 103
<211> 355
<212> PRT
<213> crab eating macaque (Macaca fascicularis)
<400> 103
Met Asp Tyr Thr Leu Asp Pro Ser Met Thr Thr Met Thr Asp Tyr Tyr
1 5 10 15
Tyr Pro Asp Ser Leu Ser Ser Pro Cys Asp Gly Glu Leu Ile Gln Arg
20 25 30
Asn Asp Lys Leu Leu Leu Ala Val Phe Tyr Cys Leu Leu Phe Val Phe
35 40 45
Ser Leu Leu Gly Asn Ser Leu Val Ile Leu Val Leu Val Val Cys Lys
50 55 60
Lys Leu Arg Asn Ile Thr Asp Ile Tyr Leu Leu Asn Leu Ala Leu Ser
65 70 75 80
Asp Leu Leu Phe Val Phe Ser Phe Pro Phe Gln Thr Tyr Tyr Gln Leu
85 90 95
Asp Gln Trp Val Phe Gly Thr Val Met Cys Lys Val Val Ser Gly Phe
100 105 110
Tyr Tyr Ile Gly Phe Tyr Ser Ser Met Phe Phe Ile Thr Leu Met Ser
115 120 125
Val Asp Arg Tyr Leu Ala Val Val His Ala Val Tyr Ala Ile Lys Val
130 135 140
Arg Thr Ile Arg Met Gly Thr Thr Leu Ser Leu Val Val Trp Leu Thr
145 150 155 160
Ala Ile Met Ala Thr Ile Pro Leu Leu Val Phe Tyr Gln Val Ala Ser
165 170 175
Glu Asp Gly Val Leu Gln Cys Tyr Ser Phe Tyr Asn Gln Gln Thr Leu
180 185 190
Lys Trp Lys Ile Phe Thr Asn Phe Glu Met Asn Ile Leu Gly Leu Leu
195 200 205
Ile Pro Phe Thr Ile Phe Met Phe Cys Tyr Ile Lys Ile Leu His Gln
210 215 220
Leu Lys Arg Cys Gln Asn His Asn Lys Thr Lys Ala Ile Arg Leu Val
225 230 235 240
Leu Ile Val Val Ile Ala Ser Leu Leu Phe Trp Val Pro Phe Asn Val
245 250 255
Val Leu Phe Leu Thr Ser Leu His Ser Met His Ile Leu Asp Gly Cys
260 265 270
Ser Ile Ser Gln Gln Leu Asn Tyr Ala Thr His Val Thr Glu Ile Ile
275 280 285
Ser Phe Thr His Cys Cys Val Asn Pro Val Ile Tyr Ala Phe Val Gly
290 295 300
Glu Lys Phe Lys Lys His Leu Ser Glu Ile Phe Gln Lys Ser Cys Ser
305 310 315 320
His Ile Phe Ile Tyr Leu Gly Arg Gln Met Pro Arg Glu Ser Cys Glu
325 330 335
Lys Ser Ser Ser Cys Gln Gln His Ser Phe Arg Ser Ser Ser Ile Asp
340 345 350
Tyr Ile Leu
355
<210> 104
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 104
Ala Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val
1 5 10
<210> 105
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 105
Val Ala Gly Leu Leu Arg Tyr Arg Phe Phe Asp Val
1 5 10
<210> 106
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 106
Val Arg Ala Leu Leu Arg Tyr Arg Phe Phe Asp Val
1 5 10
<210> 107
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 107
Val Arg Gly Ala Leu Arg Tyr Arg Phe Phe Asp Val
1 5 10
<210> 108
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 108
Val Arg Gly Leu Ala Arg Tyr Arg Phe Phe Asp Val
1 5 10
<210> 109
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 109
Val Arg Gly Leu Leu Ala Tyr Arg Phe Phe Asp Val
1 5 10
<210> 110
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 110
Val Arg Gly Leu Leu Arg Ala Arg Phe Phe Asp Val
1 5 10
<210> 111
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 111
Val Arg Gly Leu Leu Arg Tyr Ala Phe Phe Asp Val
1 5 10
<210> 112
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 112
Val Arg Gly Leu Leu Arg Tyr Arg Ala Phe Asp Val
1 5 10
<210> 113
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 113
Val Arg Gly Leu Leu Arg Tyr Arg Phe Ala Asp Val
1 5 10
<210> 114
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 114
Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Ala Val
1 5 10
<210> 115
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 115
Val Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Ala
1 5 10
<210> 116
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 116
Ala Arg Gly Leu Leu Arg Ala Arg Phe Phe Asp Val
1 5 10
<210> 117
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 117
Ala Arg Gly Leu Leu Arg Tyr Arg Phe Phe Asp Ala
1 5 10
<210> 118
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 118
Val Arg Gly Leu Leu Arg Ala Arg Phe Phe Asp Ala
1 5 10
<210> 119
<211> 12
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 119
Ala Arg Gly Leu Leu Arg Ala Arg Phe Phe Asp Ala
1 5 10
<210> 120
<211> 16
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 120
Arg Ser Ser Gln Ser Leu Leu His Ser Asn Gly Tyr Asn Tyr Leu Asp
1 5 10 15
<210> 121
<211> 7
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 121
Leu Gly Ser Asn Arg Ala Ser
1 5
<210> 122
<211> 9
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 122
Met Gln Ala Leu Gln Thr Pro Phe Val
1 5
<210> 123
<211> 10
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 123
Gly Phe Thr Phe Ser Gly Ser Ala Met His
1 5 10
<210> 124
<211> 19
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 124
Arg Ile Arg Ser Lys Ala Asn Ser Tyr Ala Thr Ala Tyr Ala Ala Ser
1 5 10 15
Val Lys Asp
<210> 125
<211> 11
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 125
Thr Arg Tyr Tyr Tyr Tyr Tyr Gly Met Asp Val
1 5 10
<210> 126
<211> 15
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 126
Arg Arg Cys Pro Leu Tyr Ile Ser Tyr Asp Pro Val Cys Arg Arg
1 5 10 15
<210> 127
<211> 19
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic peptides "
<400> 127
Arg Arg Arg Arg Cys Pro Leu Tyr Ile Ser Tyr Asp Pro Val Cys Arg
1 5 10 15
Arg Arg Arg
<210> 128
<211> 440
<212> PRT
<213> Artificial sequence
<220>
<221> Source
<223 >/comment = "description of artificial sequence: synthetic polypeptide "
<400> 128
Glu Glu Gly Lys Leu Val Ile Trp Ile Asn Gly Asp Lys Gly Tyr Asn
1 5 10 15
Gly Leu Ala Glu Val Gly Lys Lys Phe Glu Lys Asp Thr Gly Ile Lys
20 25 30
Val Thr Val Glu His Pro Asp Lys Leu Glu Glu Lys Phe Pro Gln Val
35 40 45
Ala Ala Thr Gly Asp Gly Pro Asp Ile Ile Phe Trp Ala His Asp Arg
50 55 60
Phe Gly Gly Tyr Ala Gln Ser Gly Leu Leu Ala Glu Ile Thr Pro Asp
65 70 75 80
Lys Ala Phe Gln Asp Lys Leu Tyr Pro Phe Thr Trp Asp Ala Val Arg
85 90 95
Tyr Asn Gly Lys Leu Ile Ala Tyr Pro Ile Ala Val Glu Ala Leu Ser
100 105 110
Leu Ile Tyr Asn Lys Asp Leu Leu Pro Asn Pro Pro Lys Thr Trp Glu
115 120 125
Glu Ile Pro Ala Leu Asp Lys Glu Leu Lys Ala Lys Gly Lys Ser Ala
130 135 140
Leu Met Phe Asn Leu Gln Glu Pro Tyr Phe Thr Trp Pro Leu Ile Ala
145 150 155 160
Ala Asp Gly Gly Tyr Ala Phe Lys Tyr Glu Asn Gly Lys Tyr Asp Ile
165 170 175
Lys Asp Val Gly Val Asp Asn Ala Gly Ala Lys Ala Gly Leu Thr Phe
180 185 190
Leu Ile Asp Leu Ile Lys Asn Lys His Met Asn Ala Asp Thr Asp Tyr
195 200 205
Ser Ile Ala Glu Ala Ala Phe Asn Lys Gly Glu Thr Ala Met Thr Ile
210 215 220
Asn Gly Pro Trp Ala Trp Ser Asn Ile Asp Thr Ser Lys Val Asn Tyr
225 230 235 240
Gly Val Thr Val Leu Pro Thr Phe Lys Gly Gln Pro Ser Lys Pro Phe
245 250 255
Val Gly Val Leu Ser Ala Gly Ile Asn Ala Ala Ser Pro Asn Lys Glu
260 265 270
Leu Ala Lys Glu Phe Leu Glu Asn Tyr Leu Leu Thr Asp Glu Gly Leu
275 280 285
Glu Ala Val Asn Lys Asp Lys Pro Leu Gly Ala Val Ala Leu Lys Ser
290 295 300
Tyr Glu Glu Glu Leu Ala Lys Asp Pro Arg Ile Ala Ala Thr Met Glu
305 310 315 320
Asn Ala Gln Lys Gly Glu Ile Met Pro Asn Ile Pro Gln Met Ser Ala
325 330 335
Phe Trp Tyr Ala Val Arg Thr Ala Val Ile Asn Ala Ala Ser Gly Arg
340 345 350
Gln Thr Val Asp Glu Ala Leu Lys Asp Ala Gln Thr Ser Gly Gly Gly
355 360 365
Ser Gly Gly Gly Ser Gly Gly Gly Ser Gly Gly Gly Ser His His His
370 375 380
His His His Gly Gly Gly Ser Gly Gly Gly Gly Ser Gly Gly Glu Asn
385 390 395 400
Leu Tyr Phe Gln Gly Met Asp Tyr Thr Leu Asp Leu Ser Val Thr Thr
405 410 415
Val Thr Asp Tyr Tyr Tyr Pro Asp Ile Phe Ser Ser Pro Cys Asp Ala
420 425 430
Glu Leu Ile Gln Thr Asn Gly Lys
435 440
<210> 129
<211> 4
<212> PRT
<213> Artificial sequence
<220>
<221> sources
<223 >/comment = "description of artificial sequence: synthetic peptide "
<400> 129
Ala Leu Ala Leu
1

Claims (161)

1. An isolated antibody that binds human CCR8, wherein the antibody comprises:
i) HCDR1 comprising the amino acid sequence of SEQ ID NO. 12, HCDR2 comprising the amino acid sequence of SEQ ID NO. 13, HCDR3 comprising the amino acid sequence of SEQ ID NO. 14, LCDR1 comprising the amino acid sequence of SEQ ID NO. 15, LCDR2 comprising the amino acid sequence of SEQ ID NO. 16, and LCDR3 comprising the amino acid sequence of SEQ ID NO. 17;
ii) HCDR1 comprising the amino acid sequence of SEQ ID NO. 24, HCDR2 comprising the amino acid sequence of SEQ ID NO. 25, HCDR3 comprising the amino acid sequence of SEQ ID NO. 26, LCDR1 comprising the amino acid sequence of SEQ ID NO. 27, LCDR2 comprising the amino acid sequence of SEQ ID NO. 28 and LCDR3 comprising the amino acid sequence of SEQ ID NO. 29;
iii) HCDR1 comprising the amino acid sequence of SEQ ID NO. 36, HCDR2 comprising the amino acid sequence of SEQ ID NO. 37, HCDR3 comprising the amino acid sequence of SEQ ID NO. 38, LCDR1 comprising the amino acid sequence of SEQ ID NO. 39, LCDR2 comprising the amino acid sequence of SEQ ID NO. 40, and LCDR3 comprising the amino acid sequence of SEQ ID NO. 41;
iv) HCDR1 comprising the amino acid sequence of SEQ ID NO 48, HCDR2 comprising the amino acid sequence of SEQ ID NO 49, HCDR3 comprising the amino acid sequence of SEQ ID NO 50, LCDR1 comprising the amino acid sequence of SEQ ID NO 51, LCDR2 comprising the amino acid sequence of SEQ ID NO 52 and LCDR3 comprising the amino acid sequence of SEQ ID NO 53; or
v) HCDR1 comprising the amino acid sequence of SEQ ID NO 60, HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78, HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79, LCDR1 comprising the amino acid sequence of SEQ ID NO 63, LCDR2 comprising the amino acid sequence of SEQ ID NO 64 and LCDR3 comprising the amino acid sequence of SEQ ID NO 65; or
vi) HCDR1 comprising the amino acid sequence of SEQ ID NO:84 or 100, HCDR2 comprising the amino acid sequence of SEQ ID NO:85, HCDR3 comprising the amino acid sequence of SEQ ID NO:86, LCDR1 comprising the amino acid sequence of SEQ ID NO:87, LCDR2 comprising the amino acid sequence of SEQ ID NO:88 and LCDR3 comprising the amino acid sequence of SEQ ID NO: 89.
2. The isolated antibody of claim 1, wherein the antibody comprises:
i) HCDR1 comprising the amino acid sequence of SEQ ID NO 60, HCDR2 comprising the amino acid sequence of SEQ ID NO 61, 72 or 78, HCDR3 comprising the amino acid sequence of SEQ ID NO 62, 73 or 79, LCDR1 comprising the amino acid sequence of SEQ ID NO 63, LCDR2 comprising the amino acid sequence of SEQ ID NO 64 and LCDR3 comprising the amino acid sequence of SEQ ID NO 65; or
ii) HCDR1 comprising the amino acid sequence of SEQ ID NO 84 or 100, HCDR2 comprising the amino acid sequence of SEQ ID NO 85, HCDR3 comprising the amino acid sequence of SEQ ID NO 86, LCDR1 comprising the amino acid sequence of SEQ ID NO 87, LCDR2 comprising the amino acid sequence of SEQ ID NO 88 and LCDR3 comprising the amino acid sequence of SEQ ID NO 89.
3. The isolated antibody of claim 2, wherein the antibody comprises:
i) a heavy chain variable region (VH) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 68 or 74, and a light chain variable region (VL) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 69 or 75; or
ii) a heavy chain variable region (VH) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 92 or 96, and a light chain variable region (VL) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 93 or 97.
4. The isolated antibody of claim 2 or claim 3, wherein the antibody comprises:
i) a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO:68 or 74 and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO:69 or 75; or
ii) a heavy chain variable region (VH) comprising the amino acid sequence of SEQ ID NO:92 or 96, and a light chain variable region (VL) comprising the amino acid sequence of SEQ ID NO:93 or 97.
5. The isolated antibody of any one of claims 1-4, wherein the antibody is a monoclonal antibody.
6. The isolated antibody of any one of claims 1-5, wherein the antibody is a humanized antibody.
7. The isolated antibody of any one of claims 1-6, wherein the antibody is a full-length antibody.
8. The isolated antibody of any one of claims 1-7, wherein the antibody is an IgG1 or IgG3 antibody.
9. The isolated antibody of any one of claims 1-8, wherein the antibody comprises:
i) a Heavy Chain (HC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 70 or 76, and a Light Chain (LC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 71 or 77; or
ii) a Heavy Chain (HC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 94 or 98, and a Light Chain (LC) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence of SEQ ID NO 95 or 99.
10. The isolated antibody of any one of claims 1-9, wherein the antibody comprises:
i) a Heavy Chain (HC) comprising the amino acid sequence of SEQ ID NO:70 or 76, and a Light Chain (LC) comprising the amino acid sequence of SEQ ID NO:71 or 77; or
ii) a Heavy Chain (HC) comprising the amino acid sequence of SEQ ID NO:94 or 98, and a Light Chain (LC) comprising the amino acid sequence of SEQ ID NO:95 or 99.
11. The isolated antibody of any one of claims 1-10, wherein the antibody comprises at least one modification that enhances cell killing.
12. The isolated antibody of claim 11, wherein the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC).
13. The isolated antibody of claim 11 or claim 12, wherein the at least one modification is afucosylation.
14. The isolated antibody of claim 11 or claim 12, wherein the at least one modification is one or more heavy chain constant region mutations at one or more positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334, and P396.
15. The isolated antibody of claim 14, wherein the one or more heavy chain constant region mutations are one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L.
16. The isolated antibody of claim 14, wherein the one or more heavy chain constant region mutations are selected from the group consisting of: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E.
17. The isolated antibody of claim 11 or claim 12, wherein the at least one modification is galactosylation.
18. The isolated antibody of any one of claims 1-17, wherein the antibody has an affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75pM, or less than 50pM, or less than 25pM D ) Binds to human CCR 8.
19. The isolated antibody of any one of claims 1-18, wherein the antibody has an on-cell affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75, or less than 50pM, or less than 25pM D ) Binds to human CCR 8.
20. The isolated antibody of claim 18 or 19, wherein the affinity (K) D ) As determined by kinetic exclusion assay (i.e., KinExA).
21. An isolated nucleic acid encoding the antibody of any one of claims 1 to 20.
22. A vector comprising the isolated nucleic acid of claim 21.
23. A host cell comprising the nucleic acid of claim 21 or the vector of claim 22.
24. A host cell expressing the antibody of any one of claims 1 to 20.
25. The host cell of claim 23 or claim 24, which is engineered to produce an afucosylated antibody.
26. A method of producing an antibody that binds CCR8, the method comprising culturing a host cell according to any one of claims 23-25 under conditions suitable for expression of the antibody.
27. The method of claim 26, further comprising isolating the antibody.
28. A fusion protein comprising (a) CCL1, or an active fragment thereof, or MC148, or an active fragment thereof, and (b) an Fc region.
29. The fusion protein of claim 28, wherein the Fc region is an IgG1 or IgG3Fc region.
30. The fusion protein of claim 28 or claim 29, wherein the Fc region comprises the amino acid sequence of SEQ ID No. 4.
31. The fusion protein according to any one of claims 28-30, wherein the Fc region comprises at least one modification that enhances cell killing.
32. The fusion protein of claim 31, wherein the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC).
33. The fusion protein of claim 31 or claim 32, wherein the at least one modification is afucosylation.
34. The fusion protein of claim 31 or claim 32, wherein the at least one modification is one or more Fc region mutations at one or more positions selected from L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334, and P396.
35. The fusion protein of claim 34, wherein the one or more Fc region mutations is one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L.
36. The fusion protein of claim 34, wherein the one or more Fc region mutations are selected from the group consisting of: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E.
37. The fusion protein of claim 31 or claim 32, wherein the at least one modification is galactosylation.
38. The fusion protein of any one of claims 28-37, wherein the fusion protein comprises CCL1, or an active fragment thereof.
39. The fusion protein of claim 38, wherein the CCL1, or active fragment thereof, comprises the amino acid sequence of SEQ ID No. 2 or amino acids 24-96 of SEQ ID No. 2.
40. The fusion protein of claim 38 or claim 39, wherein the fusion protein comprises the amino acid sequence of SEQ ID No. 1 or amino acids 24-316 of SEQ ID No. 1.
41. The fusion protein of any one of claims 28-37, wherein the fusion protein comprises MC148 or an active fragment thereof.
42. The fusion protein of claim 41, wherein the MC148 or active fragment thereof comprises the amino acid sequence of SEQ ID NO 6.
43. The fusion protein of claim 41 or claim 42, wherein the fusion protein comprises the amino acid sequence of SEQ ID NO 5.
44. An isolated nucleic acid encoding the fusion protein of any one of claims 28 to 43.
45. A vector comprising the isolated nucleic acid of claim 44.
46. A host cell comprising the nucleic acid of claim 44 or the vector of claim 45.
47. A host cell expressing the fusion protein of any one of claims 28 to 43.
48. The host cell of claim 46 or claim 47, which is engineered to produce an afucosylated antibody.
49. A method of producing a fusion protein, the method comprising culturing the host cell of any one of claims 46-48 under conditions suitable for expression of the fusion protein.
50. The method of claim 49, further comprising isolating the fusion protein.
51. A method of treating cancer, the method comprising administering to a subject having cancer an effective amount of an antibody of any one of claims 1-20.
52. A method of treating cancer, the method comprising administering to a subject having cancer an effective amount of the fusion protein of any one of claims 28-43.
53. The method of claim 51 or 52, wherein the subject has previously been treated for a checkpoint inhibitor (CPI), and optionally wherein the cancer is resistant to the CPI.
54. The method of any one of claims 51-53, wherein the cancer comprises tumor-infiltrating Treg cells.
55. The method of any one of claims 51-53, wherein the cancer comprises cells that express CCR 8.
56. The method of claim 55, wherein the CCR 8-expressing cell is a Treg cell.
57. The method of claim 56, wherein CCR8 is expressed on the surface of the Treg cells at less than 10,000 copies per cell (as determined by Fluorescence Activated Cell Sorting (FACS) and/or flow cytometry).
58. The method of any one of claims 51-57, wherein the cancer comprises tumor cells that express CCR 8.
59. The method of any one of claims 51-58, wherein the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma.
60. The method of any one of claims 51-59, wherein the cancer is refractory cancer or resistant to checkpoint inhibitor (CPI) therapy.
61. The method of claim 60, wherein the CPI therapy comprises anti-PDL 1 antibodies, anti-CTLA 4 antibodies, or anti-TIGIT antibodies.
62. The method of claim 61, wherein the anti-PDL 1 antibody is selected from the group consisting of Pabollizumab, nivolumab, atilizumab, Avermelimumab, Duvallisuzumab, Cemifepritumab, and Cepalizumab; wherein the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or wherein the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
63. A method of treating a hematologic cancer comprising administering to a subject having a hematologic cancer an effective amount of an antibody that binds to human CCR 8.
64. The method of claim 63, wherein the subject has previously been treated for a checkpoint inhibitor (CPI), and optionally wherein the cancer is resistant to the CPI.
65. The method of claim 63 or 64, wherein the antibody inhibits the binding of CCL1 to CCR 8.
66. The method of any one of claims 63-65, wherein the hematologic cancer expresses CCR 8.
67. The method of any one of claims 63-66, wherein the hematological cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy.
68. The method of claim 67, wherein the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody.
69. The method of claim 68, wherein the anti-PDL 1 antibody is selected from the group consisting of Pabollizumab, nivolumab, atilizumab, Avermelimumab, Duvallisuzumab, Cemifepritumab, and Cepalizumab; wherein the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or wherein the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
70. A method of selecting a subject with a hematologic cancer for treatment with an antibody that binds human CCR8, comprising detecting CCR8 expression in a sample from the subject.
71. The method of claim 70, further comprising administering an effective amount of an antibody that binds human CCR 8.
72. The method of any one of claims 63-71, wherein the antibody that binds human CCR8 is the antibody of any one of claims 1-20.
73. A method of treating a hematologic cancer, comprising administering to a subject having a hematologic cancer an effective amount of the fusion protein of any one of claims 28 to 43.
74. A method of selecting a subject with a hematologic cancer for treatment with a fusion protein comprising (a) CCL1 or an active fragment thereof, or MC148 or an active fragment thereof, and (b) an Fc region, the method comprising detecting CCR8 expression in a sample from the subject.
75. The method of claim 74, further comprising administering an effective amount of a fusion protein comprising (a) CCL1 or an active fragment thereof or MC148 or an active fragment thereof and (b) an Fc region.
76. The method of claim 75, wherein the fusion protein is according to any one of claims 28 to 43.
77. The method of any one of claims 73-76, wherein the hematologic cancer is T cell adult acute lymphocytic leukemia, T cell childhood acute lymphocytic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T cell lymphoma, T cell acute lymphocytic leukemia, adult T cell leukemia/lymphoma, T cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
78. An isolated antibody that binds human CCR8, wherein the antibody comprises HCDR3, wherein the HCDR3 comprises SEQ ID NO 86 or a variant of SEQ ID NO 86 comprising 1, 2 or 3 mutations, and wherein the antibody binds human CCR8 and has ADCC activity.
79. The isolated antibody according to claim 78, wherein the mutation is selected from a substitution, insertion or deletion.
80. The isolated antibody of claim 78 or 79, wherein the antibody comprises at least 2 substitutions in HCDR 3.
81. The isolated antibody according to any one of claims 78 to 80, wherein the 1, 2 or 3 mutations are located in at least one of amino acid positions 1-4, 6, 7 or 12 of SEQ ID NO 86.
82. The isolated antibody according to any one of claims 79 to 81, wherein the substitution is a conservative substitution.
83. The isolated antibody according to claim 82, wherein the conservative substitution is at amino acid position 1, 4 or 12 of SEQ ID NO 86.
84. The isolated antibody according to any one of claims 79 to 81, wherein the substitution is a non-conservative substitution.
85. The isolated antibody according to claim 84, wherein the non-conservative substitution is at amino acid position 7 of SEQ ID NO 86.
86. The isolated antibody according to any one of claims 79 to 81, wherein when there is more than one substitution mutation, the mutation comprises a conservative substitution and a non-conservative substitution.
87. An isolated antibody that binds to human CCR8, wherein the antibody comprises HCDR3 that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to SEQ ID NO:86, and wherein the antibody binds to human CCR8 and has ADCC activity.
88. The isolated antibody according to claim 78 or 87, wherein the HCDR3 comprises an amino acid sequence selected from any one of SEQ ID NO 104-119.
89. The isolated antibody according to any one of claims 78 to 87, wherein the HCDR3 comprises an amino acid sequence selected from any one of SEQ ID NO 86 and SEQ ID NO 104-119.
90. The isolated antibody of any one of claims 78-89, wherein the antibody comprises HCDR1 comprising SEQ ID NO 84 or SEQ ID NO 123.
91. The isolated antibody of any one of claims 78-90, wherein the antibody comprises HCDR2 comprising SEQ ID NO:85 or SEQ ID NO: 124.
92. The isolated antibody according to any one of claims 78-91, wherein the antibody comprises LCDR1 comprising SEQ ID NO 87 or SEQ ID NO 120.
93. The isolated antibody according to any one of claims 78-92, wherein the antibody comprises LCDR2 comprising SEQ ID NO:88 or SEQ ID NO: 121.
94. The isolated antibody according to any one of claims 78-93, wherein the antibody comprises LCDR3 comprising SEQ ID No. 89 or SEQ ID No. 122.
95. The isolated antibody of any one of claims 78-94, wherein the antibody comprises a heavy chain variable region (VH) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs 80, 92, and 96, and a light chain variable region (VL) comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs 81, 93, and 97.
96. The isolated antibody according to any one of claims 78-95, wherein the antibody comprises a heavy chain comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs 82, 90, 94, and 98 and a light chain comprising an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identical to the amino acid sequence of any one of SEQ ID NOs 83, 91, 95, and 99.
97. The isolated antibody according to any one of claims 78 to 96, wherein ADCC activity comprises an EC50 value of less than 200, 175, 150, 125, 100, 75, 50, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1ng/ml as measured by a bioassay based on the ADCC reporter mechanism of action (MOA).
98. The isolated antibody according to any one of claims 78 to 97, wherein the ADCC activity is more potent than the 7-B16 antibody.
99. The isolated antibody of any one of claims 78 to 98, wherein the antibody is to K of human CCR8 D Equal to or lower than the 7-B16 antibody.
100. The isolated antibody of any one of claims 78-99, wherein the antibody is to K on a cell of human CCR8 D Equal to or lower than the 7-B16 antibody.
101. The isolated antibody of claim 99 or 100, wherein the K is D As determined by a kinetic exclusion assay (i.e., KinExA).
102. The isolated antibody of any one of claims 78-101, wherein the antibody comprises at least one modification that enhances cell killing.
103. The isolated antibody of claim 102, wherein the enhanced cell killing is enhanced Antibody Dependent Cellular Cytotoxicity (ADCC) and/or Complement Dependent Cytotoxicity (CDC).
104. The isolated antibody of claim 102 or claim 103, wherein the at least one modification is afucosylation.
105. The isolated antibody of claim 102 or claim 103, wherein the at least one modification is one or more heavy chain constant region mutations at one or more positions selected from the group consisting of L234, L235, G236, S239, F243, H268, D270, R292, S298, Y300, V305, K326, a330, I332, E333, K334, and P396.
106. The isolated antibody of claim 105, wherein the one or more heavy chain constant region mutations is one or more mutations selected from the group consisting of S239D, S239M, F243L, H268D, D270E, R292P, S298A, Y300L, V305I, K326D, a330L, a330M, I332E, E333A, K334A, K334E, and P396L.
107. The isolated antibody of claim 105, wherein the one or more heavy chain constant region mutations are selected from the group consisting of: F243L/R292P/Y300L/V305I/P396L, S239D/I332E, S239D/I332E/A330L, S298A/E333A/K334A, L234Y/L235Q/G236W/S239M/H268D/D270E/S298A and D270E/K326D/A330M/K334E.
108. The isolated antibody of claim 96 or claim 97, wherein the at least one modification is galactosylation.
109. The isolated antibody of any one of claims 78-108, wherein the antibody has an affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75pM, or less than 50pM, or less than 25pM D ) Binds to human CCR 8.
110. The isolated antibody of any one of claims 78-109, wherein the antibody has an on-cell affinity (K) of less than 10nM, or less than 5nM, or less than 1nM, or less than 500pM, or less than 250pM, or less than 100pM, or less than 75, or less than 50pM, or less than 25pM D ) Binds to human CCR 8.
111. The isolated antibody of claim 109 or 110, wherein the affinity (K) D ) As determined by kinetic exclusion assay (i.e., KinExA).
112. The isolated antibody according to any one of claims 78 to 111, wherein the antibody is a monoclonal antibody.
113. The isolated antibody according to any one of claims 78 to 112, wherein the antibody is a human or humanized antibody.
114. The isolated antibody according to any one of claims 78 to 113, wherein the antibody is a full length antibody.
115. The isolated antibody of any one of claims 78-114, wherein the antibody is an IgG1 or IgG3 antibody.
116. A nucleic acid encoding the antibody of any one of claims 78-115.
117. A method of treating cancer, the method comprising administering to a subject having cancer the antibody of any one of claims 78-115.
118. The method of claim 117, wherein the cancer is a hematological cancer or a solid cancer.
119. The method of claim 117 or 118, wherein the cancer expresses CCR 8.
120. The method of any one of claims 117-119, wherein the cancer is T-cell adult acute lymphocytic leukemia, T-cell pediatric acute lymphocytic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T-cell lymphoma, T-cell acute lymphocytic leukemia, adult T-cell leukemia/lymphoma, T-cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
121. The method of any one of claims 117-119, wherein the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma.
122. The method of any one of claims 117-121, further comprising administering to the subject one or more additional therapeutic agents.
123. The method of claim 122, wherein the one or more additional therapeutic agents are selected from an anti-Trop-2 antibody (e.g., saritumumab govikang, SKB-264, JS-108(DAC-002), daptomab delutene, BAT-8003), an anti-CD 47 antibody or a CD47 blocker (e.g., mololizumab, DSP-107, AO-176, ALX-148, IBI-188, letuzumab, TTI-621, TTI-622), an anti-sirpa antibody (e.g., GS-0189), an FLT3L-Fc fusion protein (e.g., GS-3583), an anti-PD-1 antibody (palivizumab, nivolumab, palivizumab), a small molecule PD-L1 inhibitor (e.g., GS-4224), an anti-PD-L1 antibody (e.g., azib), an antibody, or a pharmaceutically acceptable salt thereof, Small molecule MCL1 inhibitors (e.g., GS-9716), small molecule HPK1 inhibitors (e.g., GS-6451), HPK1 degraders (PROTAC; e.g., ARV-766), small molecule DGKa inhibitors, small molecule CD73 inhibitors (e.g., AB680), anti-CD 73 antibodies (e.g., orlistat), dual a 2a /A 2b Adenosine receptor antagonists (e.g., itraconazole (AB928)), anti-TIGIT antibodies (e.g., tegraleigh mab, vibralizumab, donralizumab, AB308), anti-TREM 1 antibodies (e.g., PY159), anti-TREM 2 antibodies (e.g., PY314), TGF β -capturing agents (e.g., bintrafusip alpha, age-1423), anti-TGF β 1 antibodies (e.g., SRK-181), and CAR-T cell therapy (e.g., aliskiren, brij oritem, tesseram).
124. The method of claim 122, wherein the one or more additional therapeutic agents is selected from the group consisting of saritumumab gavatinib-hziy, mololizumab, GS-0189, GS-3583, cypripezumab, GS-4224, GS-9716, GS-6451, AB680, itraconazole (AB928), donnalizumab, AB308, PY159, PY314, SRK-181, aliskiren, and briguerite.
125. The method of any one of claims 122-124, wherein administration of the one or more additional therapeutic agents is concurrent with administration of the antibody.
126. The method of any one of claims 122-124, wherein administration of the one or more additional therapeutic agents is prior to or after administration of the antibody.
127. The method of any one of claims 117-126, wherein the cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy.
128. The method of claim 127, wherein the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody.
129. The method of claim 128, wherein the anti-PDL 1 antibody is selected from the group consisting of pabulizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprilinumab, and serpalizumab; wherein the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or wherein the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
130. Use of the isolated antibody of any one of claims 1 to 20 or 78 to 115 or the fusion protein of any one of claims 28 to 43 for treating cancer.
131. The use of claim 130, wherein the cancer is a hematological cancer or a solid tumor.
132. The use of claim 130 or 131, wherein the cancer expresses CCR 8.
133. The use of any one of claims 130-132, wherein the cancer is T-cell adult acute lymphocytic leukemia, T-cell childhood acute lymphocytic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T-cell lymphoma, T-cell acute lymphocytic leukemia, adult T-cell leukemia/lymphoma, T-cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
134. The use of any one of claims 130-132, wherein the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma.
135. The use of any one of claims 130-134, further comprising administering to the subject one or more additional therapeutic agents.
136. The use of claim 135, wherein the administration of the one or more additional therapeutic agents is simultaneous with the administration of the antibody.
137. The use of claim 135, wherein the administration of the one or more additional therapeutic agents is prior to or subsequent to the administration of the antibody.
138. The use of any one of claims 130-137, wherein the cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy.
139. The use of claim 138, wherein the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody.
140. The use of claim 139, wherein the anti-PDL 1 antibody is selected from the group consisting of pabulizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprilinumab, and serpalizumab; wherein the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or wherein the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
141. The isolated antibody of any one of claims 1 to 20 or 78 to 115 or the fusion protein of any one of claims 28 to 43 for use in the treatment of cancer.
142. The isolated antibody or fusion protein of claim 141, wherein the cancer is a hematological cancer or a solid tumor.
143. The isolated antibody or fusion protein of claim 141 or 142, wherein the cancer expresses CCR 8.
144. The isolated antibody or fusion protein of any one of claims 141-143, wherein the cancer is T-cell adult acute lymphocytic leukemia, T-cell childhood acute lymphocytic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T-cell lymphoma, T-cell acute lymphocytic leukemia, adult T-cell leukemia/lymphoma, T-cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
145. The isolated antibody or fusion protein of any one of claims 141-143, wherein the cancer is selected from the group consisting of breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma.
146. The isolated antibody or fusion protein of any one of claims 141-145, further comprising administering to the subject one or more additional therapeutic agents.
147. The isolated antibody or fusion protein of claim 146, wherein administration of the one or more additional therapeutic agents is concurrent with administration of the antibody.
148. The isolated antibody or fusion protein of claim 146, wherein administration of the one or more additional therapeutic agents precedes or follows administration of the antibody.
149. The isolated antibody or fusion protein of any one of claims 141-148, wherein the cancer is a refractory cancer or is resistant to checkpoint inhibitor (CPI) therapy.
150. A method of treating a refractory or checkpoint inhibitor (CPI) resistant cancer, the method comprising administering to a subject having a cancer the isolated antibody of any one of claims 1-20 or 78-115 or the fusion protein of any one of claims 28-43, wherein the subject has been previously treated with chemotherapy or CPI therapy and is not responsive to the chemotherapy or CPI therapy.
151. The method of claim 150, wherein the CPI therapy comprises an anti-PDL 1 antibody, an anti-CTLA 4 antibody, or an anti-TIGIT antibody.
152. The method of claim 151, wherein the anti-PDL 1 antibody is selected from the group consisting of pabulizumab, nivolumab, atilizumab, avilumab, bevacizumab, cimiciprilinumab, and serpalizumab; wherein the anti-CLTA 4 antibody is ipilimumab or tremelimumab; or wherein the anti-TIGIT antibody is selected from the group consisting of ibritumomab tiuxetan, bevacizumab, donralizumab, AB308, BMS-986207, and bevacizumab.
153. The method of any one of claims 150 to 152, wherein the cancer is a hematologic cancer or a solid cancer.
154. The method of any one of claims 150-153, wherein the cancer expresses CCR 8.
155. The method of any one of claims 150-154, wherein the cancer is T-cell adult acute lymphocytic leukemia, T-cell pediatric acute lymphocytic leukemia, lymphoblastic lymphoma, acute lymphocytic leukemia, cutaneous T-cell lymphoma, T-cell acute lymphocytic leukemia, adult T-cell leukemia/lymphoma, T-cell lymphoblastic leukemia/lymphoma, or anaplastic large cell lymphoma.
156. The method of any one of claims 150 to 154, wherein the cancer is selected from breast cancer, colorectal cancer, head and neck cancer, lung cancer, ovarian cancer, gastric adenocarcinoma, and thymoma.
157. The method of any one of claims 150 to 156, further comprising administering to the subject one or more additional therapeutic agents.
158. The method of claim 157, wherein the one or more additional therapeutic agents are selected from an anti-Trop-2 antibody (e.g., saritumumab govikang, SKB-264, JS-108(DAC-002), daptomab delutene, BAT-8003), an anti-CD 47 antibody or a CD47 blocker (e.g., mololizumab, DSP-107, AO-176, ALX-148, IBI-188, letuzumab, TTI-621, TTI-622), an anti-sirpa antibody (e.g., GS-0189), an FLT3L-Fc fusion protein (e.g., GS-3583), an anti-PD-1 antibody (palivizumab, nivolumab, palivizumab), a small molecule PD-L1 inhibitor (e.g., GS-4224), an anti-PD-L1 antibody (e.g., azib), an antibody, or a pharmaceutically acceptable salt thereof, Small molecule MCL1 inhibitors (e.g., GS-9716), small molecule HPK1 inhibitors (e.g., GS-6451), HPK1 degraders (PROTAC; e.g., ARV-766), small molecule DGKa inhibitors, small molecule CD73 inhibitors (e.g., AB680), anti-CD 73 antibodies (e.g., olarumab), Dual A 2a /A 2b Adenosine receptor antagonists (e.g., itraconazole (AB928)), anti-TIGIT antibodies (e.g., tegraleigh mab, vibralizumab, donralizumab, AB308), anti-TREM 1 antibodies (e.g., PY159), anti-TREM 2 antibodies (e.g., PY314), TGF-beta-capture agents (e.g., bintrafusip alpha, AGEN-1423), anti-TGF-beta 1 antibodies (e.g., SRK-181), and CAR-T cell therapy (e.g., aliskiren, brij-orenck, gamma-,Tesalasin).
159. The method of claim 157, wherein the one or more additional therapeutic agents is selected from the group consisting of safiruzumab gavitegazen-hziy, mololizumab, GS-0189, GS-3583, cyprulizumab, GS-4224, GS-9716, GS-6451, AB680, itraconazole (AB928), donnazumab, AB308, PY159, PY314, SRK-181, aliskiren, and briguel.
160. The method of any one of claims 157-159, wherein administration of the one or more additional therapeutic agents is concurrent with administration of the antibody.
161. The method of any one of claims 157-159, wherein administration of the one or more additional therapeutic agents is prior to or after administration of the antibody.
HK62023069692.9A 2020-02-14 2021-02-09 Antibodies and fusion proteins that bind to ccr8 and uses thereof HK40081400A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US62/976,869 2020-02-14
US63/130,157 2020-12-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
HK42024098476.5A Addition HK40111371A (en) 2020-02-14 2023-03-08 Antibodies and fusion proteins that bind to ccr8 and uses thereof

Publications (1)

Publication Number Publication Date
HK40081400A true HK40081400A (en) 2023-05-19

Family

ID=

Similar Documents

Publication Publication Date Title
JP7759428B2 (en) Antibodies and fusion proteins that bind to CCR8 and uses thereof
CN114555123A (en) Combination therapy for the treatment of myelodysplastic syndrome and acute myeloid leukemia
CN114599392A (en) Blood cancer therapy based on anti-CD47 and anti-CD20
US20220135682A1 (en) Anti-ICOS Antibodies for the Treatment of Cancer
US20220389394A1 (en) METHODS OF USING FLT3L-Fc FUSION PROTEINS
TWI895708B (en) Combination therapy for treating trop-2 expressing cancers
HK40081400A (en) Antibodies and fusion proteins that bind to ccr8 and uses thereof
WO2021091960A1 (en) Methods of treating cancer with anti-pd-1 antibodies
HK40075583A (en) Anti-cd47 and anti-cd20 based treatment of blood cancer
HK40074854A (en) Combination therapies for treating myelodysplastic syndromes and acute myeloid leukemia
HK40079159A (en) Methods of treating cancer with anti-pd-1 antibodies
HK40042823A (en) Anti-cd73 antibodies and methods of use thereof
HK1254148A1 (en) Immune modulation and treatment of solid tumors with antibodies that specifically bind cd38