[go: up one dir, main page]

WO2025049961A1 - Anti-cd22 chimeric antigen receptor (car) therapies - Google Patents

Anti-cd22 chimeric antigen receptor (car) therapies Download PDF

Info

Publication number
WO2025049961A1
WO2025049961A1 PCT/US2024/044747 US2024044747W WO2025049961A1 WO 2025049961 A1 WO2025049961 A1 WO 2025049961A1 US 2024044747 W US2024044747 W US 2024044747W WO 2025049961 A1 WO2025049961 A1 WO 2025049961A1
Authority
WO
WIPO (PCT)
Prior art keywords
car
cells
dose
therapy
day
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/US2024/044747
Other languages
French (fr)
Inventor
Gregg FINE
Bethany ROGERS
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cargo Therapeutics Inc
Original Assignee
Cargo Therapeutics 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 Cargo Therapeutics Inc filed Critical Cargo Therapeutics Inc
Publication of WO2025049961A1 publication Critical patent/WO2025049961A1/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/10Cellular immunotherapy characterised by the cell type used
    • A61K40/11T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/30Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
    • A61K40/31Chimeric antigen receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K40/00Cellular immunotherapy
    • A61K40/40Cellular immunotherapy characterised by antigens that are targeted or presented by cells of the immune system
    • A61K40/41Vertebrate antigens
    • A61K40/42Cancer antigens
    • A61K40/4202Receptors, cell surface antigens or cell surface determinants
    • A61K40/421Immunoglobulin superfamily
    • A61K40/4212CD22, BL-CAM, siglec-2 or sialic acid binding Ig-related lectin 2
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K40/00
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K40/00
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K40/00 characterised by the cancer treated
    • A61K2239/48Blood cells, e.g. leukemia or lymphoma

Definitions

  • the present disclosure relates generally to the use of autologous T-cells engineered to express a CAR with an anti-CD22 binding domain, to treat a disease, e.g., a hematological cancer.
  • a disease e.g., a hematological cancer.
  • the hematological cancer is a relapsed/refractory hematological cancer that recurs following treatment and a period of remission and/or that does not respond to treatment.
  • the relapsed/refractory hematological cancer recurs after treatment with standard of care chemotherapy or a CD19/CD3 bi-specific T-cell engager (BiTE).
  • a relapsed/refractory (R/R) hematological cancer in a human subject in need thereof comprising: administering to the human subject a therapeutically effective dose of autologous CAR T-cells to treat the R/R hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an anti-CD22 binding domain and (B) a portion of a CD8a extracellular domain (z.e., a CD8a hinge domain), a CD8a transmembrane domain; a 4-1BB intracellular costimulatory signaling domain; and a CD3-zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy, and/or wherein the at least one prior line of therapy comprises a bispecific T-cell engager (BiTE®) antibody therapy.
  • BiTE® bispecific T-cell engager
  • the dose of autologous CAR T-cells is less than about 0.8* 10 6 engineered CAR T-cells/kg.
  • the dose of autologous CAR T-cells is at least about 1.2* 10 6 CAR T-cells/kg.
  • the hematological cancer is a relapsed/refractory hematological cancer.
  • the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy.
  • the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager (BiTE®) antibody therapy.
  • BiTE® bispecific T-cell engager
  • the human subject has received 2 or more prior lines of therapy.
  • the human subject has received 2 or more prior lines of therapy, wherein at least one of the 2 or more prior lines of therapy comprises a bispecific T- cell engager antibody therapy.
  • the human subject has received no more than 5 prior lines of therapy.
  • the human subject has not received a prior CAR T-cell therapy.
  • the human subject has received a prior CAR T-cell therapy.
  • the human subject has received a prior CAR T-cell therapy and at least one additional therapy.
  • the prior CAR T-cell therapy is tisagenlecleucel (marketed as Kymriah®), brexucabtagene autoleucel (marketed as Tecartus®), axicabtagene cilocleucel (marketed as Yescarta®), lisocabtagene maraleucel (marketed as Breyanzi®) or any combination thereof.
  • the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises an anti-CD19 antibody, an anti-CD20 antibody, and anti-CD22 antibody or an anti-CD79b antibody.
  • At least one prior line of therapy comprises an anti-CD19 antibody-drug conjugate, an anti-CD20 antibody-drug conjugate, an anti-CD22 antibody-drug conjugate, or an anti-CD79b antibody-drug conjugate.
  • the anti-CD19 antibody comprises inebilizumab (marketed as Uplizna®), loncastuximab tesirine (marketed as Zynlonta®), or tafasitamab (marketed as Monjuvi®).
  • the anti-CD20 antibody comprises rituximab (marketed as Rituxan®), ibritumomab tiuxetan (marketed as Zevalin®), tositumomab (marketed as Bexxar®), ocrelizumab (Ocrevus®), ofatumumab (marketed as Kesimpta®), mosunetuzumab (marketed as LunsumioTM), Obinutuzumab (marketed as Gazyva®), or ublituximab (marketed as Briumvi®).
  • Rituxan® ibritumomab tiuxetan
  • Zevalin® tositumomab
  • Bexxar® ocrelizumab
  • Ocrevus® ofatumumab
  • Kesimpta® mosunetuzumab
  • Obinutuzumab marketed as Gazyva®
  • ublituximab marketed as Briumvi
  • the anti-CD22 antibody comprises moxetumomab pasudotox (marketed as Lumoxiti®), inotuzumab ozogamicin (maketed as Besponsa®), or epratuzumab.
  • the anti-CD79b antibody comprises polatuzumab vedotin (marketed as Polivy®).
  • the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
  • the bispecific T-cell engager antibody therapy comprises a T cell engager comprising an anti-CD3 domain.
  • the bispecific T-cell engaging antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, elranatamab, teclistamab, tarlatamab, talquetamab, solitomab, tebentafusp, duvortuxizumab, AFM11, REGN5458, CC- 93269, PF-06863135, TNB-383B, pdronextamab, XmAbl3676, IGM-2323, AMG 330, AMG 673, AMG 420, AMG 701, cevostamab, talquetamab, flotetuzumab, or a combination thereof.
  • the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain.
  • the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain and an anti-CD3
  • the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain and an anti-CD3 domain.
  • the bispecific T-cell engager antibody therapy comprises an anti-BCMA domaim. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-BCMA domain and an anti-CD3 domain.
  • the anti-CD22 binding domain is an scFv.
  • the anti-CD22 binding domain is an antibody or an antigen binding domain of thereof.
  • the anti-CD22 binding domain comprises the sequence:
  • the CAR comprises a linker between the anti-CD22 binding domain and a portion of a CD8a extracellular domain (z.e., a CD8a hinge domain).
  • the linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain comprises the sequence AAA.
  • the portion of a CD8a extracellular domain (z.e., a CD8a hinge domain) comprises the sequence:
  • the portion of a CD8a transmembrane domain comprises the sequence: IYIWAPLAGTCGVLLLSLVIT.
  • the CAR comprises a portion of a CD8a intracellular domain.
  • the portion of a CD8a intracellular domain comprises the sequence LYC.
  • the 4- IBB costimulatory domain comprises the sequence: KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL.
  • the CD3zeta intracellular signaling domain comprises the sequence:
  • the CAR comprises a signal peptide sequence.
  • the signal peptide sequence is derived from granulocyte macrophage colony stimulating factor receptor alpha (GMCSFRa) and comprises the sequence: MLLLVTSLLLCELPHPAFLLIP.
  • the CAR comprises the sequence:
  • the CAR comprises the sequence:
  • the hematological cancer expresses CD22.
  • the hematological cancer is Large B-Cell Lymphoma (LBCL).
  • LBCL Large B-Cell Lymphoma
  • the LBCL is diffuse LBCL (DLBCL).
  • the LBCL has been histologically confirmed.
  • the LBCL is (DLBCL) not otherwise specified (NOS).
  • the DLBCL-NOS is germinal center B-cell (GCB) type or active B-cell (ABC) type.
  • the LBCL is high-grade B-cell lymphoma with or without MYC and BCL2 and/or BCL6 rearrangements.
  • the LBCL is DLBCL associated with chronic inflammation.
  • the LBCL is primary cutaneous DLBCL, leg type.
  • the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS.
  • EBV Epstein-Barr virus
  • the LBCL is DLBCL arising from follicular lymphoma (FL).
  • the LBCL is DLBCL arising from marginal zone lymphoma (MZL).
  • MZL marginal zone lymphoma
  • the LBCL is primary mediastinal (thymic) large B-cell lymphoma.
  • the LBCL is Grade 3B FL.
  • the human subject is 18 years of age or older.
  • the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of progressive disease (PD) or stable disease (SD) after the last therapy.
  • PD progressive disease
  • SD stable disease
  • the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of disease progression after partial response (PR) or complete response (CR) after the last therapy.
  • PR partial response
  • CR complete response
  • the human subject has relapsed/refractory disease after a last therapy based on presence of viable lymphoma in a biopsy obtained after therapy.
  • the human subject has relapsed disease following a complete response (CR) or progressive disease following a PR to a bispecific T-cell engager antibody therapy.
  • the human subject has progressive disease (PD) or stable disease (SD) as best response to a bispecific T-cell engager antibody therapy.
  • PD progressive disease
  • SD stable disease
  • the dose of engineered CAR T-cells is from about O.l x lO 6 to about 4* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about O.l x lO 6 to about 0.75x l0 6 engineered CAR T-cells/kg. [0070] In some embodiments, the dose of engineered CAR T-cells is from about 0.25* 10 6 to about 0.75* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 1.2* 10 6 to about 4* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 0.8* 10 6 to about 1.2* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 2.5* 10 6 to about 3.5* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is about 0.5* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is about l > ⁇ 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is about 3-2* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is about 3* 10 6 engineered CAR T-cells/kg.
  • the method comprises administering to the human subject a dose of cyclophosphamide prior to administering the engineered CAR T-cells.
  • the dose of cyclophosphamide is about 400 mg/m 2 to about 600 mg/m 2 .
  • the dose of cyclophosphamide is about 500 mg/m 2 .
  • the dose of cyclophosphamide is administered daily for at least 2 days.
  • the dose of cyclophosphamide is administered daily for 3 days.
  • the dose of cyclophosphamide is 500 mg/m 2 administered once daily for 3 days.
  • the method comprises administering to the human subject a dose of fludarabine prior to administering the engineered CAR T-cells.
  • the dose of fludarabine is about 20 mg/m 2 to about 400 mg/m 2 .
  • the dose of fludarabine is about 25 mg/m 2 to about 35 mg/m 2 .
  • the dose of fludarabine is about 30 mg/m 2 .
  • the dose of fludarabine is administered daily for at least 2 days. [0089] In some embodiments, the dose of fludarabine is administered daily for three days. [0090] In some embodiments, the dose of fludarabine is 30 mg/m 2 administered once daily for 3 days.
  • the dose of cyclophosphamide is administered daily for three days and the dose of fludarabine is administered daily for three days.
  • At least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine.
  • a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine.
  • a second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine.
  • a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine.
  • each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered.
  • day 0 refers to the day of CAR T infusion
  • day - 1 refers to the day 1 day before CAR T infusion
  • day -2 refers to the day 2 days before CAR T infusion
  • day -3 refers to the day 3 days before CAR T infusion
  • day -4 refers to the day 4 days before CAR T infusion
  • day -5 refers to the day 5 days before CAR T infusion.
  • a dose of cyclophosphamide is administered on day -5 and day -4 and day -3 prior to CAR T infusion, and the engineered CAR T-cells are administered on day 0.
  • a dose of fludarabine is administered on day -5 and day -4 and day -3 prior to CAR T infusion, and the engineered CAR T-cells are administered on day 0.
  • a dose of cyclophosphamide is administered at 500 mg/m 2 and a dose of fludarabine is administered at 30 mg/m 2 , each administered once daily starting on day -5 for three days (z.e., day -5, day -4, day -3), followed by two days with no chemotherapy, and the engineered CAR T-cells are administered on day 0.
  • Also provided herein is a method of treating a relapsed/refractory Large B-Cell Lymphoma (LBCL) in a human subject in need thereof, the method comprising: administering to the human subject a dose of about 400 mg/m 2 to about 600 mg/m 2 cyclophosphamide on day -5, day -4 and day -3, and a dose of about 25 mg/m 2 to about 35 mg/m 2 fludarabine on day -5, day -4 and day -3; administering to the human subject a dose of from about 0.1 x 10 6 to about 4* 10 6 engineered chimeric antigen receptor (CAR) T-cells/kg on day 0; wherein the CAR comprises the sequence: MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAV
  • Also provided herein is a method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of autologous CD22-specific chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain, a CD8a transmembrane domain; a 4- IBB costimulatory domain; and a CD3zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy, and/or wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
  • CAR autologous CD22-specific chimeric antigen receptor
  • the dose of autologous CD22-specific CAR T-cells is less than about 0.8* 10 6 autologous CD22-specific CAR T-cells/kg.
  • the dose of engineered CAR T-cells is at least about 1.2* 10 6 CAR T-cells/kg.
  • Also provided herein is a method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of engineered chimeric antigen receptor (CAR) T- cells to treat the hematological cancer in the human subject; wherein the CAR comprises (i) an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain, (ii) a CD8a transmembrane domain; (iii) a 4-1BB costimulatory domain; and (iv) a CD3zeta intracellular signaling domain; wherein the dose of autologous CD22-specific CAR T-cells is less than about 0.8* 10 6 autologous CD22-specific CAR T-cells/kg, or at least about 1.2* 10 6 autologous CD22-specific CAR T-cells/kg.
  • CAR engineered chimeric antigen receptor
  • the hematological cancer is a relapsed/refractory hematological cancer.
  • the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy.
  • the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
  • Also provided herein is a method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a dose of cyclophosphamide at about 500 mg/m 2 /day to about 600 mg/m 2 /day, wherein the dose of cyclophosphamide is administered daily for three days, and a dose of fludarabine at about 30 mg/m 2 /day, wherein the dose of fludarabine is administered daily for three days; and administering to the human subject a therapeutically effective dose of engineered chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an antihuman CD22 binding domain and (B) a portion of a CD8a extracellular domain, a CD8a transmembrane domain; a 4- IBB costimulatory domain; and a CD3zeta intracellular signaling domain
  • the human subject has received 2 or more prior lines of therapy.
  • the human subject has received 2 or more prior lines of therapy, wherein at least one of the 2 or more prior lines of therapy a bispecific T-cell engager antibody therapy.
  • the human subject has received no more than 5 prior lines of therapy. [0113] In some embodiments, the human subject has not received a prior CAR T-cell therapy.
  • the human subject has received a prior CAR T-cell therapy.
  • the human subject has received a prior CAR T-cell therapy and at least one additional therapy.
  • the prior CAR T-cell therapy is tisagenlecleucel, brexucabtagene autoleucel, axicabtagene cilocleucel, aisocabtagene maraleucel or any combination thereof.
  • the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises an anti-CD19 antibody, an anti-CD20 antibody, and anti-CD22 antibody or an anti-CD79b antibody.
  • the anti-CD19 antibody, anti-CD20 antibody, anti-CD22 antibody or anti-CD79b antibody comprises an antibody-drug conjugate.
  • the anti-CD19 antibody comprises inebilizumab, loncastuximab tesirine or tafasitamab.
  • the anti-CD20 antibody comprises rituximab, ibritumomab tiuxetan, tositumomab, ocrelizumab, ofatumumab, mosunetuzumab, obinutuzumab, or ublituximab.
  • the anti-CD22 antibody comprises inotuzumab ozogamicin, or epratuzumab.
  • the anti-CD79b antibody comprises polatuzumab vedotin.
  • the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
  • the bispecific T-cell engager antibody therapy comprises a T cell engager comprising an anti-CD3 domain.
  • the bispecific T-cell engager antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, solitomab, duvortuxizumab, AFM11, REGN5458, CC-93269, TNB-383B, pdronextamab, XmAbl3676, IGM-2323, or cevostamab.
  • the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain.
  • the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain.
  • the anti-CD22 binding domain is an scFv. [0125] In some embodiments, the anti-CD22 binding domain is an antibody or an antigen binding domain of thereof.
  • the anti-CD22 binding domain comprises the sequence: QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIK (SEQ ID NO: 1).
  • the CAR comprises a linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain.
  • the linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain comprises the sequence AAA.
  • the portion of a CD8a extracellular domain comprises the sequence TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO: 2).
  • the portion of a CD8a transmembrane domain comprises the sequence IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO:4).
  • the CAR comprises a portion of a CD8a intracellular domain.
  • the portion of a CD8a intracellular domain comprises the sequence LYC.
  • the 4- IBB costimulatory domain comprises the sequence KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO:6).
  • the CD3zeta intracellular signaling domain comprises the sequence:
  • the CAR comprises a signal peptide sequence.
  • the signal peptide sequence comprises the sequence: MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 8).
  • the CAR comprises the sequence:
  • the CAR comprises the sequence:
  • the hematological cancer expresses CD22.
  • the hematological cancer is Large B-Cell Lymphoma (LBCL).
  • LBCL Large B-Cell Lymphoma
  • the LBCL is diffuse LBCL (DLBCL).
  • the LBCL has been histologically confirmed.
  • the LBCL is (DLBCL) not otherwise specified (NOS).
  • the DLBCL-NOS is germinal center B-cell (GCB) type or active B-cell (ABC) type.
  • the LBCL is high-grade B-cell lymphoma with or without MYC and BCL2 and/or BCL6 rearrangements.
  • the LBCL is DLBCL associated with chronic inflammation.
  • the LBCL is primary cutaneous DLBCL, leg type.
  • the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS.
  • EBV Epstein-Barr virus
  • the LBCL is DLBCL arising from follicular lymphoma (FL). [0150] In some embodiments, the LBCL is DLBCL arising from marginal zone lymphoma (MZL). [0151] In some embodiments, the LBCL is primary mediastinal (thymic) large B-cell lymphoma.
  • the LBCL is Grade 3B FL.
  • the human subject is at least 18 years of age.
  • the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of progressive disease (PD) or stable disease (SD) after the last therapy.
  • PD progressive disease
  • SD stable disease
  • the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of disease progression after partial response (PR) or complete response (CR) after the last therapy.
  • PR partial response
  • CR complete response
  • the human subject has relapsed/refractory disease after a last therapy based on presence of viable lymphoma in a biopsy obtained after therapy.
  • the human subject has relapsed disease following a CR or progressive disease following a PR to a bispecific T-cell engager antibody therapy.
  • the human subject has progressive disease (PD) or stable disease (SD) as best response to a bispecific T-cell engager antibody therapy.
  • PD progressive disease
  • SD stable disease
  • the dose of engineered CAR T-cells is from about O.l x lO 6 to about 4* 10 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about O.l x lO 6 to about 0.75x l0 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 0.25x l0 6 to about 0.75x l0 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 1.2x l0 6 to about 4x l0 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 0.8x l0 6 to about 1.2x l0 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is from about 2.5x l0 6 to about 3.5x l0 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is about 0.5x l0 6 engineered CAR T-cells/kg.
  • the dose of engineered CAR T-cells is about I x lO 6 engineered CAR-T cells/kg. [0167] In some embodiments, the dose of engineered CAR T-cells is about 2* 10 6 engineered CAR-T cells/kg.
  • the dose of engineered CAR T-cells is about 3* 10 6 engineered CAR T-cells/kg.
  • the method comprises administering to the human subject a dose of cyclophosphamide prior to administering the engineered CAR T-cells.
  • the dose of cyclophosphamide is about 400 mg/m 2 to about 600 mg/m 2 .
  • the dose of cyclophosphamide is about 500 mg/m 2 .
  • the dose of cyclophosphamide is administered daily for at least 2 days.
  • the dose of cyclophosphamide is administered daily for 3 days.
  • the method comprises administering to the human subject a dose of fludarabine prior to administering the engineered CAR T-cells.
  • the dose of fludarabine is about 20 mg/m 2 to about 400 mg/m 2 .
  • the dose of fludarabine is about 25 mg/m 2 to about 35 mg/m 2 .
  • the dose of fludarabine is about 30 mg/m 2 .
  • the dose of fludarabine is administered daily for at least 2 days.
  • the dose of fludarabine is administered daily for three days.
  • the dose of cyclophosphamide is administered daily for three days and the dose of fludarabine is administered daily for three days.
  • At least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine.
  • a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine.
  • a second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine.
  • a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine.
  • each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered.
  • a dose of cyclophosphamide is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells are administered on day 0.
  • a dose of fludarabine is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells are administered on day 0.
  • a dose of cyclophosphamide is administered on day -5, day -4, and day -3
  • a dose of fludarabine is administered on day -5, day -4, and day -3
  • the engineered CAR T-cells are administered on day 0.
  • Also provided herein is a method of treating a relapsed/refractory Large B-Cell Lymphoma (LBCL) in a human subject in need thereof, the method comprising administering to the human subject a dose of about 400 mg/m 2 to about 600 mg/m 2 cyclophosphamide on day -5, day -4 and day -3, and a dose of about 25 mg/m 2 to about 35 mg/m 2 fludarabine on day -5, day -4 and day -3; administering to the human subject a dose of from about 0.1 * 10 6 to about 4* 10 6 engineered chimeric antigen receptor (CAR) T-cells/kg on day 0; wherein the CAR comprises the sequence:
  • FIG. 1 shows the study design for a phase 2, multi-center Phase 2, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B- cell antigen CD22 in participants with R/R LBCL.
  • FIG. 2 shows progression free survival and overall survival of LBCL patients treated with CD22 CAR T-cell therapy.
  • FIG. 3 shows associated toxicities with our CD22 CAR T-cell therapy.
  • FIG. 4 shows an updated study design for a phase 2, multi-center Phase 2, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B-cell antigen CD22 in participants with R/R LBCL.
  • the disclosure provides a method of treating a hematological cancer, comprising administering autologous T-cells that express a CAR molecule that binds CD22.
  • the hematological cancer is lymphoma.
  • the lymphoma is LBCL.
  • the method of treating the hematological cancer comprises treating a subject who has received previous therapy.
  • the subject who has received previous therapy has relapsed or has refractory disease.
  • the term “about” in some cases refers to an amount that is approximately the stated amount, an amount that is near the stated amount by 10%, 5%, or 1%, including increments therein, or an amount that is greater or less the stated percentage by 10%, 5%, or 1%, including increments therein.
  • an autologous T cell described herein comprises a CAR molecule that binds CD22 (a “CD22 CAR”).
  • the autologous T cell is an engineered T cell, e.g., autologous CAR T cell.
  • the autologous T cell is selected from the group consisting of CD4+ T cell, CD8+ T cell, regulatory T cell, and memory T cell.
  • the CD22 CAR comprises an extracellular domain, a transmembrane (TM) domain, and an intracellular domain.
  • the extracellular domain comprises an antigen-binding domain and a hinge domain.
  • the antigen-binding domain comprises a fully human anti-CD22 antibody.
  • the hinge domain comprises a hinge domain derived from CD8alpha or CD28.
  • TM domain comprises a TM domain derived from CD8alpha or CD28.
  • the intracellular domain comprises a co-stimulatory domain, and an immunoreceptor tyrosine-based activation motif (ITAM)-containing signaling domain.
  • the co-stimulatory domain is selected from the group consisting of 4-1BB, CD28.
  • the ITAM-containing signaling domain is derived from CD3zeta.
  • the CAR molecule comprises a signal peptide.
  • the CAR molecule comprises a linker.
  • the linker is connected to/disposed between the antigen binding domain and the additional extracellular domain sequence.
  • the CAR molecule comprises a signal peptide, an antigen binding domain (e.g., an scFv comprising a VH domain linked via a linker to a VL domain), a linker, an extracellular hinge domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain.
  • the linker that connects a VH domain to a VL domain and the linker that connects the antigen binding domain to the extracellular hinge domain are different.
  • the antigen binding domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 1.
  • the antigen binding domain comprises a sequence of SEQ ID NO: 1, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the antigen binding domain comprises a sequence of: QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIK (SEQ ID NO: 1).
  • the CAR molecule comprises a CD8a extracellular domain (z.e., a CD8a hinge domain).
  • the CD8a extracellular domain comprises a sequence of: TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:2).
  • the extracellular domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:2.
  • the CD8a extracellular domain comprises a sequence of SEQ ID NO:2, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CD8a extracellular domain comprises a sequence of: TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:2).
  • the CAR molecule comprises a linker domain between the antigen binding domain and extracellular domain.
  • the linker domain comprises a sequence of AAA (SEQ ID NO:3).
  • the CAR molecule comprises a CD8a transmembrane domain.
  • the CD8a transmembrane domain comprises a sequence of: IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO:4).
  • the CD8a transmembrane domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:4.
  • the CD8a transmembrane domain comprises a sequence of SEQ ID NO:4, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CD8a transmembrane domain comprises a sequence of: IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO:4).
  • the CAR molecule comprises a portion of a CD8a intracellular domain.
  • the CD8a intracellular domain comprises the sequence of: LYC (SEQ ID NO: 5).
  • the CAR molecule comprises a 4- IBB intracellular costimulatory signaling domain.
  • the 4-1BB intracellular costimulatory signaling domain comprises a sequence of SEQ ID NO:6: KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL.
  • the 4-1BB costimulatory domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:6.
  • the 4-1BB costimulatory domain comprises a sequence of SEQ ID NO:6, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the 4-1BB costimulatory domain comprises a sequence of: KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO: 6).
  • the CAR molecule comprises a CD3zeta intracellular signaling domain.
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO:7:
  • the CD3zeta intracellular signaling domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:7.
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO:7, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 7):
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 10: RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRRKNP QEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQA LPPR.
  • the CD3zeta intracellular signaling domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 10.
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 10, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 11 : RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRRKNP QEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQA LPPR.
  • the CD3zeta intracellular signaling domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 11.
  • the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 11, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CAR molecule comprises a signal peptide sequence.
  • the signal peptide sequence comprises a sequence of SEQ ID NO:8: MLLLVTSLLLCELPHPAFLLIP.
  • the signal peptide sequence comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 8.
  • the signal peptide sequence comprises a sequence of SEQ ID NO:8, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the signal peptide sequence comprises a sequence of: MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 8).
  • the CAR molecule comprises a sequence of SEQ ID NO: 12. In some embodiments, the CAR molecule comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 40 amino acid modifications of an amino acid sequence of SEQ ID NO: 12. In some embodiments, the signal peptide sequence comprises a sequence of SEQ ID NO: 12, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CAR molecule comprises a sequence of: QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIW APLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEG GCELRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRR
  • the CAR molecule comprises a sequence of SEQ ID NO:9. In some embodiments, the CAR molecule comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 40 amino acid modifications of an amino acid sequence of SEQ ID NO:9. In some embodiments, the signal peptide sequence comprises a sequence of SEQ ID NO:9, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
  • the CAR molecule comprises a sequence of: MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVT ITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQ AEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACR PAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREE
  • the present disclosure also provides a vector in which a DNA of the present disclosure is inserted.
  • Vectors derived from retroviruses such as lentiviruses are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells.
  • the vector comprises a lentiviral vector.
  • the vector comprises a gamma retroviral vector.
  • a vector may comprise a signal sequence to facilitate secretion, a polyadenylation signal and transcription terminator (e.g., from the Bovine Growth Hormone (BGH) gene), an element allowing episomal replication and replication in prokaryotes (e.g., SV40 origin and ColEl or others known in the art) and/or elements to allow selection (e.g., an ampicillin resistance gene and/or zeocin marker).
  • the vector comprises a promoter.
  • the vector promoter is selected from the group comprising of CMV, PGK, EF-la, MND, MNDU3, MCU3, SFFV, and CBh.
  • a lentiviral vector expresses nucleotide sequences encoding the CAR sequences described herein.
  • a lentiviral vector expresses a nucleotide sequence encoding a CAR sequence comprising an antigen binding domain (e.g., an scFv comprising a VH domain linked via a linker to a VL domain), a linker, an extracellular hinge domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain.
  • an antigen binding domain e.g., an scFv comprising a VH domain linked via a linker to a VL domain
  • a linker e.g., an extracellular hinge domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain.
  • a lentiviral vector expresses a nucleotide sequence encoding a CAR sequence comprising an anti-CD22 binding domain (e.g., an anti-CD22 scFv comprising a VH domain linked via a linker to a VL domain), linked to a CD8 extracellular domain (e.g., a CD8alpha hinge domain) via linker, a CD8a transmembrane domain, a 4- IBB costimulatory domain, and a CD3zeta activation domain.
  • the nucleotide sequence encoding a CAR sequence further comprises a signal peptide sequence.
  • a lentiviral vector expresses a nucleotide sequence encoding a CAR with a sequence as described herein.
  • T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation. In some embodiments, T cells are isolated from a blood sample by apheresis.
  • the collection of blood samples or apheresis product from a subject takes place at a time period prior to lentiviral transduction and when the expanded cells as described herein in the Examples might be needed.
  • the collection of blood samples or apheresis product from a subject can be taken and then subsequently frozen for later use.
  • a source of the cells to be expanded can be collected at any time point necessary, and desired cells, such as T cells, isolated and frozen for later use in immune effector cell therapy for any number of diseases or conditions that would benefit from immune effector cell therapy, such as those described herein.
  • a blood sample or an apheresis product is taken from a generally healthy subject.
  • a blood sample or an apheresis product is taken from a generally healthy subject who is at risk of developing a disease, but who has not yet developed a disease, and the cells of interest are isolated and frozen for later use.
  • the T cells may be expanded, frozen, and used at a later time.
  • samples are collected from a patient shortly after diagnosis of a particular disease as described herein but prior to any treatments.
  • the cells are isolated from a blood sample or an apheresis product from a subject prior to any number of relevant treatment modalities, including but not limited to treatment with agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxan, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation.
  • agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD
  • T cells are obtained from a patient directly following treatment that leaves the subject with functional T cells.
  • the quality of T cells obtained may be optimal or improved for their ability to expand ex vivo.
  • these cells may be in a preferred state for enhanced engraftment and in vivo expansion.
  • apheresis product including T cells, dendritic cells, or other cells of the hematopoietic lineage, during this recovery phase.
  • mobilization for example, mobilization with GM-CSF
  • conditioning regimens can be used to create a condition in a subject wherein repopulation, recirculation, regeneration, and/or expansion of particular immune cell types is favored, especially during a defined window of time following therapy.
  • Illustrative cell types include T cells, B cells, dendritic cells, and other cells of the immune system.
  • Immune effector cells such as T cells may be activated and expanded generally using methods as described, for example, in U.S. Patent Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 2006/0121005 Al, each of which is incorporated herein by reference in its entirety for any purpose.
  • a CAR-expressing T-cell is produced according to the procedures incorporated herein by reference.
  • the hematological cancer is associated with expression of an antigen, e.g., a tumor antigen, e.g., CD22.
  • the hematological cancer comprises lymphoma.
  • the hematological cancer is a lymphoma, e.g., a relapsed and/or refractory lymphoma.
  • the hematological cancer comprises leukemia, e.g., a relapsed and/or refractory leukemia.
  • the cancer is selected from the group consisting of one or more acute leukemias including but not limited to B-cell acute lymphoid leukemia (B-ALL), T-cell acute lymphoid leukemia (TALL), small lymphocytic leukemia (SLL), acute lymphoid leukemia (ALL) (e.g., relapsing and refractory ALL); one or more chronic leukemias including but not limited to chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL).
  • B-ALL B-cell acute lymphoid leukemia
  • TALL T-cell acute lymphoid leukemia
  • SLL small lymphocytic leukemia
  • ALL acute lymphoid leukemia
  • CML chronic myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • the cancer is selected from the group consisting of additional hematologic cancers or conditions including, but not limited to, mantle cell lymphoma (MCL), B cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma (DLBCL), follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia.”
  • MCL mantle cell lymphoma
  • the disease associated with expression of CD22 is a lymphoma.
  • the lymphoma is Non-Hodgkin lymphoma (NHL).
  • NHL Non-Hodgkin lymphoma
  • the majority of NHL cases are of B-cell origin and can be further subdivided into aggressive and indolent lymphomas, each associated with different clinical outcomes and prognosis.
  • the NHL is an aggressive lymphoma selected from the group consisting of AIDS-associated lymphoma, Burkitt lymphoma, primary central nervous system lymphoma, large B cell lymphomas (LBCLs), mantle cell lymphoma, peripheral T-cell lymphoma, and T-cell lymphoblastic lymphoma.
  • the NHL is an indolent lymphoma selected from the group consisting of cutaneous T-cell lymphoma, follicular lymphoma (FL), lymphoplasmacytic lymphoma, marginal zone lymphoma, and small cell lymphocytic lymphoma.
  • the NHL lymphoma is LBCL.
  • Large B-cell lymphomas (LBCLs) are aggressive subtypes including diffuse large cell lymphoma (DLBCL), highgrade B-cell lymphomas, primary mediastinal B-cell lymphoma (PMBCL), Epstein-Barr virus-positive LBCL, follicular lymphoma (FL) grade 3B and other subtypes.
  • the LBCL is DLBCL. In some embodiments, the DLBCL has been histologically confirmed. In some embodiments, the DLBCL is not otherwise specified (NOS) type. In some embodiments, the DLBCL NOS is germinal center B-cell (GCB) type. In some embodiments, the DLBCL NOS is active B-cell (ABC) type. In some embodiments, the LBCL is DLBCL associated with chronic inflammation. In some embodiments, the LBCL is primary cutaneous DLBCL, leg type. In some embodiments, the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS. In some embodiments, the LBCL is DLBCL arising from FL. In some embodiments, the LBCL is DLBCL arising from MZL.
  • the LBCL is primary mediastinal (thymic) large B-cell lymphoma. In some embodiments, the LBCL is Grade 3B follicular large B-cell lymphoma (FL).
  • the LBCL is high-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements.
  • the subject has a relapsed/refractory disease that recurs following treatment.
  • relapsed and/or refractory disease refers to progressive disease (PD)/stable disease (SD) after the last therapy or relapsed disease following a complete response (CR) or progressive disease following a partial response (PR) after the last therapy.
  • PD progressive disease
  • SD stable disease
  • PR progressive disease following a partial response
  • the relapsed/refractory disease comprises a predicted relapse.
  • the relapsed/refractory disease comprises an identified relapse.
  • the relapsed/refractory disease comprises disease progression.
  • the subject is predicted to have a relapse (e.g., has not relapsed), has relapsed, or is identified as having relapsed.
  • the subject relapsed after having received a first-line therapy may comprise an antibody -based therapy, a cell-based therapy, a chemotherapy, or a combination thereof.
  • the antibody-based therapy may comprise CD20-targeted monoclonal antibodies.
  • the antibody -based therapy may comprise CD19-targeted monoclonal antibody therapy.
  • the cell-based therapy may comprise a CAR T-cell therapy. In some embodiments, the cell-based therapy may comprise a CD19 CAR T-cell therapy.
  • the chemotherapy comprises rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP), etoposide, erednisolone, encovin cyclophosphamide, and eydroxydaunorubicin (EPOCH), or a combination thereof.
  • the subject is 18 years or older.
  • the subject has received one or more previous treatments. In some embodiments, the subject has received about 1, about 2, about 3, about 4 or about 5 previous treatments. In some embodiments, the subject has received 1 or more previous treatments, 2 or more previous treatments, 3 or more previous treatments, 4 or more previous treatments, or 5 or more previous treatments.
  • the antibody comprises an anti-CD19 antibody and an anti-CD20 antibody. In some embodiments, the antibody comprises an anti- CD19 antibody, an anti-CD20 antibody, and an anti-CD22 antibody. In some embodiments, the antibody comprises an anti-CD19 antibody, an anti-CD20 antibody, and an anti-CD79b antibody. In some embodiments, the anti-CD19 antibody, the anti-CD20 antibody, the anti- CD22 antibody or the anti-CD79b antibody is an antibody-drug conjugate. In some embodiments, the anti-CD19 antibody comprises loncastuximab. In some embodiments, the anti-CD20 antibody comprises rituximab. In some In some embodiments, the anti-CD22 antibody-drug conjugate comprises inotuzumab ozogamicin.
  • the bispecific T-cell engager antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, elranatamab, teclistamab, tarlatamab, talquetamab, solitomab, tebentafusp, duvortuxizumab, AFM11, REGN5458, CC-93269, PF-06863135, TNB-383B, odronextamab, XmAbl3676, IGM-2323, AMG 330, AMG 673, AMG 420, AMG 701, cevostamab, talquetamab, flotetuzumab, or a combination thereof.
  • the subject has relapsed/refractory disease after the last therapy.
  • relapsed/refractory disease comprises radiographic findings comprising progressive disease/stable disease after the last therapy.
  • relapsed/refractory disease comprises radiographic disease progression following a partial response after the last therapy.
  • relapsed/refractory disease comprises radiographic disease progression following a complete response after the last therapy.
  • relapsed/refractory disease comprises radiographic disease progression after a partial response.
  • relapsed/refractory disease comprises radiographic disease progression after a complete response.
  • relapsed/refractory disease comprises viable lymphoma in a biopsy.
  • relapsed/refractory disease comprises viable lymphoma in a biopsy after therapy.
  • the subject has at least one radiographically measurable lesion.
  • the radiographically measurable lesion has at least one bidimensionally measurable nodal lesion of >1.5 cm in its longest dimension.
  • the radiographically measurable lesion has at least one bidimensionally measurable extra lesion of >1.0 cm in its longest dimension.
  • lesions that were previously irradiated are considered measurable if progression has been documented following completion of radiation therapy.
  • the subject may have a washout period following the last therapy.
  • the washout period is a time duration between receiving a therapy and receiving the next therapy.
  • the washout period is 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, or some fraction thereof.
  • the washout period is at least one half-life of the last therapy administered to the subject.
  • half-life or “half-lives” refers to one or more time periods, where each time period comprises the time required for half of something (e.g., the last therapy administered to the subject) to be metabolized or eliminated from the subject’s system.
  • the subject has previously received bendamustine (marketed as Bendeka® or Treanda®).
  • the bendamustine was last received at least 3-9 months prior to receiving the CD22-directed CAR T-cell therapy.
  • the bendamustine was last received at least 3 months, at least 3.5 months, at least 4 months, at least 4.5 months, at least 5 months, at least 5.5 months, at least 6 months, at least 6.5 months, at least 7 months, at least 7.5 months, at least 8 months, at least 8.5 months, at least 9 months or some fraction thereof prior to receiving the CD22-directed CAR T-cell therapy.
  • the bendamustine was last received at least 6 months prior to receiving the CD22-directed CAR T-cell therapy.
  • the subject has previously received CD19-directed CAR T-cell therapy.
  • the CD19-directed CAR T-cell therapy was last administered at least 10-60 days prior to receiving the CD22-directed CAR T-cell therapy.
  • the CD19-directed CAR T-cell therapy was last administered at least 10 days, at least 15 days, at least 20 days, at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, or some fraction thereof prior to receiving the CD 19- directed CAR T-cell therapy.
  • the CD19-directed CAR T-cell therapy was last administered at least 30 days prior to receiving the CD19-directed CAR T-cell therapy.
  • the subject has previously received bispecific CAR T-cell therapy.
  • the bispecific CAR T-cell therapy was last administered at least 10-60 days prior to receiving the CD22-directed CAR T-cell therapy.
  • the bispecific CAR T-cell therapy was last administered at least 10 days, at least 15 days, at least 20 days, at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, or some fraction thereof prior to receiving the CD22- directed CAR T-cell therapy.
  • the bispecific CAR T-cell therapy was last administered at least 30 days prior to receiving the CD22-directed CAR T-cell therapy.
  • the subject has previously received a bispecific T-cell engager therapy.
  • the bispecific T-cell engager therapy was last administered at least 10-60 days prior to receiving the CD22-directed CAR T-cell therapy.
  • the bispecific T-cell engager therapy was last administered at least 10 days, at least 15 days, at least 20 days, at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, or some fraction thereof prior to receiving the CD22- directed CAR T-cell therapy.
  • the bispecific T-cell engager therapy was last administered at least 30 days prior to receiving the CD22-directed CAR T-cell therapy.
  • the subject has previously received an autologous stem cell transplant.
  • the autologous stem cell transplant was last received at least 2-20 weeks prior to receiving the CD22-directed CAR T-cell therapy.
  • the autologous stem cell transplant was last received at least 2 weeks, at least 3 weeks, at least 4 weeks, at least 5 weeks, at least 6 weeks, at least 7 weeks, at least 8 weeks, at least 9 weeks, at least 10 weeks, at least 11 weeks, at least 12 weeks, at least 13 weeks, at least 14 weeks, at least 15 weeks, at least 16 weeks, at least 17 weeks, at least 18 weeks, at least 19 weeks, at least 20 weeks, or some fraction thereof prior to receiving the CD22- directed CAR T-cell therapy.
  • the autologous stem cell transplant was last received at least 6 weeks prior to receiving the CD22-directed CAR T-cell therapy.
  • the subject has previously received CNS prophylaxis.
  • the CNS prophylaxis is intrathecal methotrexate.
  • the CNS prophylaxis was last administered at least 1-20 days prior to receiving the CD22- directed CAR T-cell therapy.
  • the CNS prophylaxis was last administered at least 1 day, at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days, at least 15 days, at least 16 days, at least 17 days, at least 18 days, at least 19 days, at least 20 days, or some fraction thereof prior to receiving the CD22-directed CAR T-cell therapy.
  • the CNS prophylaxis was administered at least 7 days prior to receiving the CD22-directed CAR T- cell therapy. [0248]
  • the subject received the CD22-directed CAR T-cell therapy without a washout period.
  • a tumor tissue sample from the subject is available for analysis.
  • the tumor tissue sample is a formalin-fixed paraffin- embedded tissue block.
  • the tumor sample is resected tissue.
  • the tissue sample is fresh-frozen.
  • the tumor tissue sample is obtained following the most recent therapy.
  • the tumor tissue sample is obtained following CD 19 CAR T-cell therapy or any other line of therapy administered prior to relapse.
  • more than one tumor tissue sample is available. In some embodiments, the more than one tumor tissue sample is from the same tumor location. In some embodiments, the more than one tumor tissue sample is from more than one tumor location.
  • the subject has adequate hematologic and end-organ function.
  • the adequate hematologic and end-organ function comprises an absolute neutrophil count (ANC) greater than about 500-1500/pl, about 700-1200/pl, of about 900-1100/pl, about 800/pl, about 900/pl, about 1000/pl, about 1100/pl, or about 1200/pl.
  • the subject has an ANC of greater than or equal to about 1000/pl.
  • the adequate hematologic and end-organ function comprises a platelet count of greater than about 50, 000-100, 000/mL, about 60, 000-90, 000/mL, about 50, 000-80, 000/mL, about 50, 000/mL, about 60, 000/mL, about 70, 000/mL, about 75, 000/mL, about 80, 000/mL, about 90, 000/mL, or about 100, 000/mL.
  • the adequate hematologic and end-organ function comprises a platelet count of greater than about 75,000/pl.
  • the subject has a presence of marrow disease.
  • the adequate hematologic and end-organ function for a subject with the presence of marrow disease has a platelet count of greater than about 25, 000-75, 000/pl, about 30, 000-70, 000/pl, about 40, 000-60, 000/pl, about 25, 000/pl, 30,000/pl, about 35,000/pl, about 40,000/pl, about 45,000/pl, about 50,000/pl, about 55,000/pl, about 60,000/pl, about 65,000/pl, about 70,000/pl, or about 75,000/pl.
  • the adequate hematologic and end-organ function for a subject with the presence of marrow disease comprises a platelet count of greater than about 50,000/pl.
  • the adequate hematologic and end-organ function comprises a platelet count of greater than about 75, 000/mL.
  • the subject has a presence of marrow disease.
  • the adequate hematologic and end-organ function for a subject with the presence of marrow disease has a platelet count of greater than about 25, 000-75, 000/mL, about 30, 000-70, 000/mL, about 40, 000-60, 000/mL, about 25, 000/mL, 30, 000/mL, about 35, 000/mL, about 40, 000/mL, about 45, 000/mL, about 50, 000/mL, about 55, 000/mL, about 60, 000/mL, about 65, 000/mL, about 70, 000/mL, or about 75, 000/mL.
  • the adequate hematologic and end-organ function for a subject with the presence of marrow disease comprises a platelet count of greater than about 50, 000/mL.
  • the adequate hematologic and end-organ function comprises an absolute lymphocyte count (ALC) of greater than about 10-300/pl, about 50-150, 000/pl, about 75-125/(11, about 10/pl, 20/pl, about 30/pl, about 40/pl, about 50/pl, about 60/pl, about 70/pl, about 80/pil, about 90/pl, about 100/pl, about 110/pl, about 120/pl, about 130/pl, about 140/ .l, about 150/pl, about 160/jj.l, about 170/ .l, about 180/ .l, about 190/jj.l, about 200/pl, about 225/pl, about 250/ l, about 275/pl, or about 3 OO/JJ.1.
  • the subject has an ALC of greater than or equal to about 1 OO/JJ.1.
  • the estimated creatinine clearance is measured according to the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) method. In some embodiments, the estimated creatinine clearance is measured based on the volume of urine collected spanning about one day (e.g., about 24 hours). In some embodiments, the estimated creatinine clearance is measured in other ways.
  • CKD-EPI Chronic Kidney Disease Epidemiology Collaboration
  • the adequate hematologic and end-organ function comprises a serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) activity of less than about 1-5 times the institutional upper limit of normal (ULN), about 2-4 times ULN, about 2- 3 times the ULN, about 1 times the ULN, about 1.5 times the ULN, about 2.0 times the ULN, about 2.5 times the ULN, about 3.0 times ULN, about 3.5 times the ULN, about 4.0 times the ULN, about 4.5 times the ULN, or about 5 times the ULN.
  • ALT serum alanine aminotransferase
  • AST aspartate aminotransferase
  • the adequate hematologic and end-organ function comprises a serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) activity of less than about 2.5 times the ULN.
  • the adequate hematologic and end-organ function comprises a total bilirubin concentration of less than about 0.5-4 mg/dL, about 1-3 mg/dL, about 1-2 mg/dL, about 0.5 mg/dL, about 1.0 mg/dL, about 1.5 mg/dL, about 2.0 mg/dL, about 2.5 mg/dL, about 3.0 mg/dL, about 3.5 mg/dL, or about 4.0 mg/dL.
  • the adequate hematologic and end-organ function comprises a total bilirubin concentration of less than about 1.5 mg/dL. In some embodiments, the adequate hematologic and end-organ function comprises a total bilirubin concentration that is different for subjects with Gilbert syndrome. In some embodiments, the adequate hematologic and end-organ function comprises a total bilirubin concentration greater than 1.5 mg/dL for subjects with Gilbert syndrome.
  • the adequate hematologic and end-organ function comprises a blood oxygen saturation (SaCL) of greater than about 85-100%, about 90-95%, about 91- 93%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%.
  • the adequate hematologic and end-organ function comprises a blood oxygen saturation (SaCL) of greater than about 92%.
  • the adequate hematologic and end-organ function comprises a serum albumin concentration of greater than about 1-5 g/dL, about 2-4 g/dL, about 2-3 g/dL, about 1 g/dL, about 1.5 g/dL, about 2 g/dL, about 2.5 g/dL, about 3 g/dL, about 3.5 g/dL, about 4 g/dL, about 4.5 g/dL, or about 5 g/dL.
  • the adequate hematologic and end-organ function comprises a serum albumin concentration of greater than about 2.5 g/dL.
  • the subject is a woman of childbearing potential.
  • the women of childbearing potential may refrain from heterosexual intercourse during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the woman of childbearing potential may use contraceptive methods during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the woman of childbearing potential may use contraceptive methods that comprise a failure rate of less than about 0.1-5.0% per year, about 0.5-1.5% per year, about 0.2-2.0% per year, about 0.1% per year, about 0.2% per year, about 0.3% per year, about 0.4% per year, about 0.5% per year, about 0.6% per year, about 0.7% per year, about 0.8% per year, about 0.9% per year, about 1.0% per year, about 1.5% per year, about 2.0% per year, about 2.5% per year, about 3.0% per year, about 3.5% per year, about 4.0% per year, about 4.5% per year, or about 5.0% per year during the course of the treatment with the CAR T-cell therapy.
  • the woman of childbearing potential is post monarchal. In some embodiments, the woman of childbearing potential has not reached a postmenopausal state. In some embodiments the postmenopausal state comprises more than 12 months of amenorrhea with no identified cause other than menopause. In some embodiments, the woman of childbearing potential has not undergone surgical sterilization (removal of ovaries and/or uterus). In some embodiments, the woman of childbearing potential may comprise additional characteristics. [0264] In some embodiments, contraceptive methods may comprise bilateral tubal ligation, male sterilization, hormonal contraceptives that inhibit ovulation, hormone-releasing intrauterine devices, copper intrauterine devices, or a combination thereof.
  • the woman of childbearing potential must have a negative pregnancy test result within about 1-14 days, about 3-10 days, about 5-10 days, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days before leukapheresis.
  • the woman of childbearing potential must have a negative pregnancy test result within about 7 days before leukapheresis.
  • the subject may not have a pregnancy test.
  • the subject is male.
  • the male subject may refrain from heterosexual intercourse.
  • the male subject may refrain from heterosexual intercourse during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the male subject may use a condom with spermicide.
  • the male subject may use a condom with spermicide during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the male subject may use a condom with spermicide and the sexual partner may use a contraception method.
  • the contraception method is highly effective.
  • the male subject may refrain from donating sperm during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the male subject is vasectomized.
  • the vasectomized male subject may use a condom with spermicide.
  • the male subject may use a condom with spermicide during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the male subject may engage in sexual intercourse with a male partner.
  • the male subject that engages in sexual intercourse with a male partner may use a condom with spermicide during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after.
  • the subject is stable.
  • the subject is stable or has recovered from toxicities associated with previous chemo- or immunotherapies prior to receiving the CD22-directed CAR T-cell therapy.
  • the subject is stable or has recovered from nonhematologic toxicities due to prior therapy to grade ⁇ 1.
  • the subject has clinically non-significant toxicities prior to therapy greater than grade 1.
  • the clinically non-significant toxicity is alopecia.
  • the subject may not receive the CD22-directed CAR T-cell therapy if one or more of the following conditions apply: the subject has a malignancy other than lymphoma; the subject has an active fungal, bacterial, viral or other infection that requires intravenous antimicrobials; the subject has received prior allogenic stem cell or any other organ transplant; the subject has received a prior allogeneic CAR T-cell therapy; the subject has received prior anti-CD52 antibody therapy; the subject has a history of CNS involvement of lymphoma within 1 year prior to receiving the CD22-directed CAR T-cell therapy; the subject has ongoing cardiac involvement of lymphoma; the subject has a history of infection with human immunodeficiency virus (HIV), hepatitis B virus (HBV), as determined by positivity for hepatitis B serum antigen (HbsAg) or hepatitis B core antibody (HbcAb), or hepatitis C virus (HCV), as determined by positivity for anti-HC
  • HCV human immunodefic
  • the significant, uncontrolled concomitant disease that could affect interpretation of results from the CD22-directed CAR T-cell therapy comprises a history of myocardial infarction, cardiac angioplasty or stenting, unstable angina, serious cardiac arrhythmia, or other clinically significant cardiac disease within 12 months prior to treatment with the CD22-directed CAR T-cell therapy.
  • the significant, uncontrolled concomitant disease comprises active pulmonary disease (such as severe obstructive pulmonary disease, idiopathic pulmonary fibrosis, organizing pneumonia, drug- induced pneumonitis, idiopathic pneumonitis).
  • the significant, uncontrolled concomitant disease comprises clinically significant liver disease, including cirrhosis. In some embodiments, the significant, uncontrolled concomitant disease comprises clinically significant neurological disease (such as cerebrovascular ischemia/hemorrhage, dementia, seizure disorder, or cerebellar disease).
  • clinically significant neurological disease such as cerebrovascular ischemia/hemorrhage, dementia, seizure disorder, or cerebellar disease.
  • the modifying agents required for systemic immunosuppression do not include physiologic dose of steroid, a stable dose of thyroid replacement hormone (for subjects with a history of autoimmune-related hypothyroidism), or an insulin regimen (for subjects with controlled Type 1 diabetes mellitus).
  • the subject may have a history of immune thrombocytopenic purpura or autoimmune hemolytic anemia not requiring active treatment and thus may receive CD22- directed CAR T-cell therapy.
  • a CAR-expressing cell e.g., a CD22 CAR-expressing T cell as described herein, is administered to the subject according to a dosing regimen comprising a dose, e.g., a total dose, of cells administered to the subject by dose fractionation A.g, split dosing), e.g., one, two, three or more separate administrations of a partial dose, e.g., one, two, three, four, five or six partial doses.
  • a total dose of cells is administered as a single dose.
  • a single dose comprises the total dose.
  • a first percentage of the total dose e.g., a first partial dose
  • a second percentage of the total dose e.g., a second partial dose
  • a third percentage e.g., the remaining percentage of the total dose, e.g., a third partial dose
  • a sample of CAR-expressing cells comprises about 0.1-100% of total cells expressing at least one CD22 CAR molecule.
  • a sample of CAR-expressing cells comprises about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 2.5%, about 3%, about 3.5%, about 4%, about 4%, about 4.5%, about 5%, about 5.5%, about 5.5%, about 6%, about 6.5%, about 7%, about 7%, about 7.5%, about 8%, about 8.5%, about 8.5%, about 9%, about 9.5%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about
  • a total dose of CAR- expressing cells comprises about 0.5-20* 10 6 cells/kg, e.g., about 0.1 * 10 6 cells/kg, about 0.2* 10 6 cells/kg, about 0.3* 10 6 cells/kg, about 0.4* 10 6 cells/kg, about 0.5* 10 6 cells/kg, about 0.6*10 6 cells/kg, about 0.7* 10 6 cells/kg, about 0.8* 10 6 cells/kg, about 0.9* 10 6 cells/kg, about 1.0* 10 6 cells/kg, about 1.5* 10 6 cells/kg, about 2.0* 10 6 cells/kg, about 2.5* 10 6 cells/kg, about 3.0* 10 6 cells/kg, about 3.5* 10 6 cells/kg, about 4.0* 10 6 cells/kg, about 4.5*10 6 cells/kg, about 5.0* 10 6 cells/kg, about 5.5* 10 6 cells/kg, about 6.0* 10 6 cells/
  • the total dose of CD22 CAR-expressing cells comprises about 0.5* 10 6 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 1.0* 10 6 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 2.0* 10 6 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 3.0* 10 6 cells/kg.
  • a partial dose of CAR-expressing (e.g., CD22 CAR-expressing T cells expressing at least one CD22 CAR molecule) cells comprises about 0.2-20* 10 6 cells/kg, e.g., about 0.5-5* 10 6 cells/kg, about 1-5* 10 6 cells/kg, about 0.5-1 * 10 6 cells/kg, about 5-10* 10 6 cells/kg, about 15-20* 10 6 cells/kg, 0.6-24* 10 6 cells/kg, 0.7-23* 10 6 cells/kg, 0.8-22* 10 6 cells/kg, 0.9-21 * 10 6 cells/kg, 1-20* 10 6 cells/kg, 2-19* 10 6 cells/kg, 3-18* 10 6 cells/kg, 4-17* 10 6 cells/kg, 5-16*10 6 cells/kg, 6-15* 10 6 cells/kg, 7-14*10 6 cells/kg, 8- 13* 10 6 cells/kg, 9-12* 10 6 cells/kg, or 10-11 * 10 6 cells/kg.
  • CAR-expressing e.g., CD22 CAR
  • a partial dose of CAR-expressing cells comprises about 0.5-20* 10 6 cells/kg, e.g., about 0.1 * 10 6 cells/kg, about 0.2-* 10 6 cells/kg, about 0.3* 10 6 cells/kg, about 0.4* 10 6 cells/kg, about 0.5* 10 6 cells/kg, about 0.6* 10 6 cells/kg, about 0.7*10 6 cells/kg, about 0.8* 10 6 cells/kg, about 0.9* 10 6 cells/kg, about 1.0* 10 6 cells/kg, about 1.5* 10 6 cells/kg, about 2.0* 10 6 cells/kg, about 2.5* 10 6 cells/kg, about 3.0* 10 6 cells/kg, about 3.5* 10 6 cells/kg, about 4.0* 10 6 cells/kg, about 4.5* 10 6 cells/kg, about 5.0*10 6 cells/kg, about 5.5* 10 6 cells/kg, about 6.0* 10 6 cells/kg, about 6.5* 10 6 cells
  • the dose of CAR expressing T cells will be based on the weight of the subject. In some embodiments, the dose of the CAR expressing T cells will be partially based on the weight of the subject. In some embodiments, the subject weights less than 100 kg. In some embodiments, the subject weighs at least about 50 kg, at least about 60 kg, at least about 70 kg, at least about 80 kg, or at least about 90 kg. In some embodiments, the subject weights at least about 100 kg. In some embodiments, the subject weighs at least about 110kg, at least about 120kg, at least about 130kg, at least about 140kg, or at least about 150kg.
  • the CAR expressing T cells will be administered by IV infusion.
  • the CAR expressing T cells are formulated for IV infusion and such formulation comprises DMSO.
  • the IV infusion duration will comprise about 5-60 minutes.
  • the IV infusion duration will comprise about 5 minutes, about 10 minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes or about 60 minutes.
  • the CAR expressing T cells will be contained in an IV bag.
  • the IV bag will be attached to the subject via tubing.
  • the tubing comprises non-filtered tubing.
  • the IV bag will be agitated during the IV infusion. In some embodiments, the IV bag will be gently agitated during the IV infusion. In some embodiments, the IV bag and tubing will be rinsed after infusion of the CAR expressing T cells. In some embodiments, the IV bag and tubing will be rinsed with saline after infusion of the CAR expressing T cells. In some embodiments, the saline used to rinse the IV bag and tubing will be administered to the subject.
  • the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is frozen. In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is frozen prior to administering to the subject. In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is cryopreserved prior to administering to the subject. In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is cryopreserved in cryo-storage bags prior to administering to the subject. In some embodiments, the cryo-storage bags are stored in liquid nitrogen. In some embodiments, the cryo-storage bags are stored in the vapor phase of liquid nitrogen.
  • an additional therapy is administered in combination with the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells).
  • the additional therapy is administered before administering the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells).
  • the additional therapy is administered substantially concurrent with administering the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells).
  • the additional therapy is administered after administering the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells).
  • the additional therapy is administered to treat cytokine release syndrome (CRS).
  • CRS cytokine release syndrome
  • the additional therapy administered to treat CRS comprises tocilizumab. In some embodiments, the additional therapy administered to treat CRS comprises glucocorticoids. In some embodiments, the additional therapy administered to treat CRS comprises glucocorticoids and tocilizumab. In some embodiments, the additional therapy administered to treat CRS comprises an IL-1 receptor antagonist. In some embodiments, the IL-1 receptor antagonist comprises anakinra. In some embodiments, anakinra is administered to treat CAR T-cell associated toxicity. In some embodiments, anakinra is administered to treat immune effector cell-associated hemophagocytic lymphohistiocytosis (HLH)-like syndrome (IEC-HS).
  • HHLH immune effector cell-associated hemophagocytic lymphohistiocytosis
  • IEC-HS immune effector cell-associated hemophagocytic lymphohistiocytosis
  • the additional therapy comprises administration of treatments for infection.
  • the additional therapy comprises administration of therapeutics intended to prevent viral, fungal, bacterial or Pneumocystis infections.
  • the additional therapy comprises administration of one or more hematopoietic growth factors.
  • lymphodepletion is performed on a subject, e.g., prior to administering one or more cells that express a CAR as described herein.
  • lymphodepletion include, but may not be limited to, nonmyeloablative lymphodepleting chemotherapy, myeloablative lymphodepleting chemotherapy, total body irradiation.
  • the lymphodepletion regimen comprises administering one or more chemotherapeutics selected from the group consisting of melphalan, cytoxan, bendamustine, cyclophosphamide, and fludarabine.
  • the lymphodepletion regimen comprises administering cyclophosphamide at least one, two, three, four, five, or more days before administration of an infusion of CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells).
  • cyclophosphamide is administered once daily for 1 day, for 2 days, for 3 days, for 4 days for 5 days, for 6 days or for 7 days.
  • cyclophosphamide is administered once daily for three days beginning 5 days before administration of an infusion of CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells), on days -5, -4, and -3 prior to infusion.
  • cyclophosphamide is administered at a dosage of about 100-800 mg/m 2 /day, about 200-800 mg/m 2 /day, about 100-800 mg/m 2 /day, or about 400-600 mg/m 2 /day.
  • cyclophosphamide is administered once daily at a dosage of about 100 mg/m 2 /day, about 150 mg/m 2 /day, about 200 mg/m 2 /day, about 250 mg/m 2 /day, about 300 mg/m 2 /day, about 350 mg/m 2 /day, about 400 mg/m 2 /day, about 450 mg/m 2 /day, about 500 mg/m 2 /day, about 550 mg/m 2 /day, about 600 mg/m 2 /day, about 650 mg/ m 2 /day, about 700 mg/m 2 /day, about 750 mg/m 2 /day, or about 800 mg/m 2 /day.
  • cyclophosphamide is administered at a dosage of about 500 mg/m 2 /day. In some embodiments, cyclophosphamide is administered once daily at a dosage of about 500 mg/m 2 /day for one, two, or three days. In some embodiments, cyclophosphamide is administered once daily at a dosage of about 500 mg/m 2 /day for three days beginning 5 days before administration of an infusion of CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells), on days -5, -4, and -3 prior to infusion.
  • CAR-expressing T-cells e.g., CD22 CAR-expressing T-cells
  • cyclophosphamide is administered for 2 days at a dosage of about 200-600 mg/m 2 /day (e.g., 200-400 mg/m 2 /day, about 250-500 mg/m 2 /day, about 250- 600 mg/m 2 /day, about 300-500 mg/m 2 /day, about 300-550 mg/m 2 /day, about 300-600 mg/m 2 /day, or about 450-550 mg/m 2 /day, or about 450 mg/m 2 /day, or 500 mg/m 2 /day).
  • 200-600 mg/m 2 /day e.g., 200-400 mg/m 2 /day, about 250-500 mg/m 2 /day, about 250- 600 mg/m 2 /day, about 300-500 mg/m 2 /day, about 300-550 mg/m 2 /day, about 300-600 mg/m 2 /day, or about 450-550 mg/m 2 /day, or about 450 mg/m 2
  • cyclophosphamide is administered for 3 days at a dosage of about 200- 600 mg/m 2 /day (e.g, 200-400 mg/m 2 /day, about 250-500 mg/m 2 /day, about 250-600 mg/m 2 /day, about 300-500 mg/m 2 /day, about 300-550 mg/m 2 /day, about 300-600 mg/m 2 /day, or about 450-550 mg/m 2 /day, or about 450 mg/m 2 /day, or 500 mg/m 2 /day). In some embodiments, cyclophosphamide is administered at a dosage of about 500 mg/m 2 per day, for 3 days.
  • cyclophosphamide is administered at a dosage of about 500 mg/m 2 per day, for 3 days.
  • the lymphodepletion regimen comprises administering fludarabine.
  • fludarabine is administered for 1 day, for 2 days, for 3 days, for 4 days for 5 days, for 6 days or for 7 days.
  • fludarabine is administered at a dosage of about 10-400 mg/ m 2 /day, about 20-400 mg/ m 2 /day, about 20-200 mg/ m 2 /day, or about 25-35 mg/ m 2 /day.
  • fludarabine is administered at a dosage of about 10 mg/m 2 /day, about 15 mg/m 2 /day, about 20 mg/m 2 /day, about 25 mg/m 2 /day, about 30 mg/m 2 /day, about 35 mg/m 2 /day, about 40 mg/m 2 /day, about 45 mg/m 2 /day, about 50 mg/m 2 /day, about 55 mg/m 2 /day, about 60 mg/m 2 /day, about 65 mg/ m 2 /day, about 70 mg/m 2 /day, about 75 mg/m 2 /day, about 80 mg/m 2 /day, about 85 mg/m 2 /day, about 90 mg/m 2 /day, about 95 mg/m 2 /day, about 100 mg/m 2 /day, about 110 mg/m 2 /day, about 120 mg/m 2 /day, about 130 mg/m 2 /day, about 140 mg/m 2 /day, about 150 mg/m 2 /day
  • fludarabine is administered for 2 days at a dosage of about 15- 60 mg/m 2 /day (e.g., 20-40, 25-50, 25-60, 30-50, 30-55, 25-60, or 25-35 e.g., or about 25 mg/m 2 , or 30 mg/m 2 ). In some embodiments fludarabine is administered for 3 days at a dosage of about 15-60 mg/m 2 /day (e.g., 20-40, 25-50, 25-60, 30-50, 30-55, 25-60, or 25-35 e.g., or about 25 mg/m 2 , or 30 mg/m 2 ). In some embodiments, fludarabine is administered at a dosage of about 30 mg/m 2 per day, for 3 days.
  • the lymphodepletion regimen comprises administering cyclophosphamide prior to administering CAR T-cells. In some embodiments, the lymphodepletion regimen comprises administering fludarabine prior to administering CAR T-cells. In some embodiments, the lymphodepletion regimen comprises administering cyclophosphamide and fludarabine prior to administering CAR T-cells. In some embodiments, the lymphodepletion regimen comprises administering cyclophosphamide and fludarabine prior to administering CAR T-cells.
  • the lymphodepletion comprises administering 500 mg/m 2 cyclophosphamide once daily for 2 days and 30 mg/m 2 fludarabine once daily for 2 days. In some embodiments, the lymphodepletion regimen comprises administering 500 mg/m 2 cyclophosphamide once daily for 3 days, and 30 mg/m 2 fludarabine once daily for 3 days. In some embodiments, the lymphodepletion regimen comprises administering 500 mg/m 2 cyclophosphamide once daily for 4 days, and 30 mg/m 2 fludarabine once daily for 4 days.
  • the lymphodepletion regimen is initiated with the administration of the first dose of cyclophosphamide. In some embodiments, the lymphodepletion regimen is initiated with the administration of the first dose of fludarabine. In some embodiments, cyclophosphamide and fludarabine are administered on the same day. In some embodiments, cyclophosphamide and fludarabine are not administered on the same day. In some embodiments, the daily dosages are administered on consecutive days. [0288] In some embodiments, at least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine.
  • a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine.
  • second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine.
  • a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine.
  • each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered.
  • the first dose of cyclophosphamide is administered on day -20, -19, -18, -17, -16, -15, -14, -13, -12, -11, -10, -9, -8, -7, -6, -5, -4, -3, -2, or -1 and the engineered CAR T-cells (e.g., CD22 CAR T-cells) are administered on day 0.
  • the engineered CAR T-cells e.g., CD22 CAR T-cells
  • the engineered CAR T-cells e.g., autologous CD22 CAR T-cells
  • the first dose of cyclophosphamide and the first dose of fludarabine are administered on day -20, -19, -18, -17, -16, -15, -14, -13, -12, -11, -10, -9, -8, -7, -6, -5, -4, -3, -2, or -1 and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -10 and day -9 and day -8, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -9 and day -8 and day -7, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -8 and day -7 and day -6, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -7 and day -6 and day -5, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -6 and day -5 and day -4, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -4 and day -3 and day -2, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide is administered on day -3 and day -2 and day -1, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • the engineered CAR T-cells e.g., autologous CD22 CAR T-cells
  • a dose of fludarabine is administered on day -10 and day -9 and day -8, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -9 and day -8 and day -7, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -8 and day -7 and day -6, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -7 and day -6 and day -5, and the autologous CD22 CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -6 and day -5 and day -4, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -4 and day -3 and day -2, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of fludarabine is administered on day -3 and day -2 and day -1, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide and a dose of fludarabine are administered on day -10 and day -9 and day -8, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • the engineered CAR T-cells e.g., autologous CD22 CAR T-cells
  • a dose of cyclophosphamide and a dose of fludarabine are administered on day -9 and day -8 and day - 7, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide and a dose of fludarabine are administered on day -6 and day -5 and day -4, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • a dose of cyclophosphamide and a dose of fludarabine are administered on day -5 and day -4 and day - 3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
  • the subject is administered CAR-expressing cells (e.g., autologous CD22 CAR T-cells) about 1-14 days, e.g, 2-13, 3-12, 4-11, 5-10, 2-11, 2-6, or 1- 4 days, after completion of the lymphodepletion regimen.
  • the lymphodepletion regimen is administered to the subject about 1 week, e.g., about 6, 5, 4, 3, 2, or 1 days, prior to administration of CAR-expressing cells (e.g., autologous CD22 CAR T- cells).
  • the lymphodepletion regimen is completed, about 1 week, e.g., about 6, 5, 4, 3, 2, or 1 days, prior to administration of CAR-expressing cells (e.g., autologous CD22 CAR T-cells).
  • the lymphodepletion regimen comprises administering 500 mg/m 2 cyclophosphamide daily for 3 days, e.g., 3 doses, and 30 mg/m 2 fludarabine daily for 3 days, e.g., 3 doses.
  • the subject is administered a first lymphodepletion regimen and/or a second lymphodepletion regimen.
  • the first lymphodepletion regimen is administered before the second lymphodepletion regimen.
  • the second lymphodepletion regimen is administered before the first lymphodepletion regimen.
  • the first lymphodepletion regimen comprises cyclophosphamide and fludarabine, e.g., 500 mg/m 2 cyclophosphamide daily for 3 days, and 30 mg/m 2 fludarabine daily for 3 days
  • the allopurinol comprises a dose of about 300 mg/m 2 administered orally once per day. In some embodiments, the allopurinol comprises a dose of about 100 mg/m 2 /dose administered orally three times per day.
  • the premedication comprises acetaminophen. In some embodiments, the acetaminophen is administered orally. In some embodiments, the acetaminophen is administered by IV. In some embodiments, the acetaminophen comprises a dose of about 100-1000 mg. In some embodiments, the acetaminophen comprises a dose of about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, about 1000 mg. In some embodiments, the premedication comprises acetaminophen administered orally in a dose of about 650/mg. In some embodiments, the premedication comprises acetaminophen administered by IV with a dose of about 650 mg.
  • the premedication is administered about 5 minutes, aboutlO minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes, about 60 minutes, about 65 minutes, about 70 minutes, about 75 minutes, about 80 minutes, about 85 minutes, about 90 minutes, about 95 minutes, about 100 minutes, about 105 minutes, about 110 minutes, about
  • the premedication comprises diphenhydramine and acetaminophen before the subject receives a dose of the autologous CD22-directed CAR T-cell therapy. In some embodiments the premedication comprises diphenhydramine and acetaminophen and is administered 30-120 minutes before the subject receives a dose of the autologous CD22-directed CAR T-cell therapy.
  • a bridging therapy is administered to the subject.
  • the bridging therapy is administered after leukapheresis.
  • the bridging therapy is administered before lymphodepletion.
  • the bridging therapy is administered at least about 1-10 days before lymphodepletion.
  • the bridging therapy is administered at least about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days or about 10 days before lymphodepletion.
  • the bridging therapy is administered to the subject for about 1- 10 days. In some embodiments, the bridging therapy is administered to the subject for about 1-4 days. In some embodiments, the bridging therapy is administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days or about 10 days.
  • the bridging therapy is administered orally. In some embodiments, the bridging therapy is administered by IV. In some embodiments the bridging therapy comprises dexamethasone. In some embodiments, the dexamethasone comprises a daily dose of about 10-100 mg, or about 20-40 mg. In some embodiments, the dexamethasone comprises a daily dose of about 10-100 mg or mg equivalent.
  • the dexamethasone comprises a daily dose of about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, or about 100 mg.
  • the bridging therapy is administered orally and comprises dexamethasone comprising a daily dose of about 20-40 mg.
  • the bridging therapy is administered by IV and comprises dexamethasone comprising a daily dose of about 20-40 mg.
  • the bridging therapy is administered orally and comprises dexamethasone comprising a daily dose of about 20-40 mg for 1-4 days. In some embodiments, the bridging therapy is administered by IV and comprises dexamethasone comprising a daily dose of about 20-40 mg for 1-4 days. [0307] In some embodiments, the bridging therapy comprises radiation treatment. In some embodiments, the radiation therapy is administered to sites of symptomatic, bulky or extranodal disease.
  • the bridging therapy comprises a course of gemcitabineoxaliplatin plus rituximab (R-GemOx).
  • the time between leukapheresis and administering the CAR T-cell therapy is less than 3 weeks, and the bridging therapy is administered during that period.
  • the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks, and the bridging therapy is administered during that period.
  • the bridging therapy comprises gemcitabine-oxaliplatin plus rituximab (R-GemOx).
  • the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises R-GemOx during that time period. In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises a portion of R-GemOx during that time period. In some embodiments, the bridging therapy comprises R-polatuzumab vedotin. In some embodiments, the bridging therapy comprises a portion of R-polatuzumab vedotin.
  • the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises R-polatuzumab vedotin during that time period. In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises a portion of R-polatuzumab vedotim during that time period.
  • a response to the CAR T-cell therapy described herein comprises radiographic evidence. In some embodiments, a response to the CAR T-cell therapy described herein comprises clinical evidence. In some embodiments, a response to the CAR T-cell therapy described herein comprises a combination of radiographic evidence and clinical evidence. In some embodiments, a response to CAR T-cell therapy is determined at least in part from a positron emission tomography (PET)-computed tomography (CT) scan. In some embodiments, a response to CAR T-cell therapy is determined at least in part from a contrast enhanced CT scan. In some embodiments, the PET-CT scan detects an uptake of a radiotracer compound.
  • PET positron emission tomography
  • CT contrast enhanced CT scan
  • the radiotracer compound is fluorodeoxy glucose .
  • a subject comprises a complete response to the CAR T-cell therapy. In some embodiments, a subject comprises a partial response to the CAR T- cell therapy. In some embodiments, a subject comprises no response to the CAR T-cell therapy. In some embodiments, a subject comprises progressive disease after receiving the CAR T- cell therapy.
  • the subject has a complete response to CAR T-cell therapy.
  • the complete response to CAR T-cell therapy comprises no new lesions detected from a PET-CT scan analysis.
  • the complete response to CAR T-cell therapy comprises no evidence of fluorodeoxyglucose-avid disease in bone marrow.
  • the complete response to CAR T-cell therapy comprises a score of 1, 2 or 3 on a five point scale from a PET-CT scan analysis.
  • the score of 1, 2, or 3 on a five point scale from a PET-CT scan comprises one or residual masses.
  • the score of 1, 2, or 3 on a five point scale from a PET-CT scan comprises no or residual mass.
  • the uptake of fluorodeoxyglucose may be higher than normal.
  • the uptake of fluorodeoxy glucose may be higher than normal/baseline in mediastinum.
  • the uptake of fluorodeoxyglucose may be higher than normal/baseline in liver.
  • the uptake of fluorodeoxyglucose in sites of disease involvement may be compared to sites proximal to disease involved sites to infer the response to CAR T-cell therapy.
  • the uptake of fluorodeoxyglucose in sites of disease involvement may be at a similar level compared to sites proximal to disease involvement.
  • the complete response to CAR T-cell therapy comprises no extra lymphatic sites of disease. In some embodiments, the complete response to CAR T-cell therapy comprises no extra lymphatic sites of disease according to a CT scan analysis.
  • the complete response to CAR T-cell therapy comprises target nodes and/or nodal masses of less than about 0.5-3.0 cm in the longest diameter of a lesion (LDi), about 1-2 cm in LDi, about 0.5 cm in LDi, about 1.0 cm in LDi, about 1.5 cm in LDi, about 2.0 cm in LDi, about 2.5 cm in LDi, or about 3.0 cm in LDi. In some embodiments, the complete response to CAR T-cell therapy comprises target nodes and/or nodal masses of less than about 1.5 cm.
  • the subject comprises a partial response to CAR T-cell therapy.
  • the partial response to CAR T-cell therapy comprises a partial metabolic response score of 4 or 5.
  • the complete response to CAR T-cell therapy comprises reduced fluorodeoxyglucose uptake compared to baseline and residual mass or masses of any size based on a PET-CT scan.
  • the partial response reflects disease responding to treatment.
  • the partial response comprises residual fluorodeoxyglucose uptake in new bone marrow lesions higher than fluorodeoxyglucose uptake in normal bone marrow.
  • the partial response reflects residual disease.
  • the partial response is measured mid-treatment and reflects responding disease. In some embodiments, the partial response is measured at the end of treatment and indicates residual disease. In some embodiments, the partial response comprises a decrease of about 50% or greater in a sum of the product of the perpendicular diameters for multiple lesions determined by a CT-based analysis. In some embodiments, the partial response comprises a decrease of about 50% or greater in a sum of the product of the perpendicular diameters for up to 6 target measurable nodes and extranodal sites determined by a CT-based analysis.
  • the subject comprises no response to CAR T-cell therapy.
  • the no response comprises no metabolic response, and/or a score of 4 or 5 on a five-point scale with no significant change in fluorodeoxyglucose uptake from baseline at interim or end of treatment based on a PET-CT analysis.
  • the no response comprises less than about 50% decrease from baseline in a sum of the product of the perpendicular diameters for multiple lesions of up to 6 dominant, measurable nodes and extranodal sites based a on CT-scan analysis.
  • the no response comprises no criteria for disease progression being met.
  • the subject comprises progressive disease after receiving a CAR T-cell therapy.
  • the progressive disease comprises progressive metabolic disease based on a PET-CT scan.
  • the progressive disease comprises a score of 4 or 5 on a five-point scale with an increase in intensity of fluorodeoxyglucose uptake compared to baseline based on a PET-CT scan, or new fluorodeoxyglucose-avid foci consistent with lymphoma at interim or end of treatment assessment based on a PET-CT scan, or a combination thereof.
  • the progressive disease comprises progression in the cross product of the LDi and perpendicular diameter, new or recurrent involvement or regrowth of previously resolved lesions, a new node of greater than about 1.5 cm in any axis, a new extranodal site of greater than about 1 cm in any axis, assessable disease of any size attributed to lymphoma, new or recurrent involvement from bone marrow, or a combination thereof.
  • a node or lesion comprises an LDi of greater than about 1.5 cm, an increase of greater than about 50% compared to a cross product of the LDi and perpendicular diameter nadir and an increase in LDi or shortest diameter of a lesion (SDi), an increase in LDi or SDi of about 0.5 or greater for lesions of less than about 2 cm, an increase in LDi or SDi or about 1.0 or greater for lesions of more than about 2 cm, new or recurrent splenomegaly, new or clear progression of preexisting non-measured lesions, or some combination thereof.
  • FIG. 4 depicts the study design including subjects in Cohort 4. Subjects in Cohort 4 are treated with a target dose of IxlO 6 transduced CAR T-cells/kg targeting CD22. Participants in this cohort will not have received prior CAR T therapy (z.e., they are CAR- naive) and may have received prior bispecific T-cell engager antibody therapy.
  • Patient T-cells obtained from autologous apheresis product are transduced with the anti-CD22 lentiviral vector, expanded in vitro and then frozen for future administration.
  • Patients are given conditioning chemotherapy prior to CAR T-cell infusion with the intent of lymphodepletion.
  • Conditioning chemotherapy consists of fludarabine and cyclophosphamide as described further below. The chemotherapy is planned so that the last dose is completed 3 days prior to administration of the CAR T-cells. Alternatively, the CAR T-cells are administered 2 to 7 days after the last dose of conditioning chemotherapy.
  • the patient is, e.g., be re-treated with lymphodepleting chemotherapy prior to CAR T-cell infusion.
  • Subjects must meet certain fitness criteria to receive the CAR T-cell infusion. Subjects will be hospitalized for the product administration.
  • the CAR T-cell infusion is administered as a single infusion at a target dose of 0.5xl0 6 CAR transduced T-cells/kg based upon the subject’s weight at the time of enrollment.
  • the CAR T-cell infusion is administered as a single infusion at a target dose of l.OxlO 6 CAR transduced T-cells/kg based upon the subject’s weight at the time of enrollment.
  • Diffuse LBCL (DLBCL) not otherwise specified (NOS), including germinal center B-cell (GCB) type or active B-cell (ABC) type;
  • DLBCL arising from follicular lymphoma (FL; transformed follicular lymphoma);
  • PMBL Primary mediastinal large B-cell lymphoma
  • Central nervous system (CNS) prophylaxis e.g., intrathecal methotrexate: must be stopped at least 1 week prior to enrollment on the current study;
  • ANC Eastern Cooperative Oncology Group
  • PD-EPI Chronic Kidney Disease Epidemiology Collaboration
  • ALT serum alanine aminotransferase
  • AST aspartate aminotransferase
  • UPN upper limit of normal
  • f total bilirubin ⁇ 1.5 mg/dL (except in patients with Gilbert syndrome)
  • LVEF cardiac left ventricular ejection fraction
  • SaO2 blood oxygen saturation
  • serum albumin > 2.5 g/dL
  • patients must have previously received a CD19-directed CAR T-cell therapy. Patients may have received additional therapies after a CD19-directed CAR T-cell therapy. Subjects part of Cohort 1 must have previously received a CD19-directed CAR T- cell therapy. Additional therapies after the CD19-directed CAR T-cell therapy are permissible. Subjects have no prior exposure to bispecific T-cell engaging antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®), etc).
  • bispecific T-cell engaging antibody therapy e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®), etc.
  • patients must have received at least two prior lines of therapy with one regimen including a bispecific T-cell engager antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®).
  • a bispecific T-cell engager antibody therapy e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®).
  • a bispecific T-cell engager antibody therapy e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®.
  • they must meet one of the following conditions: a. Relapsed disease following a CR or progressive disease following a PR to bispecific T-cell engager antibody therapy
  • Subjects part of Cohort 3 must have received at least two prior lines of therapy with one regimen including a bispecific T-cell engager antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®)). In addition, they must meet the following conditions:
  • (ii) can be refractory to prior CIT or relapsed after PR or CR
  • prior bispecific T-cell engaging antibody therapy e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®)
  • mosunetuzumab marketed as Lunsumio®
  • glofitamab marketed as Columvi®
  • epcoritimab marketed as Epkinly®
  • a subject having relapsed or refractory LBCL will be ineligible to be included in the study if they meet all of the following criteria:
  • subjects who received prior bispecific T-cell engager antibody therapy may not be enrolled in cohort 1;
  • HAV human immunodeficiency virus
  • HBV hepatitis B virus
  • HbcAb hepatitis B core antibody
  • HCV hepatitis C virus
  • liver disease including cirrhosis
  • neurological disease such as cerebrovascular ischemia/hemorrhage, dementia, seizure disorder, or cerebellar disease
  • subjects with history of immune thrombocytopenic purpura or autoimmune hemolytic anemia not requiring active treatment may be eligible.
  • any in-dwelling line or drain e.g., percutaneous nephrostomy tube, in dwelling Foley catheter, biliary drain
  • Dedicated central venous access catheters such as a Port-a-Cath or Hickman catheter, are permitted
  • the subject is administered lymphodepleting chemotherapy, e.g., fludarabine (30 mg/m 2 /dayx3 days) and cyclophosphamide (500 mg/m 2 /dayx3 days).
  • the chemotherapy is planned so that the last dose is completed 2-4 days prior to infusion of the CAR T-cells.
  • Subjects may receive allopurinol orally (300 mg) once daily or at 100 mg/m 2 /dose two to three times in a day beginning the day prior to cell infusion or at the time of lymphodepletion.
  • Subjects receive diphenhydramine (25-50 mg/dose) or another Hl antihistamine.
  • Participants receive anti-CD22 CAR T-cell treatment consisting of a single infusion of CD22 CAR transduced autologous T cells administered intravenously initially at a target dose l.OxlO 6 CAR-transduced T-cells/kg.
  • the cell dose is based upon the weight at time of enrollment.
  • the dose of anti-CD22 CAR T-cells is calculated using actual body weight, capped at 100 kg. After a preliminary safety assessment, a lower dose of 0.5xl0 6 CAR- transduced T-cells/kg may be evaluated.
  • a preliminary safety assessment is performed after the first 10 patients in Cohort 1 are treated at the target dose of CRG-022 and have been followed for at least 28 days after CRG- 022 infusion. The study will continue to enroll patients during the preliminary safety assessment.
  • Example 2 Phase 1 Study of Autologous Anti-CD22 Chimeric Antigen Receptor T Cells in patients with Relapsed or Refractory Large B Cell Lymphoma
  • This example describes a phase 1, single-center Phase 1, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B-cell antigen CD22 in participants with R/R LBCL who had received previous lines of therapy. This example informed the starting dose for the Phase 2 study.
  • a cohort of 38 subjects were treated with transduced CAR T-cells targeting CD22 as part of this study. Two different dosing regimens were used for different subsets of subjects: either IxlO 6 CAR T-cell s/kg (29 subjects) or 3xl0 6 cells/kg (9 subjects).
  • cytokine release syndrome CRS
  • FIG. 3 The summary of results related to CRS are shown in FIG. 3.
  • a second type of toxicity associated with CAR T-cell therapies is immune effector cell-associated neurotoxicity syndrome (ICANS).
  • ICANS immune effector cell-associated neurotoxicity syndrome

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Provided herein are compositions and methods for treating cancer, e.g., hematological cancer, by administering a CD22 CAR-expressing cell described herein according to a regimen described herein. Also disclosed are methods of making and compositions comprising the same.

Description

ANTI-CD22 CHIMERIC ANTIGEN RECEPTOR (CAR) THERAPIES
CROSS-REFERENCE TO RELATED APPLICATION
[0001] The present application claims priority to U.S. Provisional Patent Application Serial No. 63/536,298, filed on September 1, 2023. The contents of the above-referenced application is herein expressly incorporated by reference in its entirety, including any drawings.
INCORPORATION BY REFERENCE
[0002] All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
BACKGROUND
[0003] Many patients with B cell malignancies are incurable with standard therapy and traditional treatment options often have serious side effects. Although advances have been made in developing new therapeutic modalities comprising targeted cancer immunotherapies in recent years, including bi-specific T-cell engager antibodies (BiTEs) and chimeric antigen receptor (CAR) T-cell therapies, these targeted therapies are not effective in all patients, even when they are effective patients frequently relapse for a variety of reasons including loss of tumor antigen expression, and it remains unclear how to achieve clinical effectiveness in blood cancer patients more broadly. At least one explanation for the unpredictable efficacy of these targeted immunotherapies is thought to be that the tumor antigens they target are typically derived from self and poorly immunogenic; in addition, tumors are sophisticated adversaries that have developed diverse escape mechanisms to evade detection and elimination by the patient’s immune system.
[0004] Recent developments using autologous CAR T-cell therapies show promising results in treating B cell malignancies. Besides the ability for the chimeric antigen receptor on the genetically modified T-cells to recognize and destroy the targeted cells, a successful therapeutic T-cell therapy can proliferate and persist over time, offering extended protection as a “living drug” of sorts and helping patients to avoid relapsed/refractory disease (ie., avoiding recurrence of the disease). Subjects with relapsed/refractory (R/R) large B-cell lymphoma (LBCL) often experience disease progression after receiving treatment because they become resistant to treatment, primarily by down-regulating or eliminating expression of the targeted tumor antigen or of one or more ancillary genes essential for T-cell activation and expansion. There is a need for more effective therapies that overcome treatment resistance and effectively treat the underlying disease. There is a clinical need for effective CAR T-cell therapies that can treat relap sed/refractory B cell malignancies. There is also a clinical need for effective CAR T-cell therapies that can overcome the variable quality of T cells used to manufacture autologous CAR T-cells, resulting from anergy, suppression, or exhaustion, which skilled practitioners have limited control over at this time.
SUMMARY
[0005] The present disclosure relates generally to the use of autologous T-cells engineered to express a CAR with an anti-CD22 binding domain, to treat a disease, e.g., a hematological cancer. In some cases, the hematological cancer is a relapsed/refractory hematological cancer that recurs following treatment and a period of remission and/or that does not respond to treatment. In some cases, the relapsed/refractory hematological cancer recurs after treatment with standard of care chemotherapy or a CD19/CD3 bi-specific T-cell engager (BiTE).
[0006] Provided herein are methods of treating a relapsed/refractory (R/R) hematological cancer in a human subject in need thereof, the methods comprising: administering to the human subject a therapeutically effective dose of autologous CAR T-cells to treat the R/R hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an anti-CD22 binding domain and (B) a portion of a CD8a extracellular domain (z.e., a CD8a hinge domain), a CD8a transmembrane domain; a 4-1BB intracellular costimulatory signaling domain; and a CD3-zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy, and/or wherein the at least one prior line of therapy comprises a bispecific T-cell engager (BiTE®) antibody therapy.
[0007] In some embodiments, the dose of autologous CAR T-cells is less than about 0.8* 106 engineered CAR T-cells/kg.
[0008] In some embodiments, the dose of autologous CAR T-cells is at least about 1.2* 106 CAR T-cells/kg.
[0009] Also provided herein are methods of treating a hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of autologous CAR T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an anti-CD22 binding domain and (B) a portion of a CD8a extracellular domain (z.e., a CD8a hinge domain), a CD8a transmembrane domain; a 4-1BB costimulatory domain; and a CD3zeta intracellular signaling domain; wherein the dose of engineered CAR T-cells is less than about 0.8* 106 CAR T-cells/kg, or at least about 1.2* 106 CAR T-cells/kg.
[0010] In some embodiments, the hematological cancer is a relapsed/refractory hematological cancer.
[0011] In some embodiments, the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy.
[0012] In some embodiments, the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager (BiTE®) antibody therapy.
[0013] Also provided herein are methods of treating a hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a dose of cyclophosphamide at about 500 mg/m2/day to about 600 mg/m2/day, wherein the dose of cyclophosphamide is administered daily for three days, and a dose of fludarabine at about 30 mg/m2/day; and administering to the human subject a therapeutically effective dose of autologous CAR T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an anti-CD22 binding domain and (B) a portion of a CD8a extracellular domain, a CD8a transmembrane domain; a 4- IBB costimulatory domain; and a CD3zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
[0014] In some embodiments, the human subject has received 2 or more prior lines of therapy.
[0015] In some embodiments, the human subject has received 2 or more prior lines of therapy, wherein at least one of the 2 or more prior lines of therapy comprises a bispecific T- cell engager antibody therapy.
[0016] In some embodiments, the human subject has received no more than 5 prior lines of therapy.
[0017] In some embodiments, the human subject has not received a prior CAR T-cell therapy.
[0018] In some embodiments, the human subject has received a prior CAR T-cell therapy. [0019] In some embodiments, the human subject has received a prior CAR T-cell therapy and at least one additional therapy. [0020] In some embodiments, the prior CAR T-cell therapy is tisagenlecleucel (marketed as Kymriah®), brexucabtagene autoleucel (marketed as Tecartus®), axicabtagene cilocleucel (marketed as Yescarta®), lisocabtagene maraleucel (marketed as Breyanzi®) or any combination thereof.
[0021] In some embodiments, the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises an anti-CD19 antibody, an anti-CD20 antibody, and anti-CD22 antibody or an anti-CD79b antibody.
[0022] In some embodiments, at least one prior line of therapy comprises an anti-CD19 antibody-drug conjugate, an anti-CD20 antibody-drug conjugate, an anti-CD22 antibody-drug conjugate, or an anti-CD79b antibody-drug conjugate.
[0023] In some embodiments, the anti-CD19 antibody comprises inebilizumab (marketed as Uplizna®), loncastuximab tesirine (marketed as Zynlonta®), or tafasitamab (marketed as Monjuvi®).
[0024] In some embodiments, the anti-CD20 antibody comprises rituximab (marketed as Rituxan®), ibritumomab tiuxetan (marketed as Zevalin®), tositumomab (marketed as Bexxar®), ocrelizumab (Ocrevus®), ofatumumab (marketed as Kesimpta®), mosunetuzumab (marketed as Lunsumio™), Obinutuzumab (marketed as Gazyva®), or ublituximab (marketed as Briumvi®).
[0025] In some embodiments, the anti-CD22 antibody comprises moxetumomab pasudotox (marketed as Lumoxiti®), inotuzumab ozogamicin (maketed as Besponsa®), or epratuzumab. [0026] In some embodiments, the anti-CD79b antibody comprises polatuzumab vedotin (marketed as Polivy®).
[0027] In some embodiments, the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
[0028] In some embodiments, the bispecific T-cell engager antibody therapy comprises a T cell engager comprising an anti-CD3 domain.
[0029] In some embodiments, the bispecific T-cell engaging antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, elranatamab, teclistamab, tarlatamab, talquetamab, solitomab, tebentafusp, duvortuxizumab, AFM11, REGN5458, CC- 93269, PF-06863135, TNB-383B, pdronextamab, XmAbl3676, IGM-2323, AMG 330, AMG 673, AMG 420, AMG 701, cevostamab, talquetamab, flotetuzumab, or a combination thereof. [0030] In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain and an anti-CD3 domain.
[0031] In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain and an anti-CD3 domain.
[0032] In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-BCMA domaim. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-BCMA domain and an anti-CD3 domain.
[0033] In some embodiments, the anti-CD22 binding domain is an scFv.
[0034] In some embodiments, the anti-CD22 binding domain is an antibody or an antigen binding domain of thereof.
[0035] In some embodiments, the anti-CD22 binding domain comprises the sequence:
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIK.
[0036] In some embodiments, the CAR comprises a linker between the anti-CD22 binding domain and a portion of a CD8a extracellular domain (z.e., a CD8a hinge domain).
[0037] In some embodiments, the linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain (z.e., a CD8a hinge domain) comprises the sequence AAA.
[0038] In some embodiments, the portion of a CD8a extracellular domain (z.e., a CD8a hinge domain) comprises the sequence:
TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD.
[0039] In some embodiments, the portion of a CD8a transmembrane domain comprises the sequence: IYIWAPLAGTCGVLLLSLVIT.
[0040] In some embodiments, the CAR comprises a portion of a CD8a intracellular domain.
[0041] In some embodiments, the portion of a CD8a intracellular domain comprises the sequence LYC.
[0042] In some embodiments, the 4- IBB costimulatory domain comprises the sequence: KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL. [0043] In some embodiments, the CD3zeta intracellular signaling domain comprises the sequence:
RVI<FSRSADAPAYI<QGQNQLYNELNLGRREEYDVLDI<RRGRDPEMGGI<PRRI<NPQ EGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQAL PPR.
[0044] In some embodiments, the CAR comprises a signal peptide sequence.
[0045] In some embodiments, the signal peptide sequence is derived from granulocyte macrophage colony stimulating factor receptor alpha (GMCSFRa) and comprises the sequence: MLLLVTSLLLCELPHPAFLLIP.
[0046] In some embodiments, the CAR comprises the sequence:
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIW APLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEG GCELRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRR KNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGH-
DGLYQGLSTATKDTYDALHMQALPPR
[0047] In some embodiments, the CAR comprises the sequence:
MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVT ITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQ AEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACR PAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREE YDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKG H-DGLYQGLSTATKDTYDALHMQALPPR.
[0048] In some embodiments, the hematological cancer expresses CD22.
[0049] In some embodiments, the hematological cancer is Large B-Cell Lymphoma (LBCL).
[0050] In some embodiments, the LBCL is diffuse LBCL (DLBCL).
[0051] In some embodiments, the LBCL has been histologically confirmed. [0052] In some embodiments, the LBCL is (DLBCL) not otherwise specified (NOS).
[0053] In some embodiments, the DLBCL-NOS is germinal center B-cell (GCB) type or active B-cell (ABC) type.
[0054] In some embodiments, the LBCL is high-grade B-cell lymphoma with or without MYC and BCL2 and/or BCL6 rearrangements.
[0055] In some embodiments, the LBCL is DLBCL associated with chronic inflammation.
[0056] In some embodiments, the LBCL is primary cutaneous DLBCL, leg type.
[0057] In some embodiments, the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS.
[0058] In some embodiments, the LBCL is DLBCL arising from follicular lymphoma (FL).
[0059] In some embodiments, the LBCL is DLBCL arising from marginal zone lymphoma (MZL).
[0060] In some embodiments, the LBCL is primary mediastinal (thymic) large B-cell lymphoma.
[0061] In some embodiments, the LBCL is Grade 3B FL.
[0062] In some embodiments, the human subject is 18 years of age or older.
[0063] In some embodiments, the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of progressive disease (PD) or stable disease (SD) after the last therapy.
[0064] In some embodiments, the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of disease progression after partial response (PR) or complete response (CR) after the last therapy.
[0065] In some embodiments, the human subject has relapsed/refractory disease after a last therapy based on presence of viable lymphoma in a biopsy obtained after therapy.
[0066] In some embodiments, the human subject has relapsed disease following a complete response (CR) or progressive disease following a PR to a bispecific T-cell engager antibody therapy.
[0067] In some embodiments, the human subject has progressive disease (PD) or stable disease (SD) as best response to a bispecific T-cell engager antibody therapy.
[0068] In some embodiments, the dose of engineered CAR T-cells is from about O.l x lO6 to about 4* 106 engineered CAR T-cells/kg.
[0069] In some embodiments, the dose of engineered CAR T-cells is from about O.l x lO6 to about 0.75x l06 engineered CAR T-cells/kg. [0070] In some embodiments, the dose of engineered CAR T-cells is from about 0.25* 106 to about 0.75* 106 engineered CAR T-cells/kg.
[0071] In some embodiments, the dose of engineered CAR T-cells is from about 1.2* 106 to about 4* 106 engineered CAR T-cells/kg.
[0072] In some embodiments, the dose of engineered CAR T-cells is from about 0.8* 106 to about 1.2* 106 engineered CAR T-cells/kg.
[0073] In some embodiments, the dose of engineered CAR T-cells is from about 2.5* 106 to about 3.5* 106 engineered CAR T-cells/kg.
[0074] In some embodiments, the dose of engineered CAR T-cells is about 0.5* 106 engineered CAR T-cells/kg.
[0075] In some embodiments, the dose of engineered CAR T-cells is about l >< 106 engineered CAR T-cells/kg.
[0076] In some embodiments, the dose of engineered CAR T-cells is about 3-2* 106 engineered CAR T-cells/kg.
[0077] In some embodiments, the dose of engineered CAR T-cells is about 3* 106 engineered CAR T-cells/kg.
[0078] In some embodiments, the method comprises administering to the human subject a dose of cyclophosphamide prior to administering the engineered CAR T-cells.
[0079] In some embodiments, the dose of cyclophosphamide is about 400 mg/m2 to about 600 mg/m2.
[0080] In some embodiments, the dose of cyclophosphamide is about 500 mg/m2.
[0081] In some embodiments, the dose of cyclophosphamide is administered daily for at least 2 days.
[0082] In some embodiments, the dose of cyclophosphamide is administered daily for 3 days. [0083] In some embodiments, the dose of cyclophosphamide is 500 mg/m2 administered once daily for 3 days.
[0084] In some embodiments, the method comprises administering to the human subject a dose of fludarabine prior to administering the engineered CAR T-cells.
[0085] In some embodiments, the dose of fludarabine is about 20 mg/m2 to about 400 mg/m2.
[0086] In some embodiments, the dose of fludarabine is about 25 mg/m2 to about 35 mg/m2.
[0087] In some embodiments, the dose of fludarabine is about 30 mg/m2.
[0088] In some embodiments, the dose of fludarabine is administered daily for at least 2 days. [0089] In some embodiments, the dose of fludarabine is administered daily for three days. [0090] In some embodiments, the dose of fludarabine is 30 mg/m2 administered once daily for 3 days.
[0091] In some embodiments, the dose of cyclophosphamide is administered daily for three days and the dose of fludarabine is administered daily for three days.
[0092] In some embodiments, at least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine.
[0093] In some embodiments, a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine.
[0094] In some embodiments, a second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine.
[0095] In some embodiments, a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine.
[0096] In some embodiments, each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered.
[0097] As used herein, the term “day 0” refers to the day of CAR T infusion, the term “day - 1” refers to the day 1 day before CAR T infusion, the term “day -2” refers to the day 2 days before CAR T infusion, the term “day -3” refers to the day 3 days before CAR T infusion, the term “day -4” refers to the day 4 days before CAR T infusion, and the term “day -5” refers to the day 5 days before CAR T infusion.
[0098] In some embodiments, a dose of cyclophosphamide is administered on day -5 and day -4 and day -3 prior to CAR T infusion, and the engineered CAR T-cells are administered on day 0.
[0099] In some embodiments, a dose of fludarabine is administered on day -5 and day -4 and day -3 prior to CAR T infusion, and the engineered CAR T-cells are administered on day 0. [0100] In some embodiments, a dose of cyclophosphamide is administered at 500 mg/m2 and a dose of fludarabine is administered at 30 mg/m2, each administered once daily starting on day -5 for three days (z.e., day -5, day -4, day -3), followed by two days with no chemotherapy, and the engineered CAR T-cells are administered on day 0.
[0101] Also provided herein is a method of treating a relapsed/refractory Large B-Cell Lymphoma (LBCL) in a human subject in need thereof, the method comprising: administering to the human subject a dose of about 400 mg/m2 to about 600 mg/m2 cyclophosphamide on day -5, day -4 and day -3, and a dose of about 25 mg/m2 to about 35 mg/m2 fludarabine on day -5, day -4 and day -3; administering to the human subject a dose of from about 0.1 x 106 to about 4* 106 engineered chimeric antigen receptor (CAR) T-cells/kg on day 0; wherein the CAR comprises the sequence: MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVT ITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQ AEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACR PAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREE YDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKG HDGLYQGLSTATKDTYDALHMQALPPR; wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises an anti- CD19 CAR T-cell therapy, a bispecific T-cell engager antibody therapy, or a combination thereof.
[0102] Also provided herein is a method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of autologous CD22-specific chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain, a CD8a transmembrane domain; a 4- IBB costimulatory domain; and a CD3zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy, and/or wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
[0103] In some embodiments, the dose of autologous CD22-specific CAR T-cells is less than about 0.8* 106 autologous CD22-specific CAR T-cells/kg.
[0104] In some embodiments, the dose of engineered CAR T-cells is at least about 1.2* 106 CAR T-cells/kg.
[0105] Also provided herein is a method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of engineered chimeric antigen receptor (CAR) T- cells to treat the hematological cancer in the human subject; wherein the CAR comprises (i) an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain, (ii) a CD8a transmembrane domain; (iii) a 4-1BB costimulatory domain; and (iv) a CD3zeta intracellular signaling domain; wherein the dose of autologous CD22-specific CAR T-cells is less than about 0.8* 106 autologous CD22-specific CAR T-cells/kg, or at least about 1.2* 106 autologous CD22-specific CAR T-cells/kg.
[0106] In some embodiments, the hematological cancer is a relapsed/refractory hematological cancer.
[0107] In some embodiments, the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy.
[0108] In some embodiments, the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
[0109] Also provided herein is a method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a dose of cyclophosphamide at about 500 mg/m2/day to about 600 mg/m2/day, wherein the dose of cyclophosphamide is administered daily for three days, and a dose of fludarabine at about 30 mg/m2/day, wherein the dose of fludarabine is administered daily for three days; and administering to the human subject a therapeutically effective dose of engineered chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises an extracellular domain comprising (A) an antihuman CD22 binding domain and (B) a portion of a CD8a extracellular domain, a CD8a transmembrane domain; a 4- IBB costimulatory domain; and a CD3zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
[0110] In some embodiments, the human subject has received 2 or more prior lines of therapy.
[OHl] In some embodiments, the human subject has received 2 or more prior lines of therapy, wherein at least one of the 2 or more prior lines of therapy a bispecific T-cell engager antibody therapy.
[0112] In some embodiments, the human subject has received no more than 5 prior lines of therapy. [0113] In some embodiments, the human subject has not received a prior CAR T-cell therapy.
[0114] In some embodiments, the human subject has received a prior CAR T-cell therapy. [0115] In some embodiments, the human subject has received a prior CAR T-cell therapy and at least one additional therapy.
[0116] In some embodiments, the prior CAR T-cell therapy is tisagenlecleucel, brexucabtagene autoleucel, axicabtagene cilocleucel, aisocabtagene maraleucel or any combination thereof.
[0117] In some embodiments, the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises an anti-CD19 antibody, an anti-CD20 antibody, and anti-CD22 antibody or an anti-CD79b antibody.
[0118] In some embodiments, the anti-CD19 antibody, anti-CD20 antibody, anti-CD22 antibody or anti-CD79b antibody comprises an antibody-drug conjugate.
[0119] In some embodiments, the anti-CD19 antibody comprises inebilizumab, loncastuximab tesirine or tafasitamab.
[0120] In some embodiments, the anti-CD20 antibody comprises rituximab, ibritumomab tiuxetan, tositumomab, ocrelizumab, ofatumumab, mosunetuzumab, obinutuzumab, or ublituximab.
[0121] In some embodiments, the anti-CD22 antibody comprises inotuzumab ozogamicin, or epratuzumab.
[0122] In some embodiments, the anti-CD79b antibody comprises polatuzumab vedotin. [0123] In some embodiments, the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy. In some embodiments, the bispecific T-cell engager antibody therapy comprises a T cell engager comprising an anti-CD3 domain. In some embodiments, the bispecific T-cell engager antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, solitomab, duvortuxizumab, AFM11, REGN5458, CC-93269, TNB-383B, pdronextamab, XmAbl3676, IGM-2323, or cevostamab. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain. In some embodiments, the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain.
[0124] In some embodiments, the anti-CD22 binding domain is an scFv. [0125] In some embodiments, the anti-CD22 binding domain is an antibody or an antigen binding domain of thereof.
[0126] In some embodiments, the anti-CD22 binding domain comprises the sequence: QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIK (SEQ ID NO: 1).
[0127] In some embodiments, the CAR comprises a linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain.
[0128] In some embodiments, the linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain comprises the sequence AAA.
[0129] In some embodiments, wherein the portion of a CD8a extracellular domain comprises the sequence TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO: 2).
[0130] In some embodiments, the portion of a CD8a transmembrane domain comprises the sequence IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO:4).
[0131] In some embodiments, the CAR comprises a portion of a CD8a intracellular domain.
[0132] In some embodiments, the portion of a CD8a intracellular domain comprises the sequence LYC.
[0133] In some embodiments, the 4- IBB costimulatory domain comprises the sequence KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO:6).
[0134] In some embodiments, the CD3zeta intracellular signaling domain comprises the sequence:
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQ EGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQAL PPR (SEQ ID NO: 7).
[0135] In some embodiments, the CAR comprises a signal peptide sequence.
[0136] In some embodiments, the signal peptide sequence comprises the sequence: MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 8).
[0137] In some embodiments, the CAR comprises the sequence:
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIW APLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEG GCELRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRR KNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALH MQALPPR (SEQ ID NO: 12).
[0138] In some embodiments, the CAR comprises the sequence:
MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVT ITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQ AEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACR PAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREE YDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKG HDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO:9).
[0139] In some embodiments, the hematological cancer expresses CD22.
[0140] In some embodiments, the hematological cancer is Large B-Cell Lymphoma (LBCL).
[0141] In some embodiments, the LBCL is diffuse LBCL (DLBCL).
[0142] In some embodiments, the LBCL has been histologically confirmed.
[0143] In some embodiments, the LBCL is (DLBCL) not otherwise specified (NOS).
[0144] In some embodiments, the DLBCL-NOS is germinal center B-cell (GCB) type or active B-cell (ABC) type.
[0145] In some embodiments, the LBCL is high-grade B-cell lymphoma with or without MYC and BCL2 and/or BCL6 rearrangements.
[0146] In some embodiments, the LBCL is DLBCL associated with chronic inflammation.
[0147] In some embodiments, the LBCL is primary cutaneous DLBCL, leg type.
[0148] In some embodiments, the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS.
[0149] In some embodiments, the LBCL is DLBCL arising from follicular lymphoma (FL). [0150] In some embodiments, the LBCL is DLBCL arising from marginal zone lymphoma (MZL). [0151] In some embodiments, the LBCL is primary mediastinal (thymic) large B-cell lymphoma.
[0152] In some embodiments, the LBCL is Grade 3B FL.
[0153] In some embodiments, the human subject is at least 18 years of age.
[0154] In some embodiments, the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of progressive disease (PD) or stable disease (SD) after the last therapy.
[0155] In some embodiments, the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of disease progression after partial response (PR) or complete response (CR) after the last therapy.
[0156] In some embodiments, the human subject has relapsed/refractory disease after a last therapy based on presence of viable lymphoma in a biopsy obtained after therapy.
[0157] In some embodiments, the human subject has relapsed disease following a CR or progressive disease following a PR to a bispecific T-cell engager antibody therapy.
[0158] In some embodiments, the human subject has progressive disease (PD) or stable disease (SD) as best response to a bispecific T-cell engager antibody therapy.
[0159] In some embodiments, the dose of engineered CAR T-cells is from about O.l x lO6 to about 4* 106 engineered CAR T-cells/kg.
[0160] In some embodiments, the dose of engineered CAR T-cells is from about O.l x lO6 to about 0.75x l06 engineered CAR T-cells/kg.
[0161] In some embodiments, the dose of engineered CAR T-cells is from about 0.25x l06 to about 0.75x l06 engineered CAR T-cells/kg.
[0162] In some embodiments, the dose of engineered CAR T-cells is from about 1.2x l06 to about 4x l06 engineered CAR T-cells/kg.
[0163] In some embodiments, the dose of engineered CAR T-cells is from about 0.8x l06 to about 1.2x l06 engineered CAR T-cells/kg.
[0164] In some embodiments, the dose of engineered CAR T-cells is from about 2.5x l06 to about 3.5x l06 engineered CAR T-cells/kg.
[0165] In some embodiments, the dose of engineered CAR T-cells is about 0.5x l06 engineered CAR T-cells/kg.
[0166] In some embodiments, the dose of engineered CAR T-cells is about I x lO6 engineered CAR-T cells/kg. [0167] In some embodiments, the dose of engineered CAR T-cells is about 2* 106 engineered CAR-T cells/kg.
[0168] In some embodiments, the dose of engineered CAR T-cells is about 3* 106 engineered CAR T-cells/kg.
[0169] In some embodiments, the method comprises administering to the human subject a dose of cyclophosphamide prior to administering the engineered CAR T-cells.
[0170] In some embodiments, the dose of cyclophosphamide is about 400 mg/m2 to about 600 mg/m2.
[0171] In some embodiments, the dose of cyclophosphamide is about 500 mg/m2.
[0172] In some embodiments, the dose of cyclophosphamide is administered daily for at least 2 days.
[0173] In some embodiments, the dose of cyclophosphamide is administered daily for 3 days.
[0174] In some embodiments, the method comprises administering to the human subject a dose of fludarabine prior to administering the engineered CAR T-cells.
[0175] In some embodiments, the dose of fludarabine is about 20 mg/m2 to about 400 mg/m2.
[0176] In some embodiments, the dose of fludarabine is about 25 mg/m2 to about 35 mg/m2.
[0177] In some embodiments, the dose of fludarabine is about 30 mg/m2.
[0178] In some embodiments, the dose of fludarabine is administered daily for at least 2 days.
[0179] In some embodiments, the dose of fludarabine is administered daily for three days.
[0180] In some embodiments, the dose of cyclophosphamide is administered daily for three days and the dose of fludarabine is administered daily for three days.
[0181] In some embodiments, at least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine.
[0182] In some embodiments, a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine.
[0183] In some embodiments, a second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine.
[0184] In some embodiments, a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine.
[0185] In some embodiments, each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered. [0186] In some embodiments, a dose of cyclophosphamide is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells are administered on day 0.
[0187] In some embodiments, a dose of fludarabine is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells are administered on day 0.
[0188] In some embodiments, a dose of cyclophosphamide is administered on day -5, day -4, and day -3, a dose of fludarabine is administered on day -5, day -4, and day -3, and the engineered CAR T-cells are administered on day 0.
[0189] Also provided herein is a method of treating a relapsed/refractory Large B-Cell Lymphoma (LBCL) in a human subject in need thereof, the method comprising administering to the human subject a dose of about 400 mg/m2 to about 600 mg/m2 cyclophosphamide on day -5, day -4 and day -3, and a dose of about 25 mg/m2 to about 35 mg/m2 fludarabine on day -5, day -4 and day -3; administering to the human subject a dose of from about 0.1 * 106 to about 4* 106 engineered chimeric antigen receptor (CAR) T-cells/kg on day 0; wherein the CAR comprises the sequence:
MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVT ITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQ AEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACR PAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREE YDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKG HDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 9); wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises (A) an anti-CD19 CAR T-cell therapy, (B) a bispecific T-cell engager antibody therapy, or (C) a combination thereof.
BRIEF DESCRIPTION OF THE DRAWINGS
[0190] The novel features of the disclosure are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present disclosure will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the disclosure are utilized, and the accompanying drawings of which: [0191] FIG. 1 shows the study design for a phase 2, multi-center Phase 2, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B- cell antigen CD22 in participants with R/R LBCL.
[0192] FIG. 2 shows progression free survival and overall survival of LBCL patients treated with CD22 CAR T-cell therapy.
[0193] FIG. 3 shows associated toxicities with our CD22 CAR T-cell therapy.
[0194] FIG. 4 shows an updated study design for a phase 2, multi-center Phase 2, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B-cell antigen CD22 in participants with R/R LBCL.
DETAILED DESCRIPTION
[0195] The disclosure provides a method of treating a hematological cancer, comprising administering autologous T-cells that express a CAR molecule that binds CD22. In some embodiments, the hematological cancer is lymphoma. In some embodiments, the lymphoma is LBCL. In some embodiments the method of treating the hematological cancer comprises treating a subject who has received previous therapy. In some embodiments, the subject who has received previous therapy has relapsed or has refractory disease.
Terms and Definitions
[0196] Unless otherwise defined, all technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. [0197] As used herein, the singular forms “a,” “an,” and “the” include plural references unless the context clearly dictates otherwise. Any reference to “or” herein is intended to encompass “and/or” unless otherwise stated.
[0198] As used herein, the term “about” in some cases refers to an amount that is approximately the stated amount, an amount that is near the stated amount by 10%, 5%, or 1%, including increments therein, or an amount that is greater or less the stated percentage by 10%, 5%, or 1%, including increments therein.
Chimeric Antigen Receptor molecule
[0199] In some embodiments, an autologous T cell described herein comprises a CAR molecule that binds CD22 (a “CD22 CAR”). In some embodiments, the autologous T cell is an engineered T cell, e.g., autologous CAR T cell. In some embodiments, the autologous T cell is selected from the group consisting of CD4+ T cell, CD8+ T cell, regulatory T cell, and memory T cell. In some embodiments, the CD22 CAR comprises an extracellular domain, a transmembrane (TM) domain, and an intracellular domain. In some embodiments, the extracellular domain comprises an antigen-binding domain and a hinge domain. In some embodiments, the antigen-binding domain comprises a fully human anti-CD22 antibody. In some embodiments, the hinge domain comprises a hinge domain derived from CD8alpha or CD28. In some embodiments, TM domain comprises a TM domain derived from CD8alpha or CD28. In some embodiments, the intracellular domain comprises a co-stimulatory domain, and an immunoreceptor tyrosine-based activation motif (ITAM)-containing signaling domain. In some embodiments, the co-stimulatory domain is selected from the group consisting of 4-1BB, CD28. In some embodiments, the ITAM-containing signaling domain is derived from CD3zeta. In some embodiments, the CAR molecule comprises a signal peptide. In some embodiments, the CAR molecule comprises a linker. In some embodiments, the linker is connected to/disposed between the antigen binding domain and the additional extracellular domain sequence. In some embodiments, the CAR molecule comprises a signal peptide, an antigen binding domain (e.g., an scFv comprising a VH domain linked via a linker to a VL domain), a linker, an extracellular hinge domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain. In some embodiments, the linker that connects a VH domain to a VL domain and the linker that connects the antigen binding domain to the extracellular hinge domain are different.
[0200] In some embodiments, the CAR molecule comprises an anti-CD22 binding domain In some embodiments, the anti-CD22 binding domain is an antigen binding domain. In some embodiments, the antigen binding domain recognizes CD22.. In some embodiments, the anti- CD22 binding domain is an scFv. In some embodiments, the anti-CD22 binding domain is an antibody. In some embodiments, the anti-CD22 binding domain is a fully human antibody against human CD22. In some embodiments, the anti-CD22 is a single chain variable fragment (scFv) derived from a fully human antibody against human CD22. In some embodiments, the anti-CD22 scFv also comprises a linker disposed between the heavy chain variable fragment and the light chain variable fragment comprising the scFv. In some embodiments, the antigen binding domain is an scFv and comprises a sequence of SEQ ID NO: 1 :
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIK.
[0201] In some embodiments, the antigen binding domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 1. In some embodiments, the antigen binding domain comprises a sequence of SEQ ID NO: 1, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the antigen binding domain comprises a sequence of: QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIK (SEQ ID NO: 1).
[0202] In some embodiments, the CAR molecule comprises a CD8a extracellular domain (z.e., a CD8a hinge domain). In some embodiments, the CD8a extracellular domain (z.e., a CD8a hinge domain) comprises a sequence of: TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:2). In some embodiments, the extracellular domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:2. In some embodiments, the CD8a extracellular domain (z.e., a CD8a hinge domain) comprises a sequence of SEQ ID NO:2, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the CD8a extracellular domain (z.e., a CD8a hinge domain) comprises a sequence of: TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO:2).
[0203] In some embodiments, the CAR molecule comprises a linker domain between the antigen binding domain and extracellular domain. In some embodiments, the linker domain comprises a sequence of AAA (SEQ ID NO:3).
[0204] In some embodiments, the CAR molecule comprises a CD8a transmembrane domain. In some embodiments, the CD8a transmembrane domain comprises a sequence of: IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO:4). In some embodiments, the CD8a transmembrane domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:4. In some embodiments, the CD8a transmembrane domain comprises a sequence of SEQ ID NO:4, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the CD8a transmembrane domain comprises a sequence of: IYIWAPLAGTCGVLLLSLVIT (SEQ ID NO:4).
[0205] In some embodiments, the CAR molecule comprises a portion of a CD8a intracellular domain. In some embodiments, the CD8a intracellular domain comprises the sequence of: LYC (SEQ ID NO: 5).
[0206] In some embodiments, the CAR molecule comprises a 4- IBB intracellular costimulatory signaling domain. In some embodiments, the 4-1BB intracellular costimulatory signaling domain comprises a sequence of SEQ ID NO:6: KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL. In some embodiments, the 4-1BB costimulatory domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:6. In some embodiments, the 4-1BB costimulatory domain comprises a sequence of SEQ ID NO:6, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the 4-1BB costimulatory domain comprises a sequence of: KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO: 6).
[0207] In some embodiments, the CAR molecule comprises a CD3zeta intracellular signaling domain. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO:7:
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQ EGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQAL PPR. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO:7. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO:7, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
[0208] In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 7):
RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQ EGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQAL PPR, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. [0209] In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 10: RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRRKNP QEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQA LPPR. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 10. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 10, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
[0210] In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 11 : RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPQRRKNP QEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQA LPPR. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 11. In some embodiments, the CD3zeta intracellular signaling domain comprises a sequence of SEQ ID NO: 11, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto.
[0211] In some embodiments, the CAR molecule comprises a signal peptide sequence. In some embodiments, the signal peptide sequence comprises a sequence of SEQ ID NO:8: MLLLVTSLLLCELPHPAFLLIP. In some embodiments, the signal peptide sequence comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 20 amino acid modifications of an amino acid sequence of SEQ ID NO: 8. In some embodiments, the signal peptide sequence comprises a sequence of SEQ ID NO:8, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the signal peptide sequence comprises a sequence of: MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 8).
[0212] In some embodiments, the CAR molecule comprises a sequence of SEQ ID NO: 12. In some embodiments, the CAR molecule comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 40 amino acid modifications of an amino acid sequence of SEQ ID NO: 12. In some embodiments, the signal peptide sequence comprises a sequence of SEQ ID NO: 12, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the CAR molecule comprises a sequence of: QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTYYRS KWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLEDAFDIW GQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPG KAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQ GTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIW APLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEG GCELRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRR KNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALH MQALPPR (SEQ ID NO: 12).
[0213] In some embodiments, the CAR molecule comprises a sequence of SEQ ID NO:9. In some embodiments, the CAR molecule comprises a sequence having at least 1, 2, 3, 4, or 5, but not more than 40 amino acid modifications of an amino acid sequence of SEQ ID NO:9. In some embodiments, the signal peptide sequence comprises a sequence of SEQ ID NO:9, or a sequence with at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identity thereto. In some embodiments, the CAR molecule comprises a sequence of: MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAW NWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPED TAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVT ITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQ AEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACR PAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFM RPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREE YDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKG HDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NOV).
Delivery vector
[0214] The present disclosure also provides a vector in which a DNA of the present disclosure is inserted. Vectors derived from retroviruses such as lentiviruses are suitable tools to achieve long-term gene transfer since they allow long-term, stable integration of a transgene and its propagation in daughter cells. In some embodiments, the vector comprises a lentiviral vector. In some embodiments, the vector comprises a gamma retroviral vector.
[0215] In some embodiments, a vector may comprise a signal sequence to facilitate secretion, a polyadenylation signal and transcription terminator (e.g., from the Bovine Growth Hormone (BGH) gene), an element allowing episomal replication and replication in prokaryotes (e.g., SV40 origin and ColEl or others known in the art) and/or elements to allow selection (e.g., an ampicillin resistance gene and/or zeocin marker). In some embodiments, the vector comprises a promoter. In some embodiments, the vector promoter is selected from the group comprising of CMV, PGK, EF-la, MND, MNDU3, MCU3, SFFV, and CBh.
[0216] In some embodiments, a lentiviral vector expresses nucleotide sequences encoding the CAR sequences described herein. In some embodiments, a lentiviral vector expresses a nucleotide sequence encoding a CAR sequence comprising an antigen binding domain (e.g., an scFv comprising a VH domain linked via a linker to a VL domain), a linker, an extracellular hinge domain, a transmembrane domain, a co-stimulatory domain, and an intracellular signaling domain. In some embodiments, a lentiviral vector expresses a nucleotide sequence encoding a CAR sequence comprising an anti-CD22 binding domain (e.g., an anti-CD22 scFv comprising a VH domain linked via a linker to a VL domain), linked to a CD8 extracellular domain (e.g., a CD8alpha hinge domain) via linker, a CD8a transmembrane domain, a 4- IBB costimulatory domain, and a CD3zeta activation domain. In some embodiments the nucleotide sequence encoding a CAR sequence further comprises a signal peptide sequence. In some embodiments, a lentiviral vector expresses a nucleotide sequence encoding a CAR with a sequence as described herein.
Source of Cells
[0217] In some embodiments, T cells are isolated from peripheral blood lymphocytes by lysing the red blood cells and depleting the monocytes, for example, by centrifugation. In some embodiments, T cells are isolated from a blood sample by apheresis.
[0218] In some embodiments, the collection of blood samples or apheresis product from a subject takes place at a time period prior to lentiviral transduction and when the expanded cells as described herein in the Examples might be needed. For example, in some embodiments, the collection of blood samples or apheresis product from a subject can be taken and then subsequently frozen for later use. In some embodiments, a source of the cells to be expanded can be collected at any time point necessary, and desired cells, such as T cells, isolated and frozen for later use in immune effector cell therapy for any number of diseases or conditions that would benefit from immune effector cell therapy, such as those described herein. In some embodiments, a blood sample or an apheresis product is taken from a generally healthy subject. In some embodiments, a blood sample or an apheresis product is taken from a generally healthy subject who is at risk of developing a disease, but who has not yet developed a disease, and the cells of interest are isolated and frozen for later use. In some embodiments, the T cells may be expanded, frozen, and used at a later time. In some embodiments, samples are collected from a patient shortly after diagnosis of a particular disease as described herein but prior to any treatments. In some embodiments, the cells are isolated from a blood sample or an apheresis product from a subject prior to any number of relevant treatment modalities, including but not limited to treatment with agents such as natalizumab, efalizumab, antiviral agents, chemotherapy, radiation, immunosuppressive agents, such as cyclosporin, azathioprine, methotrexate, mycophenolate, and FK506, antibodies, or other immunoablative agents such as CAMPATH, anti-CD3 antibodies, cytoxan, fludarabine, cyclosporin, FK506, rapamycin, mycophenolic acid, steroids, FR901228, and irradiation.
[0219] In a further aspect of the present disclosure, T cells are obtained from a patient directly following treatment that leaves the subject with functional T cells. In this regard, it has been observed that following certain cancer treatments, in particular treatments with drugs that damage the immune system, shortly after treatment during the period when patients would normally be recovering from the treatment, the quality of T cells obtained may be optimal or improved for their ability to expand ex vivo. Likewise, following ex vivo manipulation using the methods described herein, these cells may be in a preferred state for enhanced engraftment and in vivo expansion. Thus, it is contemplated within the context of the present disclosure to collect blood cells and/or apheresis product, including T cells, dendritic cells, or other cells of the hematopoietic lineage, during this recovery phase. Further, in certain aspects, mobilization (for example, mobilization with GM-CSF) and conditioning regimens can be used to create a condition in a subject wherein repopulation, recirculation, regeneration, and/or expansion of particular immune cell types is favored, especially during a defined window of time following therapy. Illustrative cell types include T cells, B cells, dendritic cells, and other cells of the immune system.
Activation and Expansion of Immune Cells
[0220] Immune effector cells such as T cells may be activated and expanded generally using methods as described, for example, in U.S. Patent Nos. 6,352,694; 6,534,055; 6,905,680; 6,692,964; 5,858,358; 6,887,466; 6,905,681; 7,144,575; 7,067,318; 7,172,869; 7,232,566; 7,175,843; 5,883,223; 6,905,874; 6,797,514; 6,867,041; and U.S. Patent Application Publication No. 2006/0121005 Al, each of which is incorporated herein by reference in its entirety for any purpose. [0221] Method of expansion of immune effector cells, methods of introducing CAR nucleic acid molecules into immune effector cells, and methods of detecting CAR expression are described on pages 236-246 of in International Application WO 2016/164731 filed on Apr. 8, 2016, which is incorporated by reference in its entirety for any purpose.
[0222] In some embodiments, a CAR-expressing T-cell is produced according to the procedures incorporated herein by reference.
Indications
[0223] In some embodiments, the hematological cancer is associated with expression of an antigen, e.g., a tumor antigen, e.g., CD22. In some embodiments, the hematological cancer comprises lymphoma. In some embodiments, the hematological cancer is a lymphoma, e.g., a relapsed and/or refractory lymphoma. In some embodiments, the hematological cancer comprises leukemia, e.g., a relapsed and/or refractory leukemia. In some embodiments, the cancer is selected from the group consisting of one or more acute leukemias including but not limited to B-cell acute lymphoid leukemia (B-ALL), T-cell acute lymphoid leukemia (TALL), small lymphocytic leukemia (SLL), acute lymphoid leukemia (ALL) (e.g., relapsing and refractory ALL); one or more chronic leukemias including but not limited to chronic myelogenous leukemia (CML), and chronic lymphocytic leukemia (CLL). In some embodiments, the cancer is selected from the group consisting of additional hematologic cancers or conditions including, but not limited to, mantle cell lymphoma (MCL), B cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B cell lymphoma (DLBCL), follicular lymphoma, hairy cell leukemia, small cell- or a large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, Marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom macroglobulinemia, and “preleukemia.” Preleukemia encompasses a diverse collection of hematological conditions united by ineffective production (or dysplasia) of myeloid blood cells In some embodiments, a disease as used herein includes, but is not limited to atypical and/or non-classical cancers, malignancies, precancerous conditions; and any combination thereof.
[0224] In some embodiments, the disease associated with expression of CD22 is a lymphoma. In some embodiments, the lymphoma is Non-Hodgkin lymphoma (NHL). The majority of NHL cases are of B-cell origin and can be further subdivided into aggressive and indolent lymphomas, each associated with different clinical outcomes and prognosis. In some embodiments, the NHL is an aggressive lymphoma selected from the group consisting of AIDS-associated lymphoma, Burkitt lymphoma, primary central nervous system lymphoma, large B cell lymphomas (LBCLs), mantle cell lymphoma, peripheral T-cell lymphoma, and T-cell lymphoblastic lymphoma. In some embodiments, the NHL is an indolent lymphoma selected from the group consisting of cutaneous T-cell lymphoma, follicular lymphoma (FL), lymphoplasmacytic lymphoma, marginal zone lymphoma, and small cell lymphocytic lymphoma. In some embodiments, the NHL lymphoma is LBCL. Large B-cell lymphomas (LBCLs) are aggressive subtypes including diffuse large cell lymphoma (DLBCL), highgrade B-cell lymphomas, primary mediastinal B-cell lymphoma (PMBCL), Epstein-Barr virus-positive LBCL, follicular lymphoma (FL) grade 3B and other subtypes.
[0225] In some embodiments, the LBCL is DLBCL. In some embodiments, the DLBCL has been histologically confirmed. In some embodiments, the DLBCL is not otherwise specified (NOS) type. In some embodiments, the DLBCL NOS is germinal center B-cell (GCB) type. In some embodiments, the DLBCL NOS is active B-cell (ABC) type. In some embodiments, the LBCL is DLBCL associated with chronic inflammation. In some embodiments, the LBCL is primary cutaneous DLBCL, leg type. In some embodiments, the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS. In some embodiments, the LBCL is DLBCL arising from FL. In some embodiments, the LBCL is DLBCL arising from MZL.
[0226] In some embodiments, the LBCL is primary mediastinal (thymic) large B-cell lymphoma. In some embodiments, the LBCL is Grade 3B follicular large B-cell lymphoma (FL).
[0227] In some embodiments, the LBCL is high-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements.
[0228] In some embodiments, the subject has a relapsed/refractory disease that recurs following treatment. The term “relapsed and/or refractory disease” refers to progressive disease (PD)/stable disease (SD) after the last therapy or relapsed disease following a complete response (CR) or progressive disease following a partial response (PR) after the last therapy. In some embodiments the relapsed/refractory disease comprises a predicted relapse. In some embodiments, the relapsed/refractory disease comprises an identified relapse. In some embodiments, the relapsed/refractory disease comprises disease progression. In some embodiments, the subject is predicted to have a relapse (e.g., has not relapsed), has relapsed, or is identified as having relapsed. In some embodiments, the subject relapsed after having received a front-line therapy. In some embodiments, the subject relapsed after having received a first-line therapy In some embodiments, the front-line therapy may comprise an antibody -based therapy, a cell-based therapy, a chemotherapy, or a combination thereof. In some embodiments, the antibody-based therapy may comprise CD20-targeted monoclonal antibodies. In some embodiments, the antibody -based therapy may comprise CD19-targeted monoclonal antibody therapy. In some embodiments, the cell-based therapy may comprise a CAR T-cell therapy. In some embodiments, the cell-based therapy may comprise a CD19 CAR T-cell therapy. In some embodiments, the chemotherapy comprises rituximab, cyclophosphamide, doxorubicin, vincristine and prednisolone (R-CHOP), etoposide, erednisolone, encovin cyclophosphamide, and eydroxydaunorubicin (EPOCH), or a combination thereof.
Subject treatment criteria
[0229] In some embodiments, the subject is 18 years or older.
[0230] In some embodiments, the subject has received one or more previous treatments. In some embodiments, the subject has received about 1, about 2, about 3, about 4 or about 5 previous treatments. In some embodiments, the subject has received 1 or more previous treatments, 2 or more previous treatments, 3 or more previous treatments, 4 or more previous treatments, or 5 or more previous treatments.
[0231] In some embodiments, the one or more previous treatments comprises a chemotherapy, an antibody, a cell-based treatment, or any combination thereof. In some embodiments, the subject has received a CAR T-cell therapy. In some embodiments, the subject has not received a CAR T-cell therapy. In some embodiments, the subject has received an antibody. In some embodiments, the subject has not received an antibody. In some embodiments, the subject has received a chemotherapy. In some embodiments, the subject has not received a chemotherapy. In some embodiments, the subject has received at least one or more previous treatments and have not received a CAR T-cell therapy. In some embodiments, the subject has received at least one or more previous treatments and have not received an antibody treatment. In some embodiments, the subject has received at least one or more previous treatments and have not received a chemotherapy.
[0232] In some embodiments, one or more previous treatments comprises an antibody. In some embodiments, the antibody comprises an anti-CD19 antibody, an anti-CD20 antibody, and anti-CD22 antibody or an anti-CD79b antibody. In some embodiments, the antibody comprises an anti-CD19 antibody and an anti-CD20 antibody. In some embodiments, the antibody comprises an anti-CD19 antibody and an anti-CD22 antibody. In some embodiments, the antibody comprises an anti-CD19 antibody and an anti-CD79b antibody. In some embodiments, the antibody comprises an anti-CD20 antibody and an anti-CD22 antibody. In some embodiments, the antibody comprises an anti-CD20 antibody and an anti- CD79b antibody. In some embodiments, the antibody comprises an anti-CD22 antibody and an anti-CD79b antibody. In some embodiments, the antibody comprises an anti-CD19 antibody and an anti-CD20 antibody. In some embodiments, the antibody comprises an anti- CD19 antibody, an anti-CD20 antibody, and an anti-CD22 antibody. In some embodiments, the antibody comprises an anti-CD19 antibody, an anti-CD20 antibody, and an anti-CD79b antibody. In some embodiments, the anti-CD19 antibody, the anti-CD20 antibody, the anti- CD22 antibody or the anti-CD79b antibody is an antibody-drug conjugate. In some embodiments, the anti-CD19 antibody comprises loncastuximab. In some embodiments, the anti-CD20 antibody comprises rituximab. In some In some embodiments, the anti-CD22 antibody-drug conjugate comprises inotuzumab ozogamicin.
[0233] In some embodiments, the subject may have previously received a CAR T-cell therapy. In some embodiments, the CAR T-cell therapy comprises tisagenlecleucel, brexucabtagene autoleucel, axicabtagene cilocleucel, aisocabtagene maraleucel or any combination thereof. In some embodiments, the subject may have received a CD19-directed CAR T-cell therapy. In some embodiments the CAR T-cell therapy is Abecma®. In some embodiments the CAR T-cell therapy is Kymriah™. In some embodiments the CAR T-cell therapy is Breyanzi®. In some embodiments the CAR T-cell therapy is Carvykti™. In some embodiments the CAR T-cell therapy is Tecartus™. In some embodiments the CAR T-cell therapy is Yescarta™. In some embodiments, the subject may have received additional therapies after the previously received CAR T-cell therapy. In some embodiments, the additional therapies after the previously received CAR T-cell therapy comprise a chemotherapy. In some embodiments, the chemotherapy after the previously received CAR T-cell therapy comprises rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone, or a combination thereof. In some embodiments, the chemotherapy after the previously received CAR T-cell therapy comprises etoposide, erednisolone, encovin cyclophosphamide, hydroxydaunorubicin, or a combination thereof.
[0234] In some embodiments, the one or more previous treatments comprises a bispecific T- cell engager antibody therapy. In some embodiments, the bispecific T-cell engager antibody therapy comprises T cell engager comprising an anti-CD3 domain. In some embodiments, the bispecific T-cell engager antibody therapy further comprises an anti-CD20 domain. In some embodiments, the bispecific T-cell engager antibody therapy further comprises an anti-CD19 domain. In some embodiments, the bispecific T-cell engager antibody therapy further comprises an anti-BCMA domain. In some embodiments, the bispecific T-cell engager antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, elranatamab, teclistamab, tarlatamab, talquetamab, solitomab, tebentafusp, duvortuxizumab, AFM11, REGN5458, CC-93269, PF-06863135, TNB-383B, odronextamab, XmAbl3676, IGM-2323, AMG 330, AMG 673, AMG 420, AMG 701, cevostamab, talquetamab, flotetuzumab, or a combination thereof.
[0235] In some embodiments, the subject may have received a bispecific T-cell engager antibody therapy. In some embodiments, the bispecific T-cell engager antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, elranatamab, teclistamab, tarlatamab, talquetamab, solitomab, tebentafusp, duvortuxizumab, AFM11, REGN5458, CC-93269, PF-06863135, TNB-383B, pdronextamab, XmAb 13676, IGM-2323, AMG 330, AMG 673, AMG 420, AMG 701, cevostamab, talquetamab, flotetuzumab or any combination thereof. In some embodiments, the subject may have relapsed/refractory disease following a complete recovery after receiving a bispecific CAR T-cell therapy or bispecific T-cell engager antibody therapy. In some embodiments, the subject may have progressive disease following a partial recovery after receiving a bispecific CAR T-cell therapy or bispecific T-cell engager antibody therapy. In some embodiments, the subject may have progressive disease/stable disease as a best response to a bispecific CAR T-cell therapy or bispecific T-cell engager antibody therapy.
[0236] In some embodiments, the subject has received prior treatment with a CD- 19 directed CAR T-cell therapy and no prior treatment with a bispecific T-cell engager antibody therapy. In some embodiments, the subject has received prior treatment with a CD- 19 directed CAR T-cell therapy and prior treatment with a bispecific T-cell engager antibody therapy.
[0237] In some embodiments, the subject has not received prior treatment with a CD- 19 directed CAR T-cell therapy and has been treated with no more than 2 prior lines of therapy. In some embodiments, the subject has not received prior treatment with a CAR T-cell therapy at all. In some embodiments, the patients must have received at least 1 prior chemoimmunotherapy (CIT)-containing regimen (z.e., regimen must contain a CD20-targeted therapy plus an anthracy cline). In some embodiments, the patients received prior treatment with a bispecific T-cell engager (BiTE®) antibody therapy. In some embodiments, at least 30 days must have passed from the last dose of the BiTE® to the time of leukapheresis. [0238] In some embodiments, the subject has been diagnosed with LBCL. In some embodiments, the subject has been diagnosed with LBCL through histological confirmation by a pathologist or pathology lab. In some embodiments, histological confirmation of LBCL may involve one of the following: DLBCL not otherwise specified (NOS), including germinal center B-cell (GCB) type or active B-cell (ABC) type, high-grade B-cell lymphoma with or without MYC and BCL2 and/or BCL6 rearrangements (double-hit or triple-hit lymphoma), DLBCL associated with chronic inflammation, primary cutaneous DLBCL, leg type, Epstein-Barr virus (EB V)-positive DLBCL- NOS, DLBCL arising from follicular lymphoma (FL; transformed follicular lymphoma), DLBCL arising from marginal zone lymphoma (MZL; transformed marginal zone lymphoma), primary mediastinal (thymic) large B-cell lymphoma, or FL, Grade 3B.
[0239] In some embodiments, the subject has relapsed/refractory disease after the last therapy. In some embodiments, relapsed/refractory disease comprises radiographic findings comprising progressive disease/stable disease after the last therapy. In some embodiments, relapsed/refractory disease comprises radiographic disease progression following a partial response after the last therapy. In some embodiments, relapsed/refractory disease comprises radiographic disease progression following a complete response after the last therapy. In some embodiments, relapsed/refractory disease comprises radiographic disease progression after a partial response. In some embodiments, relapsed/refractory disease comprises radiographic disease progression after a complete response. In some embodiments, relapsed/refractory disease comprises viable lymphoma in a biopsy. In some embodiments, relapsed/refractory disease comprises viable lymphoma in a biopsy after therapy.
[0240] In some embodiments, the subject has at least one radiographically measurable lesion. In some embodiments, the radiographically measurable lesion has at least one bidimensionally measurable nodal lesion of >1.5 cm in its longest dimension. In some embodiments, the radiographically measurable lesion has at least one bidimensionally measurable extra lesion of >1.0 cm in its longest dimension. In some embodiments, lesions that were previously irradiated are considered measurable if progression has been documented following completion of radiation therapy.
[0241] In some embodiments, the subject may have a washout period following the last therapy. In some embodiments, the washout period is a time duration between receiving a therapy and receiving the next therapy. In some embodiments, the washout period is 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, or some fraction thereof. In some embodiments, the washout period is at least one half-life of the last therapy administered to the subject. As used herein, the term “half-life” or “half-lives” refers to one or more time periods, where each time period comprises the time required for half of something (e.g., the last therapy administered to the subject) to be metabolized or eliminated from the subject’s system. In some embodiments, the washout period is 1 half-life, 2 half-lives, 3 half-lives, 4 half-lives, 5 halflives, 6 half-lives, 7 half-lives, 8 half-lives, 9-half-lives or 10 half-lives. In some embodiments, the washout period is 2 weeks. In some embodiments the washout period is 5 half-lives. In some embodiments, the washout period is either 2 weeks or 5 half-lives. In some embodiments, the washout period is the shorter of 2 weeks or 5 half-lives. In some embodiments, the washout period is the longer of 2 weeks or 5 half-lives.
[0242] In some embodiments, the subject has previously received bendamustine (marketed as Bendeka® or Treanda®). In some embodiments, the bendamustine was last received at least 3-9 months prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the bendamustine was last received at least 3 months, at least 3.5 months, at least 4 months, at least 4.5 months, at least 5 months, at least 5.5 months, at least 6 months, at least 6.5 months, at least 7 months, at least 7.5 months, at least 8 months, at least 8.5 months, at least 9 months or some fraction thereof prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the bendamustine was last received at least 6 months prior to receiving the CD22-directed CAR T-cell therapy.
[0243] In some embodiments, the subject has previously received CD19-directed CAR T-cell therapy. In some embodiments, the CD19-directed CAR T-cell therapy was last administered at least 10-60 days prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the CD19-directed CAR T-cell therapy was last administered at least 10 days, at least 15 days, at least 20 days, at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, or some fraction thereof prior to receiving the CD 19- directed CAR T-cell therapy. In some embodiments, the CD19-directed CAR T-cell therapy was last administered at least 30 days prior to receiving the CD19-directed CAR T-cell therapy.
[0244] In some embodiments, the subject has previously received bispecific CAR T-cell therapy. In some embodiments, the bispecific CAR T-cell therapy was last administered at least 10-60 days prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the bispecific CAR T-cell therapy was last administered at least 10 days, at least 15 days, at least 20 days, at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, or some fraction thereof prior to receiving the CD22- directed CAR T-cell therapy. In some embodiments, the bispecific CAR T-cell therapy was last administered at least 30 days prior to receiving the CD22-directed CAR T-cell therapy. [0245] In some embodiments, the subject has previously received a bispecific T-cell engager therapy. In some embodiments, the bispecific T-cell engager therapy was last administered at least 10-60 days prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the bispecific T-cell engager therapy was last administered at least 10 days, at least 15 days, at least 20 days, at least 25 days, at least 30 days, at least 35 days, at least 40 days, at least 45 days, at least 50 days, or some fraction thereof prior to receiving the CD22- directed CAR T-cell therapy. In some embodiments, the bispecific T-cell engager therapy was last administered at least 30 days prior to receiving the CD22-directed CAR T-cell therapy.
[0246] In some embodiments, the subject has previously received an autologous stem cell transplant. In some embodiments, the autologous stem cell transplant was last received at least 2-20 weeks prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the autologous stem cell transplant was last received at least 2 weeks, at least 3 weeks, at least 4 weeks, at least 5 weeks, at least 6 weeks, at least 7 weeks, at least 8 weeks, at least 9 weeks, at least 10 weeks, at least 11 weeks, at least 12 weeks, at least 13 weeks, at least 14 weeks, at least 15 weeks, at least 16 weeks, at least 17 weeks, at least 18 weeks, at least 19 weeks, at least 20 weeks, or some fraction thereof prior to receiving the CD22- directed CAR T-cell therapy. In some embodiments the autologous stem cell transplant was last received at least 6 weeks prior to receiving the CD22-directed CAR T-cell therapy.
[0247] In some embodiments, the subject has previously received CNS prophylaxis. In some embodiments the CNS prophylaxis is intrathecal methotrexate. In some embodiments, the CNS prophylaxis was last administered at least 1-20 days prior to receiving the CD22- directed CAR T-cell therapy. In some embodiments, the CNS prophylaxis was last administered at least 1 day, at least 2 days, at least 3 days, at least 4 days, at least 5 days, at least 6 days, at least 7 days, at least 8 days, at least 9 days, at least 10 days, at least 11 days, at least 12 days, at least 13 days, at least 14 days, at least 15 days, at least 16 days, at least 17 days, at least 18 days, at least 19 days, at least 20 days, or some fraction thereof prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the CNS prophylaxis was administered at least 7 days prior to receiving the CD22-directed CAR T- cell therapy. [0248] In some embodiments, the subject received the CD22-directed CAR T-cell therapy without a washout period.
[0249] In some embodiments, a tumor tissue sample from the subject is available for analysis. In some embodiments, the tumor tissue sample is a formalin-fixed paraffin- embedded tissue block. In some embodiments the tumor sample is resected tissue. In some embodiments the tissue sample is fresh-frozen. In some embodiments, the tumor tissue sample is obtained following the most recent therapy. In some embodiments, the tumor tissue sample is obtained following CD 19 CAR T-cell therapy or any other line of therapy administered prior to relapse. In some embodiments, more than one tumor tissue sample is available. In some embodiments, the more than one tumor tissue sample is from the same tumor location. In some embodiments, the more than one tumor tissue sample is from more than one tumor location. In some embodiments, the tumor tissue sample is a fine needle aspirate. In some embodiments, the tumor tissue sample is a sample of brushings. In some embodiments, the tumor tissue sample is cell pellets from effusions. In some embodiments, the tumor tissue sample is cell pellets from ascites.
[0250] In some embodiments, the subject has a performance status. In some embodiments, the performance status is according to the Eastern Cooperative Oncology Group (ECOG). In some embodiments, the subject has a performance status of 0. In some embodiments, the subject has a performance status of 1. In some embodiments, the subject has a performance status of 0 according to ECOG. In some embodiments, the subject has a performance status of 1 according to ECOG. In some embodiments, a status of 0 comprises a subject that is fully active, able to carry on all pre-disease performance without restriction. In some embodiments, a status of 1 comprises a subject that is restricted in physically strenuous activity but ambulatory and able to carry out work of a light or sedentary nature, e.g., light housework, office work.
[0251] In some embodiments, the subject has adequate hematologic and end-organ function. In some embodiments, the adequate hematologic and end-organ function comprises an absolute neutrophil count (ANC) greater than about 500-1500/pl, about 700-1200/pl, of about 900-1100/pl, about 800/pl, about 900/pl, about 1000/pl, about 1100/pl, or about 1200/pl. In some embodiments, the subject has an ANC of greater than or equal to about 1000/pl. In some embodiments, the adequate hematologic and end-organ function comprises a platelet count of greater than about 50, 000-100, 000/pl, about 60, 000-90, 000/pl, about 50,000-80,000/( 1, about 50,000/jj.l, about 60,000/ .l, about 70,000/ .l, about 75,000/pl, about 80,000/( 1, about 90,000/jil, or about 100,000/jil.
[0252] In some embodiments, the adequate hematologic and end-organ function comprises an absolute neutrophil count (ANC) greater than about 500-1500/mL, about 700-1200 mL, of about 900-1100/ mL, about 800/mL, about 900/mL, about 1000/mL, about 1100/mL, or about 1200/mL. In some embodiments, the subject has an ANC of greater than or equal to about 1000/mL. In some embodiments, the adequate hematologic and end-organ function comprises a platelet count of greater than about 50, 000-100, 000/mL, about 60, 000-90, 000/mL, about 50, 000-80, 000/mL, about 50, 000/mL, about 60, 000/mL, about 70, 000/mL, about 75, 000/mL, about 80, 000/mL, about 90, 000/mL, or about 100, 000/mL.
[0253] In some embodiments, the adequate hematologic and end-organ function comprises a platelet count of greater than about 75,000/pl. In some embodiments, the subject has a presence of marrow disease. In some embodiments, the adequate hematologic and end-organ function for a subject with the presence of marrow disease has a platelet count of greater than about 25, 000-75, 000/pl, about 30, 000-70, 000/pl, about 40, 000-60, 000/pl, about 25, 000/pl, 30,000/pl, about 35,000/pl, about 40,000/pl, about 45,000/pl, about 50,000/pl, about 55,000/pl, about 60,000/pl, about 65,000/pl, about 70,000/pl, or about 75,000/pl. In some embodiments, the adequate hematologic and end-organ function for a subject with the presence of marrow disease comprises a platelet count of greater than about 50,000/pl.
[0254] In some embodiments, the adequate hematologic and end-organ function comprises a platelet count of greater than about 75, 000/mL. In some embodiments, the subject has a presence of marrow disease. In some embodiments, the adequate hematologic and end-organ function for a subject with the presence of marrow disease has a platelet count of greater than about 25, 000-75, 000/mL, about 30, 000-70, 000/mL, about 40, 000-60, 000/mL, about 25, 000/mL, 30, 000/mL, about 35, 000/mL, about 40, 000/mL, about 45, 000/mL, about 50, 000/mL, about 55, 000/mL, about 60, 000/mL, about 65, 000/mL, about 70, 000/mL, or about 75, 000/mL. In some embodiments, the adequate hematologic and end-organ function for a subject with the presence of marrow disease comprises a platelet count of greater than about 50, 000/mL.
[0255] In some embodiments, the adequate hematologic and end-organ function comprises an absolute lymphocyte count (ALC) of greater than about 10-300/pl, about 50-150, 000/pl, about 75-125/(11, about 10/pl, 20/pl, about 30/pl, about 40/pl, about 50/pl, about 60/pl, about 70/pl, about 80/pil, about 90/pl, about 100/pl, about 110/pl, about 120/pl, about 130/pl, about 140/ .l, about 150/pl, about 160/jj.l, about 170/ .l, about 180/ .l, about 190/jj.l, about 200/pl, about 225/pl, about 250/ l, about 275/pl, or about 3 OO/JJ.1. In some embodiments, the subject has an ALC of greater than or equal to about 1 OO/JJ.1.
[0256] In some embodiments, the adequate hematologic and end-organ function comprises an absolute lymphocyte count (ALC) of greater than about 10-300/mL, about 50- 150,000/mL, about 75-125/mL, about 10/mL, 20/mL, about 30/mL, about 40/mL, about 50/mL, about 60/mL, about 70/mL, about 80/mL, about 90/mL, about 100/mL, about 110/mL, about 120/mL, about 130/mL, about 140/mL, about 150/mL, about 160/mL, about 170/mL, about 180/mL, about 190/mL, about 200/mL, about 225/mL, about 250/mL, about 275/mL, or about 300/mL. In some embodiments, the subject has an ALC of greater than or equal to about 100/mL.
[0257] In some embodiments, the adequate hematologic and end-organ function comprises an estimated creatinine clearance of greater than about 10-100 ml/minute. In some embodiments, the adequate hematologic and end-organ function comprises an estimated creatinine clearance of greater than about 10-100 ml/minute, about 30-60 ml/minute, about 40-50 ml/minute, about 10 ml/minute, about 20 ml/minute, about 30 ml/minute, about 40 ml/minute, about 45 ml/minute, about 50 ml/minute, about 55 ml/minute, about 60 ml/minute, about 65 ml/minute, about 70 ml/minute, about 75 ml/minute, about 80 ml/minute, about 85 ml/minute, about 90 ml/minute, about 95 ml/minute or about 100 ml/minute. In some embodiments, the estimated creatinine clearance is measured according to the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) method. In some embodiments, the estimated creatinine clearance is measured based on the volume of urine collected spanning about one day (e.g., about 24 hours). In some embodiments, the estimated creatinine clearance is measured in other ways.
[0258] In some embodiments, the adequate hematologic and end-organ function comprises a serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) activity of less than about 1-5 times the institutional upper limit of normal (ULN), about 2-4 times ULN, about 2- 3 times the ULN, about 1 times the ULN, about 1.5 times the ULN, about 2.0 times the ULN, about 2.5 times the ULN, about 3.0 times ULN, about 3.5 times the ULN, about 4.0 times the ULN, about 4.5 times the ULN, or about 5 times the ULN. In some embodiments, the adequate hematologic and end-organ function comprises a serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) activity of less than about 2.5 times the ULN. [0259] In some embodiments, the adequate hematologic and end-organ function comprises a total bilirubin concentration of less than about 0.5-4 mg/dL, about 1-3 mg/dL, about 1-2 mg/dL, about 0.5 mg/dL, about 1.0 mg/dL, about 1.5 mg/dL, about 2.0 mg/dL, about 2.5 mg/dL, about 3.0 mg/dL, about 3.5 mg/dL, or about 4.0 mg/dL. In some embodiments, the adequate hematologic and end-organ function comprises a total bilirubin concentration of less than about 1.5 mg/dL. In some embodiments, the adequate hematologic and end-organ function comprises a total bilirubin concentration that is different for subjects with Gilbert syndrome. In some embodiments, the adequate hematologic and end-organ function comprises a total bilirubin concentration greater than 1.5 mg/dL for subjects with Gilbert syndrome.
[0260] In some embodiments, the adequate hematologic and end-organ function comprises a cardiac left ventricular ejection fraction (LVEF) of greater than about 25-75%, about 30-60%, about 40-50%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, or about 75%. In some embodiments, the adequate hematologic and end-organ function comprises a cardiac left ventricular ejection fraction (LVEF) of greater than about 45%. In some embodiments, the LVEF comprises no evidence of pericardial effusion. In some embodiments, the adequate hematologic and end-organ function comprises a cardiac left ventricular ejection fraction (LVEF) of greater than about 45% and no evidence of pericardial effusion.
[0261] In some embodiments, the adequate hematologic and end-organ function comprises a blood oxygen saturation (SaCL) of greater than about 85-100%, about 90-95%, about 91- 93%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%. In some embodiments, the adequate hematologic and end-organ function comprises a blood oxygen saturation (SaCL) of greater than about 92%.
[0262] In some embodiments, the adequate hematologic and end-organ function comprises a serum albumin concentration of greater than about 1-5 g/dL, about 2-4 g/dL, about 2-3 g/dL, about 1 g/dL, about 1.5 g/dL, about 2 g/dL, about 2.5 g/dL, about 3 g/dL, about 3.5 g/dL, about 4 g/dL, about 4.5 g/dL, or about 5 g/dL. In some embodiments, the adequate hematologic and end-organ function comprises a serum albumin concentration of greater than about 2.5 g/dL.
[0263] In some embodiments, the subject is a woman of childbearing potential. In some embodiments, the women of childbearing potential may refrain from heterosexual intercourse during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. In some embodiments, the woman of childbearing potential may use contraceptive methods during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. In some embodiments, the woman of childbearing potential may use contraceptive methods that comprise a failure rate of less than about 0.1-5.0% per year, about 0.5-1.5% per year, about 0.2-2.0% per year, about 0.1% per year, about 0.2% per year, about 0.3% per year, about 0.4% per year, about 0.5% per year, about 0.6% per year, about 0.7% per year, about 0.8% per year, about 0.9% per year, about 1.0% per year, about 1.5% per year, about 2.0% per year, about 2.5% per year, about 3.0% per year, about 3.5% per year, about 4.0% per year, about 4.5% per year, or about 5.0% per year during the course of the treatment with the CAR T-cell therapy. In some embodiments, the woman of childbearing potential is post monarchal. In some embodiments, the woman of childbearing potential has not reached a postmenopausal state. In some embodiments the postmenopausal state comprises more than 12 months of amenorrhea with no identified cause other than menopause. In some embodiments, the woman of childbearing potential has not undergone surgical sterilization (removal of ovaries and/or uterus). In some embodiments, the woman of childbearing potential may comprise additional characteristics. [0264] In some embodiments, contraceptive methods may comprise bilateral tubal ligation, male sterilization, hormonal contraceptives that inhibit ovulation, hormone-releasing intrauterine devices, copper intrauterine devices, or a combination thereof.
[0265] In some embodiments, the woman of childbearing potential must have a negative pregnancy test result within about 1-14 days, about 3-10 days, about 5-10 days, about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days, about 11 days, about 12 days, about 13 days, or about 14 days before leukapheresis. In some embodiments, the woman of childbearing potential must have a negative pregnancy test result within about 7 days before leukapheresis. In some embodiments, the subject may not have a pregnancy test.
[0266] In some embodiments, the subject is male. In some embodiments, the male subject may refrain from heterosexual intercourse. In some embodiments, the male subject may refrain from heterosexual intercourse during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. In some embodiments, the male subject may use a condom with spermicide. In some embodiments, the male subject may use a condom with spermicide during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. In some embodiments, the male subject may use a condom with spermicide and the sexual partner may use a contraception method. In some embodiments, the contraception method is highly effective. In some embodiments, the male subject may refrain from donating sperm during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. In some embodiments, the male subject is vasectomized. In some embodiments, the vasectomized male subject may use a condom with spermicide. In some embodiments, the male subject may use a condom with spermicide during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. In some embodiments, the male subject may engage in sexual intercourse with a male partner. In some embodiments, the male subject that engages in sexual intercourse with a male partner may use a condom with spermicide during a treatment duration with the CAR T-cell therapy or the lymphodepletion regimen and for at least 6 months after. [0267] In some embodiments, the subject is stable. In some embodiments, the subject is stable or has recovered from toxicities associated with previous chemo- or immunotherapies prior to receiving the CD22-directed CAR T-cell therapy. In some embodiments, the subject is stable or has recovered from nonhematologic toxicities due to prior therapy to grade < 1. In some embodiments, the subject has clinically non-significant toxicities prior to therapy greater than grade 1. In some embodiments, the clinically non-significant toxicity is alopecia. [0268] In some embodiments, the subject may not receive the CD22-directed CAR T-cell therapy if one or more of the following conditions apply: the subject has a malignancy other than lymphoma; the subject has an active fungal, bacterial, viral or other infection that requires intravenous antimicrobials; the subject has received prior allogenic stem cell or any other organ transplant; the subject has received a prior allogeneic CAR T-cell therapy; the subject has received prior anti-CD52 antibody therapy; the subject has a history of CNS involvement of lymphoma within 1 year prior to receiving the CD22-directed CAR T-cell therapy; the subject has ongoing cardiac involvement of lymphoma; the subject has a history of infection with human immunodeficiency virus (HIV), hepatitis B virus (HBV), as determined by positivity for hepatitis B serum antigen (HbsAg) or hepatitis B core antibody (HbcAb), or hepatitis C virus (HCV), as determined by positivity for anti-HCV antibody; the subject has significant, uncontrolled concomitant disease that could affect interpretation of results from the CD22-directed CAR T-cell therapy; the subject has a history of autoimmune disease resulting in significant end-organ disease or requiring systemic immunosuppression and/or systemic disease modifying agents within 2 years of treatment; the subject has evidence of moderate to severe forms of primary immunodeficiencies; the subject has a history of severe, immediate hypersensitivity reaction attributed to aminoglycosides, DMSO, tocilizumab or any other agent that is part of the treatment regimen; the subject is pregnant; the subject is breastfeeding; the subject underwent recent major surgery other than for lymphoma treatment; the subject has an in-dwelling line or drain (e.g., percutaneous nephrostomy tube, in dwelling Foley catheter, biliary drain); the subject has a history of symptomatic deep vein thrombosis or pulmonary embolism requiring anti coagulation therapy within 6 months prior to receiving the CD22-directed CAR T-cell therapy; the subject has received treatment with a live, attenuated vaccine within 6 weeks prior to CD22-directed CAR T-cell treatment or are anticipated to need such a vaccine during the course of treatment; the subject has Richter’s transformation of CLL; the subject has T cell/histiocyte- rich large B-cell lymphoma; the subject requires urgent therapy due to mass effects of tumor of impending oncologic emergency e.g., bowel obstruction, major blood vessel compression or tumor lysis syndrome); or the subject has any other significant, uncontrolled concomitant disease that could affect compliance with treatment or interpretation of results.
[0269] In some embodiments, the significant, uncontrolled concomitant disease that could affect interpretation of results from the CD22-directed CAR T-cell therapy comprises a history of myocardial infarction, cardiac angioplasty or stenting, unstable angina, serious cardiac arrhythmia, or other clinically significant cardiac disease within 12 months prior to treatment with the CD22-directed CAR T-cell therapy. In some embodiments, the significant, uncontrolled concomitant disease comprises active pulmonary disease (such as severe obstructive pulmonary disease, idiopathic pulmonary fibrosis, organizing pneumonia, drug- induced pneumonitis, idiopathic pneumonitis). In some embodiments, the significant, uncontrolled concomitant disease comprises clinically significant liver disease, including cirrhosis. In some embodiments, the significant, uncontrolled concomitant disease comprises clinically significant neurological disease (such as cerebrovascular ischemia/hemorrhage, dementia, seizure disorder, or cerebellar disease).
[0270] In some embodiments, the modifying agents required for systemic immunosuppression do not include physiologic dose of steroid, a stable dose of thyroid replacement hormone (for subjects with a history of autoimmune-related hypothyroidism), or an insulin regimen (for subjects with controlled Type 1 diabetes mellitus). In some embodiments, the subject may have a history of immune thrombocytopenic purpura or autoimmune hemolytic anemia not requiring active treatment and thus may receive CD22- directed CAR T-cell therapy.
CAR T-cell Dosing Regimens
[0271] In some embodiments, a CAR-expressing cell, e.g., a CD22 CAR-expressing T cell as described herein, is administered to the subject according to a dosing regimen comprising a dose, e.g., a total dose, of cells administered to the subject by dose fractionation A.g, split dosing), e.g., one, two, three or more separate administrations of a partial dose, e.g., one, two, three, four, five or six partial doses. In some embodiments, a total dose of cells is administered as a single dose. In some embodiments, a single dose comprises the total dose. In some embodiments, a first percentage of the total dose, e.g., a first partial dose, is administered on a first day of treatment, a second percentage of the total dose, e.g., a second partial dose, is administered on a subsequent (e.g., second, third, fourth, fifth, sixth, or seventh or later) day of treatment, and a third percentage (e.g., the remaining percentage) of the total dose, e.g., a third partial dose, is administered on a yet subsequent (e.g., third, fourth, fifth, sixth, seventh, eighth, ninth, tenth, or later) day of treatment.
[0272] In some embodiments, a sample of CAR-expressing cells, e.g., CD22 CAR- expressing T cells, comprises about 0.1-100% of total cells expressing at least one CD22 CAR molecule. In some embodiments, a sample of CAR-expressing cells comprises about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1.5%, about 2%, about 2.5%, about 2.5%, about 3%, about 3.5%, about 4%, about 4%, about 4.5%, about 5%, about 5.5%, about 5.5%, about 6%, about 6.5%, about 7%, about 7%, about 7.5%, about 8%, about 8.5%, about 8.5%, about 9%, about 9.5%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, about 35%, about 36%, about 37%, about 38%, about 39%, about 40%, about 41%, about 42%, about 43%, about 44%, about 45%, about 46%, about 47%, about 48%, about 49%, about 50%, about 51%, about 52%, about 53%, about 54%, about 55%, about 56%, about 57%, about 58%, about 59%, about 60%, about 61%, about 62%, about 63%, about 64%, about 65%, about 66%, about 67%, about 68%, about 69%, about 70%, about 71%, about 72%, about 73%, about 74%, about 75%, about 76%, about 77%, about 78%, about 79%, about 80%, about 81%, about 82%, about 83%, about 84%, about 85%, about 86%, about 87%, about 88%, about 89%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100% of total cells expressing at least one CAR molecule.
[0273] In some embodiment, a total dose of CAR-expressing cells (e.g., CD22 CAR- expressing T cells expressing at least one CD22 CAR molecule) comprises about 0.2-20* 106 cells/kg, e.g., about 0.5-5* 106 cells/kg, about 0.1-0.4* 106 cells/kg, about 0.1-0.75* 106 cells/kg, about 0.25-0.75 * 106 cells/kg, about 0.8-1.2* 106 cells/kg, about 1.2-4.0* 106 cells/kg, about 2.5-3.5* 106 cells/kg, about 1-5* 106 cells/kg, about 0.5-1 * 106 cells/kg, about 5-10* 106 cells/kg, about 15-20* 106 cells/kg, 0.6-24* 106 cells/kg, 0.7-23* 106 cells/kg, 0.8-22* 106 cells/kg, 0.9-21 * 106 cells/kg, 1-20* 106 cells/kg, 2-19* 106 cells/kg, 3-18* 106 cells/kg, 4- 17* 106 cells/kg, 5-16* 106 cells/kg, 6-15* 106 cells/kg, 7-14* 106 cells/kg, 8-13* 106 cells/kg, 9-12* 106 cells/kg, or 10-11 * 106 cells/kg. In some embodiments, a total dose of CAR- expressing cells (e.g., CD22 CAR-expressing T cells expressing at least one CD22 CAR molecule) comprises about 0.5-20* 106 cells/kg, e.g., about 0.1 * 106 cells/kg, about 0.2* 106 cells/kg, about 0.3* 106 cells/kg, about 0.4* 106 cells/kg, about 0.5* 106 cells/kg, about 0.6*106 cells/kg, about 0.7* 106 cells/kg, about 0.8* 106 cells/kg, about 0.9* 106 cells/kg, about 1.0* 106 cells/kg, about 1.5* 106 cells/kg, about 2.0* 106 cells/kg, about 2.5* 106 cells/kg, about 3.0* 106 cells/kg, about 3.5* 106 cells/kg, about 4.0* 106 cells/kg, about 4.5*106 cells/kg, about 5.0* 106 cells/kg, about 5.5* 106 cells/kg, about 6.0* 106 cells/kg, about 6.5* 106 cells/kg, about 7.0* 106 cells/kg, about 7.5* 106 cells/kg, about 8.0* 106 cells/kg, about 8.5* 106 cells/kg, about 9.0* 106 cells/kg, about 9.5* 106 cells/kg, about 10* 106 cells/kg, about 10.5* 106 cells/kg, about l l * 106 cells/kg, about 11.5* 106 cells/kg, about 12* 106 cells/kg, about 12.5* 106 cells/kg, about 13* 106 cells/kg, about 13.5* 106 cells/kg, about 14* 106 cells/kg, about 14.5* 106 cells/kg, about 15* 106 cells/kg, about 15.5* 106 cells/kg, about 16* 106 cells/kg, about 16.5* 106 cells/kg, about 17* 106 cells/kg, about 17.5*106 cells/kg, about 18* 106 cells/kg, about 18.5* 106 cells/kg, about 19* 106 cells/kg, about 19.5* 106 cells/kg, or about 20* 106 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 0.5* 106 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 1.0* 106 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 2.0* 106 cells/kg. In some embodiments, the total dose of CD22 CAR-expressing cells comprises about 3.0* 106 cells/kg.
[0274] In some embodiments, a partial dose of CAR-expressing (e.g., CD22 CAR-expressing T cells expressing at least one CD22 CAR molecule) cells comprises about 0.2-20* 106 cells/kg, e.g., about 0.5-5* 106 cells/kg, about 1-5* 106 cells/kg, about 0.5-1 * 106 cells/kg, about 5-10* 106 cells/kg, about 15-20* 106 cells/kg, 0.6-24* 106 cells/kg, 0.7-23* 106 cells/kg, 0.8-22* 106 cells/kg, 0.9-21 * 106 cells/kg, 1-20* 106 cells/kg, 2-19* 106 cells/kg, 3-18* 106 cells/kg, 4-17* 106 cells/kg, 5-16*106 cells/kg, 6-15* 106 cells/kg, 7-14*106 cells/kg, 8- 13* 106 cells/kg, 9-12* 106 cells/kg, or 10-11 * 106 cells/kg. In some embodiments, a partial dose of CAR-expressing cells (e.g., cells expressing at least one CAR molecule) comprises about 0.5-20* 106 cells/kg, e.g., about 0.1 * 106 cells/kg, about 0.2-* 106 cells/kg, about 0.3* 106 cells/kg, about 0.4* 106 cells/kg, about 0.5* 106 cells/kg, about 0.6* 106 cells/kg, about 0.7*106 cells/kg, about 0.8* 106 cells/kg, about 0.9* 106 cells/kg, about 1.0* 106 cells/kg, about 1.5* 106 cells/kg, about 2.0* 106 cells/kg, about 2.5* 106 cells/kg, about 3.0* 106 cells/kg, about 3.5* 106 cells/kg, about 4.0* 106 cells/kg, about 4.5* 106 cells/kg, about 5.0*106 cells/kg, about 5.5* 106 cells/kg, about 6.0* 106 cells/kg, about 6.5* 106 cells/kg, about 7.0* 106 cells/kg, about 7.5* 106 cells/kg, about 8.0* 106 cells/kg, about 8.5* 106 cells/kg, about 9.0* 106 cells/kg, about 9.5* 106 cells/kg, about 10* 106 cells/kg, about 10.5*106 cells/kg, about l l * 106 cells/kg, about 11.5* 106 cells/kg, about 12* 106 cells/kg, about 12.5* 106 cells/kg, about 13* 106 cells/kg, about 13.5* 106 cells/kg, about 14* 106 cells/kg, about 14.5* 106 cells/kg, about 15* 106 cells/kg, about 15.5* 106 cells/kg, about 16* 106 cells/kg, about 16.5* 106 cells/kg, about 17* 106 cells/kg, about 17.5* 106 cells/kg, about 18* 106 cells/kg, about 18.5* 106 cells/kg, about 19* 106 cells/kg, about 19.5* 106 cells/kg, or about 20* 106 cells/kg.
[0275] In some embodiments, the dose of CAR expressing T cells will be based on the weight of the subject. In some embodiments, the dose of the CAR expressing T cells will be partially based on the weight of the subject. In some embodiments, the subject weights less than 100 kg. In some embodiments, the subject weighs at least about 50 kg, at least about 60 kg, at least about 70 kg, at least about 80 kg, or at least about 90 kg. In some embodiments, the subject weights at least about 100 kg. In some embodiments, the subject weighs at least about 110kg, at least about 120kg, at least about 130kg, at least about 140kg, or at least about 150kg. In some embodiments, the subject weighs less than 100 kg and the dose of the CAR expressing T cells will be based on the weight of the subject. In some embodiments, the subject weighs less than 100 kg and the dose of the CAR expressing T cells will be partially based on the weight of the subject. In some embodiments, the subject weighs more than 100 kg and the dose of the CAR expressing T cells will be based on the weight of the subject. In some embodiments, the subject weighs more than 100 kg and the dose of the CAR expressing T cells will be partially based on the weight of the subject.
[0276] In some embodiments, the CAR expressing T cells will be administered by IV infusion. In some embodiments, the CAR expressing T cells are formulated for IV infusion and such formulation comprises DMSO. In some embodiments, the IV infusion duration will comprise about 5-60 minutes. In some embodiments, the IV infusion duration will comprise about 5 minutes, about 10 minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes or about 60 minutes. In some embodiments, the CAR expressing T cells will be contained in an IV bag. In some embodiments, the IV bag will be attached to the subject via tubing. In some embodiments, the tubing comprises non-filtered tubing. In some embodiments, the IV bag will be agitated during the IV infusion. In some embodiments, the IV bag will be gently agitated during the IV infusion. In some embodiments, the IV bag and tubing will be rinsed after infusion of the CAR expressing T cells. In some embodiments, the IV bag and tubing will be rinsed with saline after infusion of the CAR expressing T cells. In some embodiments, the saline used to rinse the IV bag and tubing will be administered to the subject.
CAR therapy format
[0277] In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is frozen. In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is frozen prior to administering to the subject. In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is cryopreserved prior to administering to the subject. In some embodiments, the CAR T-cell therapy (e.g., a CD22 CAR T-cell therapy) is cryopreserved in cryo-storage bags prior to administering to the subject. In some embodiments, the cryo-storage bags are stored in liquid nitrogen. In some embodiments, the cryo-storage bags are stored in the vapor phase of liquid nitrogen.
[0278] In some embodiments, an additional therapy is administered in combination with the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells). In some embodiments, the additional therapy is administered before administering the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells). In some embodiments, the additional therapy is administered substantially concurrent with administering the CAR-expressing T-cells (e.g., CD22 CAR- expressing T-cells). In some embodiments, the additional therapy is administered after administering the CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells). In some embodiments, the additional therapy is administered to treat cytokine release syndrome (CRS). In some embodiments, the additional therapy administered to treat CRS comprises tocilizumab. In some embodiments, the additional therapy administered to treat CRS comprises glucocorticoids. In some embodiments, the additional therapy administered to treat CRS comprises glucocorticoids and tocilizumab. In some embodiments, the additional therapy administered to treat CRS comprises an IL-1 receptor antagonist. In some embodiments, the IL-1 receptor antagonist comprises anakinra. In some embodiments, anakinra is administered to treat CAR T-cell associated toxicity. In some embodiments, anakinra is administered to treat immune effector cell-associated hemophagocytic lymphohistiocytosis (HLH)-like syndrome (IEC-HS).
[0279] In some embodiments, the additional therapy comprises administration of treatments for infection. In some embodiments, the additional therapy comprises administration of therapeutics intended to prevent viral, fungal, bacterial or Pneumocystis infections. In some embodiments the additional therapy comprises administration of one or more hematopoietic growth factors.
Lymphodepletion
[0280] In some embodiments, lymphodepletion is performed on a subject, e.g., prior to administering one or more cells that express a CAR as described herein. Examples of lymphodepletion include, but may not be limited to, nonmyeloablative lymphodepleting chemotherapy, myeloablative lymphodepleting chemotherapy, total body irradiation. [0281] In some embodiments, the lymphodepletion regimen comprises administering one or more chemotherapeutics selected from the group consisting of melphalan, cytoxan, bendamustine, cyclophosphamide, and fludarabine. In some embodiments, the lymphodepletion regimen is also referred to as a lymphodepleting chemotherapy or a lymphodepleting therapy. In some embodiments, a subject is administered lymphodepleting chemotherapy after administration of bridging chemotherapy. In some embodiments, a subject is administered lymphodepleting chemotherapy without prior administration of bridging chemotherapy.
[0282] In some embodiments, the lymphodepletion regimen comprises administering cyclophosphamide at least one, two, three, four, five, or more days before administration of an infusion of CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells). In some embodiments, cyclophosphamide is administered once daily for 1 day, for 2 days, for 3 days, for 4 days for 5 days, for 6 days or for 7 days. In some embodiments, cyclophosphamide is administered once daily for three days beginning 5 days before administration of an infusion of CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells), on days -5, -4, and -3 prior to infusion. In some embodiments cyclophosphamide is administered at a dosage of about 100-800 mg/m2/day, about 200-800 mg/m2/day, about 100-800 mg/m2/day, or about 400-600 mg/m2/day. In some embodiments cyclophosphamide is administered once daily at a dosage of about 100 mg/m2/day, about 150 mg/m2/day, about 200 mg/m2/day, about 250 mg/m2/day, about 300 mg/m2/day, about 350 mg/m2/day, about 400 mg/m2/day, about 450 mg/m2/day, about 500 mg/m2/day, about 550 mg/m2/day, about 600 mg/m2/day, about 650 mg/ m2/day, about 700 mg/m2/day, about 750 mg/m2/day, or about 800 mg/m2/day. In some embodiments, cyclophosphamide is administered at a dosage of about 500 mg/m2/day. In some embodiments, cyclophosphamide is administered once daily at a dosage of about 500 mg/m2/day for one, two, or three days. In some embodiments, cyclophosphamide is administered once daily at a dosage of about 500 mg/m2/day for three days beginning 5 days before administration of an infusion of CAR-expressing T-cells (e.g., CD22 CAR-expressing T-cells), on days -5, -4, and -3 prior to infusion.
[0283] In some embodiments cyclophosphamide is administered for 2 days at a dosage of about 200-600 mg/m2/day (e.g., 200-400 mg/m2/day, about 250-500 mg/m2/day, about 250- 600 mg/m2/day, about 300-500 mg/m2/day, about 300-550 mg/m2/day, about 300-600 mg/m2/day, or about 450-550 mg/m2/day, or about 450 mg/m2/day, or 500 mg/m2/day). In some embodiments cyclophosphamide is administered for 3 days at a dosage of about 200- 600 mg/m2/day (e.g, 200-400 mg/m2/day, about 250-500 mg/m2/day, about 250-600 mg/m2/day, about 300-500 mg/m2/day, about 300-550 mg/m2/day, about 300-600 mg/m2/day, or about 450-550 mg/m2/day, or about 450 mg/m2/day, or 500 mg/m2/day). In some embodiments, cyclophosphamide is administered at a dosage of about 500 mg/m2 per day, for 3 days.
[0284] In embodiments, the lymphodepletion regimen comprises administering fludarabine. In embodiments, fludarabine is administered for 1 day, for 2 days, for 3 days, for 4 days for 5 days, for 6 days or for 7 days. In some embodiments fludarabine is administered at a dosage of about 10-400 mg/ m2/day, about 20-400 mg/ m2/day, about 20-200 mg/ m2/day, or about 25-35 mg/ m2/day. In some embodiments fludarabine is administered at a dosage of about 10 mg/m2/day, about 15 mg/m2/day, about 20 mg/m2/day, about 25 mg/m2/day, about 30 mg/m2/day, about 35 mg/m2/day, about 40 mg/m2/day, about 45 mg/m2/day, about 50 mg/m2/day, about 55 mg/m2/day, about 60 mg/m2/day, about 65 mg/ m2/day, about 70 mg/m2/day, about 75 mg/m2/day, about 80 mg/m2/day, about 85 mg/m2/day, about 90 mg/m2/day, about 95 mg/m2/day, about 100 mg/m2/day, about 110 mg/m2/day, about 120 mg/m2/day, about 130 mg/m2/day, about 140 mg/m2/day, about 150 mg/m2/day, about 160 mg/m2/day, about 170 mg/m2/day, about 180 mg/m2/day, about 190 mg/m2/day, about 200 mg/m2/day, about 250 mg/m2/day, about 300 mg/m2/day, about 350 mg/m2/day, or about 400 mg/m2/day. In some embodiments, fludarabine is administered at a dosage of about 30 mg/m2/day.
[0285] In some embodiments fludarabine is administered for 2 days at a dosage of about 15- 60 mg/m2/day (e.g., 20-40, 25-50, 25-60, 30-50, 30-55, 25-60, or 25-35 e.g., or about 25 mg/m2, or 30 mg/m2). In some embodiments fludarabine is administered for 3 days at a dosage of about 15-60 mg/m2/day (e.g., 20-40, 25-50, 25-60, 30-50, 30-55, 25-60, or 25-35 e.g., or about 25 mg/m2, or 30 mg/m2). In some embodiments, fludarabine is administered at a dosage of about 30 mg/m2 per day, for 3 days.
[0286] In some embodiments, the lymphodepletion regimen comprises administering cyclophosphamide prior to administering CAR T-cells. In some embodiments, the lymphodepletion regimen comprises administering fludarabine prior to administering CAR T-cells. In some embodiments, the lymphodepletion regimen comprises administering cyclophosphamide and fludarabine prior to administering CAR T-cells. In some embodiments, the lymphodepletion regimen comprises administering cyclophosphamide and fludarabine prior to administering CAR T-cells. In some embodiments, the lymphodepletion comprises administering 500 mg/m2 cyclophosphamide once daily for 2 days and 30 mg/m2 fludarabine once daily for 2 days. In some embodiments, the lymphodepletion regimen comprises administering 500 mg/m2 cyclophosphamide once daily for 3 days, and 30 mg/m2 fludarabine once daily for 3 days. In some embodiments, the lymphodepletion regimen comprises administering 500 mg/m2 cyclophosphamide once daily for 4 days, and 30 mg/m2 fludarabine once daily for 4 days.
[0287] In some embodiments, the lymphodepletion regimen is initiated with the administration of the first dose of cyclophosphamide. In some embodiments, the lymphodepletion regimen is initiated with the administration of the first dose of fludarabine. In some embodiments, cyclophosphamide and fludarabine are administered on the same day. In some embodiments, cyclophosphamide and fludarabine are not administered on the same day. In some embodiments, the daily dosages are administered on consecutive days. [0288] In some embodiments, at least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine. In some embodiments, a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine. In some embodiments, second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine. In some embodiments, a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine. In some embodiments, each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered. [0289] In some embodiments, the first dose of cyclophosphamide is administered on day -20, -19, -18, -17, -16, -15, -14, -13, -12, -11, -10, -9, -8, -7, -6, -5, -4, -3, -2, or -1 and the engineered CAR T-cells (e.g., CD22 CAR T-cells) are administered on day 0. In some embodiments, the first dose of fludarabine e-is administered on day -20, -19, -18, -17, -16, - 15, -14, -13, -12, -11, -10, -9, -8, -7, -6, -5, -4, -3, -2, or -1 and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, the first dose of cyclophosphamide and the first dose of fludarabine are administered on day -20, -19, -18, -17, -16, -15, -14, -13, -12, -11, -10, -9, -8, -7, -6, -5, -4, -3, -2, or -1 and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. [0290] In some embodiments, a dose of cyclophosphamide is administered on day -10 and day -9 and day -8, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -9 and day -8 and day -7, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -8 and day -7 and day -6, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -7 and day -6 and day -5, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -6 and day -5 and day -4, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -4 and day -3 and day -2, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide is administered on day -3 and day -2 and day -1, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
[0291] In some embodiments, a dose of fludarabine is administered on day -10 and day -9 and day -8, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -9 and day -8 and day -7, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -8 and day -7 and day -6, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -7 and day -6 and day -5, and the autologous CD22 CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -6 and day -5 and day -4, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -4 and day -3 and day -2, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of fludarabine is administered on day -3 and day -2 and day -1, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
[0292] In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -10 and day -9 and day -8, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -9 and day -8 and day - 7, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -8 and day -7 and day -6, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -7 and day -6 and day - 5, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -6 and day -5 and day -4, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -5 and day -4 and day - 3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -4 and day -3 and day -2, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0. In some embodiments, a dose of cyclophosphamide and a dose of fludarabine are administered on day -3 and day -2 and day - 1, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
[0293] In some embodiments, a dose of cyclophosphamide at 500 mg/m2/day is administered once daily on day-5, day -4, and day -3, a dose of fludarabine at 30 mg/m2/day is administered once daily on day -5, day -4, and day -3, and the engineered CAR T-cells (e.g., autologous CD22 CAR T-cells) are administered on day 0.
[0294] In some embodiments, an additional agent is administered in combination with the lymphodepletion regimen. In some embodiments, the additional agent is administered to address side effects caused by the lymphodepletion regimen. In some embodiments, the additional agent is administered to prevent hemorrhagic cystitis. In some embodiments, the additional agent comprises mesna (sodium 2-mercaptoethane sulfonate) (marketed as Mesnex®). In some embodiments, the additional agent is administered before administering the lymphodepletion regimen. In some embodiments, the additional agent is administered substantially concurrent with administering the lymphodepletion regimen. In some embodiments, the additional agent is administered after administering the lymphodepletion regimen. In some embodiments, the additional agent comprises mesna (sodium 2- mercaptoethane sulfonate) (marketed as Mesnex®) and is administered before administering the lymphodepletion regimen. In some embodiments, the additional agent comprises mesna (sodium 2-mercaptoethane sulfonate) (marketed as Mesnex®) and is administered substantially concurrent with administering the lymphodepletion regimen. In some embodiments, the additional agent comprises mesna (sodium 2-mercaptoethane sulfonate) (marketed as Mesnex®) and is administered after administering the lymphodepletion regimen.
[0295] In some embodiments, a premedication treatment is administered at the same time as the lymphodepletion regimen. In some embodiments, a premedication treatment is administered before the lymphodepletion regimen. In some embodiments, a premedication treatment is administered after the lymphodepletion regimen. In some embodiments, a premedication treatment is administered on the same day as the lymphodepletion regimen on the same day. In some embodiments, a premedication treatment is administered on a different day as the lymphodepletion regimen on the same day.
[0296] In some embodiments, the subject is administered CAR-expressing cells (e.g., autologous CD22 CAR T-cells) about 1-14 days, e.g, 2-13, 3-12, 4-11, 5-10, 2-11, 2-6, or 1- 4 days, after completion of the lymphodepletion regimen. In some embodiments, the lymphodepletion regimen is administered to the subject about 1 week, e.g., about 6, 5, 4, 3, 2, or 1 days, prior to administration of CAR-expressing cells (e.g., autologous CD22 CAR T- cells). In some embodiments, the lymphodepletion regimen is completed, about 1 week, e.g., about 6, 5, 4, 3, 2, or 1 days, prior to administration of CAR-expressing cells (e.g., autologous CD22 CAR T-cells).
[0297] In some embodiments, when the subject has LBCL, e.g., relapsed or refractory LBCL, the lymphodepletion regimen comprises administering 500 mg/m2 cyclophosphamide daily for 3 days, e.g., 3 doses, and 30 mg/m2 fludarabine daily for 3 days, e.g., 3 doses. In some embodiments, when the subject has LBCL, e.g., relapsed or refractory LBCL, the lymphodepletion regimen comprises administering a dose of 500 mg/m2 cyclophosphamide daily and a dose of 30 mg/m2 fludarabine daily for 3 days, e.g., 3 doses of each chemotherapeutic, on day -5, day -4, and day -3, followed by administration of CAR- expressing cells (e.g., autologous CD22 CAR T-cells) on day 0.
[0298] In some embodiments, the subject is administered a first lymphodepletion regimen and/or a second lymphodepletion regimen. In some embodiments, the first lymphodepletion regimen is administered before the second lymphodepletion regimen. In some embodiments, the second lymphodepletion regimen is administered before the first lymphodepletion regimen. In some embodiments, the first lymphodepletion regimen comprises cyclophosphamide and fludarabine, e.g., 500 mg/m2 cyclophosphamide daily for 3 days, and 30 mg/m2 fludarabine daily for 3 days
[0299] In some embodiments, a lymphodepleting chemotherapy is administered to the subject prior to, concurrently with, or after administration (e.g., by infusion) of CAR T-cells, e.g., cells described herein. In an example, the lymphodepleting chemotherapy is administered to the subject prior to administration of CAR T-cells. For example, the lymphodepleting chemotherapy ends 1-5 days (e.g., 1, 2, 3, 4 or 5 days) prior to CAR T-cell infusion. In some embodiments, multiple doses of CAR T-cells are administered.
Premedication treatment [0300] In some embodiments, the subject may receive one or more therapies before treatment with the CD22-directed CAR T-cell therapy as a premedication treatment. In some embodiments, the premedication treatment comprises levietiracetam. In some embodiments, the premedication comprises allopurinol. In some embodiments, the premedication comprises allopurinol administered orally. In some embodiments, the premedication comprises allopurinol administered orally daily. In some embodiments, the premedication comprises allopurinol administered orally once daily, or twice daily three times daily, four times daily, five times daily, six times daily, seven times daily, eight time daily, nine times, or ten times daily. In some embodiments, the premedication comprises allopurinol administered orally every other day, or every third day. In some embodiments, the allopurinol comprises a dose of about 50-1000 mg/m2/dose. In some embodiments, the allopurinol comprises a dose of about 50 mg/m2/dose, about 100 mg/m2/dose, about 150 mg/m2/dose, about 200 mg/m2/dose, about 250 mg/m2/dose, about 300 mg/m2/dose, about 350 mg/m2/dose, about 400 mg/m2/dose, about 450 mg/m2/dose, about 500 mg/m2/dose, about 550 mg/m2/dose, about 600 mg/m2/dose, about 650 mg/m2/dose, about 700 mg/m2/dose, about 750 mg/m2/dose, about 800 mg/m2/dose, about 850 mg/m2/dose, about 900 mg/m2/dose, about 950 mg/m2/dose, or about 100 mg/m2/dose. In some embodiments, the allopurinol comprises a dose of about 300 mg/m2 administered orally once per day. In some embodiments, the allopurinol comprises a dose of about 100 mg/m2/dose administered orally three times per day.
[0301] In some embodiments, the premedication comprises an antihistamine. In some embodiments, the antihistamine is administered orally. In some embodiments, the antihistamine is administered by IV. In some embodiments, the antihistamine comprises diphenhydramine. In some embodiments, the antihistamine comprises diphenhydramine and is administered orally. In some embodiments, the antihistamine comprises diphenhydramine and is administered by IV. In some embodiments, the diphenhydramine comprises a dose of about 10-100 mg/dose. In some embodiments, the diphenhydramine comprises a dose of about 10 mg/dose, about 15 mg/dose, about 20 mg/dose, about 25 mg/dose, about 30 mg/dose, about 35 mg/dose, about 40 mg/dose, about 45 mg/dose, about 50 mg/dose, about 55 mg/dose, about 60 mg/dose, about 65 mg/dose, about 70 mg/dose, about 75 mg/dose, about 80 mg/dose, about 85 mg/dose, about 90 mg/dose, about 95 mg/dose, or about 100 mg/dose. In some embodiments, the premedication comprises diphenhydramine administered orally with a dose of about 25-50 mg/dose. In some embodiments, the premedication comprises diphenhydramine administered by IV with a dose of about 25-50 mg/dose. [0302] In some embodiments, the premedication comprises acetaminophen. In some embodiments, the acetaminophen is administered orally. In some embodiments, the acetaminophen is administered by IV. In some embodiments, the acetaminophen comprises a dose of about 100-1000 mg. In some embodiments, the acetaminophen comprises a dose of about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 350 mg, about 400 mg, about 450 mg, about 500 mg, about 550 mg, about 600 mg, about 650 mg, about 700 mg, about 750 mg, about 800 mg, about 850 mg, about 900 mg, about 950 mg, about 1000 mg. In some embodiments, the premedication comprises acetaminophen administered orally in a dose of about 650/mg. In some embodiments, the premedication comprises acetaminophen administered by IV with a dose of about 650 mg.
[0303] In some embodiments, the premedication is administered before treatment with the autologous CD22-directed CAR T-cell therapy. In some embodiments, the premedication is administered substantially concurrent with the treatment with the autologous CD22-directed CAR T-cell therapy. In some embodiments, the premedication is administered substantially after the treatment with the autologous CD22-directed CAR T-cell therapy. In some embodiments, the premedication is administered from about 5 minutes before to about 10 hours before treatment with the autologous CD22-directed CAR T-cell therapy. In some embodiments, the premedication is administered about 5 minutes, aboutlO minutes, about 15 minutes, about 20 minutes, about 25 minutes, about 30 minutes, about 35 minutes, about 40 minutes, about 45 minutes, about 50 minutes, about 55 minutes, about 60 minutes, about 65 minutes, about 70 minutes, about 75 minutes, about 80 minutes, about 85 minutes, about 90 minutes, about 95 minutes, about 100 minutes, about 105 minutes, about 110 minutes, about
115 minutes, about 120 minutes, about 2 hours, about 2.5 hours, about 3 hours, about 3.5 hours, about 4 hours, about 4.5 hours, about 5 hours, about 5.5 hours, about 6 hours, about 6.5 hours, about 7 hours, about 7.5 hours, about 8 hours, about 8.5 hours, about 9 hours, about 9.5 hours, or about 10 hours before treatment with the autologous CD22-directed CAR T-cell therapy. In some embodiments the premedication comprises diphenhydramine and is administered 30-120 minutes before the subject receives a dose of the autologous CD22- directed CAR T-cell therapy. In some embodiments the premedication comprises acetaminophen and is administered 30-120 minutes before the subject receives a dose of the autologous CD22-directed CAR T-cell therapy. In some embodiments the premedication comprises diphenhydramine and acetaminophen before the subject receives a dose of the autologous CD22-directed CAR T-cell therapy. In some embodiments the premedication comprises diphenhydramine and acetaminophen and is administered 30-120 minutes before the subject receives a dose of the autologous CD22-directed CAR T-cell therapy.
Bridging therapy
[0304] In some embodiments, a bridging therapy is administered to the subject. In some embodiments, the bridging therapy is administered after leukapheresis. In some embodiments, the bridging therapy is administered before lymphodepletion. In some embodiments, the bridging therapy is administered at least about 1-10 days before lymphodepletion. In some embodiments, the bridging therapy is administered at least about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days or about 10 days before lymphodepletion.
[0305] In some embodiments, the bridging therapy is administered to the subject for about 1- 10 days. In some embodiments, the bridging therapy is administered to the subject for about 1-4 days. In some embodiments, the bridging therapy is administered for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days or about 10 days.
[0306] In some embodiments, the bridging therapy is administered orally. In some embodiments, the bridging therapy is administered by IV. In some embodiments the bridging therapy comprises dexamethasone. In some embodiments, the dexamethasone comprises a daily dose of about 10-100 mg, or about 20-40 mg. In some embodiments, the dexamethasone comprises a daily dose of about 10-100 mg or mg equivalent. In some embodiments the dexamethasone comprises a daily dose of about 10 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 35 mg, about 40 mg, about 45 mg, about 50 mg, about 55 mg, about 60 mg, about 65 mg, about 70 mg, about 75 mg, about 80 mg, about 85 mg, about 90 mg, about 95 mg, or about 100 mg. In some embodiments, the bridging therapy is administered orally and comprises dexamethasone comprising a daily dose of about 20-40 mg. In some embodiments, the bridging therapy is administered by IV and comprises dexamethasone comprising a daily dose of about 20-40 mg. In some embodiments, the bridging therapy is administered orally and comprises dexamethasone comprising a daily dose of about 20-40 mg for 1-4 days. In some embodiments, the bridging therapy is administered by IV and comprises dexamethasone comprising a daily dose of about 20-40 mg for 1-4 days. [0307] In some embodiments, the bridging therapy comprises radiation treatment. In some embodiments, the radiation therapy is administered to sites of symptomatic, bulky or extranodal disease.
[0308] In some embodiments, the bridging therapy comprises a course of gemcitabineoxaliplatin plus rituximab (R-GemOx). In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is less than 3 weeks, and the bridging therapy is administered during that period. In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks, and the bridging therapy is administered during that period. In some embodiments, the bridging therapy comprises gemcitabine-oxaliplatin plus rituximab (R-GemOx). In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises R-GemOx during that time period. In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises a portion of R-GemOx during that time period. In some embodiments, the bridging therapy comprises R-polatuzumab vedotin. In some embodiments, the bridging therapy comprises a portion of R-polatuzumab vedotin. In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises R-polatuzumab vedotin during that time period. In some embodiments, the time between leukapheresis and administering the CAR T-cell therapy is more than 3 weeks and the bridging therapy comprises a portion of R-polatuzumab vedotim during that time period.
LBCL and Response Criteria
[0309] In some embodiments, a response to the CAR T-cell therapy described herein comprises radiographic evidence. In some embodiments, a response to the CAR T-cell therapy described herein comprises clinical evidence. In some embodiments, a response to the CAR T-cell therapy described herein comprises a combination of radiographic evidence and clinical evidence. In some embodiments, a response to CAR T-cell therapy is determined at least in part from a positron emission tomography (PET)-computed tomography (CT) scan. In some embodiments, a response to CAR T-cell therapy is determined at least in part from a contrast enhanced CT scan. In some embodiments, the PET-CT scan detects an uptake of a radiotracer compound. In some embodiments, the radiotracer compound is fluorodeoxy glucose . [0310] In some embodiments, a subject comprises a complete response to the CAR T-cell therapy. In some embodiments, a subject comprises a partial response to the CAR T- cell therapy. In some embodiments, a subject comprises no response to the CAR T-cell therapy. In some embodiments, a subject comprises progressive disease after receiving the CAR T- cell therapy.
[0311] In some embodiments, the subject has a complete response to CAR T-cell therapy. In some embodiments, the complete response to CAR T-cell therapy comprises no new lesions detected from a PET-CT scan analysis. In some embodiments, the complete response to CAR T-cell therapy comprises no evidence of fluorodeoxyglucose-avid disease in bone marrow. In some embodiments, the complete response to CAR T-cell therapy comprises a score of 1, 2 or 3 on a five point scale from a PET-CT scan analysis. In some embodiments, the score of 1, 2, or 3 on a five point scale from a PET-CT scan comprises one or residual masses. In some embodiments, the score of 1, 2, or 3 on a five point scale from a PET-CT scan comprises no or residual mass. In some embodiments, the uptake of fluorodeoxyglucose may be higher than normal. In some embodiments, the uptake of fluorodeoxy glucose may be higher than normal/baseline in mediastinum. In some embodiments, the uptake of fluorodeoxyglucose may be higher than normal/baseline in liver. In some embodiments, the uptake of fluorodeoxyglucose in sites of disease involvement may be compared to sites proximal to disease involved sites to infer the response to CAR T-cell therapy. In some embodiments, the uptake of fluorodeoxyglucose in sites of disease involvement may be at a similar level compared to sites proximal to disease involvement. In some embodiments, the complete response to CAR T-cell therapy comprises no extra lymphatic sites of disease. In some embodiments, the complete response to CAR T-cell therapy comprises no extra lymphatic sites of disease according to a CT scan analysis. In some embodiments, the complete response to CAR T-cell therapy comprises target nodes and/or nodal masses of less than about 0.5-3.0 cm in the longest diameter of a lesion (LDi), about 1-2 cm in LDi, about 0.5 cm in LDi, about 1.0 cm in LDi, about 1.5 cm in LDi, about 2.0 cm in LDi, about 2.5 cm in LDi, or about 3.0 cm in LDi. In some embodiments, the complete response to CAR T-cell therapy comprises target nodes and/or nodal masses of less than about 1.5 cm.
[0312] In some embodiments, the subject comprises a partial response to CAR T-cell therapy. In some embodiments, the partial response to CAR T-cell therapy comprises a partial metabolic response score of 4 or 5. In some embodiments, the complete response to CAR T-cell therapy comprises reduced fluorodeoxyglucose uptake compared to baseline and residual mass or masses of any size based on a PET-CT scan. In some embodiments, the partial response reflects disease responding to treatment. In some embodiments, the partial response comprises residual fluorodeoxyglucose uptake in new bone marrow lesions higher than fluorodeoxyglucose uptake in normal bone marrow. In some embodiments, the partial response reflects residual disease. In some embodiments, the partial response is measured mid-treatment and reflects responding disease. In some embodiments, the partial response is measured at the end of treatment and indicates residual disease. In some embodiments, the partial response comprises a decrease of about 50% or greater in a sum of the product of the perpendicular diameters for multiple lesions determined by a CT-based analysis. In some embodiments, the partial response comprises a decrease of about 50% or greater in a sum of the product of the perpendicular diameters for up to 6 target measurable nodes and extranodal sites determined by a CT-based analysis.
[0313] In some embodiments, the subject comprises no response to CAR T-cell therapy. In some embodiments, the no response comprises no metabolic response, and/or a score of 4 or 5 on a five-point scale with no significant change in fluorodeoxyglucose uptake from baseline at interim or end of treatment based on a PET-CT analysis. In some embodiments, the no response comprises less than about 50% decrease from baseline in a sum of the product of the perpendicular diameters for multiple lesions of up to 6 dominant, measurable nodes and extranodal sites based a on CT-scan analysis. In some embodiments, the no response comprises no criteria for disease progression being met.
[0314] In some embodiments, the subject comprises progressive disease after receiving a CAR T-cell therapy. In some embodiments, the progressive disease comprises progressive metabolic disease based on a PET-CT scan. In some embodiments, the progressive disease comprises a score of 4 or 5 on a five-point scale with an increase in intensity of fluorodeoxyglucose uptake compared to baseline based on a PET-CT scan, or new fluorodeoxyglucose-avid foci consistent with lymphoma at interim or end of treatment assessment based on a PET-CT scan, or a combination thereof. In some embodiments, the progressive disease comprises progression in the cross product of the LDi and perpendicular diameter, new or recurrent involvement or regrowth of previously resolved lesions, a new node of greater than about 1.5 cm in any axis, a new extranodal site of greater than about 1 cm in any axis, assessable disease of any size attributed to lymphoma, new or recurrent involvement from bone marrow, or a combination thereof. In some embodiments, a node or lesion comprises an LDi of greater than about 1.5 cm, an increase of greater than about 50% compared to a cross product of the LDi and perpendicular diameter nadir and an increase in LDi or shortest diameter of a lesion (SDi), an increase in LDi or SDi of about 0.5 or greater for lesions of less than about 2 cm, an increase in LDi or SDi or about 1.0 or greater for lesions of more than about 2 cm, new or recurrent splenomegaly, new or clear progression of preexisting non-measured lesions, or some combination thereof.
EXAMPLES
Example 1: Phase 2 Study of Autologous Anti-CD22 Chimeric Antigen Receptor T Cells in patients with Relapsed or Refractory Large B Cell Lymphoma
[0315] This example describes a phase 2, multi-center Phase 2, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B-cell antigen CD22 (CRG-022) in participants with R/R LBCL.
Study design
[0316] FIG. 1 depicts the study design. Four cohorts of subjects are treated as part of this study. Subjects in Cohort 1 are treated with a target dose of IxlO6 transduced CAR T-cells/kg targeting CD22. Participants in this cohort will have received prior CD19-direct CAR T therapy. Subjects in Cohort 2 are treated with nonconforming CAR T-cells that are deemed safe to administer. Subjects in Cohort 3 are treated with a target dose of IxlO6 transduced CAR T-cells/kg targeting CD22. Participants in this cohort will have received prior bispecific T-cell engager antibody therapy.
[0317] FIG. 4 depicts the study design including subjects in Cohort 4. Subjects in Cohort 4 are treated with a target dose of IxlO6 transduced CAR T-cells/kg targeting CD22. Participants in this cohort will not have received prior CAR T therapy (z.e., they are CAR- naive) and may have received prior bispecific T-cell engager antibody therapy.
[0318] Patient T-cells obtained from autologous apheresis product are transduced with the anti-CD22 lentiviral vector, expanded in vitro and then frozen for future administration. [0319] Patients are given conditioning chemotherapy prior to CAR T-cell infusion with the intent of lymphodepletion. Conditioning chemotherapy consists of fludarabine and cyclophosphamide as described further below. The chemotherapy is planned so that the last dose is completed 3 days prior to administration of the CAR T-cells. Alternatively, the CAR T-cells are administered 2 to 7 days after the last dose of conditioning chemotherapy. If the period from chemotherapy to CRG-022 infusion is delayed 4 or more weeks, the patient is, e.g., be re-treated with lymphodepleting chemotherapy prior to CAR T-cell infusion. [0320] Subjects must meet certain fitness criteria to receive the CAR T-cell infusion. Subjects will be hospitalized for the product administration.
[0321] The CAR T-cell infusion is administered as a single infusion at a target dose of 0.5xl06 CAR transduced T-cells/kg based upon the subject’s weight at the time of enrollment.
[0322] The CAR T-cell infusion is administered as a single infusion at a target dose of l.OxlO6 CAR transduced T-cells/kg based upon the subject’s weight at the time of enrollment.
[0323] Following the infusion of the CAR T-cells, patients are evaluated for safety daily in the hospital for 7 days. Patients may be discharged on Day 8 or later. Monitoring will continue until Month 24, disease progression, or start of a subsequent anti-cancer therapy. All adverse events (including serious adverse events) will be reported during this period.
[0324] After 24 months, or earlier for patients with disease progression or who start a subsequent anti-cancer therapy, patients are moved into long-term follow-up (LTFU). Patients who remain on-study are monitored every 6 months up to Month 60 and then yearly thereafter until Year 15. Patients are monitored for disease status with imaging as clinically indicated.
[0325] The duration of the study for individual patients will vary depending on a patient’s screening requirements, response to treatment, and survival status. For a patient who completes the entire protocol from the date of ICF signature through the completion of the LTFU period, the duration of the study will take up to 15 years (± 2 months) to complete. Subject Inclusion Criteria
[0326] A subject having relapsed or refractory LBCL is eligible to be included in the study if they meet all of the following criteria:
(i) at least 18 years of age at the time of study;
(ii) have histologically confirmed LBCL as follows:
(a) Diffuse LBCL (DLBCL) not otherwise specified (NOS), including germinal center B-cell (GCB) type or active B-cell (ABC) type;
(b) High-grade B-cell lymphoma with MYC and BCL2 and/or BCL6 rearrangements (double-hit or triple-hit lymphoma);
(c) High-grade B-cell lymphoma;
(d) DLBCL arising from follicular lymphoma (FL; transformed follicular lymphoma); (e) DLBCL arising from marginal zone lymphoma (MZL; transformed marginal zone lymphoma);
(f) Primary mediastinal large B-cell lymphoma (PMBL);
(g) FL, Grade 3B/follicular large B-cell lymphoma (FLBL);
(iii) have relap sed/refractory disease after the last therapy, defined as follows:
(a) radiographic findings:
• PD/SD after the last therapy; or
• radiographic disease progression after PR/CR after the last therapy; or
(b) presence of viable lymphoma in a biopsy obtained after therapy;
(iv) have at least one radiographically measurable lesion;
(v) have a washout period of at least 2 weeks or five half-lives, whichever is shorter, since any prior cancer therapy, at the time the patient undergoes leukapheresis, except as follows:
(a) Prior bendamustine: a minimum of 6 months must have elapsed from the last infusion of bendamustine to enrollment in the current study;
(b) Prior CD19-directed CAR T-cell therapy: a minimum of 30 days must have elapsed between the CD19-directed CAR T-cell infusion and enrollment to the current study;
(c) Prior bispecific T-cell engaging antibody therapy: a minimum of 30 days must have elapsed between the last dose of bispecific T-cell engaging antibody therapy and enrollment to the current study;
(d) Autologous stem cell transplant: a minimum of 6 weeks must have elapsed from the stem cell infusion to enrollment to the current study;
(e) Central nervous system (CNS) prophylaxis (e.g., intrathecal methotrexate): must be stopped at least 1 week prior to enrollment on the current study;
(f) Prior corticosteroids: a minimum of 7 days prior to leukapheresis;
(vi) have a tumor tissue biopsy sample following the most recent therapy available for central pathology evaluation; results of this evaluation do not need to be available prior to enrollment. A formalin-fixed paraffin-embedded tissue block, sectioned tissue on unstained slides, or a fresh biopsy provided in neutral buffered formalin, along with a corresponding local pathology report indicating CD 19, CD20, and CD22 and CD79b expression is required for submission to the central pathology laboratory
(vii) have Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1; (viii) have adequate hematologic and end-organ function, defined as the following: a. ANC > 1000/pL; b. platelet count > 75,000/pL or > 50,000/pL in patients with known presence of marrow disease; c. absolute lymphocyte count (ALC) > 100/pL; d. estimated creatinine clearance > 45 mL/minute by Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) methods, or on the basis of 24-hour urine collection or other institutional standard methods; e. serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) < 2.5 times the upper limit of normal (ULN); f. total bilirubin < 1.5 mg/dL (except in patients with Gilbert syndrome); g. cardiac left ventricular ejection fraction (LVEF) >45% and no evidence of pericardial effusion; h. blood oxygen saturation (SaO2) > 92% on room air; and i. serum albumin > 2.5 g/dL;
(ix) have a negative serum pregnancy test result within 7 days prior to leukapheresis for subjects of childbearing age and capacity;
(x) have recovered from nonhematologic toxicities due to prior therapy to grade < 1 (except for clinically non-significant toxicities such as alopecia)
Cohort-Specific Criteria
[0327] For cohort 1, patients must have previously received a CD19-directed CAR T-cell therapy. Patients may have received additional therapies after a CD19-directed CAR T-cell therapy. Subjects part of Cohort 1 must have previously received a CD19-directed CAR T- cell therapy. Additional therapies after the CD19-directed CAR T-cell therapy are permissible. Subjects have no prior exposure to bispecific T-cell engaging antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®), etc). For cohort 3, patients must have received at least two prior lines of therapy with one regimen including a bispecific T-cell engager antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®). In addition, they must meet one of the following conditions: a. Relapsed disease following a CR or progressive disease following a PR to bispecific T-cell engager antibody therapy or SD/PD as best response to bispecific T-cell engager antibody therapy
[0328] Subjects part of Cohort 3 must have received at least two prior lines of therapy with one regimen including a bispecific T-cell engager antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®)). In addition, they must meet the following conditions:
(i) Relapsed disease following a CR; or
(ii) progressive disease following a PR to bispecific T-cell engager antibody therapy or SD/PD as best response to bispecific T-cell engager antibody therapy.
[0329] Subjects part of Cohort 4 must have had no prior treatment with a CAR T-cell therapy including approved and experimental autologous CAR T-cell products and no more than two prior lines of therapy. In addition, they must meet the following conditions:
(i) received at least 1 prior chemoimmunotherapy (CIT) containing regimen (z.e., regimen must contain a CD20-targeted therapy plus an anthracycline)
(ii) can be refractory to prior CIT or relapsed after PR or CR
(iii) prior bispecific T-cell engaging antibody therapy (e.g., mosunetuzumab (marketed as Lunsumio®), glofitamab (marketed as Columvi®), epcoritimab (marketed as Epkinly®)) containing regimens are allowed, however at least 30 days must have passed from the last dose to the time of leukapheresis.
Subject Exclusion Criteria
[0330] A subject having relapsed or refractory LBCL will be ineligible to be included in the study if they meet all of the following criteria:
(i) subjects who have a history of malignancy other than the lymphoma under investigation in this study, except subjects with the following malignancies/treatment characteristics are eligible:
(a) subjects with a history of malignancy that has been treated with curative intent at least 2 years prior to screening and with no evidence of relapse, if no concurrent anti-cancer therapy (except hormonal therapy) is being given;
(b) subjects with a history of malignancy with a negligible risk of metastasis or death (e.g., 5-year OS rate > 90%), such as adequately treated carcinoma in situ of the cervix, non-melanoma skin carcinoma, localized prostate cancer, ductal carcinoma in situ, or stage I uterine cancer; (c) subjects who have prostate cancer with no evidence of metastatic disease and are not on active therapy, except anti-androgen therapy;
(d) subjects with active fungal, bacterial, viral, or other infection that requires intravenous antimicrobials;
(ii) subjects with active invasive fungal infection should be excluded even if the treatment is oral antimicrobials;
(iii) subjects who received prior allogeneic stem cell or solid organ transplant;
(iv) subjects who received prior allogeneic CAR therapy;
(v) subjects who received prior anti-CD52 antibody therapy;
(vi) subjects who received prior bispecific T-cell engager antibody therapy (e.g., glofitamab, epcoritamab) may not be enrolled in cohort 1;
(vii) subjects with history of central nervous system (CNS) involvement of lymphoma within 1 year prior to enrollment;
(viii) subjects with ongoing cardiac involvement of lymphoma;
(ix) subjects with a history of CD22-directed therapy for lymphoma;
(x) subjects with a history of infection with any of the following: human immunodeficiency virus (HIV); hepatitis B virus (HBV), as determined by positivity for hepatitis B serum antigen (HBsAg) or hepatitis B core antibody (HbcAb); or hepatitis C virus (HCV), as determined by positivity for anti-HCV antibody);
(xi) a subject with a history of infection with HBV or HCV may enroll if viral load is undetectable per quantitative PCR and/or nucleic acid testing.
(xii) subjects with significant, uncontrolled concomitant disease that could affect compliance with the protocol or interpretation of results, including the following:
(a) history of myocardial infarction, cardiac angioplasty or stenting, unstable angina, serious cardiac arrhythmia or other clinically significant cardiac disease within 12 months prior to enrollment;
(b) active pulmonary disease (such as severe obstructive pulmonary disease, idiopathic pulmonary fibrosis, organizing pneumonia, drug-induced pneumonitis, idiopathic pneumonitis). History of limited radiation pneumonitis is allowed;
(c) clinically significant liver disease, including cirrhosis; (d) clinically significant neurological disease (such as cerebrovascular ischemia/hemorrhage, dementia, seizure disorder, or cerebellar disease);
(xiii) subjects with a history of autoimmune disease resulting in significant end-organ disease or requiring systemic immunosuppression and/or systemic disease modifying agents within the last 2 years:
(a) physiologic dose of corticosteroid (prednisone 7.5 mg/day or equivalent) is acceptable
(b) subjects with a history of autoimmune-related hypothyroidism on a stable dose of thyroid replacement hormone may be eligible
(c) subjects with controlled Type 1 diabetes mellitus who are on an insulin regimen are eligible for the study
(d) subjects with history of immune thrombocytopenic purpura or autoimmune hemolytic anemia not requiring active treatment may be eligible.
(xiv) subjects with evidence of moderate to severe forms of primary immunodeficiencies;
(xv) subjects with a history of severe, immediate hypersensitivity reaction attributed to aminoglycosides or any of the agents used in this study e.g., DMSO, tocilizumab)
(xvi) subjects that are pregnant or breastfeeding
(xvii) subjects who underwent recent major surgery (within 4 weeks prior to leukapheresis), other than for diagnosis;
(xviii) subjects with any in-dwelling line or drain (e.g., percutaneous nephrostomy tube, in dwelling Foley catheter, biliary drain), ommaya reservoirs. Dedicated central venous access catheters, such as a Port-a-Cath or Hickman catheter, are permitted
(xix) subjects with a history of symptomatic deep vein thrombosis or pulmonary embolism requiring anti coagulation therapy within 6 months prior to enrollment
(xx) subjects who received treatment with a live, attenuated vaccine within 6 weeks prior to initiation of study treatment or who are anticipated to need such a vaccine during the course of the study
(xxi) subj ects with Richter’ s transformation of CLL
(xxii) subjects with T cell/histiocyte-rich large B-cell lymphoma. Patients with Burkitt lymphoma. (xxiii) subjects who require urgent therapy due to mass effects of tumor of impending oncologic emergency (e.g., bowel obstruction, major blood vessel compression or tumor lysis syndrome)
(xxiv) subjects with any other significant, uncontrolled concomitant disease that could affect compliance with the protocol or interpretation of results
(xxv) subjects who, in the investigator’s judgement, are unlikely to complete all protocol required study visits or procedures, including follow up visits, or comply with the study requirements for participation
(xxvi) subjects undergoing concurrent treatment with any other investigational agent, except for the investigative agent under study. Examples of other investigational agents that would be exclusionary include supportive care agents like investigational antifungals.
(xxvii) Subjects who require growth factor support or platelet transfusion within the past 7 days to satisfy ANC or platelet count eligibility criteria
Lymphodepleting Chemotherapy
[0331] The subject is administered lymphodepleting chemotherapy, e.g., fludarabine (30 mg/m2/dayx3 days) and cyclophosphamide (500 mg/m2/dayx3 days). The chemotherapy is planned so that the last dose is completed 2-4 days prior to infusion of the CAR T-cells.
Premedication for Treatment Regimen
[0332] Subjects may receive allopurinol orally (300 mg) once daily or at 100 mg/m2/dose two to three times in a day beginning the day prior to cell infusion or at the time of lymphodepletion.
[0333] Subjects receive diphenhydramine (25-50 mg/dose) or another Hl antihistamine.
[0334] Subjects receive acetaminophen (650 mg/dose).
Treatment Regimen
[0335] Participants receive anti-CD22 CAR T-cell treatment consisting of a single infusion of CD22 CAR transduced autologous T cells administered intravenously initially at a target dose l.OxlO6 CAR-transduced T-cells/kg. The cell dose is based upon the weight at time of enrollment. The dose of anti-CD22 CAR T-cells is calculated using actual body weight, capped at 100 kg. After a preliminary safety assessment, a lower dose of 0.5xl06 CAR- transduced T-cells/kg may be evaluated.
[0336] A preliminary safety assessment is performed after the first 10 patients in Cohort 1 are treated at the target dose of CRG-022 and have been followed for at least 28 days after CRG- 022 infusion. The study will continue to enroll patients during the preliminary safety assessment.
Example 2: Phase 1 Study of Autologous Anti-CD22 Chimeric Antigen Receptor T Cells in patients with Relapsed or Refractory Large B Cell Lymphoma
[0337] This example describes a phase 1, single-center Phase 1, open-label study evaluating the efficacy and safety of an autologous CAR T-cell product targeting the pan B-cell antigen CD22 in participants with R/R LBCL who had received previous lines of therapy. This example informed the starting dose for the Phase 2 study.
Study design
[0338] A cohort of 38 subjects were treated with transduced CAR T-cells targeting CD22 as part of this study. Two different dosing regimens were used for different subsets of subjects: either IxlO6 CAR T-cell s/kg (29 subjects) or 3xl06 cells/kg (9 subjects).
Results
[0339] The overall rate of progression free survival (PFS) at 6 months was 47% and the median PFS was 3.0 months (95% CI 1.7-28.7). The median survival in this clinical trial was 14.1 months in the overall population (FIG. 2).
[0340] Subjects treated with CD22 CAR T-cell therapy experienced an immune toxicity referred to as cytokine release syndrome (CRS). The summary of results related to CRS are shown in FIG. 3. The majority of patients treated with CD22 CAR T-cell therapy had mild CRS, reported as Grades 1 or 2. Only a single patient experienced Grade 3 CRS at DL2. This is in contrast to outcomes reported with approved CAR T treatments, where up to 23% of patients experienced Grade 3 or higher CRS. One subject treated with CD22 CAR T-cell therapy had Grade 2 CRS and developed septicemia, deemed possibly related to CD22 CAR T-cell therapy, leading to multi-organ failure and death at day 41.
[0341] A second type of toxicity associated with CAR T-cell therapies is immune effector cell-associated neurotoxicity syndrome (ICANS). The summary of results related to ICANS are shown in FIG. 3. In this CD22 CAR T-cell therapy trial, 13% of patients experienced ICANS of Grades 1 or 2 severity. There were no reports of patients with ICANS of Grades 3 or above. By contrast, up to 28% of patients treated with CD19 CAR T-cell therapy developed ICANS of Grade 3 or above during clinical testing.

Claims

CLAIMS WHAT IS CLAIMED IS:
1. A method of treating a relap sed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of autologous CD22-specific chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises
(i) an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain,
(ii) a CD8a transmembrane domain;
(iii) a 4-lBB costimulatory domain; and
(iv) a CD3zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy and
(a) has not received a prior CAR T-cell therapy, and/or
(b) wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
2. The method according to claim 1, wherein the dose of autologous CD22-specific CAR T- cells is less than about 0.8* 106 autologous CD22-specific CAR T-cells/kg.
3. The method according to claim 1, wherein the dose of engineered CAR T-cells is at least about 1.2* 106 CAR T-cells/kg.
4. A method of treating a relap sed/refractory hematological cancer in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective dose of engineered chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises
(i) an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain,
(ii) a CD8a transmembrane domain;
(iii) a 4-lBB costimulatory domain; and
(iv) a CD3zeta intracellular signaling domain; wherein the dose of autologous CD22-specific CAR T-cells is
(a) less than about 0.8* 106 autologous CD22-specific CAR T-cells/kg, or
(b) at least about 1.2x 106 autologous CD22-specific CAR T-cells/kg.
5. The method according to claim 4, wherein the hematological cancer is a relapsed/refractory hematological cancer.
6. The method according to claim 4, wherein the human subject has received at least one prior line of therapy and has not received a prior CAR T-cell therapy.
7. The method according to claim 4, wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
8. A method of treating a relapsed/refractory hematological cancer in a human subject in need thereof, the method comprising
(a) administering to the human subject
(i) a dose of cyclophosphamide at about 500 mg/m2/day to about 600 mg/m2/day, wherein the dose of cyclophosphamide is administered daily for three days, and
(ii) a dose of fludarabine at about 30 mg/m2/day, wherein the dose of fludarabine is administered daily for three days; and
(b) administering to the human subject a therapeutically effective dose of engineered chimeric antigen receptor (CAR) T-cells to treat the hematological cancer in the human subject; wherein the CAR comprises
(i) an extracellular domain comprising (A) an anti-human CD22 binding domain and (B) a portion of a CD8a extracellular domain,
(ii) a CD8a transmembrane domain;
(iii)a 4-1BB costimulatory domain; and
(iv)a CD3zeta intracellular signaling domain; wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
9. The method according to any one of claims 1-8, wherein the human subject has received 2 or more prior lines of therapy.
10. The method according to any one of claims 1-9, wherein the human subject has received 2 or more prior lines of therapy, wherein at least one of the 2 or more prior lines of therapy a bispecific T-cell engager antibody therapy.
11. The method according to any one of claims 1-10, wherein the human subject has received no more than 5 prior lines of therapy.
12. The method according to any one of claims 1-11, wherein the human subject has not received a prior CAR T-cell therapy.
13. The method according to any one of claims 1-11, wherein the human subject has received a prior CAR T-cell therapy.
14. The method according to any one of claims 1-11, wherein the human subject has received a prior CAR T-cell therapy and at least one additional therapy.
15. The method according to any one of claims 1-11 and 13-14, wherein the prior CAR T-cell therapy is tisagenlecleucel, brexucabtagene autoleucel, axicabtagene cilocleucel, aisocabtagene maraleucel or any combination thereof.
16. The method according to any one of claims 1-15, wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises an anti-CD19 antibody, an anti-CD20 antibody, and anti-CD22 antibody or an anti- CD79b antibody.
17. The method according to claim 16, wherein the anti-CD19 antibody, anti-CD20 antibody, anti-CD22 antibody or anti-CD79b antibody comprises an antibody-drug conjugate.
18. The method according to any one of claims 16-17, wherein the anti-CD19 antibody comprises inebilizumab, loncastuximab tesirine or tafasitamab.
19. The method according to any one of claims 16-17, wherein the anti-CD20 antibody comprises rituximab, ibritumomab tiuxetan, tositumomab, ocrelizumab, ofatumumab, mosunetuzumab, obinutuzumab, or ublituximab.
20. The method according to any one of claims 16-17, wherein the anti-CD22 antibody comprises inotuzumab ozogamicin, or epratuzumab.
21. The method according to any one of claims 16-17, wherein the anti-CD79b antibody comprises polatuzumab vedotin.
22. The method according to any one of claims 1-21, wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises a bispecific T-cell engager antibody therapy.
23. The method according to claim 22, wherein the bispecific T-cell engager antibody therapy comprises a T cell engager comprising an anti-CD3 domain.
24. The method according to any one of claims 22-23, wherein the bispecific T-cell engager antibody therapy comprises mosunetuzumab, glofitamab, epcoritamab, blinatumomab, solitomab, duvortuxizumab, AFM11, REGN5458, CC-93269, TNB-383B, pdronextamab, XmAbl3676, IGM-2323, or cevostamab.
25. The method according to any one of claims 22-24, wherein the bispecific T-cell engager antibody therapy comprises an anti-CD20 domain.
26. The method according to any one of claims 22-24, wherein the bispecific T-cell engager antibody therapy comprises an anti-CD19 domain.
27. The method according to any one of claims 1-26, wherein the anti-CD22 binding domain is an scFv.
28. The method according to any one of claims 1-27, wherein the anti-CD22 binding domain is an antibody or an antigen binding domain of thereof.
29. The method according to any one of claims 1-28, wherein the anti-CD22 binding domain comprises the sequence:
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTY YRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLE DAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLN WYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQ QSYSIPQTFGQGTKLEIK (SEQ ID NO:1).
30. The method according to any one of claims 1-29, wherein the CAR comprises a linker between the anti-CD22 binding domain and the portion of a CD8a extracellular domain.
31. The method according to any one of claims 1-30, wherein the linker between the anti- CD22 binding domain and the portion of a CD8a extracellular domain comprises the sequence AAA.
32. The method according to any one of claims 1-31, wherein the portion of a CD8a extracellular domain comprises the sequence TTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACD (SEQ ID NO: 2).
33. The method according to any one of claims 1-32, wherein the portion of a CD8a transmembrane domain comprises the sequence IYIWAPLAGTCGVLLLSLVIT (SEQ ID N0:4).
34. The method according to any one of claims 1-33, wherein the CAR comprises a portion of a CD8a intracellular domain.
35. The method according to any one of claims 1-34, wherein the portion of a CD8a intracellular domain comprises the sequence LYC.
36. The method according to any one of claims 1-35, wherein the 4-1BB costimulatory domain comprises the sequence KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL (SEQ ID NO:6).
37. The method according to any one of claims 1-36, wherein the CD3zeta intracellular signaling domain comprises the sequence: RVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKN PQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALH MQALPPR (SEQ ID NO: 7).
38. The method according to any one of claims 1-37, wherein the CAR comprises a signal peptide sequence.
39. The method according to any one of claims 1-38, wherein the signal peptide sequence comprises the sequence: MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO:8).
40. The method according to any one of claims 1-39, wherein the CAR comprises the sequence:
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWLGRTY YRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCAREVTGDLE DAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSASVGDRVTITCRASQTIWSYLN WYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGTDFTLTISSLQAEDFATYYCQ QSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIASQPLSLRPEACRPAAGGAVH TRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTT QEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDV LDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGH DGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 12).
41. The method according to any one of claims 1-40, wherein the CAR comprises the sequence:
MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAA WNWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSV TPEDTAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQSPSSLSAS VGDRVTITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVPSRFSGRGSGT DFTLTISSLQAEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTTPAPRPPTPAPTIAS QPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCKRG RKKLLYIFKQPFMRP VQTTQEEDGC SCRFPEEEEGGCELRVKF SRS ADAP AYKQG QNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMA EAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR (SEQ ID NO: 9).
42. The method according to any one of claims 1-41, wherein the hematological cancer expresses CD22.
43. The method according to any one of claims 1-42, wherein the hematological cancer is Large B-Cell Lymphoma (LBCL).
44. The method according to any one of claims 1-43, wherein the LBCL is diffuse LBCL (DLBCL).
45. The method according to any one of claims 1-44, wherein the LBCL has been histologically confirmed.
46. The method according to any one of claims 1-45, wherein the LBCL is (DLBCL) not otherwise specified (NOS).
47. The method according to any one of claims 1-46, wherein the DLBCL-NOS is germinal center B-cell (GCB) type or active B-cell (ABC) type.
48. The method according to any one of claims 1-47, wherein the LBCL is high-grade B-cell lymphoma with or without MYC and BCL2 and/or BCL6 rearrangements.
49. The method according to any one of claims 1-48, wherein the LBCL is DLBCL associated with chronic inflammation.
50. The method according to any one of claims 1-49, wherein the LBCL is primary cutaneous DLBCL, leg type.
51. The method according to any one of claims 1-50, wherein the LBCL is Epstein-Barr virus (EBV)-positive DLBCL-NOS.
52. The method according to any one of claims 1-51, wherein the LBCL is DLBCL arising from follicular lymphoma (FL).
53. The method according to any one of claims 1-52, wherein the LBCL is DLBCL arising from marginal zone lymphoma (MZL).
54. The method according to any one of claims 1-53, wherein the LBCL is primary mediastinal (thymic) large B-cell lymphoma.
55. The method according to any one of claims 1-54, wherein the LBCL is Grade 3B FL.
56. The method according to any one of claims 1-55, wherein the human subject is at least 18 years of age.
57. The method according to any one of claims 1-56, wherein the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of progressive disease (PD) or stable disease (SD) after the last therapy.
58. The method according to any one of claims 1-57, wherein the human subject has relapsed/refractory disease after a last therapy based on a radiographic finding of disease progression after partial response (PR) or complete response (CR) after the last therapy.
59. The method according to any one of claims 1-58, wherein the human subject has relapsed/refractory disease after a last therapy based on presence of viable lymphoma in a biopsy obtained after therapy.
60. The method according to any one of claims 1-59, wherein the human subject has relapsed disease following a CR or progressive disease following a PR to a bispecific T-cell engager antibody therapy.
61. The method according to any one of claims 1-60, wherein the human subject has progressive disease (PD) or stable disease (SD) as best response to a bispecific T-cell engager antibody therapy.
62. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is from about 0.1 * 106 to about 4* 106 engineered CAR T-cells/kg.
63. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is from about 0.1 * 106 to about 0.75* 106 engineered CAR T-cells/kg.
64. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is from about 0.25* 106 to about 0.75* 106 engineered CAR T-cells/kg.
65. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is from about 1.2* 106 to about 4* 106 engineered CAR T-cells/kg.
66. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is from about 0.8* 106 to about 1.2* 106 engineered CAR T-cells/kg.
67. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is from about 2.5* 106 to about 3.5* 106 engineered CAR T-cells/kg.
68. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is about 0.5* 106 engineered CAR T-cells/kg.
69. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is about l >< 106 engineered CAR-T cells/kg.
70. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is about 2* 106 engineered CAR-T cells/kg.
71. The method according to any one of claims 1-61, wherein the dose of engineered CAR T- cells is about 3* 106 engineered CAR T-cells/kg.
72. The method according to any one of claims 1-70, wherein the method comprises administering to the human subject a dose of cyclophosphamide prior to administering the engineered CAR T-cells.
73. The method according to any one of claims 1-72, wherein the dose of cyclophosphamide is about 400 mg/m2 to about 600 mg/m2.
74. The method according to any one of claims 1-72, wherein the dose of cyclophosphamide is about 500 mg/m2.
75. The method according to any one of claims 1-74, wherein the dose of cyclophosphamide is administered daily for at least 2 days.
76. The method according to any one of claims 1-74, wherein the dose of cyclophosphamide is administered daily for 3 days.
77. The method according to any one of claims 1-76, wherein the method comprises administering to the human subject a dose of fludarabine prior to administering the engineered CAR T-cells.
78. The method according to any one of claims 1-77, wherein the dose of fludarabine is about 20 mg/m2 to about 400 mg/m2.
79. The method according to any one of claims 1-77, wherein the dose of fludarabine is about 25 mg/m2 to about 35 mg/m2.
80. The method according to any one of claims 1-77, wherein the dose of fludarabine is about 30 mg/m2.
81. The method according to any one of claims 1-80, wherein the dose of fludarabine is administered daily for at least 2 days.
82. The method according to any one of claims 1-80, wherein the dose of fludarabine is administered daily for three days.
83. The method according to any one of claims 1-82, wherein the dose of cyclophosphamide is administered daily for three days and the dose of fludarabine is administered daily for three days.
84. The method according to any one of claims 1-83, wherein at least one dose of cyclophosphamide is administered on the same day as a dose of fludarabine.
85. The method according to any one of claims 1-84, wherein a first dose of cyclophosphamide is administered on the same day as a first dose of fludarabine.
86. The method according to any one of claims 1-85, wherein a second dose of cyclophosphamide is administered on the same day as a second dose of fludarabine.
87. The method according to any one of claims 1-86, wherein a third dose of cyclophosphamide is administered on the same day as a third dose of fludarabine.
88. The method according to any one of claims 1-87, wherein each dose of cyclophosphamide administered is administered on the same day that a dose of fludarabine is administered.
89. The method according to any one of claims 1-88, wherein a dose of cyclophosphamide is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells are administered on day 0.
90. The method according to any one of claims 1-89, wherein a dose of fludarabine is administered on day -5 and day -4 and day -3, and the engineered CAR T-cells are administered on day 0.
91. The method according to any one of claims 1-88, wherein a dose of cyclophosphamide is administered on day -5, day -4, and day -3, a dose of fludarabine is administered on day - 5, day -4, and day -3, and the engineered CAR T-cells are administered on day 0.
92. A method of treating a relap sed/refractory Large B-Cell Lymphoma (LBCL) in a human subject in need thereof, the method comprising:
(a) administering to the human subject a dose of about 400 mg/m2 to about 600 mg/m2 cyclophosphamide on day -5, day -4 and day -3, and a dose of about 25 mg/m2 to about 35 mg/m2 fludarabine on day -5, day -4 and day -3;
(b) administering to the human subject a dose of from about 0.1 * 106 to about
4* 106 engineered chimeric antigen receptor (CAR) T-cells/kg on day 0; wherein the CAR comprises the sequence: MLLLVTSLLLCELPHPAFLLIPQVQLQQSGPGLVKPSQTLSLTCAISGDSVSSN SAAWNWIRQSPSRGLEWLGRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSL QLNSVTPEDTAVYYCAREVTGDLEDAFDIWGQGTMVTVSSGGGGSDIQMTQ SPSSLSASVGDRVTITCRASQTIWSYLNWYQQRPGKAPNLLIYAASSLQSGVP SRFSGRGSGTDFTLTISSLQAEDFATYYCQQSYSIPQTFGQGTKLEIKAAATTT PAPRPPTPAPTIASQPLSLRPEACRPAAGGAVHTRGLDFACDIYIWAPLAGTCG VLLLSLVITLYCKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCE LRVKFSRSADAPAYKQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPR RKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDT
YDALHMQALPPR (SEQ ID NO: 9); wherein the human subject has received at least one prior line of therapy, wherein the at least one prior line of therapy comprises
(A) an anti-CD19 CAR T-cell therapy, (B) a bispecific T-cell engager antibody therapy, or
(C) a combination thereof.
PCT/US2024/044747 2023-09-01 2024-08-30 Anti-cd22 chimeric antigen receptor (car) therapies Pending WO2025049961A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202363536298P 2023-09-01 2023-09-01
US63/536,298 2023-09-01

Publications (1)

Publication Number Publication Date
WO2025049961A1 true WO2025049961A1 (en) 2025-03-06

Family

ID=93010706

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2024/044747 Pending WO2025049961A1 (en) 2023-09-01 2024-08-30 Anti-cd22 chimeric antigen receptor (car) therapies

Country Status (1)

Country Link
WO (1) WO2025049961A1 (en)

Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US6905874B2 (en) 2000-02-24 2005-06-14 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US20060121005A1 (en) 2000-02-24 2006-06-08 Xcyte Therapies, Inc. Activation and expansion of cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2018178378A1 (en) * 2017-03-31 2018-10-04 Cellectis Sa New universal chimeric antigen receptor t cells specific for cd22
WO2020069405A1 (en) * 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6887466B2 (en) 1988-11-23 2005-05-03 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US5883223A (en) 1988-11-23 1999-03-16 Gray; Gary S. CD9 antigen peptides and antibodies thereto
US7232566B2 (en) 1988-11-23 2007-06-19 The United States As Represented By The Secretary Of The Navy Methods for treating HIV infected subjects
US6534055B1 (en) 1988-11-23 2003-03-18 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US7144575B2 (en) 1988-11-23 2006-12-05 The Regents Of The University Of Michigan Methods for selectively stimulating proliferation of T cells
US6905680B2 (en) 1988-11-23 2005-06-14 Genetics Institute, Inc. Methods of treating HIV infected subjects
US5858358A (en) 1992-04-07 1999-01-12 The United States Of America As Represented By The Secretary Of The Navy Methods for selectively stimulating proliferation of T cells
US7175843B2 (en) 1994-06-03 2007-02-13 Genetics Institute, Llc Methods for selectively stimulating proliferation of T cells
US6905681B1 (en) 1994-06-03 2005-06-14 Genetics Institute, Inc. Methods for selectively stimulating proliferation of T cells
US6352694B1 (en) 1994-06-03 2002-03-05 Genetics Institute, Inc. Methods for inducing a population of T cells to proliferate using agents which recognize TCR/CD3 and ligands which stimulate an accessory molecule on the surface of the T cells
US7172869B2 (en) 1995-05-04 2007-02-06 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US6692964B1 (en) 1995-05-04 2004-02-17 The United States Of America As Represented By The Secretary Of The Navy Methods for transfecting T cells
US7067318B2 (en) 1995-06-07 2006-06-27 The Regents Of The University Of Michigan Methods for transfecting T cells
US6867041B2 (en) 2000-02-24 2005-03-15 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US6905874B2 (en) 2000-02-24 2005-06-14 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
US20060121005A1 (en) 2000-02-24 2006-06-08 Xcyte Therapies, Inc. Activation and expansion of cells
US6797514B2 (en) 2000-02-24 2004-09-28 Xcyte Therapies, Inc. Simultaneous stimulation and concentration of cells
WO2016164731A2 (en) 2015-04-08 2016-10-13 Novartis Ag Cd20 therapies, cd22 therapies, and combination therapies with a cd19 chimeric antigen receptor (car) - expressing cell
WO2018178378A1 (en) * 2017-03-31 2018-10-04 Cellectis Sa New universal chimeric antigen receptor t cells specific for cd22
WO2020069405A1 (en) * 2018-09-28 2020-04-02 Novartis Ag Cd22 chimeric antigen receptor (car) therapies

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
GHOBADI A ET AL: "Phase 2 open label, multicenter study evaluating CRG-022, a CD22-directed autologous CAR T-cell therapy, in patients (pts) with relapsed/refractory (R/R) large B-cell lymphoma (LBCL) after CD19-directed CAR T-cell therapy.", JOURNAL OF CLINICAL ONCOLOGY, vol. 42, no. 16_suppl, TPS7085, 29 May 2024 (2024-05-29), ASCO Annual Meeting; Chicago, IL, USA; 31 May-4 June 2024, XP093226383, ISSN: 0732-183X, DOI: 10.1200/JCO.2024.42.16_suppl.TPS7085 *

Similar Documents

Publication Publication Date Title
Kirkman et al. A randomized prospective trial of anti-Tac monoclonal antibody in human renal transplantation
AU2020380366B2 (en) Chimeric antigen receptor T cell therapy
AU2020267378B2 (en) Methods of administering chimeric antigen receptor immunotherapy
Li et al. Advances in the treatment of extramedullary disease in multiple myeloma
TWI841961B (en) Chimeric antigen receptor t cell therapy
JP2025507678A (en) BCMA-targeted CAR-T cell therapy for multiple myeloma
JP2025513108A (en) Combination antibody therapy for treating colorectal cancer
JP2022508513A (en) Combination therapy for cancer treatment
JP2025118642A (en) Methods for treating or preventing graft-versus-host disease
US20040175827A1 (en) Methods of generating human cd4+ th2 cells and uses thereof
WO2025049961A1 (en) Anti-cd22 chimeric antigen receptor (car) therapies
UA126313C2 (en) Combination therapy
JP2024534926A (en) Methods and compositions for inducing tolerance
Spinner et al. New treatment algorithms in Hodgkin lymphoma: too much or too little?
US20250152643A1 (en) Combinations of checkpoint inhibitors and oncolytic virus for treating cancer
Lepik et al. Safety and efficacy of nivolumab applied at different dosage in the patients with relapsing Hodgkin lymphoma after allogeneic hematopoietic stem cell transplantation
EP1351713B1 (en) Anti-tenascin monoclonal antibody therapy for lymphoma
US20250268941A1 (en) Bcma/cd19 car for treating multiple myeloma
Nanni Analysis of efficacy and safety of CAR T-cell therapy in relapsed/refractory lymphomas: current indications and future perspectives
WO2024256823A1 (en) Treatment of trbc1-positive t cell malignancies
JPWO2022117068A5 (en)
US20230279132A1 (en) Treatment of b cell malignancies
AU2023367198A1 (en) Improving efficacy and durable response of immunotherapy
Cairo Targeted immunotherapy in the treatment of childhood and adolescent mature B-cell lymphoma
TW202517679A (en) Treatment of cancer with anti-ilt2 antibodies

Legal Events

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

Ref document number: 24786188

Country of ref document: EP

Kind code of ref document: A1