CANCER TREATMENT USING CD38 INHIBITOR AND/OR LENALIDOMIDE AND T-CELLS EXPRESSING A CHIMERIC ANTIGEN RECEPTOR

Information

  • Patent Application
  • 20220202859
  • Publication Number
    20220202859
  • Date Filed
    December 22, 2021
    2 years ago
  • Date Published
    June 30, 2022
    a year ago
Abstract
Combined therapy for treating multiple myeloma (MM), comprising (a) a population of genetically engineered T cells, which may express a chimeric antigen receptor (CAR) that binds B-cell maturation antigen (BCMA), and (b) an anti-CD38 antibody such as daratumumab or lenalidomide or a derivative thereof.
Description
SEQUENCE LISTING

The application contains a Sequence Listing that has been filed electronically in the form of a text file, created Dec. 20, 2021, and named “095136-0492-039US1_SEQ.TXT” (66,013 bytes), the contents of which are incorporated by reference herein in their entirety.


BACKGROUND OF THE INVENTION

Multiple myeloma (MM) is a malignancy of terminally differentiated plasma cells in the bone marrow. MM results from the secretion of a monoclonal immunoglobulin protein (also known as M-protein or monoclonal protein) or monoclonal free light chains by abnormal plasma cells, and is differentiated on the spectrum of plasma cell dyscrasias by characteristic bone marrow biopsy findings as well as symptoms attributable to end organ damage related to plasma cell proliferation (hypercalcemia, renal insufficiency, anemia, fractures) (Kumar 2017a). MM represents about 10% of all hematologic malignancies and is the second most common hematologic malignancy after Non-Hodgkin lymphoma (NHL) (Kumar 2017a, Rajkumar and Kumar 2016). For most patients, MM is an incurable disease that ultimately leads to death. There is an unmet need for effective therapies for treating MM, particularly relapsed/refractory MM.


SUMMARY OF THE INVENTION

The present disclosure is based, at least in part, on the unexpected discoveries that an anti-CD38 antibody (daratumumab), which is an exemplary NK cell inhibitor, successfully depleted NK cells both in vitro and in vivo but did not affect T cell numbers, including numbers of genetically engineered T cells expressing a chimeric antigen receptor (CAR), and did not activate CAR T cells. Further, it was found that, unexpectedly, daratumumab pre-treatment significantly reduced NK cell-mediated CAR T cell lysis (e.g., by approximately 50%) and preserves the viability and number of allogeneic CAR T cells. Moreover, combined therapy of daratumumab and CAR-T cells exhibited synergistic effect in reducing tumor burden and extending survival rates in a xenograft mouse model, even in the presence of NK cells.


Further, the present disclosure is also based, at least in part, on the unexpected discovery that combined use of lenalidomide and CAR-T cells specific to B-cell maturation antigen (BCMA) such as CTX120 cells showed substantially enhanced anti-tumor effects as relative to lenalidomide or the anti-BCMA CAR-T cells alone as observed in a multiple myeloma mouse model. Further, it was observed, surprisingly, that lenalidomide did not enhance immune recognition of allogeneic CAR-T cells.


Accordingly, the present disclosure features, in some aspects, a method for treating multiple myeloma (MM), the method comprising: (i) administering to a subject in need thereof an effective amount of one or more lymphodepleting chemotherapeutic agents; (ii) administering to the subject a first dose of a population of genetically engineered T cells after step (i); and

  • (iii) administering to the subject an effective amount of lenalidomide, an effective amount of daratumumab, or a combination thereof. In some embodiments, step (ii) may be performed 2-7 days after step (i).


In some embodiments, step (i) may comprise co-administering to the subject fludarabine at about 30 mg/m2 and cyclophosphamide at about 300 mg/m2 to about 500 mg/m2 intravenously per day for three days. For example, cyclophosphamide may be administered at about 300 mg/m2. In other examples, cyclophosphamide may be administered at about 500 mg/m2.


In some embodiments, the first dose of the population of genetically engineered T cells in step (ii) ranges from about 5.0×107 to about 1.05×109 CAR+ T cells. For example, the first dose of the population of genetically engineered T cells in step (ii) may range from about 5.0×107 to about 7.5×108 CAR+ T cells. In some examples, the first dose of the population of genetically engineered T cells is about 5×107 CAR+ T cells, about 1.5×108 CAR+ T cells, about 4.5×108 CAR+ T cells, about 6×108 CAR+ T cells, about 7.5×108 CAR+ T cells, or about 9×108 CAR+ T cells. In some examples, the first dose of the population of genetically engineered T cells in step (ii) ranges from about 5.0×107 to about 1.5×108 CAR+ T cells, about 1.5×108 to about 4.5×108 CAR+ T cells, about 4.5×108 to about 6.0×108 CAR+ T cells, about 6.0×108 to about 7.5×108 CAR+ T cells, about 7.5×108 to about 9×108 CAR+ T cells, or about 9×108 to about 1.05×109 CAR+ T cells.


In some examples, the effective amount of the population of genetically engineered T cells is sufficient to achieve one or more of the following: (a) decrease soft tissue plasmacytomas sizes (SPD) by at least 50% in the subject; (b) decrease serum M-protein levels by at least 25%, optionally by 50% in the subject; (c) decrease 24-hour urine M-protein levels by at least 50%, optionally by 90% in the subject; (d) decrease differences between involved and uninvolved free light chain (FLC) levels by at least 50% in the subject; (e) decrease plasma cell counts by at least 50% in the subject, optionally wherein baseline BM plasma-cell percentage is ≥30%, (f) decrease kappa-to-lambda light chain ratios (κ/λ ratios) to 4:1 or lower in the subject, who has myeloma cells that produce kappa light chains; and (g) increase kappa-to-lambda light chain ratios (κ/λ ratios) to 1:2 or higher in the subject, who has myeloma cells that produce lambda light chains.


In some examples, the effective amount of the population of genetically engineered T cells is sufficient to decrease serum M-protein levels by at least 90% and 24-hour urine M-protein levels to less than 100 mg in the subject, and/or wherein the effective amount of the population of genetically engineered T cells is sufficient to decrease serum M-proteins, urine M-proteins, and soft tissue plasmacytomas to undetectable levels, and plasma cell counts to less than 5% of bone marrow (BM) aspirates in the subject. In some examples, the effective amount of the population of genetically engineered T cells is sufficient to achieve Stringent Complete Response (sCR), Complete Response (CR), Very Good Partial Response (VGPR), Partial Response (PR), Minimal Response (MR), or Stable Disease (SD).


In some embodiments, an effective amount of lenalidomide is administered to the subject in step (iii). In some examples, step (iii) may comprise administering to the subject about 10 mg lenalidomide orally per day for 21 days. In some examples, the first dose of lenalidomide in step (iii) starts on the third day of the administration of the lymphodepleting chemotherapeutic agents. In some examples, the method further comprises performing one or more cycles of treatment comprising lenalidomide to the subject after step (ii). For example, the first cycle starts 28 days after step (ii). In some instances, the subject exhibits stable disease or better when receiving the one or more cycles of lenalidomide treatment. In some examples, the one or more cycles of treatment comprising lenalidomide are up to five cycles, each of which comprises a daily dose of lenalidomide for 21 days, followed by a 7-day resting period. In some instances, the daily dose of lenalidomide is 5 mg. In some examples, the method further comprising terminating the one or more cycles of the treatment comprising lenalidomide when the subject exhibits disease progression and/or unacceptable toxicity.


In some embodiments, an effective amount of daratumumab is administered to the subject in step (iii). For example, about 16 mg/kg daratumumab is administered to the subject by intravenous infusion within 3 days prior to step (ii). In some instances, the dose of about 16 mg/kg of daratumumab can be split to 8 mg/kg over two consecutive days. Alternatively, about 1800 mg of daratumumab can be administered to the subject by subcutaneous injection. In some instances, the daratumumab is injected together with hyaluronidase, e.g., at an amount of about 30,000 units. In some examples, the daratumumab is administered to the subject no more than 14 days prior to step (ii). In some examples, the subject can be administered multiple doses of daratumumab once per month, for example, up to 5 monthly doses, when the subject exhibits stable disease or better.


In some examples, the daratumumab is terminated when the subject exhibits disease progression and/or unacceptable toxicity. In some examples, the subject is administered corticosteroid, antipyretic, antihistamine, or a combination thereof, prior to the administration of the daratumumab. In some examples, the subject is administered methylprednisolone at about 100 mg by intravenous infusion or about 60 mg by intravenous infusion or by oral administration, acetaminophen at about 650-1000 mg by oral administration, and diphenhydramine hydrochloride at about 20-50 mg by intravenous infusion or oral administration.


In some embodiments, an effective amount of lenalidomide and an effective amount of daratumumab are administered to the subject in step (iii). For example, about 10 mg lenalidomide is administered to the subject orally per day for 21 days. In some instances, the first dose of lenalidomide starts on the third day of the administration of the lymphodepleting chemotherapeutic agents. In some examples, the method may further comprise performing one or more cycles of treatment comprising lenalidomide to the subject after step (ii). For example, the first cycle starts 28 days after step (ii), optionally when the subject exhibits stable disease or better. In some instances, the one or more cycles of treatment comprising lenalidomide (e.g., 5 mg per day) are up to five cycles, each of which comprises a daily dose of lenalidomide for 21 days, followed by a 7-day resting period. In some instances, the method further comprising terminating the one or more cycles of the treatment comprising lenalidomide when the subject exhibits disease progression and/or unacceptable toxicity.


Alternatively or in addition, about 16 mg/kg daratumumab is administered to the subject by intravenous infusion within 3 days prior to step (ii), which may be split to 8 mg/kg over two consecutive days. In other examples, about 1800 mg of daratumumab is administered to the subject by subcutaneous injection, which may be co-administered with hyaluronidase, e.g., at an amount of about 30,000 units. In some examples, the daratumumab is administered to the subject no more than 14 days prior to step (ii). In some examples, the subject is administered multiple doses of daratumumab once per month, e.g., up to 5 monthly doses, when the subject exhibits stable disease or better. In some examples, treatment of the daratumumab is terminated when the subject exhibits disease progression and/or unacceptable toxicity.


The population of genetically engineered T cells used in any of the methods disclosed herein comprise T cells, which comprise a nucleic acid comprising a nucleotide sequence encoding a chimeric antigen receptor (CAR) that binds B-cell maturation antigen (BCMA), a disrupted TRAC gene, and a disrupted β2M gene; and wherein the nucleic acid encoding the CAR is inserted into the disrupted TRAC gene. In some embodiments, ≥30% of the genetically engineered T cells are CAR+, ≤0.4% of the genetically engineered T cells are TCR+, and/or ≤30% of the genetically engineered T cells are B2M+.


In some embodiments, the CAR that binds BCMA comprises: (i) an ectodomain comprising an anti-BCMA single chain variable fragment (scFv); (ii) a CD8a transmembrane domain; and (iii) an endodomain comprising a 4-1BB co-stimulatory domain and a CD3ζ signaling domain. In some embodiments, the anti-BCMA scFv comprises a heavy chain variable domain (VH) comprising SEQ ID NO: 42 and a light chain variable domain (VL) comprising SEQ ID NO: 43. In some examples, the anti-BCMA scFv comprises SEQ ID NO: 41. In some specific examples, the CAR that binds BCMA comprises the amino acid sequence of SEQ ID NO: 40, which may be encoded by a nucleic acid encoding the anti-BCMA CAR comprises the nucleotide sequence of SEQ ID NO: 33.


In some embodiments, the disrupted TRAC gene can be produced by a CRISPR/Cas9 gene editing system, which comprises a guide RNA comprising a spacer sequence of SEQ ID NO: 4. In some examples, the disrupted TRAC gene has a deletion comprising the SEQ ID NO: 10. For example, the disrupted TRAC gene comprises the nucleotide sequence of SEQ ID NO:30, which substitutes for the deletion comprising SEQ ID NO:10.


In some embodiments, the disrupted β2M gene is produced by a CRISPR/Cas9 gene editing system, which comprises a guide RNA comprising a spacer sequence of SEQ ID NO: 8. In some examples, the disrupted β2M gene comprises at least one of SEQ ID NOs: 21-26.


In some embodiments, the population of genetically engineered T cells is derived from one or more healthy human donors. The population of genetically engineered T cells may be suspended in a cryopreservation solution. In some examples, the population of genetically engineered T cells is administered by intravenous infusion.


Any of the methods disclosed herein may further comprise (iv) monitoring the human patient for development of acute toxicity after step (ii). In some embodiments, the acute toxicity comprises infusion reactions, febrile reactions, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), tumor lysis syndrome, hemophagocytic lymphohistiocytosis (HLH), Cytopenias, GvHD, hypotention, renal insufficiency, viral encephalitis, neutropenia, thrombocytopenia, or a combination thereof. In some examples, the subject is subject to toxicity management if development of toxicity is observed.


In any of the methods disclosed herein, the subject is a human patient, who optionally is 18 years of age or older. Such a human patient may have relapsed and/or refractory MM. In some embodiments, the subject has undergone at least two prior therapies for MM, which optionally comprise an immunomodulatory agent, a proteasome inhibitor, an anti-CD38 antibody, or a combination thereof. For example, the subject may be refractory to one or more prior therapies comprising an immunomodulatory agent, a proteasome inhibitor, and/or an anti-CD38 antibody. In some examples, the subject may be double-refractory to prior therapies comprising an immunomodulatory agent and a proteasome inhibitor. In other examples, the subject may be triple-refractory to prior therapies comprising an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 antibody. In some instances, the subject relapsed after an autologous stem cell transplant (SCT), and wherein optionally the relapse occurs within 12 months after the SCT. In some instances, the subject has received prior lenalidomide treatment.


In some examples, the subject is a human patient having one or more of the following features: (a) measurable disease, (b) Eastern Cooperative Oncology Group performance status 0 or 1, (c) adequate organ function, (d) free of a prior allogeneic stem cell transplantation (SCT), (e) free of autologous SCT within 60 days prior to step (i), (f) free of plasma cell leukemia, non-secretory MM, Waldenstrom's macroglobulinemia, POEM syndrome, and/or amyloidosis with end organ involvement and damage, (g) free of prior gene therapy, anti-BCMA therapy, and non-palliative radiation therapy within 14 days prior to step (i), (h) free of contraindication to lenalidomide, daratumumab, cyclophosphamide, and/or fludarabine, (i) free of central nervous system involvement by MM, (j) free of history or presence of clinically relevant CNS pathology, cerebrovascular ischemia and/or hemorrhage, dementia, a cerebellar disease, an autoimmune disease with CNS involvement, (h) free of unstable angina, arrhythmia, and/or myocardial infarction within 6 month prior to step (i), (i) free of uncontrolled infections, optionally wherein the infection is caused by HIV, HBV, or HCV, (j) free of previous or concurrent malignancy, provided that the malignancy is not basal cell or squamous cell skin carcinoma, adequately resected and in situ carcinoma of cervix, or a previous malignancy that was completely resected and has been in remission for ≥5 years, (k) free of live vaccine administration within 28 days prior to step (i), (l) free of systemic anti-tumor therapy within 14 days prior to step (i), and (m) free of primary immunodeficiency disorders or autoimmune disorders that require immunosuppressive therapy.


In some embodiments, prior to step (i), the human patient does not show one or more of the following features: (a) significant worsening of clinical status, (b) requirement for supplemental oxygen to maintain a saturation level of greater than about 91%, (c) uncontrolled cardiac arrhythmia, (d) hypotension requiring vasopressor support, (e) active infection, and (f) neurological toxicity that increases risk of immune effector cell-associated neurotoxicity syndrome (ICANS).


In some embodiments, prior to step (ii) and after step (i), the human patient does not show one or more of the following features: (a) active uncontrolled infection, (b) worsening of clinical status compared to the clinical status prior to step (i), and (c) neurological toxicity that increases risk of immune effector cell-associated neurotoxicity syndrome (ICANS).


Any of the methods disclosed herein may further comprise administering to the subject a second dose of the population of genetically engineered T cells about 4 to 12 weeks after the first dose of the population of genetically engineered T cells. The subject may achieve stable disease or better response after the first dose, optionally assessed on Day 28 after the first dose. In some examples, the subject is treated by the lymphodepleting chemotherapeutic agents 2-7 days prior to the second dose of the population of genetically engineered T cells. In some instances, the subject is administered fludarabine at about 30 mg/m2 and cyclophosphamide at about 300 mg/m2 to about 500 mg/m2, optionally at about 300 mg/m2, intravenously per day for three days. In some instances, the second dose of the population of genetically engineered T cells is not accompanied with lymphodepleting therapy when the subject is experiencing significant cytopenias.


Also within the scope of the present disclosure are any of the genetically engineered T cells disclosed herein, targeting BCMA, for use in treating multiple myeloma, consurrently with an NK inhibitor such as an anti-CD38 antibody (e.g., daratumumab), lenalidomide or a derivative thereof, or a combination thereof. Also provided herein are uses of the genetically engineered anti-BCMA CAR-T cells as disclosed herein, concurrently with NK inhibitor such as an anti-CD38 antibody (e.g., daratumumab), lenalidomide or a derivative thereof, or a combination thereof, for manufaring a medicament for use in treating multiple myeloma.


The details of one or more embodiments of the invention are set forth in the description below. Other features or advantages of the present invention will be apparent from the following drawings and detailed description of several embodiments, and also from the appended claims.





BRIEF DESCRIPTION OF THE DRAWINGS

The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure, which can be better understood by reference to the drawing in combination with the detailed description of specific embodiments presented herein.



FIG. 1 is a diagram depicting the percentage of TCR, β2M, anti-BCMA CAR+ and TCR/β2M/anti-BCMA CAR+ cells in a population of genetically engineered T cells (CTX120 cells), as measured by flow cytometry.



FIGS. 2A-2B include diagrams depicting the percentage of CD4+ (FIG. 2A) or CD8+ (FIG. 2B) T cells within a population of genetically engineered cells (CTX120 cells) or unedited cells as measured by flow cytometry.



FIG. 3 is a diagram depicting the volume of subcutaneous BCMA-expressing human MM tumors (MM.1S tumors) measured over time in immunocompromised mice that were untreated or treated with CTX120 cells on day 0. Circles depict the growth of primary tumors inoculated in the right flank in treated or untreated animals, with all untreated animals requiring euthanasia due to tumor burden and all treated animals rejecting primary tumors. Surviving treated animals were re-challenged with tumor cells on day 29 by inoculation with tumors cells in the left flank. Open triangles depict the growth of re-challenge tumors in treated animals, while closed triangles depict growth of tumors inoculated in the left flank of a new cohort of untreated animals.



FIG. 4 is a diagram depicting the volume of subcutaneous BCMA-expressing human MM tumors (RPMI-8226 tumors) measured over time in immunocompromised mice that were untreated or treated with CTX120 cells on day 1.



FIGS. 5A-5B include charts depicting production of interferon-gamma (IFNγ) (FIG. 5A) or interleukin-2 (IL-2) (FIG. 5B) by effector CTX120 cells following in vitro co-culture with tumor cells positive for surface expression of BCMA (MM.1S and JeKo-1) or negative for expression of BCMA (K562).



FIGS. 6A-6C include diagrams depicting the percentage of target cells characterized as dead/dying by flow cytometry following in vitro co-culture with unedited cells or edited CTX120 cells at different T cell to target cell ratios. The target cells were high BCMA-expressing MM.1S cells (FIG. 6A), low BCMA-expressing JeKo-1 cells (FIG. 6B), or BCMA-negative K562 cells (FIG. 6C).



FIGS. 7A-7B include charts depicting the production of IFNγ (FIG. 7A) or IL-2 (FIG. 7B) by effector CTX120 cells following in vitro co-culture with primary cells derived from human tissues, including B cells that contain BCMA-expressing cells, as compared to BCMA-expressing JeKo-1 cells as a positive control.



FIG. 8 is a diagram depicting the viability of an ex vivo culture of edited CTX120 cells over time as measured by cell counting when grown in complete media (serum+cytokines), media with serum (no cytokines), or media lacking serum and cytokines.



FIG. 9 is a diagram depicting survival of mice over time following exposure to a dosage of radiation and treatment with vehicle-only (no T cells), unedited T cells or edited CTX120 cells.



FIG. 10 is a chart depicting proliferation of unedited T cells or edited TRAC−/B2M− T cells following in vitro co-culture with peripheral blood mononuclear cells (PBMCs) derived from the same donor (autologous PBMCs) or a different donor (allogeneic PBMCs). As a positive control, T cells were stimulated with phytohaemagglutinin-L (PHA) to induce proliferation.



FIGS. 11A-11D are graphs showing the effect of daratumumab (Dara) on normal immune cells (PBMCs) collected from a healthy donor 96 hours after culture in either media alone or media supplemented with 10% complement. Daratumumab was used at doses of 0.01, 0.1, or 1 μg/mL. Some cells were treated with control isotype mAb (Hu IgG1k). FIG. 11A shows the frequency of NK cells after these treatments. FIG. 11B shows the number of NK cells after these treatments. FIG. 11C shows the frequency of T cells after these treatments. FIG. 11D shows the number of T cells after these treatments.



FIGS. 12A-12B are graphs showing the frequency and number of anti-BCMA CAR T cells after 72 hours culture with daratumumab (Dara) or control isotype mAb (Hu IgG1k), with or without 10% complement. Daratumumab was used at doses of 0.01, 0.1, or 1 μg/mL (from left to right for each indicated group). FIG. 12A shows the frequency of anti-BCMA CAR T cells after these treatments. FIG. 12B shows the number of anti-BCMA CAR T cells after these treatments.



FIGS. 13A-13B provide diagrams showing CAR T cell lysis in the presence of NK cells. FIG. 13A shows the frequency anti-BCMA CAR T cell lysis in a co-culture of anti-BCMA CAR T cells and purified NK cells from a normal donor that were pre-treated with daratumumab or isotype control mAb at 0.01, 0.1 or 1 μg/mL. Error bars represent standard error of mean (SEM) where n=3. FIG. 13B is a flow cytometry plot showing the levels of TCRa/b and β2M expression in the anti-BCMA CAR T cells prior to co-culture with NK cells as described in FIG. 13A.



FIGS. 14A-14C provide diagrams NK-cell mediated CAR T cell lysis in the presence or absence of daratumumab. FIG. 14A shows anti-BCMA CAR T cell frequency of CAR T cells after a 24 hour co-culture with daratumumab at 0.1, 1, or 10 μg/mL. Error bars represent standard error of mean (SEM) where n=3. FIG. 14B shows the protection from NK mediated cell lysis in co-cultures of anti-BCMA CAR T cells deficient in B2M and daratumumab-treated NK cells at a 1:1 ratio. The NK cells were from a normal donor and were pre-treated for 60 hours with daratumumab or isotype control mAb at 0.1, 1, or 10 μg/mL. FIG. 14C shows the protection from NK mediated cell lysis in co-cultures with a 3:1 ratio of daratumumab-treated NK cells to anti-BCMA CAR T cells. Error bars represent standard error of mean (SEM) where n=3.



FIGS. 15A-15B are graphs showing NK and CAR T cells numbers after CAR T cells anti-BCMA CAR T cells deficient in B2M were co-cultured with purified NK cells that were pre-treated for 60 hours with either daratumumab at concentrations of 0.01, 0.1, 1, 10, 100 or 300 μg/mL. Error bars represent standard error of mean (SEM) where n=3. FIG. 15A shows NK cell numbers after co-culturing for 72 hours. FIG. 15B shows T cell numbers after co-culturing for 72 hours.



FIG. 16A-16E are graphs showing tumor volume and survival of immune-deficient mice intravenously injected with 5×106 MM.1S cells/mouse, and treated with daratumumab, anti-BCMA CAR-T cells, or a combination thereof. FIGS. 16A and 16B are graphs showing tumor volume (16A) and survival (16B) of mice treated with a low dose of anti-BCMA CAR-T cells (0.8×106 CAR+ T cells) alone or in combination with daratumumab (15 mg/kg). FIGS. 16C and 16D are graphs showing tumor volume (16C) and survival (16D) of mice treated with a high dose of anti-BCMA CAR-T cells (2.4×106 CAR+ T cells) alone or in combination with daratumumab (15 mg/kg). FIG. 16E is a graph showing tumor volume at day 26 of mice treated with a high dose of anti-BCMA CAR-T cells alone or in combination with daratumumab.



FIGS. 17A-17C are graphs showing that Lenalidomide (Len) addition demonstrates beneficial effect on multiple aspects of BCMA directed CAR-T cells in vitro. FIG. 17A is a graph showing that Lenalidomide enhances proliferation of BCMA directed CAR-T cells in vitro. FIG. 17B is a graph showing that Lenalidomide reduces the expression of a senescence marker in BCMA directed CAR-T cell in vitro. FIG. 17C includes graphs showing that Lenalidomide enhances secretion of effector cytokines following antigen stimulation of BCMA directed CAR-T cell in vitro.



FIGS. 18A-18C are graphs that show that Lenalidomide (Len) enhances BCMA directed CAR-T cell activity in vivo. FIG. 18A is a graph showing that combination of BCMA directed CAR-T cells & lenalidomide enhance tumor regression. Top panel: 1.5 mg/ml lenalidomide. Bottom panel: 10 mg/ml lenalidomide. FIG. 18B is a graph showing that combination of BCMA directed CAR-T cells & lenalidomide prolongs mouse survival. Top panel: 1.5 mg/ml lenalidomide. Bottom panel: 10 mg/ml lenalidomide. FIG. 18C is a graph showing that combination of BCMA directed CAR-T cells with lenalidomide enhances CAR-T expansion in mice.



FIGS. 19A-19C are graphs showing that Lenalidomide does not enhance immune recognition of allogenic T cells. FIG. 19A is a graph showing that Lenalidomide does not enhance NK cytotoxicity towards TRAC−/B2M− T cells. FIG. 19B includes graphs showing that Lenalidomide does not enhance secretion of cytokines by NK cells upon stimulation by Allo T cells. FIG. 19C are graphs that show that reduced allo reactivity towards TRAC−/B2M− allogenic T cells is maintained in the presence of Lenalidomide.



FIG. 20 includes graphs showing that BCMA directed CAR-T cells produced in the presence of Lenalidomide exhibit increased cytokine secretion upon antigen stimulation. Top left: IFN-γ. Top middle: TNF-α. Top right: MIP1-α. Bottom left: IL-6. Bottom middle: MCP-1. Bottom right MIP1-β.



FIGS. 21A and 21B are graphs showing impact of Lenalidomide on CAR-T cell editing efficiency and CD4/CD8 cell ratio. FIG. 21A is a graph showing the CAR+%, TRAC−%, and B2M−% of anti-BCMA CAR-T cells on day 8. FIG. 21B is a graph showing CD4% and CD8% from anti-BCMA CAR-T cells expanded at small and medium scale on day 8.



FIG. 22 is a schematic illustration showing an exemplary schedule for a combined treatment comprising CTX120 cells and daratumumab. Subjects in Cohort 1 receive an IV infusion of daratumumab (single dose of 16 mg/kg) followed by LD chemotherapy (co-administration of fludarabine 30 mg/m2 and cyclophosphamide 300 mg/m2 IV daily for 3 days). Daratumumab may be administered as a subcutaneous injection rather than an IV infusion. Cyclophosphamide may be administered at a dose of up to 500 mg/m2 IV daily for 3 days. Daratumumab infusion is administered within 3 days prior to starting LD chemotherapy and no more than 14 days prior to CTX120 infusion. CTX120 is administered 48 hours to 7 days after LD chemotherapy. For subjects who achieve stable disease or better on Day 28, up to 5 additional monthly doses of daratumumab (16 mg/kg IV or SC equivalent) continue unless disease progression or unacceptable toxicity occurs. D: day; Dara: daratumumab; DLT: dose-limiting toxicity; IV: intravenously; LD: lymphodepleting; M: month.



FIG. 23 is a schematic illustration showing an exemplary treatment schedule for a combined therapy of CTX120 cells and lenalidomide. Subjects in Cohort 2 receive lenalidomide 10 mg administered orally once daily for 21 days beginning on the third day of LD chemotherapy (co-administration of fludarabine 30 mg/m2 and cyclophosphamide 300 mg/m2 IV daily for 3 days), continuing through CTX120 infusion. Cyclophosphamide may be administered at a dose of up to 500 mg/m2 IV daily for 3 days. For subjects who achieve stable disease or better on Day 28 post-CTX120 infusion and have met other criteria specified in the protocol, a 28-day cycle (21 days on and 7 days off) of 5 mg lenalidomide administration continue for up to 5 additional cycles unless disease progression or unacceptable toxicity occurs. D: day; DLT: dose-limiting toxicity; LD: lymphodepleting; M: month.



FIG. 24 is a schematic illustration showing an exemplary treatment schedule for a combined therapy of CTX120 cells, daratumumab, and lenalidomide. Subjects in Cohort 3 receive an IV infusion of daratumumab (single dose of 16 mg/kg) followed by LD chemotherapy (co-administration of fludarabine 30 mg/m2 and cyclophosphamide 300 mg/m2 IV daily for 3 days). Daratumumab may be administered as a subcutaneous injection rather than an IV infusion. Lenalidomide 10 mg is administered orally once daily for 21 days beginning on the third day of LD chemotherapy (co-administration of fludarabine 30 mg/m2 and cyclophosphamide 300 mg/m2 IV daily for 3 days), continuing through CTX120 infusion. Cyclophosphamide may be administered at a dose of up to 500 mg/m2 IV daily for 3 days. For subjects who achieve stable disease or better on Day 28, up to 5 additional monthly doses of daratumumab (16 mg/kg IV or SC equivalent) may continue unless disease progression or unacceptable toxicity occurs. For subjects who achieve stable disease or better on Day 28 post-CTX120 infusion and have met other criteria specified in the protocol, a 28-day cycle (21 days on and 7 days off) of 5 mg lenalidomide administration may continue for up to 5 additional cycles unless disease progression or unacceptable toxicity occurs. D: day; Dara: daratumumab; DLT: dose-limiting toxicity; LD: lymphodepleting; M: month; SC: subcutaneously.



FIG. 25 is a chart showing estimated daratumumab plasma concentration after a single dose or 3 consecutive doses. Dashed line indicates the approximate 90% effective concentration (EC90) for natural killer cell cytotoxicity.



FIG. 26 is a diagram showing NK cell depletion and recovery time frame in patients receiving CTX120, CTX120+daratumumab, or CTX120+lenalidomide. For CTX120, the patients received the dose of DL3 or DL4.



FIG. 27 is a diagram showing lymphocyte suppression in in patients receiving CTX120, CTX120+daratumumab, or CTX120+lenalidomide. For CTX120, the patients received the dose of DL3 or DL4.



FIG. 28 is a diagram showing CTX120 cell expansion in patients receiving CTX120, CTX120+daratumumab, or CTX120+lenalidomide. For CTX120, the patients received the dose of DL3 or DL4.





DETAILED DESCRIPTION OF THE INVENTION

B-cell maturation antigen (BCMA), also known as tumor necrosis factor receptor superfamily member 17 (TNFRSF17), is an antigenic determinant expressed by mature B cells. However, BCMA is differentially expressed in certain types of hematologic malignancies, wherein expression of BCMA is higher on malignant tumor cells than healthy cells. For example, BCMA is selectively expressed on the surface of multiple myeloma (MM) plasma cells and differentiated plasma cells, but not on memory B cells, naïve B cells, CD34+ hematopoietic stem cells, and other normal tissue cells (Cho, et al., (2018) Front Immuno., 9:1821). Without being bound by theory, BCMA is thought to promote the proliferation and survival of MM cells, as well as promote an immunosuppressive bone marrow microenvironment that protects the MM cells from immune detection.


Chimeric antigen receptor (CAR) T-cell therapy uses genetically-modified T cells to more specifically and efficiently target and kill cancer cells. After T cells have been collected from the blood, the cells are engineered to include CARs on their surface. The CARs may be introduced into the T cells using CRISPR/Cas9 gene editing technology. When these CAR T cells are injected into a patient, the receptors enable the T cells to kill cancer cells.


Without wishing to be bound by theory, it is believed that CAR T cells with disrupted MHC Class I are not able to provide the required MHC Class I-NK KIR receptor binding that prevents NK-cells from eliminating MHC-Class I sufficient cells, i.e., self-cells. Thus, allogeneic CAR T cells with disrupted MHC Class I are susceptible to elimination by NK cell-mediated immune surveillance. It was discovered that the administration of an NK cell inhibitor, such as anti-CD38 monoclonal antibody daratumumab, resulted in a reduction of NK cell numbers. The depletion of NK cells, in turn, protects the allogeneic CAR T cell from host NK-mediated cell lysis. The combination of CAR T cell therapy and NK cell inhibitors such as daratumumab thus presents an improvement over the existing CAR T cell therapy.


It was demonstrated that T cells isolated from PBMCs also express CD38 protein on the cell surface. Surprisingly, the addition of an anti-CD38 monoclonal antibody at doses that depleted NK cells did not affect T cell numbers, even after multi-day culture with an anti-CD38 monoclonal antibody. Nor does the addition of anti-CD38 monoclonal antibody at doses that depleted NK cell numbers induce CAR T cell activation. Accordingly, without wishing to be bound by theory, it is believed that anti-CD38 monoclonal antibody treatment is NK cell-specific and induces reduction of NK cells without causing undesirable non-specific CAR T cell activation or elimination. The addition of an NK cell inhibitor, such as an anti-CD38 monoclonal antibody, represents an improvement to existing CAR T cell therapy. See also International Patent Application No. PCT/IB2020/056085, the relevant discloses of which are incorporated by reference for the subject matter and purpose referenced herein.


It was further demonstrated that the effect of the anti-CD38 antibody on NK cells was not complement-dependent, as the addition of complement to co-culture of anti-CD38 antibody and PBMC did not affect the magnitude of NK cell depletion. More importantly, the addition of complement did not result in the depletion of T cells or affected CAR T cell activation status. Accordingly, without wishing to be bound by theory, it is believed that administration of an NK cell inhibitor, such as an anti-CD38 antibody, in combination with a CAR T cell therapy improves CAR T cell persistence and efficacy. Moreover, it was observed in an animal model that an anti-CD38 antibody successfully enhanced the anti-tumor effect of CAR-T cells targeting a tumor antigen (e.g., CD19 or BCMA). Without wishing to be bound by theory, it is believed that the combination therapy improves clinical response in the subject, for example, by increasing anti-tumor activity of the CAR T cell therapy.


Lenalidomide is a small molecule compounds that modulate the substrate activity of the CRL4CRBN E3 ubiquitin ligase. Lenalidomide is deemed as immunomodulatory drugs since they can increase IL-2 production in T lymphocytes and decrease pro-inflammatory cytokines. It is reported that lenalidomide can stimulate both T cells and NK cells, which could target both diseased cells and foreign cells. As such, there are concerns in the art that co-use of lenalidomide with allogeneic therapeutic cells may enhance immune recognition of the allogeneic therapeutic cells, thereby reducing the expected therapeutic effects.


The present disclosure reports that anti-BCMA CAR-T cells such as CTX120 cells successfully inhibited tumor growth as observed in an MM mouse model. Administration of the genetically engineered anti-BCMA CAR-T cells, having disrupted endogenous TRAC and β2M genes and expressing an anti-BCMA CAR, successfully eradicated human MM tumors that express BCMA as observed in animal models. Significantly, it has been observed that administration of the anti-BCMA CAR-T cells eliminated tumor burden and protected animals from re-challenge with tumors cells. Further, the genetically engineered anti-BCMA CAR-T cells, having disrupted endogenous TRAC and β2M genes did not induce graft versus host disease (GvHD) or host versus graft disease (HvGD) in animal models. Accordingly, the allogenic anti-BCMA CAR-T therapy disclosed herein are expected to be highly effective and safe in treating cancer such as MM in human patients.


Further, the present disclosure reports that the co-use of lenalidomide and the anti-BCMA CAR-T cells exhibited significantly higher anti-tumor effects as compared with the single agent in an MM mouse model. Surprisingly, lenalidomide did not enhance immune recognition of the allogeneic anti-BCMA-CAR T cells.


Accordingly, provided herein are methods for treating BCMA+ cancers using a combined therapy of (a) anti-BCMA CAR+ T cells (e.g., CTX120 cells disclosed herein) and (b) NK inhibitors such as anti-CD38 antibodies (preferably daratumumab) and/or lenalidomide or a derivative thereof as disclosed herein.


I. Genetically Engineered Anti-BCMA CAR-T Cells

In some aspects, the present disclosure provides a population of genetically engineered T cells expressing a CAR that specifically binds to BCMA (an anti-BCMA CAR or anti-BMCA CAR-T cells). In some embodiments, at least a portion of the genetically engineered T cells comprise: a nucleic acid encoding an anti-BCMA CAR; a disrupted gene associated with graft-versus-host disease (GvHD); and/or a disrupted gene associated with host-versus-graft (HvG) response. Methods of producing and using anti-BCMA CAR T cells are described in WO/2019/097305 and WO/2019/215500, the relevant disclosures of each of which are incorporated by reference herein for the purpose and subject matter referenced herein.


(i) Chimeric Antigen Receptor (CAR) Targeting BCMA (Anti-BCMA CAR)


A chimeric antigen receptor (CAR) refers to an artificial immune cell receptor that is engineered to recognize and bind to an antigen expressed by undesired cells, for example, disease cells such as cancer cells. A T cell that expresses a CAR polypeptide is referred to as a CAR T cell. CARs have the ability to redirect T-cell specificity and reactivity toward a selected target in a non-MHC-restricted manner The non-MHC-restricted antigen recognition gives CAR-T cells the ability to recognize an antigen independent of antigen processing, thus bypassing a major mechanism of tumor escape. Moreover, when expressed on T-cells, CARs advantageously do not dimerize with endogenous T-cell receptor (TCR) alpha and beta chains.


The anti-BCMA CAR disclosed herein refers to a CAR capable of binding to a BCMA molecule, preferably a BCMA molecule expressed on cell surfaces. The human and murine amino acid and nucleic acid sequences of BCMA can be found in a public database (e.g., GenBank, UniProt, or Swiss-Prot). See, e.g., UniProt/Swiss-Prot Accession Nos. Q02223 (human BCMA) and O88472 (murine BCMA). In general, an anti-BCMA CAR is a fusion polypeptide comprising an extracellular domain (ectodomain) that recognizes BCMA (e.g., a single chain fragment (scFv) of an antibody or other antibody fragment) and an intracellular domain (endodomain) comprising a signaling domain of the T-cell receptor (TCR) complex (e.g., CD3ζ) and, in most cases, a co-stimulatory domain. (Enblad et al., Human Gene Therapy. 2015; 26(8):498-505). The anti-BCMA CAR disclosed herein may further comprise a hinge and transmembrane domain between the extracellular domain and the intracellular domain, as well as a signal peptide at the N-terminus for surface expression. Examples of signal peptides include MLLLVTSLLLCELPHPAFLLIP (SEQ ID NO: 54) and MALPVTALLLPLALLLHAARP (SEQ ID NO: 55). Other signal peptides may be used. In some examples, the anti-BCMA CAR may further comprise an epitope tag such as a GST tag or a FLAG tag.


(a) Antigen Binding Extracellular Domain


The antigen-binding extracellular domain is the region of a CAR polypeptide that is exposed to the extracellular fluid when the CAR is expressed on cell surface. In some instances, a signal peptide may be located at the N-terminus to facilitate cell surface expression. In some embodiments, the antigen binding domain can be a single-chain variable fragment (scFv), which may include an antibody heavy chain variable region (VH) and an antibody light chain variable region (VL) (in either orientation). In some instances, the VH and VL fragment may be linked via a peptide linker. The linker, in some embodiments, includes hydrophilic residues with stretches of glycine and serine for flexibility as well as stretches of glutamate and lysine for added solubility. The linker peptide may be about 10 to about 25 amino acids. In specific examples, the linker peptide comprises a sequence set forth in SEQ ID NO: 53 (Table 5). The scFv fragment retains the antigen-binding specificity of the parent antibody, from which the scFv fragment is derived. In some embodiments, the scFv may comprise humanized VH and/or VL domains. In other embodiments, the VH and/or VL domains of the scFv are fully human


The antigen-binding extracellular domain of the anti-BCMA CAR disclosed herein is capable of binding to a BCMA molecule, preferably a BCMA molecule expressed on cell surface. The antigen-binding extracellular domain can be an antibody specific to BCMA or an antigen-binding fragment thereof. In some embodiments, the antigen-binding extracellular domain (the BCMA-binding domain) comprises a single-chain variable fragment (scFv), which may be derived from a suitable antibody, for example, a murine antibody, a rat antibody, a rabbit antibody, a human antibody, or a chimeric antibody. In some instances, the scFv is derived from a human anti-BCMA antibody. In other instances, the anti-BCMA scFv is humanized (e.g., fully humanized). For example, the anti-BCMA scFv is humanized and comprises one or more residues from complementarity determining regions (CDRs) of a non-human species, e.g., from mouse, rat, or rabbit.


In some embodiments, the anti-BCMA scFv comprises an antibody heavy chain variable region (VH) and an antibody light chain variable region (VL) (in either orientation), which comprise the same heavy chain complementary determining regions (CDRs) as the VH of SEQ ID NO:42 and the same light chain CDRs as the VL of SEQ ID NO:43. Two antibodies having the same VH and/or VL CDRs means that their CDRs are identical when determined by the same approach (e.g., the Kabat approach, the Chothia approach, the AbM approach, the Contact approach, or the IMGT approach as known in the art. See, e.g., bioinf.org.uk/abs/). For example, the anti-BCMA scFv may comprise the heavy chain and light chain CDR1s, CDR2s, and CDR3s provided in Table 5 below, following the Kabat approach. Alternative, the anti-BCMA scFv may comprise the heavy chain and light chain CDR1s, CDR2s, and CDR3s provided in Table 5 below, following the Chothia approach.


In other examples, the anti-BCMA scFv used in any of the anti-BCMA CAR constructs disclosed herein may be a functional variant of an anti-BCMA scFv comprising the amino acid sequence of SEQ ID NO:41 (exemplary anti-BCMA scFv). Such functional variants are substantially similar to the exemplary antibody, both structurally and functionally. A functional variant comprises substantially the same VH and VL CDRs as the exemplary anti-BCMA antibody. For example, it may comprise only up to 8 (e.g., 8, 7, 6, 5, 4, 3, 2, or 1) amino acid residue variations in the total CDR regions of the exemplary anti-BCMA scFv and binds the same epitope of BCMA with substantially similar affinity (e.g., having a KD value in the same order).


For example, an anti-BCMA scFv disclosed herein may comprises: a) a VL CDR1 comprising SEQ ID NO: 44, or a sequence having 1 to 3 amino acid substitutions relative to SEQ ID NO: 44; b) a VL CDR2 comprising SEQ ID NO: 45, or a sequence having 1 amino acid substitution relative to SEQ ID NO: 45; c) a VL CDR3 comprising SEQ ID NO: 46, or a sequence having 1 to 2 amino acid substitutions relative to SEQ ID NO: 46; and/or d) a VH CDR1 comprising SEQ ID NO: 47, or a sequence having 1 amino acid substitution relative to SEQ ID NO: 47; e) a VH CDR2 comprising SEQ ID NO: 48, or a sequence having 1 to 3 amino acid substitutions relative to SEQ ID NO: 48; f) a VH CDR3 comprising SEQ ID NO: 49, or a sequence having 1 to 2 amino acid substitutions relative to SEQ ID NO: 49, or any combination thereof. See Table 5. In some examples, the anti-BCMA scFv comprises: a VL CDR1 comprising SEQ ID NO: 44, a VL CDR2 comprising SEQ ID NO: 45, a VL CDR3 comprising SEQ ID NO: 46, a VH CDR1 comprising SEQ ID NO: 47, a VH CDR2 comprising SEQ ID NO: 48, and a VH CDR3 comprising SEQ ID NO: 49.


In other examples, the anti-BCMA scFv may comprise: a) a VL CDR1 comprising SEQ ID NO: 44, or a sequence having 1 to 3 amino acid substitutions relative to SEQ ID NO: 44; b) a VL CDR2 comprising SEQ ID NO: 45, or a sequence having 1 amino acid substitution relative to SEQ ID NO: 45; c) a VL CDR3 comprising SEQ ID NO: 46, or a sequence having 1 to 2 amino acid substitutions relative to SEQ ID NO: 46; and/or d) a VH CDR1 comprising SEQ ID NO: 50, or a sequence having 1 amino acid substitution relative to SEQ ID NO: 50; e) a VH CDR2 comprising SEQ ID NO: 51, or a sequence having 1 amino acid substitution relative to SEQ ID NO: 51; f) a VH CDR3 comprising SEQ ID NO: 52, or a sequence having 1 to 2 amino acid substitutions relative to SEQ ID NO: 52, or any combination thereof (Table 5). In some embodiments, the anti-BCMA scFv comprises: a VL CDR1 comprising SEQ ID NO: 44, a VL CDR2 comprising SEQ ID NO: 45, a VL CDR3 comprising SEQ ID NO: 46, a VH CDR1 comprising SEQ ID NO: 50, a VH CDR2 comprising SEQ ID NO: 51, and a VH CDR3 comprising SEQ ID NO: 52.


In some instances, the amino acid residue variations or substitution in one or more of the CDRs disclosed herein can be conservative amino acid residue substitutions. As used herein, a “conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made. Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references which compile such methods, e.g. Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989, or Current Protocols in Molecular Biology, F. M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York. Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) M, I, L, V; (b) F, Y, W; (c) K, R, H; (d) A, G; (e) S, T; (f) Q, N; and (g) E, D.


In some embodiments, the anti-BCMA scFv disclosed herein may comprise heavy chain CDRs that are at least 80% (e.g., 85%, 90%, 95%, or 98%) sequence identity, individually or collectively, as compared with the VH CDRs of the exemplary anti-BCMA scFv of SEQ ID NO:41. Alternatively or in addition, the anti-BCMA scFv may comprise light chain CDRs that are at least 80% (e.g., 85%, 90%, 95%, or 98%) sequence identity, individually or collectively, as compared with the VL CDRs as the exemplary anti-BCMA scFv. As used herein, “individually” means that one CDR of an antibody shares the indicated sequence identity relative to the corresponding CDR of the exemplary antibody. “Collectively” means that three VH or VL CDRs of an antibody in combination share the indicated sequence identity relative the corresponding three VH or VL CDRs of the exemplary antibody in combination.


In some examples, the anti-BCMA scFv may comprise a VH domain that comprises an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 42 (Table 5). Alternatively or in addition, the anti-BCMA scFv may comprise a VL domain that comprises an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 43 (Table 5). In some examples, the linker peptide connects the N-terminus of the anti-BCMA VH with the C-terminus of the anti-BCMA VL. Alternatively, the linker peptide connects the C-terminus of the anti-BCMA VH with the N-terminus of the anti-BCMA VL.


In some examples, the anti-BCMA scFv may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 41.


The “percent identity” of two amino acid sequences is determined using the algorithm of Karlin and Altschul Proc. Natl. Acad. Sci. USA 87:2264-68, 1990, modified as in Karlin and Altschul Proc. Natl. Acad. Sci. USA 90:5873-77, 1993. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. J. Mol. Biol. 215:403-10, 1990. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to the protein molecules of interest. Where gaps exist between two sequences, Gapped BLAST can be utilized as described in Altschul et al., Nucleic Acids Res. 25(17):3389-3402, 1997. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.


(b) Transmembrane Domain


The CAR polypeptide disclosed herein may contain a transmembrane domain, which can be a hydrophobic alpha helix that spans the membrane. As used herein, a “transmembrane domain” refers to any protein structure that is thermodynamically stable in a cell membrane, preferably a eukaryotic cell membrane. The transmembrane domain can provide stability of the CAR containing such.


In some embodiments, the transmembrane domain of a CAR as provided herein can be a CD8 transmembrane domain. In other embodiments, the transmembrane domain can be a CD28 transmembrane domain. In yet other embodiments, the transmembrane domain is a chimera of a CD8 and CD28 transmembrane domain. Other transmembrane domains may be used as provided herein. In some embodiments, the transmembrane domain is a CD8a transmembrane domain containing the sequence of FVPVFLPAKPTTTPAPRPPTPAPTIASQPLSLRPEACRPAAGG AVHTRGLDFACDIYIWAPLAGTCGVLLLSLVITLYCNHRNR (SEQ ID NO: 60) or IYIWAPLAGTCGVLLLSLVITLY (SEQ ID NO: 56). In some embodiments, the CD8a transmembrane domain may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 56. Other transmembrane domains may be used.


(c) Hinge Domain


In some embodiments, the anti-BCMA CAR further comprises a hinge domain, which may be located between the extracellular domain (comprising the antigen binding domain) and the transmembrane domain of the CAR, or between the cytoplasmic domain and the transmembrane domain of the CAR. A hinge domain can be any oligopeptide or polypeptide that functions to link the transmembrane domain to the extracellular domain and/or the cytoplasmic domain in the polypeptide chain. A hinge domain may function to provide flexibility to the CAR, or domains thereof, or to prevent steric hindrance of the CAR, or domains thereof.


In some embodiments, a hinge domain may comprise up to 300 amino acids (e.g., 10 to 100 amino acids, or 5 to 20 amino acids). In some embodiments, one or more hinge domain(s) may be included in other regions of a CAR. In some embodiments, the hinge domain may be a CD8 hinge domain. Other hinge domains may be used.


In some embodiments, the hinge domain comprises about 5 to about 300 amino acids, e.g., about 5 to about 250, about 10 to about 250, about 10 to about 200, about 15 to about 200, about 15 to about 150, about 20 to about 150, about 20 to about 100, about 25 to about 100, about 25 to about 75, or about 30 to about 750 amino acids. In some embodiments, the anti-BCMA hinge domain comprises a CD8a hinge domain and, optionally, an extension comprising an additional 1-10 amino acids (e.g., 4 amino acids) at the N-terminus of the hinge domain In some examples, the extension comprises amino acid sequence SAAA.


(d) Intracellular Signaling Domains


Any of the CAR constructs contain one or more intracellular signaling domains (e.g., CD3ζ, and optionally one or more co-stimulatory domains), which are the functional end of the receptor. Following antigen recognition, receptors cluster and a signal is transmitted to the cell.


CD3ζ is the cytoplasmic signaling domain of the T cell receptor complex. CD3ζ contains three (3) immunoreceptor tyrosine-based activation motif (ITAM)s, which transmit an activation signal to the T cell after the T cell is engaged with a cognate antigen. In many cases, CD3ζ provides a primary T cell activation signal but not a fully competent activation signal, which requires a co-stimulatory signaling. In some embodiments, the CD3ζ signaling domain comprises an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, or at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 59 (Table 5).


In some embodiments, the CAR polypeptides disclosed herein may further comprise one or more co-stimulatory signaling domains. For example, the co-stimulatory domains of CD28 and/or 4-1BB may be used to transmit a full proliferative/survival signal, together with the primary signaling mediated by CD3ζ. In some examples, the CAR disclosed herein comprises a CD28 co-stimulatory molecule. In other examples, the CAR disclosed herein comprises a 4-1BB co-stimulatory molecule. In some embodiments, a CAR includes a CD3ζ signaling domain and a CD28 co-stimulatory domain. In other embodiments, a CAR includes a CD3ζ signaling domain and 4-1BB co-stimulatory domain. In still other embodiments, a CAR includes a CD3ζ signaling domain, a CD28 co-stimulatory domain, and a 4-1BB co-stimulatory domain.


In some examples, the anti-BCMA CAR comprises a 4-1BB co-stimulatory domain. The 4-1BB co-stimulatory domain may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 57 (Table 5).


In some examples, the anti-BCMA CAR comprises a CD28 co-stimulatory domain. The CD28 co-stimulatory domain may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 58 (Table 5).


(e) Exemplary Anti-BCMA CAR


In some examples, the anti-BCMA CAR disclosed herein comprises, from the N-terminus to the C-terminus, a CD8 signaling peptide (e.g., SEQ ID NO:55), an anti-BCMA scFv (e.g., SEQ ID NO:41), a CD8a transmembrane domain (e.g., SEQ ID NO:56), a 4-1BB co-stimulatory domain (e.g., SEQ ID NO: 57), and a CD3z signaling domain (e.g., SEQ ID NO:59). Such an anti-BCMA CAR may comprise an amino acid sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 40 (Table 5). The anti-BCMA CAR may be encoded by a nucleic acid comprising a sequence at least 80%, at least 85%, at least 90%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to a sequence set forth in SEQ ID NO: 33 (Table 4).


In specific examples, the anti-BCMA CAR is CTX-166b, which comprises the amino acid sequence of SEQ ID NO: 40 (Table 5).


It should be understood that methods described herein encompasses more than one suitable CAR that can be used to produce genetically engineered T cells expressing the CAR, for example, those known in the art or disclosed herein. Examples can be found in WO2019/097305 and WO/2019/215500, the relevant disclosures of each of which are incorporated by reference for the purpose and subject matter referenced herein.


Expression of any of the anti-BCMA CAR (e.g., CTX-166b) can be driven by an endogenous promoter at the integration site. Alternatively, expression of the anti-BCMA CAR can be driven by an exogenous promoter. For example, an exogenous EF1α promoter (e.g., comprising the nucleotide sequence of SEQ ID NO: 38; see Table 4) can be located directly upstream of the nucleic acid sequence encoding the anti-BCMA CAR. In some embodiments, the anti-BCMA CAR expression cassette may further comprise an exogenous enhancer, an insulator, an internal ribosome entry site, a sequence encoding 2A peptides, a 3′ polyadenylation (poly A) signal, or a combination thereof. In specific examples, the 3′ poly A signal comprises a nucleotide sequence set forth in SEQ ID NO: 39 (Table 4).


(ii) Genetic Modification of TRAC and B2M Endogenous Genes


The anti-BCMA CAR-T cells may be further modified genetically to disrupt an endogenous gene associated with GvHD (e.g., a gene encoding a component of TCR such as a TRAC gene), an endogenous gene associated with HvGD (e.g., a β2M gene).


It should be understood that gene disruption encompasses gene modification through gene editing (e.g., using CRISPR/Cas gene editing to insert or delete one or more nucleotides). As used herein, the term “a disrupted gene” refers to a gene containing one or more mutations (e.g., insertion, deletion, or nucleotide substitution, etc.) relative to the wild-type counterpart so as to substantially reduce or completely eliminate the activity of the encoded gene product. The one or more mutations may be located in a non-coding region, for example, a promoter region, a regulatory region that regulates transcription or translation; or an intron region. Alternatively, the one or more mutations may be located in a coding region (e.g., in an exon). In some instances, the disrupted gene does not express or expresses a substantially reduced level of the encoded protein. In other instances, the disrupted gene expresses the encoded protein in a mutated form, which is either not functional or has substantially reduced activity. In some embodiments, a disrupted gene is a gene that does not encode functional protein. In some embodiments, a cell that comprises a disrupted gene does not express (e.g., at the cell surface) a detectable level (e.g. by antibody, e.g., by flow cytometry) of the protein encoded by the gene. A cell that does not express a detectable level of the protein may be referred to as a knockout cell. For example, a cell having a β2M gene edit may be considered a β2M knockout cell if β2M protein cannot be detected at the cell surface using an antibody that specifically binds β2M protein.


Disrupted TRAC Gene


GvHD is commonly seen in the setting of allogeneic stem cell transplantation (SCT). Immunocompetent donor T cells (the graft) recognize the recipient (the host) as foreign and become activated to attack the recipient to eliminate “foreign antigen-bearing” host cells. Clinically, GvHD is divided into acute, chronic, and overlap syndrome based upon clinical manifestations and the time of incidence relative to administration of allogeneic donor cells. Symptoms of acute GvHD (aGvHD) can include maculopapular rash; hyperbilirubinemia with jaundice due to damage to the small bile ducts, leading to cholestasis; nausea, vomiting, and anorexia; and watery or bloody diarrhea and cramping abdominal pain (Zeiser, R. et al. (2017) N Engl J Med 377:2167-79). The severity of aGvHD is based upon clinical manifestations and is readily evaluated by one skilled in the art using widely accepted grading parameters as defined, for example, in Table 17.


In some embodiments, the anti-BCMA CAR-T cells have a disrupted endogenous gene associated with GvHD, for example, an endogenous TRAC gene, to reduce the risk or eliminate GvHD when the anti-BCMA CAR-T cells are administered to a recipient. In some embodiments, the disrupted TRAC gene may comprise a deletion, a nucleotide residue substation, an insertion, or a combination thereof. Structure of a disrupted TRAC gene would depend on the gene editing method used to disrupt the endogenous TRAC gene. For example, the TRAC gene may be disrupted by the CRISPR/Cas9 system using a suitable guide RNA (e.g., those disclosed herein. See Table 1 and Example 1 below). Such a gene editing approach may create deletions, insertions, and/or nucleotide substitutions nearby the gene locus targeted by the guide RNA (gRNA).


In some embodiments, the genetically engineered anti-BCMA CAR-T cell comprises a disrupted TRAC gene, which comprises an insertion and/or a deletion. In some examples, the insertion and/or deletion is within Exon 1. In specific examples, the disrupted TRAC gene has a deletion of a fragment comprising SEQ ID NO: 10. Alternatively or in addition, the disrupted TRAC gene may comprise an insertion of a nucleic acid, which comprises a nucleotide sequence encoding any of the anti-BCMA CAR. In some examples, the anti-BCMA CAR-encoding sequence may be flanked by a left homology arm and a right homology arm, which comprise homologous sequences flanking the region targeted by the gene editing method for use in disrupting the TRAC gene in the T cells. In some instances, the left homology arm and the right homology arm comprise sequences homologous to a 5′ end and a 3′ end site nearby the region of SEQ ID NO:10, respectfully, such that via homologous recombination, the nucleic acid encoding an anti-BCMA CAR is inserted into the disrupted TRAC locus. In specific examples, an exogenous nucleic acid comprising the nucleotide sequence of SEQ ID NO: 33 (encoding an anti-BCMA CAR comprising the amino acid sequence of SEQ ID NO:40) can be inserted into the TRAC gene, for example, inserted at or nearby the region of SEQ ID NO:10. The exogenous nucleic acid may further comprise a promoter in operative linkage to the coding sequence of the anti-BCMA CAR to drive expression of the anti-BCMA CAR in the genetically engineered T cells as disclosed herein. In some examples, the promoter can be an EF-la promoter, which may comprise the nucleotide sequence of SEQ ID NO: 38. Alternatively or in addition, the exogenous nucleic acid may further comprise a poly A sequence downstream of the anti-BCMA CAR coding sequence.


Disrupted B2M Gene


HvGD refers to the immune rejection of donor cells, for example, tumor-targeting CAR T cells, by the recipient's immune system. Risk of tumor relapse with tumor-targeting CAR T cell therapy is thought to be due, in part, to limited persistence of CAR T cells in a subject following administration (Maude, S., et al. (2014) N Engl J Med. 371:1507-17; Turtle, C. et al., (2016) J Clin Invest. 126:2123-38) Elimination of allogeneic antigens from CAR T cells prior to transplantation can eliminate or reduce the risk of host rejections (e.g., a HvG response), thereby increasing persistence following administration.


In some embodiments, the genetically engineered anti-BCMA CAR-T cells may comprise a genetic disruption in a gene associated with HvGD, either alone or in combination with disruption of a gene associated with GvHD (e.g., TRAC gene disclosed herein). In some embodiments, the gene associated with HvGD encodes a component of major histocompatibility (MHC) class I molecules, for example, the β2M gene. Disruption of the gene associated with HvGD, e.g., disruption of the β2M gene, minimizes the risk of HvGD. Alternatively or in addition, the disruption of the β2M gene improves persistence of the CAR T cells.


In some embodiments, the genetically engineered anti-BCMA CAR-T cells comprise a disrupted β2M gene, either alone or in combination with a disrupted TRAC gene, comprises a genetic modification, which can be a deletion, an insertion, a nucleotide residue substitution, or a combination thereof. Structure of a disrupted β2M gene would depend on the gene editing method used to disrupt the endogenous β2M gene. For example, the β2M gene may be disrupted by the CRISPR/Cas9 system using a suitable guide RNA (e.g., those disclosed herein. See Table 1 and Example 1 below). Such a gene editing approach may create deletions, insertions, and/or nucleotide substitutions nearby the gene locus targeted by the guide RNA (gRNA).


In some examples, the disrupted β2M gene comprises a deletion, an insertion, a substitution, or a combination thereof in SEQ ID NO: 12 (Table 1). In examples, the disrupted β2M gene comprises at least one nucleotide sequence of any one of SEQ ID NO:21-26 (Table 3).


(iii) Population of Anti-BCMA CAR-T Cells


The present disclosure also provides a population of genetically engineered anti-BCMA CAR-T cells disclosed herein, which express an anti-BCMA CAR and have a disrupted endogenous TRAC gene, an endogenous β2M gene, or both. In some embodiments, the population of the genetically engineered anti-BCMA CAR-T cells is heterogeneous, i.e., comprising genetically engineered T cells having different or different combination of the genetic modifications as disclosed herein (i.e., expression of anti-BCMA CAR, disrupted endogenous TRAC gene, and disrupted endogenous β2M gene). For example, the population of genetically engineered T cells may comprise a first group of T cells expressing the anti-BCMA CAR as disclosed herein and having a disrupted TRAC gene and a second group of T cells expressing the anti-BCMA CAR and a disrupted β2M gene. The first group and second group of the T cells may overlap. In some examples, a portion of the T cell population disclosed herein comprises all of the three genetic modifications, including expression of an anti-BCMA CAR, disrupted TRAC gene, and disrupted β2M gene.


In some embodiments, a portion of the population of genetically engineered T cells express an anti-BCMA CAR and comprise a disrupted TRAC gene, which may comprise an insertion, a deletion, a substitution, or a combination thereof. In some embodiments, the disruption of the TRAC gene eliminates or decreases expression of the TCR in the genetically engineered T cells. In some examples, 50% or less of the T cells express a TCR (TCR+), for example, 45% or less, 40% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, 9% or less, 8% or less, 7% or less, 6% or less, 5% or less, 4% or less, 3% or less, 2% or less, 1% or less, 0.9% or less, 0.8% or less, 0.7% or less, 0.6% or less, 0.5% or less, 0.4% or less, 0.3% or less, 0.2% or less, or 0.1% or less. In some examples, 0.05%-50% of the genetically engineered T cells express a TCR, for example, 10%-50%, 20%-50%, 30%-50%, 40%-50%, 0.05%-40%, 10%-40%, 20%-40%, 30%-40%, 0.05%-30%, 10%-30%, 20%-30%, 0.05%-20%, 10%-20%, or 0.05%-10% of the genetically engineered T cells express a TCR. In some examples, 0.4% or less of the genetically engineered T cells express a TCR.


In some embodiments, the population of genetically engineered T cells elicits no clinical manifestations of GVHD response in a subject. For example, the genetically engineered T cells elicits no clinical manifestations of aGvHD (e.g., steroid-refractory aGvHD) in the subject. In some examples, the genetically engineered T cells elicits no clinically significant (e.g., grade 2-4) aGvHD in the subject. In some examples, the genetically engineered T cells elicits only mild aGvHD response (e.g., below clinical grade 2, 1, or 0) in the subject. In some examples, the genetically engineered T cells elicit clinically significant (e.g., grade 2-4) aGvHD (e.g., steroid-refractory aGvHD) in less than 18% of the subjects, e.g., less than 16%, less than 14%, less than 12%, less than 10%, less than 8%, less than 6%, less than 5%, less than 4%, less than 3%, less than 2%, or less than 1%.


In some embodiments, risk of GvHD (e.g., clinically significant aGvHD) elicited by the population of genetically engineered T cells as disclosed herein are reduced compared to a T cell population where at least 50% of the T cells express a TCR, e.g., at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%. In some examples, the reduction in clinically significant aGvHD (e.g., grade 2-4) is at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or 100%.


In some embodiments, symptoms of aGvHD is observed for up to 36 days after administration of the population of genetically engineered T cells disclosed herein, e.g., up to 21 days, up to 24 days, up to 28 days, up to 30 days, or up to 35 days. In some examples, symptoms of aGvHD is observed for about 20 to about 50 days, about 25 to about 70 days, or about 28 to about 100 days after administration of the T cell population.


Alternatively or in addition, a portion of the genetically engineered T cells express an anti-BCMA CAR and comprise a disrupted β2M gene, which may comprise an insertion, a deletion, a substitution, or a combination thereof. In some embodiments, the disruption of the β2M gene eliminates or decreases expression of β2 microglobulin, leading to a loss of function of the MHC I complex. In some examples, 50% or less of the genetically engineered T cell population express β2 microglobulin, e.g., 45% or less, 40% or less, 35% or less, 30% or less, 25% or less, 20% or less, 15% or less, 10% or less, or 5% or less. In some examples, about 5% to about 50% of the genetically engineered T cells in the T cell population express β2 microglobulin, e.g., about 10%-50%, 10%-45%, 15%-45%, 15%-40%, 20%-40%, 20%-35%, or 25%-35%. In some examples, 30% or less of the genetically engineered T cells express β2 microglobulin.


In some embodiments, the genetic disruption of the gene associated with HvG (e.g., the β2M gene) eliminates or reduces the risk of HvGD response. Alternatively or in addition, the genetic disruption of the gene associated with HvGD (e.g., the β2M gene) increases the persistence of the allogeneic T cells in the subject. In some examples, a subject receiving the genetically engineered T cell population disclosed herein has no clinical manifestations of HvGD response. In some examples, the genetically engineered T cells are detectable in a tissue (e.g., in peripheral blood) of the subject at least 1 day after administration, e.g., at least 2, 4, 5, 7, 10, 14, 15, 20, 21, 25, 28, 30, or 35 days. The tissue may be obtained from peripheral blood, cerebrospinal fluid, tumor, skin, bone, bone marrow, breast, kidney, liver, lung, lymph node, spleen, gastrointestinal tract, tonsils, thymus, prostate, or a combination thereof.


Detectable is defined in terms of the limit of detection of a method of analysis. Persistence is the duration of time after administration where a detectable quantity of allogeneic T cells is measured. Methods for detecting or quantity T cells in a tissue of interest are known to those of skill in the art. Such methods include, but are not limited to, reverse transcription polymerase chain reaction (RT-PCR), competitive RT-PCR, real-time RT-PCR, RNase protection assay (RPA), quantitative immunofluorescence (QIF), flow cytometry, northern blotting, nucleic acid microarray using DNA, western blotting, enzyme-linked immunosorbent assay (ELISA), radioimmunoassay (RIA), tissue immunostaining, immunoprecipitation assay, complement fixation assay, fluorescence-activated cell sorting (FACS), mass spectrometry, magnetic bead-antibody immunoprecipitation, or protein chip.


In specific examples, the population of genetically engineered anti-BCMA CAR-T cells are CTX120 cells (see also Example 1 below), which are produced using CRISPR/Cas technology to disrupt targeted genes (TRAC and β2M), and adeno-associated virus (AAV) transduction to deliver the CAR construct of SEQ ID NO:40 CRISPR-Cas9-mediated gene editing involves two guide RNAs (sgRNAs): TA-1 sgRNA (SEQ ID NO: 1), which targets the TRAC locus, and B2M-1 sgRNA (SEQ ID NO: 5), which targets the β2M locus. The anti-BCMA CAR of the CTX120 cells is composed of an anti-BCMA single-chain antibody fragment (scFv) specific for BCMA, followed by a CD8 hinge and transmembrane domain that is fused to an intracellular co-signaling domain of 4-1BB and a CD3 signaling domain. The anti-BCMA scFv comprises the amino acid sequence of SEQ ID NO:41 and the anti-BCMA CAR comprises the amino acid sequence of SEQ ID NO: 40. Sequences of the other components in the anti-BCMA CAR are provided in Tables 4 and 5 below.


At least a portion of the CTX120 cells comprises anti-BCMA CAR-expressing T cells with a disrupted TRAC gene, in which the fragment of SEQ ID NO:10 is deleted. An exogenous nucleic acid configured for expressing the anti-BCMA CAR can be inserted into the TRAC gene. The exogenous nucleic acid comprises a promoter sequence (e.g., EF-1a promoter, which may comprise the nucleotide sequence of SEQ ID NO: 38), a nucleotide sequence coding for an anti-BCMA CAR (e.g., SEQ ID NO: 33, coding for the anti-BCMA CAR comprising the amino acid sequence of SEQ ID NO: 40), and a poly A sequence (e.g., SEQ ID NO: 39) downstream of the coding sequence. The promoter sequence is in operable linkage to the coding sequence such that it drives expression of the anti-BCMA CAR in the CTX120 cells. At least a portion of the CTX120 cells comprise, collectively, a population of disrupted β2M genes, which may comprise one or more of nucleotide sequence of SEQ ID Nos: 21-26. See also FIG. 1 and Example 1 below.


Further, at least 30% of the T cells in the CTX120 cell population express the anti-BCMA CAR (CAR+ cells). In some examples, about 40% to about 80% (e.g., about 40%-75%, about 45%-75%, about 50%-70%, or about 50%-60%) of the T cells in the CTX120 cell population are CAR+. In addition, less than 35% (e.g., ≤30%) at of the T cells in the CTX120 cell population express a detectable level of β2M surface protein. For example, about 70% to about 85% of the T cells in the CTX120 cell population do not express a detectable level of β2M surface protein. Moreover, less than about 1% (e.g., less than about 0.8%, less than 0.5%, or less than 4%) of the T cells in the CTX120 cell population express functional TCR.


At least a portion of the CTX120 T cells (e.g., at least 35%) are triple-modified CAR T cells, which refer to a genetically engineered T cell expressing the anti-BCMA CAR and having disrupted endogenous TRAC gene and endogenous β2M gene, e.g., produced by the CRISPR/Cas9 approach disclosed above and AAV-mediated delivery of the CAR construct. In some examples, about 35% to about 70% (e.g., about 40% to about 70% or about 50% to about 65%) of the T cells in the CTX120 cell population are triple-modified CAR T cells.


(iv) Pharmaceutical Compositions


In some aspects, the present disclosure provides pharmaceutical compositions comprising any of the genetically engineered anti-BCMA CAR T cells as disclosed herein, for example, CTX120 cells, and a pharmaceutically acceptable carrier. Such pharmaceutical compositions can be used in cancer treatment in human patients, which is also disclosed herein.


As used herein, the term “pharmaceutically acceptable” refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues, organs, and/or bodily fluids of the subject without excessive toxicity, irritation, allergic response, or other problems or complications commensurate with a reasonable benefit/risk ratio. As used herein, the term “pharmaceutically acceptable carrier” refers to solvents, dispersion media, coatings, antibacterial agents, antifungal agents, isotonic and absorption delaying agents, or the like that are physiologically compatible. The compositions can include a pharmaceutically acceptable salt, e.g., an acid addition salt or a base addition salt. See, e.g., Berge et al., (1977) J Pharm Sci 66:1-19.


In some embodiments, the pharmaceutical composition further comprises a pharmaceutically acceptable salt. Non-limiting examples of pharmaceutically acceptable salts include acid addition salts (formed from a free amino group of a polypeptide with an inorganic acid (e.g., hydrochloric or phosphoric acids), or an organic acid such as acetic, tartaric, mandelic, or the like). In some embodiments, the salt formed with the free carboxyl groups is derived from an inorganic base (e.g., sodium, potassium, ammonium, calcium or ferric hydroxides), or an organic base such as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, or the like).


In some embodiments, the pharmaceutical composition disclosed herein comprises a population of the genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) suspended in a cryopreservation solution (e.g., CryoStor® C55). In some instances, the cryopreservation solution may contain about 2-10% dimethyl sulfoxide (DMSO). For example, the cryopreservation solution may contain about 2%, about 3%, about 4%, about 5%, about 6%, about 7%, about 8%, about 9%, or about 10% DMSO. In specific examples, the cryopreservation solution may contain about 5% DMSO.


In addition to DMSO, a cryopreservation solution for use in the present disclosure may also comprise adenosine, dextrose, dextran-40, lactobionic acid, sucrose, mannitol, a buffer agent such as N-)2-hydroxethyl) piperazine-N′-(2-ethanesulfonic acid) (HEPES), one or more salts (e.g., calcium chloride, magnesium chloride, potassium chloride, postassium bicarbonate, potassium phosphate, etc.), one or more base (e.g., sodium hydroxide, potassium hydroxide, etc.), or a combination thereof. Components of a cryopreservation solution may be dissolved in sterile water (injection quality). Any of the cryopreservation solution may be substantially free of serum (undetectable by routine methods).


In some instances, a pharmaceutical composition comprising a population of genetically engineered anti-BCMA CAR-T cells such as the CTX120 cells suspended in a cryopreservation solution (e.g., comprising about 5% DMSO and optionally substantially free of serum) may be placed in storage vials. In some examples, each storage vial may contain about 25-85×106 cells/ml of the T cells (e.g., CTX120). In some examples, each storage vial may contain about 50×106 cells/ml. Among the cells in a storage vial, ≥30% are CAR+ T cells, ≤0.4% are TCR+ T cells, and ≤30% are B2M+ T cells.


Any of the pharmaceutical compositions disclosed herein, comprising a population of genetically engineered anti-BCMA CAR T cells as also disclosed herein (e.g., CTX120 cells), which optionally may be suspended in a cryopreservation solution (e.g., comprising about 5% DMSO and optionally substantially free of serum), may be stored in an environment that does not substantially affect viability and bioactivity of the T cells for future use, e.g., under conditions commonly applied for storage of cells and tissues. In some examples, the pharmaceutical composition may be stored in the vapor phase of liquid nitrogen at ≤−135° C. No significant changes were observed with respect to appearance, cell count, viability, % CAR+ T cells, % TCR+ T cells, and % B2M+ T cells after the cells have been stored under such conditions for a period of time.


II. Preparation of Genetically Engineered Anti-BCMA CAR-T Cells

Any suitable gene editing methods known in the art can be used for making the genetically engineered anti-BCMA CAR T cells disclosed herein, for example, nuclease-dependent targeted editing using zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), or RNA-guided CRISPR-Cas9 nucleases (CRISPR/Cas9; Clustered Regular Interspaced Short Palindromic Repeats Associated 9).


(a) Sources of T Cells


In some embodiments, primary T cells isolated from one or more donors may be used for making the genetically engineered anti-BCMA CAR-T cells. For example, primary T cells may be isolated from a suitable tissue of one or more healthy human donors, e.g., peripheral blood mononuclear cells (PBMCs), bone marrow, lymph nodes tissue, cord blood, thymus issue, tissue from a site of infection, ascites, pleural effusion, spleen tissue, or a combination thereof. In some embodiments, a subpopulation of primary T cells expressing TCRαβ, CD3, CD4, CD8, CD27 CD28, CD38, CD45RA, CD45RO, CD62L, CD127, CD122, CD95, CD197, CCR7, KLRG1, MHC-I proteins, MHC-II proteins, or a combination thereof may be further enriched, using a positive or negative selection technique, which is known in the art. In some embodiments, the T cell subpopulation express TCRαβ, CD4, CD8, or a combination thereof. In some embodiments, the T cell subpopulation express CD3, CD4, CD8, or a combination thereof. In some embodiments, the primary T cells for use in making the genetic edits disclosed herein may comprise at least 40%, at least 50%, or at least 60% CD27+CD45RO− T cells.


In other embodiments, the T cells for use in generating the genetically engineered T cells disclosed herein may be derived from a T cell bank. A T cell bank may comprise T cells with genetic editing of certain genes (e.g., genes involved in cell self renewal, apoptosis, and/or T cell exhaustion or replicative senescence) to improve T cell persistence in cell culture. A T cell bank may be produced from bona fide T cells, for example, non-transformed T cells, terminally differentiated T cells, T cells having stable genome, and/or T cells that depend on cytokines and growth factors for proliferation and expansion. Alternatively, such a T cell bank may be produced from precursor cells such as hematopoietic stem cells (e.g., iPSCs), e.g., in vitro culture. In some examples, the T cells in the T cell bank may comprise genetic editing of one or more genes involved in cell self-renewal, one or more genes involved in apoptosis, and/or one or more genes involved in T cell exhaustion, so as to disrupt or reduce expression of such genes, leading to improved persistence in culture. Examples of the edited genes in a T cell bank include, but are not limited to, Tet2, Fas, CD70, Reg1, or a combination thereof. Compared with the non-edited T counterpart, T cells in a T cell bank may have enhanced expansion capacity in culture, enhanced proliferation capacity, greater T cell activation, and/or reduced apoptosis levels. Additional information of T cell bank may be found in International Application No. PCT/IB2020/058280, the relevant disclosures of which are incorporated by reference for the subject matter and purpose referenced herein.


In some embodiments, parent T cells for use in making the genetically engineered CAR T cells (e.g., any of the T cells derived from primary T cell sources) may be undergone one or more rounds of stimulation, activation, expansion, or a combination thereof. In some embodiments, the parent T cells are activated and stimulated to proliferate in vitro before gene editing. In some embodiments, the T cells are activated, expanded, or both, before or after gene editing. In some embodiments, the T cells are activated and expanded at the same time as gene editing. In some embodiments, the T cells are activated and expanded for about 1-4 days, e.g., about 1-3 days, about 1-2 days, about 2-3 days, about 2-4 days, about 3-4 days, about 1 day, about 2 days, about 3 days, or about 4 days. In some embodiments, the allogeneic T cells are activated and expanded for about 4 hours, about 6 hours, about 12 hours, about 18 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours, or about 72 hours. Non-limiting examples of methods to activate and/or expand T cells are described in U.S. Pat. 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; and 6,867,041.


(ii) CRISPR-Cas9-Mediated Gene Editing System


Any of the parent T cells may be subject to one or more genetic editing/modification steps to introduce the gene editing events disclosed herein, i.e., disrupt endogenous TRAC gene, disrupt endogenous β2M gene, and/or introducing a nucleic acid coding for any of the anti-BCMA CAR as disclosed herein. Conventional genetically engineering approaches, such as gene editing approaches (e.g., those disclosed herein) can be used. In some examples, the genetic modifications of the T cells can be implemented by a CRISPR/Cas9-mediated gene editing system.


The CRISPR-Cas9 system is a naturally-occurring defense mechanism in prokaryotes that has been repurposed as an RNA-guided DNA-targeting platform used for gene editing. It relies on the DNA nuclease Cas9, and two noncoding RNAs, crisprRNA (crRNA) and trans-activating RNA (tracrRNA), to target the cleavage of DNA. CRISPR is an abbreviation for Clustered Regularly Interspaced Short Palindromic Repeats, a family of DNA sequences found in the genomes of bacteria and archaea that contain fragments of DNA (spacer DNA) with similarity to foreign DNA previously exposed to the cell, for example, by viruses that have infected or attacked the prokaryote. These fragments of DNA are used by the prokaryote to detect and destroy similar foreign DNA upon re-introduction, for example, from similar viruses during subsequent attacks. Transcription of the CRISPR locus results in the formation of an RNA molecule comprising the spacer sequence, which associates with and targets Cas (CRISPR-associated) proteins able to recognize and cut the foreign, exogenous DNA. Numerous types and classes of CRISPR/Cas systems have been described (see, e.g., Koonin et al., (2017) Curr Opin Microbiol 37:67-78).


crRNA drives sequence recognition and specificity of the CRISPR-Cas9 complex through Watson-Crick base pairing typically with a 20 nucleotide (nt) sequence in the target DNA. Changing the sequence of the 5′ 20nt in the crRNA allows targeting of the CRISPR-Cas9 complex to specific loci. The CRISPR-Cas9 complex only binds DNA sequences that contain a sequence match to the first 20 nt of the crRNA, if the target sequence is followed by a specific short DNA motif (with the sequence NGG) referred to as a protospacer adjacent motif (PAM).


TracrRNA hybridizes with the 3′ end of crRNA to form an RNA-duplex structure that is bound by the Cas9 endonuclease to form the catalytically active CRISPR-Cas9 complex, which can then cleave the target DNA.


Once the CRISPR-Cas9 complex is bound to DNA at a target site, two independent nuclease domains within the Cas9 enzyme each cleave one of the DNA strands upstream of the PAM site, leaving a double-strand break (DSB) where both strands of the DNA terminate in a base pair (a blunt end).


After binding of CRISPR-Cas9 complex to DNA at a specific target site and formation of the site-specific DSB, the next key step is repair of the DSB. Cells use two main DNA repair pathways to repair the DSB: non-homologous end joining (NHEJ) and homology-directed repair (HDR).


NHEJ is a robust repair mechanism that appears highly active in the majority of cell types, including non-dividing cells. NHEJ is error-prone and can often result in the removal or addition of between one and several hundred nucleotides at the site of the DSB, though such modifications are typically <20 nt. The resulting insertions and deletions (indels) can disrupt coding or noncoding regions of genes. Alternatively, HDR uses a long stretch of homologous donor DNA, provided endogenously or exogenously, to repair the DSB with high fidelity. HDR is active only in dividing cells and occurs at a relatively low frequency in most cell types. In many embodiments of the present disclosure, NHEJ is utilized as the repair operant.


(a) Cas9


In some embodiments, the Cas9 (CRISPR associated protein 9) endonuclease is used in a CRISPR method for making the genetically engineered T cells as disclosed herein. The Cas9 enzyme may be one from Streptococcus pyogenes, although other Cas9 homologs may also be used. It should be understood, that wild-type Cas9 may be used or modified versions of Cas9 may be used (e.g., evolved versions of Cas9, or Cas9 orthologues or variants), as provided herein. In some embodiments, Cas9 comprises a Streptococcus pyogenes-derived Cas9 nuclease protein that has been engineered to include C- and N-terminal SV40 large T antigen nuclear localization sequences (NLS). The resulting Cas9 nuclease (sNLS-spCas9-sNLS) is a 162 kDa protein that is produced by recombinant E. coli fermentation and purified by chromatography. The spCas9 amino acid sequence can be found as UniProt Accession No. Q99ZW2, which is provided herein as SEQ ID NO: 61.











Amino acid sequence of Cas9



nuclease (SEQ ID NO: 61):



MDKKYSIGLDIGTNSVGWAVITDEYKVPSKKEKVLG






NTDRHSIKKNLIGALLEDSGETAEATRLKRTARRR






YTRRKNRICYLQEIFSNEMAKVDDSFFHRLEESFL






VEEDKKHERHPIFGNIVDEVAYHEKYPTIYHLRKK






LVDSTDKADLRLIYLALAHMIKFRGHFLIEGDLNP






DNSDVDKLFIQLVQTYNQLFEENPINASGVDAKAI






LSARLSKSRRLENLIAQLPGEKKNGLFGNLIALSL






GLIPNEKSNEDLAEDAKLQLSKDTYDDDLDNLLAQ






IGDQYADLFLAAKNLSDAILLSDILRVNTEITKAP






LSASMIKRYDEHHQDLTLLKALVRQQLPEKYKEIF






FDQSKNGYAGYIDGGASQEEFYKFIKPILEKMDGT






EELLVKLNREDLLRKQRTFDNGSIPHQIHLGELHA






ILRRQEDFYPFLKDNREKIEKILTFRIPYYVGPLA






RGNSRFAWMTRKSEETITPWNFEEVVDKGASAQSF






IERMTNEDKNLPNEKVLPKHSLLYEYFTVYNELTK






VKYVTEGMRKPAFLSGEQKKAIVDLLEKTNRKVIV






KQLKEDYFKKIECEDSVEISGVEDRFNASLGTYHD






LLKIIKDKDFLDNEENEDILEDIVLTLTLFEDREM






IEERLKTYAHLFDDKVMKQLKRRRYTGWGRLSRKL






INGIRDKQSGKTILDELKSDGFANRNFMQLIHDDS






LTEKEDIQKAQVSGQGDSLHEHIANLAGSPAIKKG






ILQTVKVVDELVKVMGRHKPENIVIEMARENQTTQ






KGQKNSRERMKRIEEGIKELGSQILKEHPVENTQL






QNEKLYLYYLQNGRDMYVDQELDINRLSDYDVDHI






VPQSFLKDDSIDNKVLTRSDKNRGKSDNVPSEEVV






KKMKNYWRQLLNAKLITQRKEDNLIKAERGGLSEL






DKAGFIKRQLVETRQITKHVAQILDSRMNTKYDEN






DKLIREVKVITLKSKLVSDFRKDFQFYKVREINNY






HHAHDAYLNAVVGTALIKKYPKLESEFVYGDYKVY






DVRKMIAKSEQEIGKATAKYFFYSNIMNFFKTEIT






LANGEIRKRPLIETNGETGEIVWDKGRDFATVRKV






LSMPQVNIVKKTEVQTGGFSKESILPKRNSDKLIA






RKKDWDPKKYGGFDSPTVAYSVLVVAKVEKGKSKK






LKSVKELLGITIMERSSFEKNPIDFLEAKGYKEVK






KDLIIKLPKYSLFELENGRKRMLASAGELQKGNEL






ALPSKYVNFLYLASHYEKLKGSPEDNEQKQLFVEQ






HKHYLDEIIEQISEFSKRVILADANLDKVLSAYNK






HRDKPIREQAENIIHLFTLTNLGAPAAFKYFDTTI






DRKRYTSTKEVLDATLIHQSITGLYETRIDLSQLG






GD






(b) Guide RNAs (gRNAs)


CRISPR-Cas9-mediated gene editing as described herein includes the use of a guide RNA or a gRNA. As used herein, a “gRNA” refers to a genome-targeting nucleic acid that can direct the Cas9 to a specific target sequence within a TRAC gene or a β2M gene for gene editing at the specific target sequence. A guide RNA comprises at least a spacer sequence that hybridizes to a target nucleic acid sequence within a target gene for editing, and a CRISPR repeat sequence.


Exemplary gRNAs targeting a TRAC gene may comprise a nucleotide sequence provided in any one of SEQ ID NOs: 1-4. See WO 2019/097305A2, the relevant disclosures of which are incorporated by reference herein for the subject matter and purpose referenced herein. Other gRNA sequences may be designed using the TRAC gene sequence located on chromosome 14 (GRCh38: chromosome 14: 22,547,506-22,552,154; Ensembl; ENSG00000277734). In some embodiments, gRNAs targeting the TRAC genomic region and Cas9 create breaks in the TRAC genomic region resulting Indels in the TRAC gene disrupting expression of the mRNA or protein.


Exemplary gRNAs targeting a β2M gene may comprise a nucleotide sequence provided in any one of SEQ ID NOs: 5-8. See also WO 2019/097305A2, the relevant disclosures of which are incorporated by reference herein for the purpose and subject matter referenced herein. Other gRNA sequences may be designed using the β2M gene sequence located on Chromosome 15 (GRCh38 coordinates: Chromosome 15: 44,711,477-44,718,877; Ensembl: ENSG00000166710). In some embodiments, gRNAs targeting the β2M genomic region and RNA-guided nuclease create breaks in the β2M genomic region resulting in Indels in the β2M gene disrupting expression of the mRNA or protein.


In Type II systems, the gRNA also comprises a second RNA called the tracrRNA sequence. In the Type II gRNA, the CRISPR repeat sequence and tracrRNA sequence hybridize to each other to form a duplex. In the Type V gRNA, the crRNA forms a duplex. In both systems, the duplex binds a site-directed polypeptide, such that the guide RNA and site-direct polypeptide form a complex. In some embodiments, the genome-targeting nucleic acid provides target specificity to the complex by virtue of its association with the site-directed polypeptide. The genome-targeting nucleic acid thus directs the activity of the site-directed polypeptide.


As is understood by the person of ordinary skill in the art, each guide RNA is designed to include a spacer sequence complementary to its genomic target sequence. See Jinek et al., Science, 337, 816-821 (2012) and Deltcheva et al., Nature, 471, 602-607 (2011).


In some embodiments, the genome-targeting nucleic acid (e.g., gRNA) is a double-molecule guide RNA. In some embodiments, the genome-targeting nucleic acid (e.g., gRNA) is a single-molecule guide RNA.


A double-molecule guide RNA comprises two strands of RNA molecules. The first strand comprises in the 5′ to 3′ direction, an optional spacer extension sequence, a spacer sequence and a minimum CRISPR repeat sequence. The second strand comprises a minimum tracrRNA sequence (complementary to the minimum CRISPR repeat sequence), a 3′ tracrRNA sequence and an optional tracrRNA extension sequence.


A single-molecule guide RNA (referred to as a “sgRNA”) in a Type II system comprises, in the 5′ to 3′ direction, an optional spacer extension sequence, a spacer sequence, a minimum CRISPR repeat sequence, a single-molecule guide linker, a minimum tracrRNA sequence, a 3′ tracrRNA sequence and an optional tracrRNA extension sequence. The optional tracrRNA extension may comprise elements that contribute additional functionality (e.g., stability) to the guide RNA. The single-molecule guide linker links the minimum CRISPR repeat and the minimum tracrRNA sequence to form a hairpin structure. The optional tracrRNA extension comprises one or more hairpins. A single-molecule guide RNA in a Type V system comprises, in the 5′ to 3′ direction, a minimum CRISPR repeat sequence and a spacer sequence.


The “target sequence” is in a target gene that is adjacent to a PAM sequence and is the sequence to be modified by Cas9. The “target sequence” is on the so-called PAM-strand in a “target nucleic acid,” which is a double-stranded molecule containing the PAM-strand and a complementary non-PAM strand. One of skill in the art recognizes that the gRNA spacer sequence hybridizes to the complementary sequence located in the non-PAM strand of the target nucleic acid of interest. Thus, the gRNA spacer sequence is the RNA equivalent of the target sequence.


For example, if the TRAC target sequence is 5′-AGAGCAACAGTGCTGTGGCC-3′ (SEQ ID NO: 10), then the gRNA spacer sequence is 5′-AGAGCAACAGUGCUGUGGCC-3′ (SEQ ID NO: 4). In yet another example, if the 132M target sequence is 5′-GCTACTCTCTCTTTCTGGCC-3′ (SEQ ID NO: 12), then the gRNA spacer sequence is 5′-GCUACUCUCUCUUUCUGGCC-3′ (SEQ ID NO: 4). The spacer of a gRNA interacts with a target nucleic acid of interest in a sequence-specific manner via hybridization (i.e., base pairing). The nucleotide sequence of the spacer thus varies depending on the target sequence of the target nucleic acid of interest.


In a CRISPR/Cas system herein, the spacer sequence is designed to hybridize to a region of the target nucleic acid that is located 5′ of a PAM recognizable by a Cas9 enzyme used in the system. The spacer may perfectly match the target sequence or may have mismatches. Each Cas9 enzyme has a particular PAM sequence that it recognizes in a target DNA. For example, S. pyogenes recognizes in a target nucleic acid a PAM that comprises the sequence 5′-NRG-3′, where R comprises either A or G, where N is any nucleotide and N is immediately 3′ of the target nucleic acid sequence targeted by the spacer sequence.


In some embodiments, the target nucleic acid sequence has 20 nucleotides in length. In some embodiments, the target nucleic acid has less than 20 nucleotides in length. In some embodiments, the target nucleic acid has more than 20 nucleotides in length. In some embodiments, the target nucleic acid has at least: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides in length. In some embodiments, the target nucleic acid has at most: 5, 10, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30 or more nucleotides in length. In some embodiments, the target nucleic acid sequence has 20 bases immediately 5′ of the first nucleotide of the PAM. For example, in a sequence comprising 5′-NNNNNNNNNNNNNNNNNNNNNRG-3′, the target nucleic acid can be the sequence that corresponds to the Ns, wherein N can be any nucleotide, and the underlined NRG sequence is the S. pyogenes PAM.


A spacer sequence in a gRNA is a sequence (e.g., a 20 nucleotide sequence) that defines the target sequence (e.g., a DNA target sequences, such as a genomic target sequence) of a target gene of interest. An exemplary spacer sequence of a gRNA targeting a TRAC gene is provided in SEQ ID NO: 4. An exemplary spacer sequence of a gRNA targeting a β2M gene is provided in SEQ ID NO: 8.


The guide RNA disclosed herein may target any sequence of interest via the spacer sequence in the crRNA. In some embodiments, the degree of complementarity between the spacer sequence of the guide RNA and the target sequence in the target gene can be about 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99%, or 100%. In some embodiments, the spacer sequence of the guide RNA and the target sequence in the target gene is 100% complementary. In other embodiments, the spacer sequence of the guide RNA and the target sequence in the target gene may contain up to 10 mismatches, e.g., up to 9, up to 8, up to 7, up to 6, up to 5, up to 4, up to 3, up to 2, or up to 1 mismatch.


Non-limiting examples of gRNAs that may be used as provided herein are provided in WO 2019/097305A2, and WO/2019/215500, the relevant disclosures of each of the prior applications are herein incorporated by reference for the purposes and subject matter referenced herein. For any of the gRNA sequences provided herein, those that do not explicitly indicate modifications are meant to encompass both unmodified sequences and sequences having any suitable modifications.


The length of the spacer sequence in any of the gRNAs disclosed herein may depend on the CRISPR/Cas9 system and components used for editing any of the target genes also disclosed herein. For example, different Cas9 proteins from different bacterial species have varying optimal spacer sequence lengths. Accordingly, the spacer sequence may have 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50, or more than 50 nucleotides in length. In some embodiments, the spacer sequence may have 18-24 nucleotides in length. In some embodiments, the targeting sequence may have 19-21 nucleotides in length. In some embodiments, the spacer sequence may comprise 20 nucleotides in length.


In some embodiments, the gRNA can be a sgRNA, which may comprise a 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence. In some embodiments, the sgRNA may comprise a less than 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence. In some embodiments, the sgRNA may comprise a more than 20 nucleotide spacer sequence at the 5′ end of the sgRNA sequence. In some embodiments, the sgRNA comprises a variable length spacer sequence with 17-30 nucleotides at the 5′ end of the sgRNA sequence.


In some embodiments, the sgRNA comprises no uracil at the 3′ end of the sgRNA sequence. In other embodiments, the sgRNA may comprise one or more uracil at the 3′ end of the sgRNA sequence. For example, the sgRNA can comprise 1-8 uracil residues, at the 3′ end of the sgRNA sequence, e.g., 1, 2, 3, 4, 5, 6, 7, or 8 uracil residues at the 3′ end of the sgRNA sequence.


Any of the gRNAs disclosed herein, including any of the sgRNAs, may be unmodified. Alternatively, it may contain one or more modified nucleotides and/or modified backbones. For example, a modified gRNA such as an sgRNA can comprise one or more 2′-O-methyl phosphorothioate nucleotides, which may be located at either the 5′ end, the 3′ end, or both.


In certain embodiments, more than one guide RNAs can be used with a CRISPR/Cas nuclease system. Each guide RNA may contain a different targeting sequence, such that the CRISPR/Cas system cleaves more than one target nucleic acid. In some embodiments, one or more guide RNAs may have the same or differing properties such as activity or stability within the Cas9 RNP complex. Where more than one guide RNA is used, each guide RNA can be encoded on the same or on different vectors. The promoters used to drive expression of the more than one guide RNA is the same or different.


It should be understood that more than one suitable Cas9 and more than one suitable gRNA can be used in methods described herein, for example, those known in the art or disclosed herein. In some embodiments, methods comprise a Cas9 enzyme and/or a gRNA known in the art. Examples can be found in, e.g., WO 2019/097305A2, and WO/2019/215500, the relevant disclosures of each of the prior applications are herein incorporated by reference for the purposes and subject matter referenced herein.


(iii) AAV Vectors for Delivery of CAR Constructs to T Cells


A nucleic acid encoding any of the anti-BCMA CAR construct can be delivered to a cell using an adeno-associated virus (AAV). AAVs are small viruses which integrate site-specifically into the host genome and can therefore deliver a transgene, such as CAR. Inverted terminal repeats (ITRs) are present flanking the AAV genome and/or the transgene of interest and serve as origins of replication. Also present in the AAV genome are rep and cap proteins which, when transcribed, form capsids which encapsulate the AAV genome for delivery into target cells. Surface receptors on these capsids which confer AAV serotype, which determines which target organs the capsids will primarily bind and thus what cells the AAV will most efficiently infect. There are twelve currently known human AAV serotypes. In some embodiments, the AAV for use in delivering the CAR-coding nucleic acid is AAV serotype 6 (AAV6).


Adeno-associated viruses are among the most frequently used viruses for gene therapy for several reasons. First, AAVs do not provoke an immune response upon administration to mammals, including humans. Second, AAVs are effectively delivered to target cells, particularly when consideration is given to selecting the appropriate AAV serotype. Finally, AAVs have the ability to infect both dividing and non-dividing cells because the genome can persist in the host cell without integration. This trait makes them an ideal candidate for gene therapy.


A nucleic acid encoding an anti-BCMA CAR can be designed to insert into a genomic site of interest in the host T cells. In some embodiments, the target genomic site can be in a safe harbor locus.


In some embodiments, a nucleic acid encoding an anti-BCMA CAR (e.g., via a donor template, which can be carried by a viral vector such as an adeno-associated viral (AAV) vector) can be designed such that it can insert into a location within a TRAC gene to disrupt the TRAC gene in the genetically engineered T cells and express the CAR polypeptide. Disruption of TRAC leads to loss of function of the endogenous TCR. For example, a disruption in the TRAC gene can be created with an endonuclease such as those described herein and one or more gRNAs targeting one or more TRAC genomic regions. Any of the gRNAs specific to a TRAC gene and the target regions can be used for this purpose, e.g., those disclosed herein.


In some examples, a genomic deletion in the TRAC gene and replacement by an anti-BCMA CAR coding segment can be created by homology directed repair or HDR (e.g., using a donor template, which may be part of a viral vector such as an adeno-associated viral (AAV) vector). In some embodiments, a disruption in the TRAC gene can be created with an endonuclease as those disclosed herein and one or more gRNAs targeting one or more TRAC genomic regions and inserting a CAR coding segment into the TRAC gene.


A donor template as disclosed herein can contain a coding sequence for an anti-BCMA CAR. In some examples, the anti-BCMA CAR-coding sequence may be flanked by two regions of homology to allow for efficient HDR at a genomic location of interest, for example, at a TRAC gene using CRISPR-Cas9 gene editing technology. In this case, both strands of the DNA at the target locus can be cut by a CRISPR Cas9 enzyme guided by gRNAs specific to the target locus. HDR then occurs to repair the double-strand break (DSB) and insert the donor DNA coding for the CAR. For this to occur correctly, the donor sequence is designed with flanking residues which are complementary to the sequence surrounding the DSB site in the target gene (hereinafter “homology arms”), such as the TRAC gene. These homology arms serve as the template for DSB repair and allow HDR to be an essentially error-free mechanism. The rate of homology directed repair (HDR) is a function of the distance between the mutation and the cut site so choosing overlapping or nearby target sites is important. Templates can include extra sequences flanked by the homologous regions or can contain a sequence that differs from the genomic sequence, thus allowing sequence editing. Examples of the donor template, including flanking homology sequences, are provided in Table 4 below.


Alternatively, a donor template may have no regions of homology to the targeted location in the DNA and may be integrated by NHEJ-dependent end joining following cleavage at the target site.


A donor template can be DNA or RNA, single-stranded and/or double-stranded, and can be introduced into a cell in linear or circular form. If introduced in linear form, the ends of the donor sequence can be protected (e.g., from exonucleolytic degradation) by methods known to those of skill in the art. For example, one or more dideoxynucleotide residues are added to the 3′ terminus of a linear molecule and/or self-complementary oligonucleotides are ligated to one or both ends. See, for example, Chang et al., (1987) Proc. Natl. Acad. Sci. USA 84:4959-4963; Nehls et al., (1996) Science 272:886-889. Additional methods for protecting exogenous polynucleotides from degradation include, but are not limited to, addition of terminal amino group(s) and the use of modified internucleotide linkages such as, for example, phosphorothioates, phosphoramidates, and 0-methyl ribose or deoxyribose residues.


A donor template can be introduced into a cell as part of a vector molecule having additional sequences such as, for example, replication origins, promoters and genes encoding antibiotic resistance. Moreover, a donor template can be introduced into a cell as naked nucleic acid, as nucleic acid complexed with an agent such as a liposome or poloxamer, or can be delivered by viruses (e.g., adenovirus, AAV, herpesvirus, retrovirus, lentivirus and integrase defective lentivirus (IDLV)).


A donor template, in some embodiments, can be inserted at a site nearby an endogenous promoter (e.g., downstream or upstream) so that its expression can be driven by the endogenous promoter. In other embodiments, the donor template may comprise an exogenous promoter and/or enhancer, for example, a constitutive promoter, an inducible promoter, or tissue-specific promoter to control the expression of the CAR gene. In some embodiments, the exogenous promoter is an EF1α promoter. Other promoters may be used.


Furthermore, exogenous sequences may also include transcriptional or translational regulatory sequences, for example, promoters, enhancers, insulators, internal ribosome entry sites, sequences encoding 2A peptides and/or polyadenylation signals.


The resultant T cells expressing an anti-BCMA CAR and having a disrupted TRAC and/or β2M genes may be collected and expanded in vitro. In some examples, the resultant T cells are subject to further purification to enrich the cells having the desired genetic modifications. For example, CAR+ T cells can be positively selected and TCR+ and/or B2M+ T cells can be excluded. In some embodiments, TCR+ T cells are removed. Non-limiting examples of methods of removal include cell sorting (e.g., fluorescence-activated cell sorting), immunomagnetic separation, chromatography, or microfluidic cell sorting. In some embodiments, TCR+ cells are removed using immunomagnetic separation. In some embodiments, TCR+ cells are labeled using a biotinylated antibody targeting the TCR and removed using anti-biotin magnetic beads.


(iv) Characterization of the Genetically Engineered Anti-BCMA CAR-T Cells


The genetically engineered anti-BCMA CAR-T cells, prepared by the methods disclosed herein or common approaches, can be characterized by routine approaches for features such as levels of surface protein of interest (e.g., TCR, β2M, anti-BCMA CAR, or a combination thereof), cell viability, cell bioactivity, impurity, etc.


In some embodiments, the surface protein of interest can be labeled, e.g., with an antibody and a tag such as a fluorescent tag. Flow cytometry can be used to detect the presence of the surface protein of interest, to quantify the level of surface marker expression, to quantify the fraction of T cells expressing the surface marker, or a combination thereof.


In some embodiments, insertion of the anti-BCMA CAR into the TRAC gene is assessed using digital droplet PCR (ddPCR). Digital PCR quantifies DNA concentration in a sample, comprising a) fractionating a PCR reaction; b) PCR amplifying the fractions; and c) analyzing the PCR amplifications of the fractions, wherein a fraction comprising a probe and a target molecule yields an amplification product and a fraction comprising no PCR probe yields no amplification product. The fraction containing amplification products is fitted to a Poisson distribution to determine the absolute copy number of target DNA molecules per given volume of the unfractionated sample (i.e., copies per microliter of sample) (see Hindson, B. et al., (2011) Anal Chem. 83:8604-10). Digital droplet PCR is a variation of digital PCR that can be used to provide absolute quantifications of DNA in samples, analyze copy number variations, and/or assess gene editing efficiencies. The sample of nucleic acids is fractionated into droplets using a water-oil emulsion; the PCR amplification is performed on the droplets collectively; and a fluidics system is used to separate the droplets and provide analysis of each individual droplet. In some embodiments, ddPCR is used to determine an absolute quantification of anti-BCMA CAR copies per sample composition. In some embodiments, ddPCR is used to assess HDR efficiency of inserting the anti-BCMA CAR sequences into the TRAC gene.


In some embodiments, the genetically engineered anti-BCMA CAR T cells can be assessed for cytokine-independent proliferation. The T cells are expected to only proliferate in the presence of a stimulatory cytokine, and proliferation in the absence of the stimulatory cytokine is indicative of a tumorigenic potential. The T cells may be cultured in the presence of a stimulatory cytokine for at least 1 day, e.g., at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 days, and proliferation of the T cells can be determined by conventional approaches. In some examples, the stimulatory cytokine comprises IL-2, IL-7, or both. T cell proliferation may be assessed at the end of the culture period. Alternatively, T cell proliferation may be assessed during the culture period, for example, on the 1st, 2nd, 3rd, 4th, 5th, or 6th day of the culture period. In some examples, T cell proliferation can be assessed about every 1 day, about every 2 days, about every 3 days, about every 4 days, about every 5 days, about every 6 days, about every 7 days, or about every 8 days.


In some embodiments, viable T cells can be counted using a conventional method, for example, flow cytometry, microscopy, optical density, metabolic activity, or a combination thereof. In some embodiments, the genetically engineered anti-BCMA CAR-T cells disclosed herein do not proliferate in the absence of any of the stimulatory cytokines or a combination thereof (and is defined as lacking tumorigenic potential). No proliferation can be defined as the number of viable T cells at the end of the culture period being less than 150% of the number of viable T cells at the beginning of the culture period, e.g., less than 140%, less than 130%, less than 120%, less than 110%, less than 100%, less than 90%, less than 80%, less than 70%, less than 60%, less than 50%, less than 40%, less than 30%, less than 20%, or less than 10%.


In some embodiments, a population of the genetically modified anti-BCMA CAR-T cells disclosed herein may show no growth in the absence of one or more stimulatory cytokines when assessed at 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days or 20 days following culture. In some examples, the T cells do not proliferate in the absence of cytokine, growth factor, antigen, or a combination thereof.


III. NK Cell Inhibitors

NK cells play an important role in both innate and adaptive immunity—including mediating anti-tumor and anti-viral responses. Because NK cells do not require prior sensitization or priming to mediate its cytotoxic function, they are the first line of defense against virus-infected and malignant cells that have missing or nonfunctioning MHC class I (e.g., disrupted MHC class I, or disrupted MCH Class I subunits). NK cells recognize “non-self” cells without the need for antibodies and antigen-priming. MHC class I-specific inhibitory receptors on NK cells negatively regulate NK cell function. Engagement of NK cell inhibitory receptors with their MHC class I ligand checks NK cell-mediated lysis. When MHC class I-disrupted cells fail to bind inhibitory NK receptors (e.g., KIRs), the cells become susceptible to NK cell-mediated lysis. This phenomenon is also referred to as the “missing self recognition.” See e.g., Malmberg K J et al., Immunogenetics (2017), 69:547-556; Cruz-Munoz M E et al., J. Leukoc. Biol. (2019), 105:955-971.


Therefore, engineered human CAR T cells comprising disrupted MHC class I as described herein are susceptible to NK cell-mediated lysis, thus reducing the persistence and subsequent efficacy of the engineered human CAR T cells. Accordingly, in some embodiments the present disclosure provides NK cell inhibitors for use in combination with CAR T cell therapy comprising a population of engineered human CAR T cells as described herein.


The NK cell inhibitor to be used in the methods described herein can be a molecule that blocks, suppresses, or reduces the activity or number of NK cells, either directly or indirectly. The term “inhibitor” implies no specific mechanism of biological action whatsoever, and is deemed to expressly include and encompass all possible pharmacological, physiological, and biochemical interactions with NK cells whether direct or indirect. For the purpose of the present disclosure, it will be explicitly understood that the term “inhibitor” encompasses all the previously identified terms, titles, and functional states and characteristics whereby the NK cell itself, a biological activity of the NK cell (including but not limited to its ability to mediate cell killing), or the consequences of the biological activity, are substantially nullified, decreased, or neutralized in any meaningful degree, e.g., by at least 20%, 50%, 70%, 85%, 90%, 100%, 150%, 200%, 300%,or 500%, or by 10-fold, 20-fold, 50-fold, 100-fold, 1000-fold, or 104-fold.


NK cell inhibitors may be a small molecule compound, a peptide or polypeptide, a nucleic acid, etc. Such NK cell inhibitors may be found in, for example, in International Patent Application No. PCT/IB2020/056085, the relevant discloses of which are incorporated by reference for the subject matter and purpose referenced herein. In some embodiments, the NK cell inhibitor disclosed herein is an antibody specific to CD38.


A. Antibodies that Bind CD38 (Anti-CD38 Antibodies)


In some embodiments, the present disclosure provides antibodies that specifically bind CD38 (anti-CD38 antibodies) for use in the methods described herein. CD38, also known as cyclic ADP ribose hydrolase, is a 46-kDa type II transmembrane glycoprotein that synthesizes and hydrolyzes cyclic adenosine 5′-diphosphate-ribose, an intracellular calcium ion mobilizing messenger. A multifunctional protein, CD38 is also involved in receptor-mediated cell adhesion and signaling. An amino acid sequence of an exemplary human CD38 protein is provided in SEQ ID NO: 62 (NCBI Reference Sequence: NP001766.2). See Table 6 below. Methods for generating antibodies that specifically bind human CD38 are known to those of ordinary skill in the art.


An antibody (interchangeably used in plural form) as used herein is an immunoglobulin molecule capable of specific binding to a target, such as a carbohydrate, polynucleotide, lipid, polypeptide, etc., through at least one antigen recognition site, located in the variable region of the immunoglobulin molecule. As used herein, the term “antibody” encompasses not only intact (i.e., full-length) monoclonal antibodies, but also antigen-binding fragments (such as Fab, Fab′, F(ab′)2, Fv, single chain variable fragment (scFv)), mutants thereof, fusion proteins comprising an antibody portion, humanized antibodies, chimeric antibodies, diabodies, linear antibodies, single chain antibodies, single domain antibodies (e.g., camel or llama VHH antibodies), multi-specific antibodies (e.g., bispecific antibodies) and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity, including glycosylation variants of antibodies, amino acid sequence variants of antibodies, and covalently modified antibodies.


A typical antibody molecule comprises a heavy chain variable region (VH) and a light chain variable region (VL), which are usually involved in antigen binding. These regions/residues that are responsible for antigen-binding can be identified from amino acid sequences of the VH/VL sequences of a reference antibody (e.g., an anti-CD38 antibody as described herein) by methods known in the art. The VH and VL regions can be further subdivided into regions of hypervariability, also known as “complementarity determining regions” (“CDR”), interspersed with regions that are more conserved, which are known as “framework regions” (“FR”). Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The extent of the framework region and CDRs can be precisely identified using methodology known in the art, for example, by the Kabat definition, the Chothia definition, the AbM definition, and/or the contact definition, all of which are well known in the art. As used herein, a CDR may refer to the CDR defined by any method known in the art. Two antibodies having the same CDR means that the two antibodies have the same amino acid sequence of that CDR as determined by the same method. See, e.g., Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242, Chothia et al., (1989) Nature 342:877; Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917, Al-lazikani et al (1997) J. Molec. Biol. 273:927-948; and Almagro, J. Mol. Recognit. 17:132-143 (2004). See also hgmp.mrc.ac.uk and bioinf.org.uk/abs.


An antibody includes an antibody of any class, such as IgD, IgE, IgG, IgA, or IgM (or sub-class thereof), and the antibody need not be of any particular class. Depending on the antibody amino acid sequence of the constant domain of its heavy chains, immunoglobulins can be assigned to different classes. There are five major classes of immunoglobulins: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2. The heavy-chain constant domains that correspond to the different classes of immunoglobulins are called alpha, delta, epsilon, gamma, and mu, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.


The antibodies to be used as provided herein can be murine, rat, human, or any other origin (including chimeric or humanized antibodies). In some examples, the antibody comprises a modified constant region, such as a constant region that is immunologically inert, e.g., does not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).


In some embodiments, an antibody of the present disclosure is a humanized antibody. Humanized antibodies refer to forms of non-human (e.g., murine) antibodies that are specific chimeric immunoglobulins, immunoglobulin chains, or antigen-binding fragments thereof that contain minimal sequence derived from non-human immunoglobulin. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat, or rabbit having the desired specificity, affinity, and capacity. In some instances, Fv framework region (FR) residues of the human immunoglobulin are replaced by corresponding non-human residues. Furthermore, the humanized antibody may comprise residues that are found neither in the recipient antibody nor in the imported CDR or framework sequences, but are included to further refine and optimize antibody performance In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence. A humanized antibody optimally also will comprise at least a portion of an immunoglobulin constant region or domain (Fc), typically that of a human immunoglobulin. Other forms of humanized antibodies have one or more CDRs (one, two, three, four, five, and/or six) which are altered with respect to the original antibody, which are also termed one or more CDRs “derived from” one or more CDRs from the original antibody. Humanized antibodies may also involve affinity maturation.


In some embodiments, an antibody of the present disclosure is a chimeric antibody, which can include a heavy constant region and a light constant region from a human antibody. Chimeric antibodies refer to antibodies having a variable region or part of variable region from a first species and a constant region from a second species. Typically, in these chimeric antibodies, the variable region of both light and heavy chains mimics the variable regions of antibodies derived from one species of mammals (e.g., a non-human mammal such as mouse, rabbit, and rat), while the constant portions are homologous to the sequences in antibodies derived from another mammal such as human. In some embodiments, amino acid modifications can be made in the variable region and/or the constant region.


In some embodiments, an antibody of the present disclosure specifically binds a target antigen (e.g., human CD38). An antibody that “specifically binds” (used interchangeably herein) to a target or an epitope is a term well understood in the art, and methods to determine such specific binding are also well known in the art. A molecule is said to exhibit “specific binding” if it reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with a particular target antigen than it does with alternative targets. An antibody “specifically binds” to a target antigen if it binds with greater affinity, avidity, more readily, and/or with greater duration than it binds to other substances. For example, an antibody that specifically (or preferentially) binds to a CD38 epitope, or is an antibody that binds this epitope with greater affinity, avidity, more readily, and/or with greater duration than it binds to other epitopes of the same antigen or a different antigen. It is also understood by reading this definition that, for example, an antibody that specifically binds to a first target antigen may or may not specifically or preferentially bind to a second target antigen. As such, “specific binding” or “preferential binding” does not necessarily require (although it can include) exclusive binding. Generally, but not necessarily, reference to binding means preferential binding.


Also within the scope of the present disclosure are functional variants of any of the exemplary antibodies as disclosed herein. A functional variant may contain one or more amino acid residue variations in the VH and/or VL, or in one or more of the HC CDRs and/or one or more of the VL CDRs as relative to a reference antibody, while retaining substantially similar binding and biological activities (e.g., substantially similar binding affinity, binding specificity, inhibitory activity, anti-tumor activity, or a combination thereof) as the reference antibody.


In some instances, the amino acid residue variations can be conservative amino acid residue substitutions. As used herein, a “conservative amino acid substitution” refers to an amino acid substitution that does not alter the relative charge or size characteristics of the protein in which the amino acid substitution is made. Variants can be prepared according to methods for altering polypeptide sequence known to one of ordinary skill in the art such as are found in references which compile such methods, e.g., Molecular Cloning: A Laboratory Manual, J. Sambrook, et al., eds., Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1989, or Current Protocols in Molecular Biology, F. M. Ausubel, et al., eds., John Wiley & Sons, Inc., New York. Conservative substitutions of amino acids include substitutions made amongst amino acids within the following groups: (a) A→G, S; (b) R→K, H; (c) N→Q, H; (d) D→E, N; (e) C→S, A; (f) Q→N; (g) E→D, Q; (h) G→A; (i) H→Q; (j) I→L, V; (k) L→I, V; (l) K→R, H; (m) M→L, I, Y; (n) F→Y, M, L; (o) P→A; (p) S→T; (q) T→S; (r) W→Y, F; (s) Y→W, F; and (t) V→I, L.


Anti-CD38 antibodies have been tested in various pre-clinical and clinical studies, e.g., for NK/T cell lymphoma, or T-cell acute lymphoblastic leukemia. Exemplary anti-CD38 antibodies tested for anti-tumor properties include SAR650984 (also referred to as isatuximab, chimeric mAb), which is in phase I clinical trials in patients with CD38+ B-cell malignancies (Deckert J. et al., Clin. Cancer. Res. (2014): 20(17):4574-83), MOR202 (also referred to as MOR03087, fully human mAb), and TAK-079 (fully human mAb).


In some embodiments, an anti-CD38 antibody for use in the present disclosure includes SAR650984 (Isatuximab), MOR202, Ab79, Ab10, HM-025, HM-028, HM-034; as well as antibodies disclosed in U.S. Pat. Nos. 9,944,711, 7,829,673, WO2006/099875, WO 2008/047242, WO2012/092612, and EP 1 720 907 B1, herein incorporated by reference. In some embodiments, the anti-CD38 antibody disclosed herein may be a functional variant of any of the reference antibodies disclosed herein. Such a functional variant may comprise the same heavy chain and light chain complementary determining regions as the reference antibody. In some examples, the functional variant may comprise the same heavy chain variable region and the same light chain variable region as the reference antibody.


In some embodiments, the anti-CD38 antibody for use in the present disclosure is daratumumab. Daratumumab (also referred to as Darzalex®, HuMax-CD38, or IgG1-005) is a fully human IgGκ monoclonal antibody that targets CD38 and has been approved for treating multiple myeloma. It is used as a monotherapy or as a combination therapy for treating newly diagnosed or previously treated multiple myeloma patients. Daratumumab is described in U.S. Pat. No. 7,829,673 and WO2006/099875.


Daratumumab binds an epitope on CD38 that comprises two β-strands located at amino acids 233-246 and 267-280 of CD38. Experiments with CD38 mutant polypeptides show that the 5274 amino acid residue is important for daratumumab binding. (van de Donk N W C J et al., Immunol. Rev. (2016) 270:95-112). Daratumumab's binding orientation to CD38 allows for Fc-receptor mediated downstream immune processes.


Mechanisms of action attributed to Daratumumab as a lymphoma and multiple myeloma therapy includes Fc-dependent effector mechanisms such as complement-dependent cytotoxicity (CDC), natural killer (NK)-cell mediated antibody-dependent cellular cytotoxicity (ADCC) (De Weers M, et al., J. Immunol. (2011) 186:1840-8), antibody-mediated cellular phagocytosis (ADCP) (Overdijk M B et al., MAbs (2015), 7(2):311-21), and apoptosis after cross-linking (van de Donk N W C J and Usmani S Z, Front. Immunol. (2018), 9:2134).


The full heavy chain amino acid sequence of daratumumab is set forth in SEQ ID NO: 63 and the full light chain amino acid sequence of daratumumab is set forth in SEQ ID NO: 65. The amino acid sequence of the heavy chain variable region of daratumumab is set forth in SEQ ID NO: 64 and the amino acid sequence of the light chain variable region of daratumumab is set forth in SEQ ID NO: 66. Daratumumab includes the heavy chain complementary determining regions (HCDRs) 1, 2, and 3 (SEQ ID NOs: 67, 68, and 69, respectively), and the light chain CDRs (LCDRs) 1, 2, and 3 (SEQ ID NOs. 70, 71, and 72, respectively). See Table 6 below. In some embodiments, these sequences can be used to produce a monoclonal antibody that binds CD38. For example, methods for making daratumumab are described in U.S. Pat. No. 7,829,673 (incorporated herein by reference for the purpose and subject matter referenced herein).


In some embodiments, an anti-CD38 antibody for use in the present disclosure is daratumumab, an antibody having the same functional features as daratumumab, or an antibody which binds to the same epitope as daratumumab or competes against daratumumab from binding to CD38.


In some embodiments, the anti-CD38 antibody comprises: (a) an immunoglobulin heavy chain variable region and (b) an immunoglobulin light variable region, wherein the heavy chain variable region and the light chain variable region defines a binding site (paratope) for CD38. In some embodiments, the heavy chain variable region comprises an HCDR1 comprising the amino acid sequence set forth in SEQ ID NO: 67, an HCDR2 comprising the amino acid sequence set forth in SEQ ID NO: 68; and an HCDR3 comprising the amino acid sequence in SEQ ID NO: 69. The HCDR1, HCDR2, and HCDR3 sequences are separated by the immunoglobulin framework (FR) sequences.


In some embodiments, the anti-CD38 antibody comprises: (a) an immunoglobulin light chain variable region and (b) an immunoglobulin heavy chain variable region, wherein the light chain variable region and the heavy chain variable region defines a binding site (paratope) for CD38. In some embodiments, the light chain variable region comprises an LCDR1 comprising the amino acid sequence set forth in SEQ ID NO: 70, an LCDR2 comprising the amino acid sequence set forth in SEQ ID NO: 71; and an LCDR3 comprising the amino acid sequence in SEQ ID NO: 72. The LCDR1, LCDR2, and LCDR3 sequences are separated by the immunoglobulin framework (FR) sequences.


In some embodiments, the anti-CD38 antibody comprises an immunoglobulin heavy chain variable region (VH) comprising the amino acid sequence set forth in SEQ ID NO: 64, and an immunoglobulin light chain variable region (VL). In some embodiments, the anti-CD38 antibody comprises an immunoglobulin light chain variable region (VL) comprising the amino acid sequence set forth in SEQ ID NO: 66, and an immunoglobulin heavy chain variable region (VH). In some embodiments, the anti-CD38 antibody comprises a VH comprising an amino acid sequence that is at least 70%, 75%, 70%, 85%, 90%, 95%, 96%, 97%, 98%, and 99% identical to the amino acid sequence set forth in SEQ ID NO: 64, and comprises an VL comprising an amino acid sequence that is at least 70%, 75%, 70%, 85%, 90%, 95%, 96%, 97%, 98%, and 99% identical to the amino acid sequence set forth in SEQ ID NO: 66.


The “percent identity” of two amino acid sequences is determined using the algorithm of Karlin and Altschul Proc. Natl. Acad. Sci. USA 87:2264-68, 1990, modified as in Karlin and Altschul Proc. Natl. Acad. Sci. USA 90:5873-77, 1993. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. J. Mol. Biol. 215:403-10, 1990. BLAST protein searches can be performed with the XBLAST program, score=50, wordlength=3 to obtain amino acid sequences homologous to the protein molecules of the invention. Where gaps exist between two sequences, Gapped BLAST can be utilized as described in Altschul et al., Nucleic Acids Res. 25(17):3389-3402, 1997. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used.


CD38 is expressed on NK cells and infusion of daratumumab results in a reduction of NK cells in peripheral blood and bone marrow. The reduction of NK cells is due to NK-cell killing via ADCC, in which NK cells mediate cytotoxic killing of neighboring NK cells. Administration of daratumumab has also been shown to decrease cell numbers of myeloid derived suppressor cells, regulatory T cells, and regulatory B cells. The elimination of regulatory immune cells results in increased T cell responses and increased T cell numbers (J Krejcik et al., Blood (2016), 128(3):384-394.


Accordingly, in some embodiments, the anti-CD38 antibody (e.g., daratumumab) reduces absolute NK cell numbers. In some embodiments, the anti-CD38 antibody reduces NK cell percentage in PBMCs. In some embodiments, the anti-CD38 antibody inhibits NK cell activity through Fc-mediated mechanisms. In other embodiments, the anti-CD38 antibody mediates the killing of NK cells through CDC. In other embodiments, the anti-CD38 antibody mediates the killing of NK cells through ADCC. In other embodiments, the anti-CD38 antibody enhances phagocytosis of NK cells. In other embodiments, the anti-CD38 antibody enhances apoptosis induction after FcγR-mediated cross-linking.


In some embodiments, the anti-CD38 antibody is daratumumab or an antibody having the same functional features as daratumumab, for example, a functional variant of daratumumab. In some examples, a functional variant comprises substantially the same VH and VL CDRs as daratumumab. For example, it may comprise only up to 8 (e.g., 8, 7, 6, 5, 4, 3, 2, or 1) amino acid residue variations in the total CDR regions of the antibody and binds the same epitope of CD38 with substantially similar affinity (e.g., having a KD value in the same order) as daratumumab. In some instances, the functional variants may have the same heavy chain CDR3 as daratumumab, and optionally the same light chain CDR3 as daratumumab. Alternatively or in addition, the functional variants may have the same heavy chain CDR2 as daratumumab. Such an anti-CD38 antibody may comprise a VH fragment having CDR amino acid residue variations in only the heavy chain CDR1 as compared with the VH of daratumumab. In some examples, the anti-CD38 antibody may further comprise a VL fragment having the same VL CDR3, and optionally same VL CDR1 or VL CDR2 as daratumumab. Alternatively or in addition, the amino acid residue variations can be conservative amino acid residue substitutions (see above disclosures).


In some embodiments, the anti-CD38 antibody may comprise heavy chain CDRs that are at least 80% (e.g., 85%, 90%, 95%, or 98%) sequence identity, individually or collectively, as compared with the VH CDRs of daratumumab. Alternatively or in addition, the anti-CD38 antibody may comprise light chain CDRs that are at least 80% (e.g., 85%, 90%, 95%, or 98%) sequence identity, individually or collectively, as compared with the VL CDRs as daratumumab. As used herein, “individually” means that one CDR of an antibody shares the indicated sequence identity relative to the corresponding CDR of daratumumab. “Collectively” means that three VH or VL CDRs of an antibody in combination share the indicated sequence identity relative the corresponding three VH or VL CDRs of daratumumab.


In some embodiments, the anti-CD38 antibody binds to the same epitope bound by daratumumab on human CD38. In some embodiments, the anti-CD38 antibody competes with daratumumab for binding to human CD38.


Competition assays for determining whether an antibody binds to the same epitope as daratumumab, or competes with daratumumab for binding to CD38, are known in the art. Exemplary competition assays include immunoassays (e.g., ELISA assay, RIA assays), surface plasmon resonance, (e.g., BIAcore analysis), bio-layer interferometry, and flow cytometry.


A competition assay typically involves an immobilized antigen (e.g., CD38), a test antibody (e.g., CD38-binding antibody) and a reference antibody (e.g., daratumumab). Either one of the reference or test antibody is labeled, and the other unlabeled. In some embodiments, competitive binding is determined by the amount of a reference antibody bound to the immobilized antigen in increasing concentrations of the test antibody. Antibodies that compete with a reference antibody include antibodies that bind the same or overlapping epitopes as the reference antibody. In some embodiments, the test antibodies bind to adjacent, non-overlapping epitopes such that the proximity of the antibodies causes a steric hindrance sufficient to affect the binding of the reference antibody to the antigen.


A competition assay can be conducted in both directions to ensure that the presence of the label or steric hindrance does not interfere or inhibit binding to the epitope. For example, in the first direction, the reference antibody is labeled and the test antibody is unlabeled. In the second direction, the test antibody is labeled, and the reference antibody is unlabeled. In another embodiment, in the first direction, the reference antibody is bound to the immobilized antigen, and increasing concentrations of the test antibody are added to measure competitive binding. In the second direction, the test antibody is bound to the immobilized antigen, and increasing concentrations of the reference antibody are added to measure competitive binding.


In some embodiments, two antibodies can be determined to bind to the same epitope if essentially all amino acid mutations in the antigen that reduce or eliminate the binding of one antibody reduce or eliminate binding of the other. Two antibodies can be determined to bind to overlapping epitopes if only a subset of the mutations that reduce or eliminate the binding of one antibody reduces or eliminates the binding of the other.


In some embodiments, the heavy chain of any of the anti-CD38 antibodies as described herein (e.g., daratumumab) may further comprise a heavy chain constant region (CH) or a portion thereof (e.g., CH1, CH2, CH3, or a combination thereof). The heavy chain constant region can of any suitable origin, e.g., human, mouse, rat, or rabbit. Alternatively or in addition, the light chain of the anti-CD38 antibody may further comprise a light chain constant region (CL), which can be any CL known in the art. In some examples, the CL is a kappa light chain. In other examples, the CL is a lambda light chain. Antibody heavy and light chain constant regions are well known in the art, e.g., those provided in the IMGT database (www.imgt.org) or at www.vbase2.org/vbstat.php., both of which are incorporated by reference herein.


Any of the anti-CD38 antibodies, including human antibodies or humanized antibodies, can be prepared by conventional approaches, for example, hybridoma technology, antibody library screening, or recombinant technology. See, for example, Harlow and Lane, (1998) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, WO 87/04462, Morrison et al., (1984) Proc. Nat. Acad. Sci. 81:6851, and Queen et al., Proc. Natl. Acad. Sci. USA, 86:10029-10033 (1989).


It should be understood that the described antibodies are only exemplary and that any anti-CD38 antibodies can be used in the compositions and methods disclosed herein. Methods for producing antibodies are known to those of skill in the art.


IV. Combined Therapy of Anti-BCMA CAR-T Cells

In some aspects, the present disclosure features combined therapy of (a) anti-BCMA CAR-T cells and (b) an NK cell inhibitor such as an anti-CD38 antibody (preferably daratumumab), lenalidomide or a derivative thereof, or a combination thereof for treating a BCMA+ tumor, for example, multiple myeloma (e.g., refractory and/or relapsed MM). In some embodiments, the combined therapy comprises anti-BCMA CAR-T cells such as CTX120 cells and an NK cell inhibitor such as an anti-CD38 antibody (e.g., daratumumab). In other embodiments, the combined therapy comprises comprises anti-BCMA CAR-T cells such as CTX120 cells and lenalidomide or a derivative thereof. In yet other emdobiments, the combined therapy comprises anti-BCMA CAR-T cells such as CTX120 cells, an NK cell inhibitor such as an anti-CD38 antibody (e.g., daratumumab), and lenalidomide or a derivative thereof.


Lenalidomide is a small molecule compounds that modulates the substrate activity of the CRL4CRBN E3 ubiquitin ligase. Lenalidomide has a structure of:




embedded image


Any of the genetically engineered anti-BCMA CAR-T cells disclosed herein may be used for therapeutic purposes, for example, in treating BCMA+ cancers. Accordingly, provided herein are methods of treating cancer (e.g., hematologic malignancies involving BCMA+ cancer cells) comprising administering an effective amount of a population of the genetically engineered anti-BCMA CAR-T cells disclosed herein (e.g., CTX120 cells) and an effective amount of lenalidomide, an effective amount of daratumumab, or a combination thereof, to a subject in need of the treatment. In some embodiments, the cancer is MM, including refractory and/or relapsed MM.


(i) Multiple Myeloma


MM is a malignancy of terminally differentiated plasma cells in the bone marrow that represents about 10% of all hematologic malignancies, and is the second most common hematologic malignancy after non-Hodgkin lymphoma (Kumar et al., 2017, Leukemia 31, 2443-2448; and Rajkumar and Kumar, 2016, Mayo Clin Proc 91, 101-119).


MM is a result of secretion of a monoclonal immunoglobulin protein (also known as monoclonal protein or M-protein) or monoclonal free light chains by abnormal plasma cells. MM exists on a spectrum of plasma cell dyscrasias and results from the stepwise progression from premalignant monoclonal gammopathy of undetermined significance (MGUS) to asymptomatic smoldering MM to symptomatic MM. Importantly, diagnosis of MM is determined and differentiated from smoldering MM and MGUS by characteristic bone marrow biopsy findings as well as symptoms attributable to end organ damage related to plasma cell proliferation (hypercalcemia, renal insufficiency, anemia, fractures) (Kumar et al., 2017, Leukemia 31, 2443-2448; and Kumar et al., 2017, Nat Rev Dis Primers 3, 17046). Whereas patients with MGUS and smoldering MM are typically observed or enroll in exploratory clinical trials, patients with symptomatic MM require treatment.


With the discovery and approval of agents such as proteasome inhibitors (PIs; e.g., bortezomib, carfilzomib), immunomodulatory drugs (IMiDs; e.g., lenalidomide, pomalidomide), and more recently, monoclonal antibodies (mAbs; e.g., daratumumab, elotuzumab), the survival of patients with MM has improved significantly over the past decade. Nevertheless, although combinations of these agents along with autologous stem cell transplant (SCT) have improved responses, progression-free survival (PFS), and overall survival (OS), for most patients MM remains an incurable disease that ultimately leads to death. The only potentially curative approach remains allogeneic SCT, which is rarely used due to its high transplant-related mortality.


Patients with MM that is double-refractory to both PI and IMiD agents have a poor prognosis. A multicenter, retrospective study of patients with relapsed MM showed that these double-refractory patients had a median PFS of 5 months and OS of 9 months (Kumar et al., 2012, Leukemia 26, 149-157). A more recent study of this population reflecting the use of second-generation PI and IMiD agents also demonstrated poor prognosis, with median PFS of 5 months and OS of 13 months (Kumar et al., 2017, Nat Rev Dis Primers 3, 17046).


Early relapse (≤12 months) after autologous SCT is seen in nearly 20% of transplanted MM patients and identifies a high-risk population with poor outcomes. In one study comparing patients who experienced early relapse to non-early relapse (>12 months or disease-free) patients, early relapse was associated with a significantly shorter median OS from diagnosis (26.6 vs 90.7 months) and from autologous SCT (20.1 vs 82.5 months). Among patients who relapsed after SCT (n=345), median OS from relapse was 10.8 months for the early relapse group versus 41.8 months for the rest (Kumar et al., 2008). A more recent analysis showed early relapse after SCT to be a major predictor of poor survival (median OS of 20 months vs 93 months for early versus non-early relapse, respectively) despite the advent of PIs, IMiDs, and other novel agents (Jimenez-Zepeda et al., 2015). Together these findings support aggressive treatment strategies for MM patients with early relapse after SCT, including clinical trials of agents with alternative mechanisms of action.


Thus, there is a need to develop effective treatment approaches targeting refractory and/or relapsed MM.


(ii) Patient Population


The subject to be treated by the combined therapy disclosed herein (e.g., allogeneic anti-BCMA CAR-T cells such as CTX120 cells, NK cell inhibitor such as daratumumab, and/or lenalidomide or a derivative thereof such as those disclosed herein) can be a mammal, for example, a human patient, who may be 18 years or older. In some examples, the subject is a human patient having a cancer that involves BCMA+ cancer cells. For example, the subject may be a human patient having MM, including symptomatic MM and asymptomatic MM. In specific examples, the human patient has refractory MM. In other specific examples, the human patient has relapsed MM. In other examples, the subject may have monoclonal gammopathy of unknown significance (MUGS) or asymptomatic smoldering MM. Alternatively, the subject may be a human patient who is diagnosed with a high risk of developing MM, e.g., a subtype disclosed herein such as symptomatic MM.


A subject having MM can be diagnosed via routine medical practice. Methods of diagnosing MM are known in the art. Non-limiting examples include analysis of bone marrow biopsy, analysis of end organ damage related to plasma cell proliferation (e.g., hypercalcemia, renal insufficiency, anemia, destructive bone lesions), or both. See e.g., Kumar, et al. (2017) Leukemia 31:2443-48; Kumar, et al., (2016) Lancet Oncol 17: e328-46; and NCCN Guidelines v.2.2019 (2018) National Comprehensive Cancer Network Clinical Practice Guidelines for Multiple Myeloma. In some embodiments, the subject has MGUS.


In some embodiments, the subject (e.g., a human patient) has MM cells expressing an elevated level of BCMA. Methods of quantifying expression of BCMA mRNA and protein in cells or tissues are known in the art. For example, expression of BCMA mRNA can be measured using reverse transcription polymerase chain reaction (RT-PCR), quantitative PCR (qPCR), multiplex-PCR, digital PCR, and/or whole transcriptome shotgun sequencing; and expression of BCMA protein can be measured using mass spectrometry, enzyme-linked immunosorbent assay (ELISA), protein immunoprecipitation, immunoelectrophoresis, western blot, and/or immunostaining (e g , immunofluorescence staining, immunohistochemical staining) with analysis by flow cytometry or microscopy.


In some embodiments, the subject (e.g., a human patient) has relapsed from or is refractory to a prior MM therapy. As used herein, “refractory” refers to MM that does not respond to or becomes resistant to a treatment. As used herein, “relapsed” or “relapses” refers to MM that returns or progresses following a period of improvement (e.g., a partial or complete response) with treatment. In some embodiments, relapse occurs during the treatment. In some embodiments, relapse occurs after the treatment. A lack of response may be measured, for example, as a lack of change in serum M-protein levels, urine M-protein levels, bone marrow plasma cell counts, bone lesion sizes, bone lesion numbers, or a combination thereof. A return or progression in MM may be measured, for example, as an increase in serum creatinine levels, serum M-protein levels, urine M-protein levels, bone marrow plasma cell counts, bone marrow plasmacytomas sizes, bone marrow plasmacytomas numbers, bone lesion sizes, bone lesion numbers, calcium levels unexplained by other conditions, red blood cell counts, organ damage, or a combination thereof.


In some embodiments, the prior MM therapy comprises a steroid, chemotherapy, a proteasome inhibitor (PI), an immunomodulatory drug (IMiD), a monoclonal antibody, an autologous stem cell transplant (SCT), or a combination thereof (see e.g., NCCN Guidelines v.2.2019 (2018) National Comprehensive Cancer Network Clinical Practice Guidelines for Multiple Myeloma). Non-limiting examples of steroids include dexamethasone and prednisone. Non-limiting examples of chemotherapies include bendamustine, cisplatin, cyclophosphamide, doxorubicin hydrochloride, doxorubicin hydrochloride liposome, etoposide, and melphalan. Non-limiting examples of PIs include bortezomib, ixazomib, and carfilzomib. In some embodiments, the PI comprises bortezomib, carfilzomib, or both. Non-limiting examples of IMiDs include lenalidomide, pomalidomide, thalidomide. In some embodiments, the IMiD therapy comprises lenalidomide, pomalidomide, or both. Non-limiting examples of monoclonal antibodies include CD38-directed monoclonal antibodies (e.g., daratumumab, and isatuximab), and elotuzumab (binding to CD319). In some embodiments, the monoclonal antibody comprises a CD38-directed monoclonal antibody such as daratumumab.


In some embodiments, the prior MM therapy comprises more than one line of therapy. In some embodiments, the prior MM therapy comprises two or more lines of therapy, e.g., three lines of prior therapy, four lines of prior therapy, etc. In some embodiments, the two or more lines of therapy are administered separately. In some embodiments, the two or more lines of therapy are administered in combination. In some embodiments, the prior MM therapy comprises an IMiD, a PI, a CD38-directed monoclonal antibody, or a combination thereof. In some embodiments, the prior MM therapy comprises IMiD and PI. In some embodiments, the IMiD is administered before the PI. In some embodiments, the IMiD is administered after the PI.


In some examples, the prior MM therapy comprises two lines of therapy, e.g., an IMiD, and a PI. A MM patient who is refractory to two prior MM therapies may be referred to as “double-refractory.” In some embodiments, a double-refractory MM patient has disease progression on or within 60 days of treatment with the two lines of therapy. In some instances, the two lines of therapy may be part of the same regimen. In other instances, the two lines of therapy may be part of different treatment regimens. A double-refractory MM patient may have disease progression on or within 60 days of the last treatment regimen.


In some examples, the prior MM therapy comprises three lines of therapy, e.g., an IMiD, a PI, and a CD38-directed monoclonal antibody. A MM patient who is refractory to three prior MM therapies may be referred to as “triple-refractory.” In some embodiments, a triple-refractory MM patient has disease progression on or within 60 days of treatment with the three lines of therapy. In some instances, the three lines of therapy may be part of the same regimen. In other instances, the three lines of therapy may be part of different treatment regimens. A triple-refractory MM patient may have disease progression on or within 60 days of the last treatment regimen.


In some embodiments, relapsed or refractory MM is detected at least 10 days, at least 20 days, at least 30 days, at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months, at least 1 year, at least 2 years, at least 3 years, at least 4 years, or at least 5 years after the prior MM therapy. In some embodiments, relapsed or refractory MM is detected within 10-100 days after the prior MM therapy, e.g., within 10-90 days, 20-90 days, 20-80 days, 30-80 days, 30-70 days, 40-70 days, 40-60 days, or 50-60 days. In some embodiments, relapsed or refractory MM is detected within about 100 days after the prior MM therapy, e.g., within about 90 days, within about 80 days, within about 70 days, within about 60 days, within about 50 days, within about 40 days, within about 30 days, within about 20 days, or within about 10 days after the prior MM therapy.


In some embodiments, relapsed MM is detected in the subject during an autologous SCT. In some embodiments, relapsed MM is detected in the subject after an autologous SCT. In some embodiments, relapsed or refractory MM is detected at least 10 days, at least 20 days, at least 30 days, at least 2 months, at least 4 months, at least 6 months, at least 8 months, at least 10 months, at least 1 year, at least 2 years, at least 2 years, at least 3 years, at least 4 years, or at least 5 years after the autologous SCT. In some embodiments, relapsed or refractory MM is detected within about 18 months after the autologous SCT, e.g., within about 17 months, within about 16 months, within about 15 months, within about 14 months, within about 13 months, within about 12 months, within about 11 months, within about 10 months, within about 9 months, within about 8 months, within about 7 months, within about 6 months, within about 5 months, within about 4 months, within about 3 months, within about 2 months, or within about 1 month after the autologous SCT. In some embodiments, relapsed or refractory MM is detected between about 1-18 months after the autologous SCT, e.g., about 2-18 months, about 2-16 months, about 3-16 months, about 3-14 months, about 4-14 months, about 4-12 months, about 5-12 months, about 5-10 months, about 6-10 months, or about 6-8 months after the autologous SCT.


In some embodiments, the subject is a human MM patient having one or more of the following features: adequate organ function, free of a prior allogeneic stem cell transplantation (SCT), free of autologous SCT within 60 days prior to the enrollment into the allogenic T cell therapy disclosed herein, free of plasma cell leukemia, non-secretory MM, Waldenstrom's macroglobulinemia, POEM syndrome, and/or amyloidosis with end organ involvement and damage, free of prior gene therapy, anti-BCMA therapy, and non-palliative radiation therapy within 14 days prior to enrollment into the allogenic T cell therapy, free of central nervous system involvement by MM, free of history or presence of clinically relevant CNS pathology, cerebrovascular ischemia and/or hemorrhage, dementia, a cerebellar disease, an autoimmune disease with CNS involvement, free of unstable angina, arrhythmia, and/or myocardial infarction within 6 month prior to enrollment into the allogenic T cell therapy, free of uncontrolled infections (e.g., infections is caused by HIV, HBV, or HCV), free of previous or concurrent malignancy, provided that the malignancy is not basal cell or squamous cell skin carcinoma, adequately resected and in situ carcinoma of cervix, or a previous malignancy that was completely resected and has been in remission for ≥5 years, free of live vaccine administration within 28 days prior to enrollment into the allogenic T cell therapy, free of systemic anti-tumor therapy within 14 days prior to enrollment into the allogenic T cell therapy, and free of primary immunodeficiency disorders or autoimmune disorders that require immunosuppressive therapy. In some embodiments, the subject is a human patient having Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1. The human patient may be free of contraindication to lymphodepleting agents such as cyclophosphamide and/or fludarabine.


In some embodiments, the subject is a human MM patient (e.g., refractory and/or relapsed MM patient) who has received prior treatment comprising daratumumab. In some instances, the subject may be free of contraindication to daratumumab. Alternatively or in addition, the subject is a human MM patient (e.g., refractory and/or relapsed MM patient) who has received prior treatment comprising lenalidomide. In some instances, the subject may be free of contraindication to lenalidomide.


In some examples, the subject is a human patient who meets one or more of the inclusion and/or exclusion criteria disclosed in Example 16 below. In some examples, the subject may meet all of the inclusion and/or exclusion criteria disclosed in Example 16 below.


(iii) NK Cell Inhibitor Treatment


An NK cell inhibitor such as daratumumab may be formulated in a pharmaceutical composition and given to a suitable subject as disclosed herein at a suitable time point relative to the LD and/or allogeneic anti-BCMA CAR-T cell (e.g., CTX120) therapy. For example, the daratumumab may be given to a subject no more than 14 days prior to the first dose of the anti-BCMA CAR-T cells. A pharmaceutical composition comprising daratumumab and one or more pharmaceutically acceptable carriers may be administered to the subject via a suitable route, for example, orally, parenterally, by inhalation spray, rectally, nasally, buccally, vaginally or via an implanted reservoir.


In some embodiments, the pharmaceutical composition comprising daratumumab is to be administered by injection, for example, intravenous infusion or subcutaneous injection. A sterile injectable composition, e.g., a sterile injectable aqueous or oleaginous suspension, can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as Tween® 80) and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents. Other commonly used surfactants such as Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation.


The pharmaceutical compositions as described herein can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions. Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Williams and Wilkins, Ed. K. E. Hoover. Such carriers, excipients or stabilizers may enhance one or more properties of the active ingredients in the compositions described herein, e.g., bioactivity, stability, bioavailability, and other pharmacokinetics and/or bioactivities.


Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; benzoates, sorbate and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, serine, alanine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™ (polysorbate), PLURONICS™ (nonionic surfactants), or polyethylene glycol (PEG).


In some embodiments, an effective amount of daratumumab (e.g., about 10-20 mg/kg such as about 16 mg/kg) may be given to the subject via a suitable route (e.g., intravenous infusion). The effective amount of daratumumab may split into two parts (e.g., equally) and be administered to the subject on two consecutive days. In some examples, administration of daratumumab may be performed prior to the LD therapy. In specific examples, administration of daratumumab may be performed within 3 days prior to the LD therapy. Alternatively or in addition, administration of daratumumab may be performed no more than 14 days prior to the treatment with the anti-BCMA CAR-T cells such as CTX120 cells.


In some embodiments, an effective amount of daratumumab may be administered subcutaneously. In some examples, the subcutaneous injection of daratumumab may be accompanied with hyaluronidase. For example, about 1800 mg of daratumumab may be administered to a subject together with about 30,000 unit hyaluronidase to facilitate delivery of daratumumab.


In some instances, daratumumab treatment may be repeated on a monthly basis. For example, the subject may be administered daratumumab for additional five doses, once per month, when a patient shows stable disease or better after infusion of the anti-BCMA CAR-T cell (e.g., CTX120 cell) therapy. In some examples, the additional doses of daratumumab may start on at least 21 days post CAR-T cell infusion, e.g., at least 28 days post CAR-T cell infusion. In one example, the additional doses of daratumymab may start on Day 28 post CAR-T infusion. The doses of daratumumab post infusion of the anti-BCMA CAR-T cells may be the same as the dose of daratumumab given to the patient prior to the LD and anti-BCMA CAR-T cell therapy (the first dose), for example, 16 mg/kg, via intravenous infusion. Alternatively, the additional doses of daratumumab may be lower than that the first dose. The additional doses of daratumumab may vary as determined by a medical practitioner. If the subject exhibits disease progress or severe toxicity, the additional daratumumab treatment may be terminated.


(iv) Conditioning Regimen (Lymphodepleting Therapy)


Any human patients suitable for the allogeneic anti-BCMA CAR-T cell therapy as disclosed herein may receive a lymphodepleting therapy prior to infusion of the anti-BCMA CAR-T cells to reduce or deplete the endogenous lymphocyte of the subject. In any of the combined therapy disclosed herein, the LD therapy may be performed after daratumumab administration, for example, within 3 days post daratumumab infusion.


Lymphodepletion (LD) refers to the destruction of endogenous lymphocytes and/or T cells, which is commonly used prior to immunotransplantation and immunotherapy. Lymphodepletion can be achieved by irradiation and/or chemotherapy. A “lymphodepleting agent” can be any molecule capable of reducing, depleting, or eliminating endogenous lymphocytes and/or T cells when administered to a subject. In some embodiments, the lymphodepleting agents are administered in an amount effective in reducing the number of lymphocytes by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 96%, 96%, 97%, 98%, or at least 99% as compared to the number of lymphocytes prior to administration of the agents. In some embodiments, the lymphodepleting agents are administered in an amount effective in reducing the number of lymphocytes such that the number of lymphocytes in the subject is below the limits of detection. In some embodiments, the subject is administered at least one (e.g., 2, 3, 4, 5 or more) lymphodepleting agents.


In some embodiments, the lymphodepleting agents are cytotoxic agents that specifically kill lymphocytes. Examples of lymphodepleting agents include, without limitation, fludarabine, cyclophosphamide, bendamustin, 5-fluorouracil, gemcitabine, methotrexate, dacarbazine, melphalan, doxorubicin, vinblastine, cisplatin, oxaliplatin, paclitaxel, docetaxel, irinotecan, etopside phosphate, mitoxantrone, cladribine, denileukin diftitox, or DAB-IL2. In some instances, the lymphodepleting agent may be accompanied with low-dose irradiation. The lymphodepletion effect of the conditioning regimen can be monitored via routine practice.


In some embodiments, the method described herein involves a conditioning regimen that comprises one or more lymphodepleting agents, for example, fludarabine and cyclophosphamide. A human patient to be treated by the method described herein may receive multiple doses of the one or more lymphodepleting agents for a suitable period (e.g., 1-5 days) in the conditioning stage. The patient may receive one or more of the lymphodepleting agents once per day during the lymphodepleting period. In one example, the human patient receives fludarabine at about 20-50 mg/m2 (e.g., 30 mg/m2) per day for 2-4 days (e.g., 3 days) and cyclophosphamide at about 300-600 mg/m2 (e.g., 300 mg/m2 or 500 mg/m2) per day for 2-4 days (e.g., 3 days).


In one example, the human patient receives fludarabine at about 30 mg/m2 per day for 3 days and cyclophosphamide at about 300 mg/m2 per day for 3 days. In other examples, the human patient receives fludarabine at about 30 mg/m2 per day for 3 days and cyclophosphamide at about 500 mg/m2 per day for 3 days.


In some embodiments, the LD chemotherapy increases a serum level of IL-7, IL-15, IL-2, IL-21, IL-10, IL-5, IL-8, MCP-1, PLGF, CRP, sICAM-1, sVCAM-1, or a combination thereof in the subject. In some embodiments, the LD chemotherapy decreases a serum level of perforin, MIP-1b, or both in the subject. In some embodiments, the LD chemotherapy is associated with lymphopenia in the subject. In some embodiments, the LD chemotherapy is associated with a decrease of regulatory T cells in the subject.


Before the LD chemotherapy, the subject may be examined for conditions that may suggest delay of the LD chemotherapy. Exemplary conditions include: significant worsening of clinical status, requirement for supplemental oxygen to maintain a saturation level of greater than about 90%, uncontrolled cardiac arrhythmia, hypotension requiring vasopressor support, active infection, and/or grade≥2 acute neurological toxicity. If one or more of the conditions occur, LC chemotherapy to a subject should be delayed until improvement of the conditions.


(v) Lenalidomide Treatment


Lenalidomide may be formulated in a pharmaceutical composition and given to a suitable subject as disclosed herein at a suitable time point relative to the LD and/or allogeneic anti-BCMA CAR-T cell (e.g., CTX120) therapy. A pharmaceutical composition comprising lenalidomide and one or more pharmaceutically acceptable carriers may be administered to the subject via a suitable route, for example, orally, parenterally, by inhalation spray, rectally, nasally, buccally, vaginally or via an implanted reservoir.


In some embodiments, the pharmaceutical composition comprising lenalidomide is to be administered by injection, for example, intravenous infusion or subcutaneous injection. A sterile injectable composition, e.g., a sterile injectable aqueous or oleaginous suspension, can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as Tween® 80) and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides). Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents. Other commonly used surfactants such as Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation.


In some embodiments, the pharmaceutical composition comprising lenalidomide is formulated for oral administration. A composition for oral administration can be any orally acceptable dosage form including, but not limited to, capsules, tablets, emulsions and aqueous suspensions, dispersions and solutions. In the case of tablets for oral use, carriers which are commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added. A nasal aerosol or inhalation composition can be prepared according to techniques well known in the art of pharmaceutical formulation. An oxadiazole compound-containing composition can also be administered in the form of suppositories for rectal administration.


The pharmaceutical compositions as described herein can comprise pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions. Remington: The Science and Practice of Pharmacy 20th Ed. (2000) Lippincott Williams and Wilkins, Ed. K. E. Hoover. Such carriers, excipients or stabilizers may enhance one or more properties of the active ingredients in the compositions described herein, e.g., bioactivity, stability, bioavailability, and other pharmacokinetics and/or bioactivities.


Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations used, and may comprise buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; benzoates, sorbate and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, serine, alanine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as TWEEN™ (polysorbate), PLURONICS™ (nonionic surfactants), or polyethylene glycol (PEG).


In some embodiments, an effective amount of lenalidomide (e.g., about 5-15 mg such as about 5-10 mg) may be given to the subject via a suitable route (e.g., orally) on a daily basis for a suitable period of time (e.g., 15-30 days, such as 21 days)—the first course of lenalidomide treatment. In some examples, the first dose may start after the LD therapy and before the CAR-T cell administration. In other examples, the first dose may start during the LD therapy, for example, concurrently with the first dose of the LD therapy or after the first dose of the LD therapy. In one example, the first dose of lenalidomide is starting on the same date as the third dose of the LD therapy.


In some instances, lenalidomide treatment may be repeated for one or more additional cycles after the first course of treatment, for example, when a patient shows stable disease or better after infusion of the anti-BCMA CAR-T cell (e.g., CTX120 cell) therapy. In some examples, the additional cycles of lenalidomide may start on at least 21 days post CAR-T cell infusion, e.g., at least 28 days post CAR-T cell infusion. In one example, the additional cycles of lenalidomide may start on Day 28 post CAR-T infusion. Such additional lenalidomide treatment cycles may be up to five cycles. Each cycle may be 28 days in total, including a 21-day treatment period during which the subject is administered a daily dose of lenalidomide, followed by a 7-day resting period (lenalidomide free period). The daily dose of lenalidomide in the additional cycles may be the same as in the first course of treatment (e.g., 10 mg daily if the patient tolerated it). Alternatively, the daily dose of lenalidomide in the additional cycles may be lower than that used in the first course of treatment. The daily dose of lenalidomide in each of the additional cycles may vary as determined by a medical practitioner. If the subject exhibits disease progress or severe toxicity, the additional cycles of lenalidomide treatment may be terminated.


In one example, a human patient having refractory or relapsed MM and meets one or more of the inclusion and/or exclusion criteria listed in Example 11 below can be selected for the combined therapy disclosed herein. For example, the human patient may have previously received lenalidomide and meet one of the following conditions: (a) have had at least 2 prior lines of therapy, including an IMiD (e.g., lenalidomide, or pomalidomide), PI (e.g., bortezomib, or carfilzomib), and a CD38-directed monoclonal antibody (e.g., daratumumab); (b) multiple myeloma that is triple-refractory (e.g., progression on or within 60 days of treatment with PI, IMiD, and anti-CD38 antibody, as part of the same or different regimens) or multiple myeloma that is double-refractory to PI and IMiD, as part of the same or different regimens); and (c) multiple myeloma relapsed within 12 months after autologous SCT.


The human patient is first subject to a lymphodepleting (LD) chemotherapy, which may comprise co-administration of fludarabine at 30 mg/m2 and cyclophosphamide at 300 mg/m2 via intravenous infusion each day for three days. On the 3rd day, the patient may start the lenalidomide treatment, for example, by oral administration of 10 mg lenalidomide once daily for 21 days. 2-7 days after the LD chemotherapy, the human patient is administered CTX120 cells at a dose of 5×107 to 7.5×108 CAR+ cells via intravenous infusion, for example, 5×107, 1.5×108, 4.5×108, 6.0×108, or 7.5×108 CAR+ T cells. In some examples, the dose of CTX120 used in this method is 4.5×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 7.5×108 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 6.0×108 CAR+ T cells. The patient can be monitored for disease status. If the patient achieves stable disease or better on Day 28 post-CTX120 infusion, a 28-day cycle (21 days treatment and 7 days resting) of 5 mg lenalidomide by oral administration may be performed to the patient for up to five cycles. The lenalidomide treatment may be terminated if the patient exhibits disease progression or unacceptable toxicity.


Lenalidomide may be stopped at any point if the subject develops grade≥3 CRS, grade≥2 ICANS, acute kidney injury, etc., or a combination thereof. See also Example 11 below. The subject may be examined weekly to monitor cytopenia resolution. For example, the patient would be suitable for lenalidomide treatment if he or she has ANC≥1000/μL and platelets≥30,000/μL. If ANC, platelets, and/or complete blood count are below the standard, lenalidomide treatment may be postponed, for example, to 6 weeks post-CTX120 infusion.


(vi) Allogenic Anti-BCMA CAR-T Cell Therapy


After a subject has been conditioned for receiving allogenic CAR-T cell therapy (e.g., have undergone the LD chemotherapy), an effective amount of the population of genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) or a pharmaceutical composition comprising such as disclosed herein (e.g., comprising CTX120 cells suspended in a cryopreservation solution, which may comprise about 5% DMSO) may be given to the subject (e.g., a human MM patient) via suitable route and schedule. In some examples, the T cells are administered via intravenous infusion. “Allogenic T cell therapy” means that the T cells given to a recipient is derived from one or more donors of the species but not from the recipient. In the allogenic cell therapy disclosed herein, the genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) may be derived from one or more health human donors and are given to a human MM patient.


In some embodiments, the genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) can be administered to a subject (e.g., a human MM patient) at least 24 hours (one day) after the subject receives the LD chemotherapy. For example, administration of the genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) may be 2-7 days after the LD chemotherapy. In some embodiments, the allogeneic T cells are administered no more than ten days after administration of the LD chemotherapy, e.g., no more than nine days, no more than eight days, no more than seven days, no more than six days, no more than five days, no more than four days, no more than three days, no more than two days, or no more than one day. In some embodiments, the allogeneic T cells are administered within 24 hours to ten days, 24 hours to nine days, 30 hours to nine days, 30 hours to eight days, 36 hours to eight days, 36 hours to seven days, or 48 hours to seven days, after administration of the LD chemotherapy. In some embodiments, the allogeneic T cells are administered within 48 hours to seven days after administration of the LD chemotherapy.


After the LD chemotherapy and before administration of the genetically engineered anti-BCMA CAR-T cells, the subject (e.g., a human MM patient) may be examined for conditions that may suggest delay of the allogenic T cell administration. Exemplary conditions include: active uncontrolled infection, worsening of clinical status compared to the clinical status prior to the LD chemotherapy, and/or grade≥2 acute neurological toxicity. Administration of the anti-BCMA CAR-T cells should be delayed if one or more of such conditions occur until improvement is observed. If the delay extends beyond a certain period after the LD chemotherapy (e.g., at least 10 days, at least 12 days, at least 15 days, or at least 21 days after the LD chemotherapy), the LD chemotherapy may be repeated before administration of the anti-BCMA CAR-T cells.


To perform the allogenic T cell therapy, an effective amount of the population of genetically engineered anti-BCMA CAR-T cells as disclosed herein, for example, CTX120 cells, can be administered to a suitable subject (e.g., a human MM patient), who meets the requirements disclosed herein. The genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) may be suspended in a cryopreservation solution, which may comprise about 2-10% DMSO (e.g., about 5% DMSO), and optionally substantially free of serum. As used herein, the term “an effective amount” refers to an amount sufficient to provide a desired effect in treating MM. Non-limiting examples of the desired effects include preventing development of MM; reducing likelihoods of developing MM; slowing, delaying, arresting or reversing progression of MM; inhibiting, reducing, ameliorating, or alleviating a symptom of MM, or a combination thereof in the subject. The effective amount of a given case can be determined by one of ordinary skill in the art using routine experimentation, for example, by accessing a change in a relevant target level (e.g., by at least 10%), need for hospitalization or other medical interventions.


In some embodiments, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 2.5×107 to about 1.05×109 CAR+ T cells, such as 2.5×107 to about 7.5×108 CAR+ T cells, are administered to a human MM patient (e.g., those disclosed herein) via intravenous infusion. For example, about 5×107 to about 1.05×109 CAR+ T cells expressing the anti-BCMA CAR (e.g., CTX120 cells), such as about 5×107 to about 7.5×108 CAR+ T cells expressing the anti-BCMA CAR (e.g., CTX120), may be administered to the patient by intravenous infusion. Exemplary effective amount of CAR+ T cells for use in the allogenic T cell therapy disclosed herein include about 2.5×107, about 3×107, about 4×107, about 5×107, about 6×107, about 7×107, about 8×107, about 9×107, about 1×108, about 2×108, about 3×108, about 4×108, about 5×108, about 6×108, about 7.5×108, about 9×108, or about 1.05×109 CAR+ T cells. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 2.5×107 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 5×107 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 1.5×108 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 4.5×108 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 6×108 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 7.5×108 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 9.0×108 CAR+ T cells are administered to the patient by intravenous infusion. In some examples, a population of genetically engineered anti-BCMA CAR-T cells such as CTX120 cells comprising about 1.05×109 CAR+ T cells are administered to the patient by intravenous infusion.


In some embodiments, an effective amount of the genetically engineered T cell population as disclosed herein (e.g., the CTX120 cells) may range from about 1.5×108 to about 71.05×109 CAR+ T cells, for example, about 1.5×108 to about 7.5×108 CAR+ T cells, for example, about 1.5×108 to about 4.5×108 CAR+ T cells, about 1.5×108 to about 6.0×108 CAR+ T cells, about 4.5×108 to about 6.0×108 CAR+ T cells, about 4.5×108 to about 7.5×108 CAR+ T cells, about 6.0×108 to about 7.5×108 CAR+ T cells, about 7.5×108 to about 9.0×108 CAR+ T cells, or about 9.0×108 to about 1.05×109 CAR+ T cells. In specific embodiments, an effective amount of the genetically engineered T cell population as disclosed herein (e.g., the CTX120 cells) may range from about 4.5×108 to about 6×108 CAR+ T cells, or about 6×108 to about 7.5×108 CAR+ T cells. In specific examples, an effective amount of the genetically engineered T cell population as disclosed herein (e.g., the CTX120 cells) may be about 4.5×108 CAR+ T cells. In other specific examples, an effective amount of the genetically engineered T cell population as disclosed herein (e.g., the CTX120 cells) may be about 7.5×108 CAR+ T cells, which may be decreasd to 6.0×108 CAR+ T cells under certain circumstances (see Example 16 below).


In some examples, the effective amout of the genetically engineered T cells as disclosed herein (e.g., CTX120 cells) is at least 1.5×108 CAR+ T cells. In some examples, the effective amout of the genetically engineered T cells as disclosed herein (e.g., CTX120 cells) is at least 4.5×108 CAR+ T cells. In some examples, the effective amout of the genetically engineered T cells as disclosed herein (e.g., CTX120 cells) is at least 6.0×108 CAR+ T cells. In some examples, the effective amout of the genetically engineered T cells as disclosed herein (e.g., CTX120 cells) is at least 7.5×108 CAR+ T cells.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease serum M-protein levels by at least 25% in the subject, e.g., by at least 30%, by at least 35%, by at least 40%, by at least 45%, by at least 50%, by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, or by at least 95% in the subject.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease 24-hour urine M-protein levels by at least 50% in the subject, e.g., by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, or by at least 95% in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-protein levels by at least 25%, 24-hour urine M-protein levels by at least 50%, or both in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-protein levels by at least 25% and 24-hour urine M-protein levels by at least 50% in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-protein levels by at least 50%, 24-hour urine M-protein levels by at least 90%, or both in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-protein levels by at least 50% and 24-hour urine M-protein levels by at least 90% in the subject.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease 24-hour urine M-protein levels to less than 200 mg in the subject, e.g., to less than 190 mg, to less than 180 mg, to less than 170 mg, to less than 160 mg, to less than 150 mg, to less than 140 mg, to less than 130 mg, to less than 120 mg, to less than 110 mg, to less than 100 mg, to less than 90 mg, to less than 80 mg, to less than 70 mg, to less than 60 mg, or to less than 50 mg in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-protein levels by at least 90%, 24-hour urine M-protein levels to less than 100 mg, or both in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-protein levels by at least 90% and 24-hour urine M-protein levels to less than 100 mg in the subject.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease soft tissue plasmacytomas sizes (SPD) by at least 30% in the subject, e.g., by at least 35%, by at least 40%, by at least 45%, by at least 50%, by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, or by at least 95% in the subject. In some embodiments, the effective dosage is sufficient to decrease soft tissue plasmacytomas sizes (SPD) by at least 50% in the subject. In some embodiments, the effective dosage is sufficient to decrease soft tissue plasmacytomas to undetectable levels.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease plasma cell counts by at least 20% in the subject, e.g., by at least 25%, by at least 30%, by at least 35%, by at least 40%, by at least 45%, by at least 50%, by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, or by at least 95% in the subject. In some embodiments, the effective dosage is sufficient to decrease plasma cell counts by at least 50% in the subject.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease plasma cell counts to less than 10% of bone marrow (BM) aspirates in the subject, e.g., less than 9%, less than 8%, less than 7%, less than 6%, less than 5%, less than 4%, or less than 3% of BM aspirates in the subject. In some embodiments, the effective dosage is sufficient to decrease plasma cell counts to less than 5% of BM aspirates in the subject. In some embodiments, the effective dosage is sufficient to decrease serum M-proteins, urine M-proteins, and soft tissue plasmacytomas to undetectable levels, and plasma cell counts to less than 5% of BM aspirates in the subject.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells is sufficient to decrease differences between involved and uninvolved free light chain (FLC) levels by at least 20% in the subject, e.g., by at least 25%, by at least 30%, by at least 35%, by at least 40%, by at least 45%, by at least 50%, by at least 55%, by at least 60%, by at least 65%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, or by at least 95% in the subject. In some embodiments, the effective dosage is sufficient to decrease differences between involved and uninvolved FLC levels by at least 50% in the subject.


In some embodiments, the subject has myeloma cells that produce kappa (κ) light chains, and the effective dosage is sufficient to decrease kappa-to-lambda light chain ratios (κ/λ ratios) to 6:1 or lower, e.g., 11:2 or lower, 11:2 or lower, 5:1 or lower, 9:2 or lower, 4:1 or lower, 7:2 or lower, 3:1 or lower, 5:2 or lower, 2:1 or lower, 3:2 or lower, or 1:1 or lower. In some embodiments, the subject has myeloma cells that produce κ light chains, and the effective dosage is sufficient to decrease κ/λ ratios to 4:1 or lower.


In some embodiments, the subject has myeloma cells that produce lambda (λ) light chains, and the effective dosage is sufficient to increase kappa-to-lambda light chain ratios (κ/λ ratios) to 1:4 or higher, e.g., 2:7 or higher, 1:3 or higher, 2:5 or higher, 1:2 or higher, 1:1 or higher, 3:2 or higher, or 2:1 or higher. In some embodiments, the subject has myeloma cells that produce λ light chains, and the effective dosage is sufficient to increase κ/λ ratios to 1:2 or higher.


In some embodiments, the effective amount of the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells comprises 1×106 or less TCR+ T cells/kg (subject), e.g., 8×105 or less, 6×105 or less, 4×105 or less, 2×105 or less, 1×105 or less, 8×104 or less, 6×104 or less, 4×104 or less, 2×104 or less, or 1×104 or less TCR+ T cells/kg (subject). In some embodiments, the effective dosage comprises about 1×104 to about 1×106 TCR+ T cells/kg (subject), e.g., about 1×104 to about 1×106, about 2×104 to about 1×106, about 2×104 to about 8×105, about 4×104 to about 8×105, about 4×104 to about 6×105, about 6×104 to about 6×105, about 6×104 to about 4×105, about 8×104 to about 4×105, or about 1×105 to about 2×105 TCR+ T cells/kg (subject). In some embodiments, the effective dosage comprises 1×105 or less TCR+ T cells/kg (subject). In some embodiments, the effective dosage comprises 7×104 or less TCR+ T cells/kg (subject).


In some embodiments, the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells T cells are injected, for example, infused intravenously. Non-limiting examples of routes of administration include intravenous, intrathecal, intraperitoneal, intraspinal, intracereberal, spinal, and intrasternal infusion. In some embodiments, the route is intravenous. In some embodiments, the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells are administered directly into a target site, tissue, or organ. In some embodiments, the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells are administered systemically (e.g., into the subject's circulatory system). In some embodiments, the systemic route comprises intraperitoneal administration, intravenous administration, or both. In some embodiments, the genetically engineered anti-BCMA CAR-T cells disclosed herein such as CTX120 cells are administered as a single intravenous infusion. In some embodiments, the allogeneic T cells are administered as two or more intravenous infusions.


After the allogenic T cell therapy disclosed herein, the subject shall be monitored for development of acute toxicity, for example, infusion reactions, cytokine release syndrome (CRS), febrile reactions, neurotoxicity (e g , immune effector cell-associated neutotoxicity syndrome or ICANS), tumor lysis syndrome, hemophagocytic lymphohistiocytosis (HLH), Cytopenias, GvHD, hypotention, renal insufficiency, viral encephalitis (e.g., via HHV6 infection), neutropenia, thrombocytopenia, or a combination thereof. Toxicity management known to those medical practitioners shall be performed to the subject if toxicity is observed after administration of the genetically engineered anti-BCMA CAR-T cells such as CTX120 cells. See Example 16 for more details regarding toxicity management.


In some instances, a pharmacokinetic (PK) profile of the genetically engineered anti-BCMA CAR-T cells such as CTX120 cells in a human recipient after administration may be examined. The PK profile may evaluate an effectiveness of the allogenic T cell therapy on a human MM patient.


The genetically engineered CAR-T cells may undergo an expansion phase following administration to a subject. Expansion is a response to antigen recognition and signal activation (Savoldo, B. et al. (2011) J Clin Invest. 121:1822; van der Stegen, S. et al. (2015) Nat Rev Drug Discov. 14:499-509). Following expansion, the genetically engineered CAR-T cells undergo a contraction phase, where short-lived effector CAR-T cells are eliminated and that are long-lived memory CAR-T cells remain. The duration of the persistence phase provides a measure of the longevity of the CAR-T cells following expansion and contraction.


In some embodiments, the PK profile comprises the quantity of the genetically engineered anti-BCMA CAR-T cells in a tissue over time. Exemplary tissues suitable for this analysis include peripheral blood. The tissue sample may be collected daily or weekly. Alternatively or in addition, the tissue sample may be collected starting on day 1, day 2, day 3, or day 4 after T cell administration. Collection of the tissue sample may end not earlier than day 5 after the T cell administration, e.g., not earlier than day 8, not earlier than day 10, not earlier than day 15, or not earlier than day 20 after T cell administration. In some embodiments, collection of the tissue sample is performed at least once per week after T cell administration, e.g., at least twice, or at least 3 times per week after T cell administration. In some embodiments, collection of the tissue sample is performed for up to 16 weeks after T cell administration, e.g., up to 15 weeks, up to 12 weeks, up to 10 weeks, up to 8 weeks, or up to 6 weeks.


In some embodiments, evaluating the PK profile comprising obtaining a baseline measurement, which may be obtained before administration of the genetically engineered anti-BCMA CART cells, for example, no more than 15 days before T cell administration, e.g., no more than 10 days, no more than 5 days, no more than 1 day before T cell administration. In some embodiments, the baseline measurement is obtained within 0.25 to 48 hours before T cell administration, e.g., within 0.5-24 hours, within 1 to 36 hours, within 1-12 hours, or within 2-12 hours.


In some embodiments, the time course of the quantity of the genetically engineered anti-BCMA CAR-T cells in the tissue is measured by an area under the curve (AUC). A method of calculating an AUC is known to one skilled in the art and is comprised of approximating an AUC by a series of trapezoids, computing the area of the trapezoids, and summing the area of the trapezoids to determine the AUC. In some embodiments, an AUC is defined for a PK profile wherein the quantity of the genetically engineered anti-BCMA CAR-T cells is measured for a given tissue type over time. In some embodiments, an AUC is defined for a PK profile from one designated time point to another designated time point (i.e., AUC10-80 refers to the total area under a quantity-time curve depicting quantity from day 10 to day 80 following administration). In some embodiments, an AUC is determined for a preselected time period extending from time of administration (e.g., day 1) to a time ending on a day that is 1-7, 10-20 days, 15-45 days, 20-70 days, 25-100 days, or 40-180 days following administration. In some embodiments, an AUC measured for a PK profile in a recipient is indicative of a response in the recipient (e.g., CR or PR). In some embodiments, an AUC measured for a PK profile in a recipient is indicative of a risk of relapse in the recipient.


In some embodiments, the genetically engineered anti-BCMA CAR-T cells do not induce toxicity in non-cancer cells in the subject. Alternatively, the genetically engineered anti-BCMA CAR-T cells do not trigger complement mediated lysis, or does not stimulate antibody-dependent cell mediated cytotoxicity (ADCC).


In one example, a human patient having refractory or relapsed MM and meets one or more of the inclusion and/or exclusion criteria listed in Example 16 below can be selected for the combined therapy disclosed herein. For example, the human patient may have previously received daratumumab and meet one of the following conditions: (a) have had at least 2 prior lines of therapy, including an IMiD (e.g., lenalidomide, or pomalidomide), PI (e.g., bortezomib, or carfilzomib), and a CD38-directed monoclonal antibody (e.g., daratumumab); (b) multiple myeloma that is triple-refractory (e.g., progression on or within 60 days of treatment with PI, IMiD, and anti-CD38 antibody, as part of the same or different regimens) or multiple myeloma that is double-refractory to PI and IMiD, as part of the same or different regimens); and (c) multiple myeloma relapsed within 12 months after autologous SCT.


The patient can be monitored for disease status. If the patient achieves stable disease or better on Day 28 post-CTX120 infusion, up to five monthly doses of daratumumab may be given to the patient (e.g., 16 mg/kg via intravenous infusion). The daratumumab treatment may be terminated if the patient exhibits disease progression or unacceptable toxicity. For example, disease response may be assessed pursuant to the IMWG response criteria disclosed in Example 16 below before report dosing with daratumumab. Redosing would not be permitted if the patient exhibits severe adverse effects related to daratumumab.


In some embodiments, the patient can be premedicated with corticosteroids, antipyretics, and/or antihistamines prior to daratumumab infusion to reduce the risk of infusion reactions. More details are provided in Example 16 below. The patient can be monitored during the infusion process for infusion reaction of any grade and/or severity and the infusion may be interrupted if any of such occurs. Alternatively or in addition, the patient may be subject to antiviral prophylaxis after daratumumab infusion, which may be continued for a suitable period.


In some embodiments, the allogenic anti-BCMA CAR-T cell therapy may be in combination with one or more anti-cancer therapies, for example, therapies commonly applied to multiple myeloma. Alternatively or in addition, multiple doses of the allogenic anti-BCMA CAR-T cells such as CTX120 cells disclosed herein may be administered to a human patient.


In some instances, the patient may receive up to four doses of the allogenic anti-BCMA CAR-T cells such as CTX120 cells disclosed herein. For example, a second dose of the anti-BCMA CAR-T cells may be given to the patient within about 4 to 12 weeks after the first dose, when the patient shows stable disease or better responses (based on IMWG criteria). In some examples, each of the additional doses can be accompanied with a lyphodepleting treatment as disclosed herein 2-7 days prior to the CAR-T cell infusion. In other examples, an additional dose may not be accompanied with a lyphodepleting treatment, for example, when the patient experiences significant cytopenias. Any of the patients may also receive additional doses of the anti-BCMA CAR-T cells, which may be accomapneid with lyphodepleting treatment, after the patient shows progressed disease (PD), if the patient had prior response (PR) or better responses based on the IMWG criteria.


The amount of the anti-BCMA CAR-T cells such as the CTX120 cells used in the redosing may range from 5×107 to 1.05×108 CAR+ cells (e.g., 5×107 to 7.5×108 CAR+ cells) via intravenous infusion, for example, 5×107, 1.5×108, 4.5×108, 6.0×108, 7.5×108 CAR+ T cells, 9×108 CAR+ T cells, or 1.05×109 CAR+ T cells. It may be the same as the first dose. Alternatively, it may be higher or lower than the first dose, depending upon the patient's disease status and response to the first dose, which would be within the knowledge of a medical practioner.


(vii) Exemplary Combined Therapy Regimens


Provided herein a few specific treatment regimens, which serve as examples of the combined therapy disclosed herein.


In one examples, the combined therapy disclosed herein may be performed as follows. An eligible multiple myelanoma human patient (e.g., meeting one or more inclusion and exclusion criteria disclosed in Example 16 below) can first be treated with daratumumab at a dose of 16 mg/kg (which may slit 8 mg/kg for two consecutive days) via intravenous infusion. Alernatively, the daratumumab may be given via subcutaneous injection at about 1800 mg per 30,000 units of hyaluronidase. Within up to three days after daratumumab administration, the patient may be subject to a lymphodepleting (LD) chemotherapy, which may comprise co-administration of fludarabine at 30 mg/m2 and cyclophosphamide at 300 mg/m2 via intravenous infusion each day for three days. Alternatively, the LD chemotherapy may comprise fludarabine at 30 mg/m2 and cyclophosphamide at 500 mg/m2 via intravenous infusion each day for three days. 2-7 days after the LD chemotherapy, the human patient is administered CTX120 cells at a dose of 5×107 to 1.05×108 CAR+ cells (e.g., 5×107 to 7.5×108 CAR+ cells) via intravenous infusion, for example, 5×107, 1.5×108, 4.5×108, 6.0×108, 7.5×108 CAR+ T cells, 9×108 CAR+ T cells, or 1.05×109 CAR+ T cells. In some examples, the dose of CTX120 used in this method is 4.5×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 7.5×108 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 6.0×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 1.05×109 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 9.0×108 CAR+ T cells. See FIG. 22 and Cohort 1 disclosed in Example 16.


In another example, the eligible human patient having MM is first subject to a lymphodepleting (LD) chemotherapy, which may comprise co-administration of fludarabine at 30 mg/m2 and cyclophosphamide at 300 mg/m2 via intravenous infusion each day for three days. Alternatively, the LD chemotherapy may comprise fludarabine at 30 mg/m2 and cyclophosphamide at 500 mg/m2 via intravenous infusion each day for three days. On the 3rd day, the patient may start the lenalidomide treatment, for example, by oral administration of 10 mg lenalidomide once daily for 21 days. 2-7 days after the LD chemotherapy, the human patient is administered CTX120 cells at a dose of 5×107 to 1.05×108 CAR+ cells (e.g., 5×107 to 7.5×108 CAR+ cells) via intravenous infusion, for example, 5×107, 1.5×108, 4.5×108, 6.0×108, 7.5×108 CAR+ T cells, 9×108 CAR+ T cells, or 1.05×109 CAR+ T cells. In some examples, the dose of CTX120 used in this method is 4.5×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 7.5×108 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 6.0×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 1.05×109 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 9.0×108 CAR+ T cells. The patient can be monitored for disease status. If the patient achieves stable disease or better on Day 28 post-CTX120 infusion, a 28-day cycle (21 days treatment and 7 days resting) of 5 mg lenalidomide by oral administration may be performed to the patient for up to five cycles. The lenalidomide treatment may be terminated if the patient exhibits disease progression or unacceptable toxicity. See FIG. 23 and Cohort 2 disclosed in Example 16.


In yet another example, an eligible multiple myelanoma human patient can first be treated with daratumumab at a dose of 16 mg/kg (which may slit 8 mg/kg for two consecutive days) via intravenous infusion. Alernatively, the daratumumab may be given via subcutaneous injection at about 1800 mg per 30,000 units of hyaluronidase. Within up to three days after daratumumab administration, the patient may be subject to a lymphodepleting (LD) chemotherapy, which may comprise co-administration of fludarabine at 30 mg/m2 and cyclophosphamide at 300 mg/m2 via intravenous infusion each day for three days. Alternatively, the LD chemotherapy may comprise fludarabine at 30 mg/m2 and cyclophosphamide at 500 mg/m2 via intravenous infusion each day for three days. 2-7 days after the LD chemotherapy, the human patient is administered CTX120 cells at a dose of 5×107 to 1.05×108 CAR+ cells (e.g., 5×107 to 7.5×108 CAR+ cells) via intravenous infusion, for example, 5×107, 1.5×108, 4.5×108, 6.0×108, 7.5×108 CAR+ T cells, 9×108 CAR+ T cells, or 1.05×109 CAR+ T cells. In some examples, the dose of CTX120 used in this method is 4.5×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 7.5×108 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 6.0×108 CAR+ T cells. In other examples, the dose of CTX120 used in this method is 1.05×109 CAR+ T cells. When needed, the dose of CTX120 may be adjusted to 9.0×108 CAR+ T cells. On the 3rd day, the patient may start the lenalidomide treatment, for example, by oral administration of 10 mg lenalidomide once daily for 21 days. If the patient achieves stable disease or better on Day 28 post-CTX120 infusion, a 28-day cycle (21 days treatment and 7 days resting) of 5 mg lenalidomide by oral administration may be performed to the patient for up to five cycles. The lenalidomide treatment may be terminated if the patient exhibits disease progression or unacceptable toxicity. See FIG. 24 and Cohort 3 disclosed in Example 16.


Any of the specific treatment regimens disclosed herein may further comprise a second dose of the CTX120 cells, and optionally a third and fourth doses of the CTX120 cells, following the re-dosing conditions disclosed herein (e.g., see Example 16 below).


III. Kit for Combined Allogeneic Anti-BCMA CAR-T Cell and NK Cell Inhibitor Therapy

The present disclosure also provides kits for use of a population of anti-BCMA CAR T cells such as CTX120 T cells and an NK cell inhibitor such as an anti-CD38 antibody (e.g., daratumumab) as described herein in methods for treating multiple myeloma, such as refractory and/or relapsed multiple myeloma. Such kits may include a first container comprising a first pharmaceutical composition that comprises any of the populations of genetically engineered anti-BCMA CAR T cells (e.g., those described herein such as CTX120 cells), and a pharmaceutically acceptable carrier, and optionally a second container comprising a second pharmaceutical composition comprising the NK cell inhibitor such as daratumumab. The anti-BCMA CAR-T cells may be suspended in a cryopreservation solution such as those disclosed herein. Optionally, the kit may further comprise a third container comprising a third pharmaceutical composition that comprises one or more lymphodepleting agents.


In some embodiments, the kit can comprise instructions for use in any of the methods described herein. The included instructions can comprise a description of administration of the first, the second, and/or the third pharmaceutical compositions to a subject to achieve the intended activity in a human MM patient. The kit may further comprise a description of selecting a human MM patient suitable for treatment based on identifying whether the human patient is in need of the treatment. In some embodiments, the instructions comprise a description of administering the first, the second, and/or the third pharmaceutical compositions to a human patient who is in need of the treatment.


The instructions relating to the use of a population of anti-BCMA CAR-T cells such as CTX120 T cells described herein generally include information as to dosage, dosing schedule, and route of administration for the intended treatment. The instructions may also include information relating to the use of daratumumab, for example, dosage, dosing schedule, and route of administration for the intended treatment. The containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. Instructions supplied in the kits of the disclosure are typically written instructions on a label or package insert. The label or package insert indicates that the population of genetically engineered T cells is used for treating, delaying the onset, and/or alleviating a symptom of MM in a subject.


The kits provided herein are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging, and the like. Also contemplated are packages for use in combination with a specific device, such as an inhaler, nasal administration device, or an infusion device. A kit may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The container may also have a sterile access port. At least one active agent in the pharmaceutical composition is a population of the anti-BCMA CAR-T cells such as the CTX120 T cells as disclosed herein.


Kits optionally may provide additional components such as buffers and interpretive information. Normally, the kit comprises a container and a label or package insert(s) on or associated with the container. In some embodiment, the disclosure provides articles of manufacture comprising contents of the kits described above.


General Techniques

The practice of the present disclosure will employ, unless otherwise indicated, conventional techniques of molecular biology (including recombinant techniques), microbiology, cell biology, biochemistry, and immunology, which are within the skill of the art. Such techniques are explained fully in the literature, such as Molecular Cloning: A Laboratory Manual, second edition (Sambrook, et al., 1989) Cold Spring Harbor Press; Oligonucleotide Synthesis (M. J. Gait, ed. 1984); Methods in Molecular Biology, Humana Press; Cell Biology: A Laboratory Notebook (J. E. Cellis, ed., 1989) Academic Press; Animal Cell Culture (R. I. Freshney, ed. 1987); Introuction to Cell and Tissue Culture (J. P. Mather and P. E. Roberts, 1998) Plenum Press; Cell and Tissue Culture: Laboratory Procedures (A. Doyle, J. B. Griffiths, and D. G. Newell, eds. 1993-8) J. Wiley and Sons; Methods in Enzymology (Academic Press, Inc.); Handbook of Experimental Immunology (D. M. Weir and C. C. Blackwell, eds.): Gene Transfer Vectors for Mammalian Cells (J. M. Miller and M. P. Calos, eds., 1987); Current Protocols in Molecular Biology (F. M. Ausubel, et al. eds. 1987); PCR: The Polymerase Chain Reaction, (Mullis, et al., eds. 1994); Current Protocols in Immunology (J. E. Coligan et al., eds., 1991); Short Protocols in Molecular Biology (Wiley and Sons, 1999); Immunobiology (C. A. Janeway and P. Travers, 1997); Antibodies (P. Finch, 1997); Antibodies: a practice approach (D. Catty., ed., IRL Press, 1988-1989); Monoclonal antibodies: a practical approach (P. Shepherd and C. Dean, eds., Oxford University Press, 2000); Using antibodies: a laboratory manual (E. Harlow and D. Lane (Cold Spring Harbor Laboratory Press, 1999); The Antibodies (M. Zanetti and J. D. Capra, eds. Harwood Academic Publishers, 1995); DNA Cloning: A practical Approach, Volumes I and II (D. N. Glover ed. 1985); Nucleic Acid Hybridization (B. D. Hames & S. J. Higgins eds. (1985; Transcription and Translation (B. D. Hames & S. J. Higgins, eds. (1984»; Animal Cell Culture (R. I. Freshney, ed. (1986; Immobilized Cells and Enzymes (IRL Press, (1986; and B. Perbal, A practical Guide To Molecular Cloning (1984); F. M. Ausubel et al. (eds.).


Without further elaboration, it is believed that one skilled in the art can, based on the above description, utilize the present invention to its fullest extent. The following specific embodiments are, therefore, to be construed as merely illustrative, and not limitative of the remainder of the disclosure in any way whatsoever. All publications cited herein are incorporated by reference for the purposes or subject matter referenced herein.


EXAMPLE 1
Preparation of Anti-BCMA CAR T Cells

Genetically engineered T cells expressing a CAR specific for the BCMA antigen (e.g., CTX120 cells) were prepared from healthy donor PBMCs obtained via a standard leukapheresis procedure as described in WO2019/097305 and WO/2019/215500, the relevant disclosures of each of which are incorporated by reference for the purpose and subject matter referenced herein.


Briefly, mononuclear cells were enriched for T cells and activated with anti-CD3/CD28 antibody-coated beads. The enriched and activated T cells were then genetically modified using CRISPR/Cas9 to disrupt (e.g., generate a gene knockout) the coding sequences of the TRAC gene and the β2M gene, with simultaneous insertion of a CAR specific to BCMA that is expressed by human MM cells. The insertion of the CAR occurred by HDR of a DNA DSB generated by Cas9/gRNA. The CAR was encoded by donor DNA with left and right flanking homology arms that were specific to the TRAC gene, thus enabling insertion of the CAR into a DNA DSB generated at the TRAC gene. The CAR homology donor DNA was administered using rAAV6. Disruption of the TRAC gene yielded loss of function of the TCR and renders the gene-edited T cell non-alloreactive and suitable for allogeneic transplantation by minimizing the risk of GVHD, while disruption of the β2M gene yielded loss of expression of MHC I and prevents susceptibility of the gene-edited T cells to a HVG response. Insertion of an anti-BCMA CAR into the TRAC gene provides T cells that are reactive to MM tumor cells that express BCMA surface antigen.


To perform the gene-editing, primary human T cells were first electroporated with Cas9-sgRNA RNP complexes targeting the TRAC and β2M genes. Cas9 nuclease was mixed with TA-1 sgRNA (SEQ ID NO: 1, targeting TCR) and with B2M-1 sgRNA (SEQ ID NO:5, targeting β2M) in separate microcentrifuge tubes. Each solution was incubated for no less than 10 minutes at room temperature to form each ribonucleoprotein complex. The two Cas9/gRNA mixtures were combined, and mixed with the cells, bringing Cas9, TA-1 and B2M-1 to a final concentration of 0.3 mg/mL, 0.08 mg/mL and 0.2 mg/mL, respectively. Cells were electroporated with the Cas9-sgRNA RNP. Following electroporation, cells were treated with rAAV6 encoding an anti-BCMA CAR with flanking left and right 800-bp homology arms specific to the TRAC locus. The encoded CAR was operably linked to a 5′ elongation factor EF-1α to function as a promoter and a 3′ polyadenylation sequence to promote mRNA transcription stability. The CAR comprised a humanized scFv derived from a murine antibody specific for human BCMA, a hinge region and transmembrane domain, a signaling domain comprising CD3-ζ, and a 4-1BB co-stimulatory domain.


The target gene sequences, and sgRNAs, and the spacer sequences encoded by the sgRNAs are provided in Table 1 below.


A disrupted TRAC gene produced by a TRAC sgRNA in Table 1 above may comprise one of the edited TRAC gene sequences provided in Table 2 below (“-” indicates deletion and residues in boldface indicate mutation or insertion):


A portion of the genetically engineered anti-BCMA CAR-T cells may comprise an edited TRAC gene, a fragment of which may be replaced by the nucleotide sequence encoding the anti-BCMA CAR via homologous recombination at the regions corresponding to the left and right homology arms (see Table 4 below). As such, a portion of the genetically engineered anti-BCMA CAR-T cells disclosed herein (e.g., CTX120 cells) may comprise a disrupted TRAC gene, which has a deletion of at least the AGAGCAACAGTGCTGTGGCC (SEQ ID NO: 10) fragment. A nucleic acid comprising a nucleotide sequence encoding the anti-BCMA CAR (e.g., SEQ ID NO: 33; see Table 4 below) may be inserted into the TRAC gene locus. The CAR-coding sequence is in operably linkage to a EF-1a promoter such as SEQ ID NO: 38. A poly A sequence (e.g., SEQ ID NO: 39) can be located downstream of the coding sequence. See Table 4 below.


Further, a portion of the genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) comprise a plurality of disrupted β2M genes, which collectively may comprise one or more of the edited β2M gene sequence listed in Table 3 below (“-” indicates deletion and residues in boldface indicate mutation or insertion):


The components of the rAAV encoding the anti-BCMA CAR, including nucleotide sequences and amino acid sequences are provided in Table 4 and Table 5, respectively below.


At least a portion of the resultant genetically engineered anti-BCMA CAR-T cells (e.g., CTX120 cells) may comprise a disrupted TRAC gene, which has a deletion of at least the sequence of SEQ ID NO: 10, a disrupted β2M gene, and express an anti-BCMA CAR (e.g., SEQ ID NO: 40). Further, a portion of the cells in the CTX120 cell population may comprise a plurality of disrupted β2M genes, which collectively may comprise one or more of the sequences of SEQ ID NOs: 21-26. Further, the genetically engineered anti-BCMA CAR-T cells comprise the nucleotide sequence coding for the anti-BCMA CAR. In some examples, the CAR-coding sequence may be inserted into the TRAC gene locus (e.g., SEQ ID NO: 33, coding for the anti-BCMA CAR of SEQ ID NO: 40). The anti-BCMA CAR coding sequence is in operable linkage to an EF-1a promoter, which may comprise the nucleotide sequence of SEQ ID NO: 38. Further, a poly A sequence (e.g., SEQ ID NO: 39) is located downstream of the coding sequence.


The resultant genetically engineered T cells were characterized for incorporation of the desired gene edits: loss of TCR, loss of MHC I expression, and expression of an anti-BCMA CAR. Approximately one week after gene-editing, allogeneic T cells were assessed for surface expression of TCR, β2M, and anti-BCMA CAR using flow cytometry. The allogeneic cells were stained with biotinylated recombinant human BCMA (Acro Biosystems Cat: #BC7-H82F0) and tagged with fluorescent streptavidin and with fluorescent antibodies targeting cell surface markers. The percentage of cells that were TCR, β2M, and anti-BCMA CAR+ was determined. Nine lots of CTX120 cells were prepared from eight healthy donors.


As shown in FIG. 1, reduction in TCR expression was nearly quantitative (96-99% of cells TCR); reduction in β2M expression was also high (72-86% of cells β2M); and anti-BCMA CAR incorporation ranged 46-79%. The percentage of CTX120 cells including the triple gene edits (TCR, β2M, and anti-BCMA CAR+) was between 38% and 67%.


The percentage of the CTX120 cells that were CD4+ or CD8+ was also determined by flow cytometry. As shown in FIGS. 2A-2B, the percentage of CD4+ T cells (FIG. 2A) or CD8+ T cells (FIG. 2B) remained unchanged after the gene-editing process.


EXAMPLE 2
Anti-BCMA CAR T Cells Reduces Tumor Volume and Protects Against Re-Challenge in the MM.1S Tumor Model

The ability of CTX120 cells to limit growth of human BCMA-expressing MM tumors was evaluated in immunocompromised mice. The efficacy of CTX120 cells against the subcutaneous MM.1S tumor xenograft model in NOG mice (NOD.Cg PrkdcscidIl2rgtm1Sug/JicTac) was evaluated. In brief, 5 to 8-week old female NOG mice were individually housed in ventilated microisolator cages and maintained under pathogen-free conditions. The animals each received a subcutaneous inoculation in the right flank of 5×106 MM.1S cells in 50% Matrigel. When the mean tumor volume reached 100 mm3 (approximately 75 to 125 mm3), the mice were randomized into two groups with 5 mice per group. One group was untreated, while the second group was dosed by intravenous injection of 8×106 CTX120 CAR+ T cells.


Tumor volume and body weights were measured twice weekly and individual mice were euthanized when their tumor volume reached ≥2000 mm3. By day 15, animals treated with CTX120 cells showed tumor regression from the starting volumes while animals in the control group had tumors averaging greater than 1000 mm3. By day 29, all animals in the control group had reached the tumor volume endpoint of ≥2000 mm3, whereas all treated animals had rejected the primary tumor burden (FIG. 3).


On day 29, all mice from the group receiving CTX120 treatment were further subjected to a second inoculation of MM.1S tumor cells (e.g., a tumor re-challenge). The mice received a second subcutaneous inoculation in the left flank of 5×106 MM.1S cells in 50% Matrigel. Given that the first untreated group succumbed to tumor burden, a second cohort of tumor-free animals was administered the re-challenge inoculation in the left flank as a positive control.


All mice were monitored for tumor growth in both the initial right flank tumor and the re-challenge tumor in the left flank. Animals treated with CTX120 cells successfully eliminated tumor growth in both the initial right flank tumor and in the re-challenge left flank tumor for the duration of the study, while untreated animals succumbed to tumor burden when given an inoculation of tumor cells in either the right or the left flank (FIG. 3).


EXAMPLE 3
Eradication of RPMI-8226 Tumors with Treatment of Anti-BCMA CAR T Cells

The efficacy of CTX120 was further evaluated in a second model of BCMA-expressing human MM, using the RPMI-8226 tumor xenograft model in NOG mice. In brief, 5 to 8-week old female, NOG (NOD.Cg-PrkdcscidIl2rgtm1Sug/JicTac) mice were individually housed in ventilated microisolator cages and maintained under pathogen-free conditions. At 10 days prior to treatment, the mice received a subcutaneous inoculation of 10×106 RPMI-8226 cells/mouse in the right flank. On day 1, the mice were randomized into groups (n=5 mice per group) and were either untreated or dosed with an intravenous injection of 0.8×106 CAR-expressing CTX120 cells.


Tumor volume was measured twice weekly. Animals treated with CTX120 cells demonstrated complete eradication of tumor burden, while tumors in untreated animals reached a tumor volume exceeding 1500 mm3 by the end of the study duration (FIG. 4).


EXAMPLE 4
Evaluation of Safety and Tolerability of the Anti-BCMA CAR T Cells

The selectivity of CTX120 cells for activation in response to BCMA-expressing cells and tissues was evaluated. To do so, the humanized mouse antibody, from which the scFv portion of the CTX120 CAR was derived, was evaluated for cross-reactivity to human tissues. Briefly, a standard panel of 32 human tissues (Adrenal, Bladder, Blood cells, Bone Marrow, Breast, Brain—cerebellum, Brain—cerebral cortext, Colon, Endothelium—blood vessels, eye, fallopian tube, GI: Tract: stomach, GI Tract: small intestine, Heart, Kidney—glomerulus, Kidney—tubule, Liver, lung, lymph node, Nerve—peripheral, ovary, pancreas, parathyroid, parotid (salivary) gland, Pituitary, placenta, prostate, skin, spinal cord, spleen, striated muscle, testis, thymus, thyroid, tonsil, ureter, uterus—cervix, uterus—endometrium) was evaluated for binding of the antibody following exposure to two concentrations of antibody: an optimal concentration (5.0 μg/mL) and a high concentration (50.0 μg/mL). Binding was evaluated by an immunohistochemistry-based assay, wherein tissue staining was evaluated by a pathologist and positive staining was indicative of reactivity of the antibody to the tissue. As a positive control, staining was evaluated against purified BCMA protein absorbed to a tissue slide. For each tissue tested for antibody binding, tissue sections from three different human donors were evaluated. While robust staining was observed against the purified BCMA protein, no positive staining was observed in any of the human tissues. Thus, the antigen-binding scFv of the anti-BCMA CAR is highly-selective for tissues expressing BCMA.


The selectivity of CTX120 cells for activation in response to BCMA-expressing cell lines was evaluated in vitro. To do so, CTX120 cells were co-cultured for 24 hours with 50,000 target cells with high BCMA expression (MM.1S cells), low BCMA expression (Jeko-1 cells), or no BCMA expression (K562 cells) at a ratio of 2:1 CAR T cells to target cells. Levels of IFNγ and IL-2 that were produced by activated anti-BCMA CAR T cells were measured in the co-culture supernatant using a Luminex-based assay (Milliplex, Millipore Sigma, MA, USA). Cytokine production in response to co-culture with target cells was evaluated for CTX120 cells derived from four individual donors, with the average±the standard error shown in FIGS. 5A-5B. As shown, no cytokine expression was measured when CTX120 cells were co-cultured with K562 cells that lack BCMA expression. In contrast, significant levels of both IFNγ and IL-2 were measured in co-cultures of CTX120 cells co-cultured with BCMA-expressing MM.1S or JeKo-1 cells (FIGS. 5A-5B).


Further, the selectivity of CTX120 cells for inducing target cell killing of BCMA-expressing cell lines was evaluated in vitro. To do so, CTX120 cells or unedited T cells were co-cultured for 24 hours with 50,000 target cells (e.g., MM.1S, JeKo-1 or K562 cells) at a ratio of 8:1, 4:1, 2:1, 1:1, or 0.5:1 T cells to target cells. Prior to co-culture, the target cells were labeled with 5 μM efluor670 (eBiosciences). Following co-culture, the cells were washed, suspended in 200 μL media containing a 1:500 dilution of 4′,6-diamidino-2-phenylindole (DAPI, Molecular Probes) for enumeration of dead/dying cells. 25 μL of CountBright beads (Life technologies) was added per sample. Cells were assessed for labeling by flow cytometry, and the percentage of target cells succumbing to cell lysis was determined using the following calculation:





Cells/μL=((number of live target cell events)/(number of bead events))×((Assigned bead count of lot (beads/50 μL))/(volume of sample))


Total target cells were calculated by multiplying cells/μL×the total volume of cells. The percent cell lysis was then calculated with the following equation:





% Cell lysis=(1−((Total Number of Target Cells in Test Sample)/(Total Number of Target Cells in Control Sample))×100.


Cell killing was evaluated for unedited and edited T cells derived from four different donors, with the average % cell lysis±the standard deviation shown in FIGS. 6A-6C. For cells expressing BCMA (MM.1S in FIG. 6A and JeKo-1 in FIG. 6B), cell lysis induced by edited CTX120 cells was significantly higher than that induced by unedited T cells, even at low T cell to target cell ratios. In contrast, no difference in cell lysis was observed between unedited and edited T cells for K562 cells lacking BCMA expression (FIG. 6C). Thus, cytotoxicity induced by CTX120 cells is dependent upon expression of BCMA by the target cell.


The potential for primary non-tumor human cells to activate CTX120 cells was further evaluated. Of the primary human cells, only B cells are expected to comprise BCMA expressing cells. Activation of CTX120 cells was measured by quantifying levels of IFNγ and IL-2 following co-culture with primary human cells listed in Table 7 below.









Table 7







Primary Human Cells Evaluated for the Ability to Activate CTX120 Cells








Organ
Cell Type





Central Nervous System
Astrocytes



Neurons



Schwann


Heart
Cardiac fibroblasts



Aortic endothelial



Cardiac myocytes


Kidney
Renal epithelial


Lung
Microvascular endothelial cells



Lung fibroblasts



Bronchial smooth muscle



Small airway epithelial


Liver
Hepatocytes


Bone
Chondrocytes



Osteoblasts


Skin
Epidermal keratinocytes



Epidermal melanocytes


Skeletal muscle
Skeletal muscle myoblasts


Intestinal
Intestinal smooth muscle cells


Blood
PBMCs



B cells









To do so, primary human cells were seeded at 25,000 cells per well in 96-well flat-bottom plates in preferred media and incubated overnight. After 24 hours, the primary cell media was removed, and 50,000 CTX120 cells were added in T cell growth media. Co-cultures were incubated for 24 hours and assayed for production of IFNγ and IL-2 using a Luminex-based assay (Milliplex, Millipore Sigma, MA, USA). As a positive control, activation of CTX120 cells was evaluated in response to cells with low BCMA expression (e.g., Jeko-1 cells). The average±the standard deviation production of IFNγ and IL-2 is shown in FIG. 7A and FIG. 7B respectively. Open bars indicated that the values were below the limit of quantification. As shown in FIG. 7A, no co-culture between primary human cells and CTX120 cells resulted in significant secretion of IFNγ when compared to the Jeko-1 positive control cell line, except for co-culture with primary B cells that are known to comprise CD19+/BCMA+ cells. As shown in FIG. 7B, no-culture resulted in significant IL-2 production as compared to the Jeko-1 positive control. Based upon this outcome, CTX120 cells are not activated in the presence of normal, non-BCMA expressing human cells.


Transformed cells proliferate in a cytokine-independent manner Thus, to determine whether gene-editing results in oncogenic transformation, CTX120 cells were evaluated for the ability to grow in the absence of cytokines. To do so, the growth of CTX120 cells in ex vivo culture was evaluated over 27 days in complete media comprising serum and the cytokines IL-2 and IL-7, in media comprising serum but lacking cytokines (e.g., no IL-2 or IL-7), or in media lacking both serum and cytokines (e.g., no serum, IL-2, or IL-7). 5×106 CTX120 cells were plated at approximately 2 weeks following gene-editing (day 0). At various time points, the number of viable CTX120 cells was enumerated using flow cytometry. While T cell growth plateaued when cultured in complete media, the number of viable T cells decreased over time when grown in media lacking cytokines (either with or without serum) as shown in FIG. 8. Shown is the average number of viable cells±the standard error for CTX120 cells derived from four different donors. Thus, the gene-editing approach used to generate CTX120 cells does not result in undesirable oncogenic transformation.


EXAMPLE 5
Analysis of Immune Reactivity with Administration of Anti-BCMA CAR T Cells

The potential for unedited T cells and edited CTX120 cells to cause GvHD following a single dosage was evaluated in mice. Edited CTX120 cells were prepared as described in Example 1. The CTX120 anti-BCMA CAR does not recognize mouse BCMA. However, evaluation for GvHD symptoms in mice (e.g., weight loss, decreased survival, and/or increased morbidity) in response to treatment with unedited or edited T cells is indicative of a GvHD toxicity induced by off-target reactivity of the T cells (e.g., due to TCR reactivity towards alloantigens). As a positive control, mice were treated with unedited allogeneic T cells that cause GvHD toxicity due to reactivity of the TCR with mouse tissue antigens. Treatment with allogeneic CTX120 cells that have very low expression of TCR was evaluated for inducing GvHD toxicity.


To evaluate a GvHD response, NSG mice (NOD.Cg-PrkdcscidIl2rgtm1Whl/SzJ) were first exposed to total body irradiation (total irradiation dosage of 200 cGy), then treated with vehicle only (e.g., no T cells), unedited T cells, or edited CTX120 cells (e.g., TCRβ2MCAR+ T cells) as shown in Table 8. T cells were administered approximately 6 hours post radiation on day 1 in a 250 μL volume of phosphate-buffered saline (PBS) via an intravenous slow bolus injection. Radiation was delivered at a rate of 160 cGy/min.









TABLE 8







Design of an In Vivo Study to Evaluate GvHD Response to CTX120












T cell
T cell

Number of



dosage
concen-
Total
animals



(cells/
tration
irradiation
(Male/


Group
mouse)
(cells/mL)
dosage
Female)





Vehicle-no radiation
0
0
  0 cGy
5/5


Vehicle-radiation
0
0
200 cGy
15/15


Unedited T cells
1 × 107
4 × 107

15/15


CTX120 cells
4 × 107
8 × 107

15/15









Following treatment, the animals were evaluated for up to 84 days after radiation for survival, appearance of GvHD symptoms, and body weight. GvHD symptoms were defined as changes to the skin (e.g., pallor and/or redness), decreased activity, hunched back posture, slight to moderate thinness, and increased respiratory rate.


No mortality was observed in untreated animals or animals exposed to radiation alone or radiation combined with a dosage of CTX120 cells. However, significant mortality was observed for animals receiving radiation in combination with a dosage of unedited T cells as shown in FIG. 9. Additionally, weight loss was observed in several animals treated with unedited T cells, but not in animals treated with vehicle or CTX120 cells. Additionally, no GvHD symptoms were observed in animals treated with CTX120 cells. Thus, these results confirm that CTX120 cells edited to eliminate TCR-expressing cells do not induce off-target reactivity that results in a GvHD response.


Alloreactivity towards human cells was compared for unedited T cells and T cells edited to be TCR and β2M negative according to the gene-editing methods described in Example 1. Specifically, primary human T cells were electroporated with Cas9-sgRNA RNP complexes targeting the TRAC and β2M gene loci. However, the cells were not treated with rAAV encoding an anti-BCMA CAR, thus providing a population of cells comprising T cells with a disrupted TRAC and β2M gene (TRAC/β2M T cells) for use in evaluating the effect of a TCR knockout on alloreactivity.


To evaluate alloreactivity, unedited T cells or edited T cells were incubated with PBMCs that were derived from the same donor (e.g., autologous or matched PBMCs) or a different donor (e.g., allogeneic or unmatched PBMCs) and activation was evaluated by measuring T cell proliferation using a flow cytometry-based assay measuring incorporation of 5-ethynyl-2′-deoxyuridine (EdU: Invitrogen) according to the manufacturer's protocol. As a positive control, T cells were treated with phytohaemagglutinin-L (PHA) that functions to cross-link the TCR and induce T cell activation. Treatment with PHA resulted in robust proliferation in unedited T cells, but as expected, not in edited T cells that lack TCR expression (FIG. 10). Also, as expected, neither edited nor unedited T cells proliferated in the presence of autologous PBMCs. However, unedited T cells proliferate in the presence of allogenic PBMCs, indicating alloreactivity to unmatched human cells. In contrast, edited T cells demonstrated no proliferation in response to allogeneic PBMCs (FIG. 10). Thus, loss of TCR expression in edited T cells corresponds to lack of activation in response to unmatched human cells.


EXAMPLE 6
Daratumumab Treatment Depleted NK Cells While T Cell Numbers Remained Unaffected

Based on the expression levels of CD38 on NK and T cells, the effect of an anti-CD38 antibody, daratumumab (TAB-236, Creative Biolabs), on such cells was assessed. PBMCs from a healthy donor were cultured for 96 hours in media containing 0.01, 0.1, or 1 μg/mL of daratumumab. The effect of 10% complement on the cell cultures was also tested. Untreated cells and cells treated with 0.01, 0.1 or 1 μg/mL isotype control mAb (human IgG1k) (cat #403501, BioLegend) were used as controls. After 96 hours of culture, NK and T cell frequency and numbers were measured.


In vitro culture of daratumumab resulted in a dose-dependent decrease of NK cell frequency and numbers (FIGS. 11A-11B). At the highest dose tested, 1 μg/mL, daratumumab reduced NK cell numbers by approximately 75% after 96 hours. This effect is specific to daratumumab, as treatment with an isotype control mAb did not affect NK cell numbers. The reduction in NK cells is not complement dependent under these culture conditions, as the addition of 10% complement to the cell culture did not alter daratumumab's effect of NK cells.


In a second experiment PBMCs from a different donor, daratumumab reduced NK cell numbers ˜57% after only 72 hours (data not shown). These data demonstrate that daratumumab has similar effects on NK cells from different donor populations.


Contrary to its effect on NK cells, daratumumab did not affect T cell numbers or frequency (FIGS. 11C-11D). Although CD38 expression was detected on T cells and in vitro culture of PBMC resulted in upregulation of CD38 surface expression in T cells, T cell numbers were surprisingly unaffected by the addition of daratumumab to the culture media.


EXAMPLE 7
Daratumumab Treatment Does Not Affect CAR T Growth and Activation

To assess whether daratumumab treatment affects CAR T cells with a disrupted β2M gene, anti-BCMA CAR T cells generated in Example 1 were treated with daratumumab with or without 10% complement. After a 72 hour culture period, anti-BCMA CAR T cell numbers and frequency were measured in a flow cytometry assay as described in Example 3 (FIGS. 12A-12B). Although a majority (70.5%) of anti-BCMA CAR T cells expressed CD38, treatment with daratumumab, with or without 10% complement, did not affect anti-BCMA CAR T cells numbers or frequency.


It was also found that daratumumab treatment did not induce activation of CAR T cells.


EXAMPLE 8
Daratumumab Pre-Treatment Reduced NK Cell-Induced CAR T Cell Lysis

To determine if daratumumab blunts NK-cell mediated CAR T cell lysis, anti-BCMA CAR T cells were co-cultured with purified NK cells that were pre-treated for 60 hours with either daratumumab or isotype control mAb at concentrations of 0.01, 0.1, or 1 μg/mL (FIG. 13A). At the end of the 60 hour pre-treatment period, 50,000 efluor-labelled anti-BCMA CAR T cells were added to the plate containing 150,000 NK cells and Dara/isotype control, and incubated for an additional 24 hours. At the end of the 24-hour co-culture period, anti-BCMA CAR T cell lysis was measured in a cell-kill assay with DAPI.


Specifically, the anti-BCMA CAR T cells were labeled with 5 μM efluor670 (Cat #65-0840-90; ThermoFisher Scientific), washed and incubated in co-cultures with the NK cells at a 3:1 (NK:T) ratio. The co-culture was incubated 24 hr. After incubation, wells were washed and media was replaced with 150 μL of 1× FACS buffer containing a 1:500 dilution of 5 mg/mL DAPI (Molecular Probes) and 12.5 μL of CountBright beads (C36950; ThermoFisher Scientific). The cells were analyzed for cell viability by flow cytometry (i.e., viable cells being negative for DAPI staining) Pre-treatment with daratumumab resulted in a reduced anti-BCMA CAR T cell lysis in a dose-dependent manner (FIG. 13A). NK cells pretreated for 60 hours with 1 μg/mL daratumumab showed a 50% reduction in their ability to cause anti-BCMA CAR T cell lysis. This effect is daratumumab-specific, as anti-BCMA CAR T cells that were co-cultured with NK cells pretreated with isotype control mAb did not affect change in NK cell-mediated CAR T cell lysis.


EXAMPLE 9
Effect of High Concentrations and Increased Dosage of Daratumumab on NK and CAR T Cells

To determine if higher concentrations of daratumumab (10 μg/mL) activates CAR T cells and causes subsequent proliferation or activation-induced cell death, anti-BCMA CAR T cells deficient in B2M were cultured with daratumumab at concentrations of 0.1, 1 or 10 μg/mL for 24 hours. Untreated cells or anti-BCMA CAR T cells deficient in B2M treated with IgG1k isotype control mAb were used as controls. FIG. 14A demonstrates that increasing the concentration of daratumumab to 10 μg/mL did not significantly reduce B2M deficient CAR T cells numbers.


To determine if 10 μg/mL daratumumab blunts NK-cell mediated CAR T cell lysis, anti-BCMA CAR T cells deficient in B2M were co-cultured with purified NK cells that were pre-treated for 60 hours with either daratumumab or isotype control mAb at concentrations of 0.1, 1 or 10 μg/mL. Briefly, NK cells were plated at 50,000 or 150,000 cells per well and treated with daratumumab or the isotype control at concentrations of 0, 0.1, 1 and 10 μg/mL. After 60 hours of treatment of NK cells with daratumumab, the anti-BCMA CAR T cells were labeled with 5 μM efluor670 (Cat #65-0840-90; ThermoFisher Scientific), washed and seeded at 50,000 cells per well in co-cultures with the daratumumab-treated NK cells to make 1:1 or 3:1 (NK:T) ratio. The co-culture was incubated for further 24 hr. After incubation, wells were washed and media was replaced with 150 μL of 1× FACS buffer containing a 1:500 dilution of 5 mg/mL DAPI (Molecular Probes) and 12.5 μL of CountBright beads (C36950; ThermoFisher Scientific). The cells were analyzed for cell viability by flow cytometry (i.e., viable cells being negative for DAPI staining) Pre-treatment with daratumumab protected anti-BCMA CAR T cell from NK induced cell lysis in a dose-dependent manner (FIGS. 14B-14C). When CAR T cells were co-cultured with NK cells at a 1:1 ratio, pretreatment of the NK cells with 0.1 μg/mL daratumumab showed a maximal protective effect of 91% against anti-BCMA CAR T cell lysis (FIG. 14B). When the ratio of NK:CAR T cells increased to 3:1, daratumumab still produced a significant protective effect from NK cell lysis (85% protection) at a slightly higher dose of 1 μg/mL (FIG. 14C).


Daratumumab is prescribed in the clinic at a dose of 16 mg/kg (225 μg/mL equivalent). To determine the effect of daratumumab at high concentrations on NK and CAR T cells, anti-BCMA CAR T cells deficient in B2M were co-cultured with purified NK cells that were pre-treated for 60 hours with either human IgGl-k or daratumumab, each at concentrations of 0.01, 0.1, 1, 10, 100 or 300 μg/mL using methods as described in the previous examples. Flow cytometry was used to assess NK and CAR T cells numbers 72 hours after co-culturing with pre-treated NK cells using methods as described in the previous examples.



FIG. 15A demonstrates that increasing doses of daratumumab decreased NK cell number 72 hours after exposure. A 29% decrease in NK cells is seen after exposure with 1 μg/mL of daratumumab, while 300 μg/mL daratumumab results is a further 38% decrease in NK cells. In contrast, the BCMA CAR T cell numbers were unaffected by the high daratumumab concentrations (FIG. 15B).


EXAMPLE 10
Daratumumab Enhances the Anti-Tumor Activity of Anti-BCMA CAR-T Cells and Prolongs Survival in a Xenograft Mouse Model of Multiple Myeloma

The effect of combining daratumumab with anti-BCMA CAR-T cell treatment was tested in a subcutaneous MM.1S xenograft model in immunocompromised NOG mice (NOD.Cg-PrkdcscidIl2rgtm1Sug/JicTac). In brief, 5 to 8-week old female NOG mice were individually housed in ventilated microisolator cages and maintained under pathogen-free conditions. The animals each received a subcutaneous inoculation in the right flank of 5×106 MM.1S cells in 50% Matrigel. When the mean tumor volume reached 150 mm3 (approximately 125 to 175 mm3), the mice were randomized into groups with 5 mice per group. Tested groups included an untreated arm, daratumumab only treatment, anti-BCMA CAR-T cell only treatment (low dose or high dose), and anti-BCMA CAR-T cell (low dose or high dose) in combination with daratumumab. Anti-BCMA CAR-T cells were dosed by intravenous injection of 0.8×106 (low dose) or 2.4×106 (high dose) CAR+ T cells at day 0. Daratumumab was dosed IP at 15 mg/kg, twice weekly, starting 2 days prior to anti-BCMA CAR-T cell dosing.


Tumor volume and body weights were measured twice weekly and individual mice were euthanized when their tumor volume reached ≥2000 mm3. In both doses of anti-BCMA CAR-T cells tested, the highest efficacy in tumor inhibition was observed in the combination arm, as compared to each single arm treatment. Additionally, prolonged survival was observed in the combination arm, in both low dose (FIGS. 16A and 16B) and high dose (FIGS. 16C and 16D) anti-BCMA CAR-T cell treatments, compared to either single arm treatment of daratumumab or anti-BCMA CAR-T cells. Tumor volume at day 26 is shown in FIG. 16C. Animals treated with either high dose of anti-BCMA CAR-T cells or with daratumumab only, showed mean tumor volume of 1500 mm3 (1486 mm3 and 1475 mm3, respectively), while mean tumor volume in the combination arm showed a mean of 668 mm3 (FIG. 16C).


In sum, these results demonstrate that the combination of anti-BCMA CAR-T cells and daratumumab showed increase efficacy in both tumor inhibition and increased survival in a mouse model of multiple myeloma compared to either anti-BCMA CAR-T cells or daratumumab alone.


EXAMPL 11
Lenalidomide Showed Beneficial Effect on Multiple Aspects of BCMA Directed CAR-T Cells In-Vitro

Anti-BCMA CAR-T cells were used in this Example as exemplary CAR-T cell. The anti-BCMA CAR-T cells express an anti-BCMA CAR comprising the amino acid sequence of SEQ ID NO: 40, a disrupted TRAC gene having the anti-BCMA CAR coding sequence inserted, and a disrupted β2M gene.


The CAR-T cells were thawed and expanded in-vitro in the presence or absence of Lenalidomide. Multiple concentrations of Lenalidomide were added to the culture media, to evaluate the activity of Lenalidomide across a wide range of concentrations, from 0.5 uM to 10 uM. In all tested concentrations, Lenalidomide enhanced the proliferation of the anti-BCMA CAR-T cells, showing 5-30 fold higher expansion in the tested time period (FIG. 17A). The anti-BCMA CAR-T cells expanded in the presence of Lenalidomide showed decreased senescence as evident by reduced expression of CD57 in the cell population in all the tested concentrations of Lenalidomide (FIG. 17B, tested after 10 day culture with Lenalidomide).


In addition to enhancing CAR-T cell expansion, Lenalidomide enhanced effector cytokine secretion upon antigen stimulation in all the Lenalidomide concentrations tested. FIG. 17C shows the level of multiple cytokines following an overnight culture of the anti-BCMA CAR-T cells with a cell line which expresses low levels of BCMA (JeKo-1), at a ratio of 2:1 effector to target cell. Addition of Lenalidomide to the co-culture media led to enhanced cytokine secretion of multiple effector cytokines, among them IFN-γ and TNF-α following CAR-T cell engagement by the BCMA expressing target cell line (FIG. 1C).


EXAMPLE 12
Lenalidomide Enhanced BCMA Directed CAR-T Cell Activity In-Vivo in Mice

The effect of a combination treatment of the anti-BCMA CAR-T cells described in Example 1 above and Lenalidomide was tested in mice using an MM.1S subcutaneous tumor model. Mice were inoculated with MM.1S cells, and the tumor was allowed to reach a mean volume of 150 mm3. Once tumors reached target volume, mice were treated with:


a) 3 million anti-BCMA CAR-T cells,


b) Lenalidomide at a dose of 1.5 mg/kg daily for 21 days, followed by 3 days off and QD4 till end,


c) Lenalidomide at a dose of 10 mg/kg daily for 14 days, followed by 3 days off and QD4 till end,


d) combination of anti-BCMA CAR-T cells and Lenalidomide at a dose of 1.5 mg/kg using the schedule described in b, or


e) combination of anti-BCMA CAR-T cells and Lenalidomide at a dose of 10 mg/kg using the schedule described in c.


The effect of each treatment on tumor regression and mouse survival was monitored throughout the study. Single arm treatment of either the anti-BCMA CAR-T cells or Lenalidomide in both tested doses showed a minimal effect on both tumor regression and mouse survival compared to the no treatment arm. However, the combination arm showed a potent inhibition of tumor growth in both Lenalidomide doses tested, with complete tumor clearance of 5/5 mice in the low Lenalidomide dose, and 4/5 in the higher lenalidomide dose (FIG. 18A). This led to prolonged mouse survival in the combination arm, and while in the single treatment arms all mice were sacrificed due to reaching max tumor volume by day 32, in the combination arms 5/5 mice survived at day 64 in the low Lenalidomide dose, and 4/5 mice survived in the high Lenalidomide dose at day 64. FIG. 18B.


Examination of the anti-BCMA CAR-T cells expansion in peripheral blood, revealed that co-administration of Lenalidomide enhanced the expansion of the CAR-T cells following dosing in mice. Presence of human cells in mouse blood was evaluated using staining for human CD45+, and the number of human cells per ul of mouse blood was calculated using BD TruCount vials per manufacturer's protocol. Human T cells were quantified in mouse blood ˜1, 2 & 3 weeks after the CAR-T cells dosing to mice. Lenalidomide was found to significantly increase the numbers of the CAR-T cells in mouse blood in a dose dependent manner, 2 & 3 weeks after CAR-T dosing, with maximal increase from 10 cells/ul in the absence of Lenalidomide to ˜70 cells/ul in the presence of 10 mg/kg Lenalidomide, 2 weeks post dosing (FIG. 19C).


EXAMPLE 13
Lenalidomide Did Not Enhance Immune Recognition of Allogenic T Cells

Since Lenalidomide has been shown to have a co-stimulatory effect on T cells, and stimulate NK cells, the ability of allogenic T cells (B2M−/TRAC− cells) to stimulate immune recognition of allogenic cells was assessed. Two modes of allogenic immune recognition were tested: immune recognition of B2Mneg cells by NK cells, and immune recognition by allogenic T cells. Examination of the cytotoxic activity of NK cells towards B2Mneg cells was tested following overnight (ON) co-culture in varying concentrations of NK to T cells, and varying concentrations of Lenalidomide. Increasing concentration of NK to B2Mneg T cells led to an increase in the cytotoxic activity of NK cells towards B2Mneg T cells. Surprisingly, adding Lenalidomide in a wide range of concentrations did not lead to an increased cell killing of the B2Mneg T cells (FIG. 19A).


Additionally, cytokine secretion was tested at the end of the co-culture described above, following co-culture of NK cells with B2Mneg T cells, K562 cells (a B2Mneg cell line, commonly used as a positive control for activation of NK cells due to lack of B2M expression), and unedited T cells (used as a negative control for NK cells activation). Analysis of cytokine secretion following co-culture with NK cells, showed that several cytokines were upregulated upon co-culture of NK cells with K562 cells. This included cytokines previously shown to be upregulated upon NK cell activation, such as IL-6, MCP-1, IFN-γ and TNF-α. Upregulation of secretion of several cytokines has been observed upon addition of Lenalidomide to the co-culture of NK cell with K562, which is consistent with the known role of Lenalidomide in enhancing NK cell activation (FIG. 19B). However, when examining the cytokine secretion upon co-culture of NK cells with B2Mneg T cells, the levels of several cytokines were much lower compared to the co-culture with K562, and minimal changes were observed upon addition of Lenalidomide, in the concentrations tested (FIG. 19B). This indicated that, although in some case cytotoxic activity of NK cells can be enhanced in the presence of Lenalidomide, enhanced NK recognition of allo T cells does not seem to be a concern, following addition of Lenalidomide.


Next, allo-reactivity towards edited T cells (B2Mneg/TCRneg) was tested using an MLR assay (mixed lymphocyte reaction). In this assay PBMCs (“responder cells”) were mixed with irradiated auto or allo T cells (“stimulator cells”). At the end of the assay, the activation of the responder cells was evaluated by measuring the cell proliferation in the co-culture, with cell proliferation serving as a proxy for immune activation. In the assay shown in FIG. 19C, both auto (donor 1) & allo (donor 2 & 3) were evaluated for immune activation following co-culture. As shown in FIG. 19C, in the allo setting, proliferation was observed upon co-culture of unedited T cells with PBMCs from 2 individual donors. As expected, immune activation was reduced upon deletion of B2M & TRAC from the T cells, as evident by the reduced proliferation in both donors tested. Addition of Lenalidomide may in some cases enhance allo reactivity towards unedited T cells (see donor 2 panel FIG. 19C, unedited T cells, in the various Lenalidomide concentrations tested). However, the proliferation observed upon allo co-culture with edited T cells remained low, with minimal changes upon addition of Lenalidomide to the co-culture, indicating that the allo reactivity towards edited T cells was unaffected by the addition of Lenalidomide.


Taken together, results from this Example show that, unexpectedly, Lenalidomide did not enhance immune recognition of allogenic T cells.


EXAMPLE 14
BCMA Directed CAR-T Cells Produced in the Presence of Lenalidomide Exhibited Increased Cytokine Secretion Upon Antigen Stimulation

PBMCs were thawed and activated by T cell activation agents to enrich for T cells. After 3 days, T cells were edited for B2M and TRAC knock-out using a CRISPR/Cas gene editing system. An anti-BCMA expression cassette (as an exemplary CAR construct) was knocked into the TRAC locus to produce anti-BCMA CAR-T cells. Following the editing procedure, resulting T cells were expanded in the presence of absence of Lenalidomide in a concentration of 0.5, 2, & 10 uM for approximately 10 days. The resulting cells were later evaluated for cytokine secretion following antigen stimulation, in the absence of Lenalidomide.


Lenalidomide addition during production of the anti-BCMA CAR-T cells was found to enhance effector cytokine secretion upon antigen stimulation, in the absence of continued presence of Lenalidomide. FIG. 20 shows the level of multiple cytokines following an overnight culture of the CAR-T cells with a cell line which expresses low levels of BCMA (JeKo-1), at a ratio of 0.5:1 effector to target cell. The inclusion of Lenalidomide to the co-culture media led to enhanced cytokine secretion of multiple effector cytokines, among them IFN-γ and TNF-α, upon CAR-T engagement by the BCMA expressing target cell line (FIG. 20). This indicated that inclusion of Lenalidomide during the manufacturing process could serve as a means to not only enhance CAR-T cell proliferation, but also enhance the potency of the CAR-T cells, by programming them to a state with enhanced cytokine secretion upon antigen engagement.


EXAMPLE 15
Impact of Lenalidomide on CAR-T Cell Features

This Example investigates the effects of Lenalidomide on various CAR-T features.


Editing Efficiency


Editing efficiency, including TRAC−%, B2M−% and CAR+% were assessed at day 7/8 and/or day 13/14 with anti-BCMA CAR-T cells. FIG. 21A shows the CAR+%, TRAC−% and B2M−% from the anti-BCMA CAR-T cells on day 8. Anti-BCMA CAR-T cells were not harvested around day 14 due to slower growth rate. About 51-58% of CAR+%, 96% TRAC−% and 75-77% of B2M−% were detected from anti-BCMA CAR-T cells with or without Lenalidomide treatment.


CD4 and CD8 Ratio


CD4% and CD8% were assessed at day 7/8 and/or day 13/14 with the anti-BCMA CAR-T cells disclosed above. FIG. 21B shows CD4% and CD8% from the anti-BCMA CAR-T cells expanded at small and medium scale on day 8. The anti-BCMA CAR-T cells were not assessed and harvested around day 14 due to slower expansion. Compared with Lenalidomide untreated anti-BCMA CAR-T cells, there was dose-dependent increase of CD8 positive cells, ranging from 7-15%. However, the overall distribution of CD4 and CD8 cells was not significantly altered. Expansion scale (small or medium) didn't impact the CD4 and CD8 phonotype.


EXAMPLE 16
A Phase I Dose Escalation and Cohort Expansion Study of Safety and Efficacy of Anti-BCMA Allogenic CRISPR-Cas9-Engineered T Cells (CTX120) in Subjects with Relapsed or Refractory Multiple Myeloma

This study evaluates the safety, efficacy, pharmacokinetics, and pharmacodynamic effects of CTX120, an allogeneic chimeric antigen receptor (CAR) T cell therapy directed towards B cell maturation antigen (BCMA) in subjects with relapsed or refractory multiple myeloma. Multiple myeloma is a malignancy of terminally differentiated plasma cells in the bone marrow that represents about 10% of all hematologic malignancies and is the second most common hematologic malignancy after non-Hodgkin lymphoma (Kumar et al., Leukemia 31, 2443-2448; 2017; Rajkumar et al., Mayo Clin. Proc 91, 101-119; 2016). CTX120 is a BCMA-directed T cell immunotherapy comprised of allogeneic T cells that are genetically modified ex vivo using CRISPR-Cas9 gene editing components (sgRNA and Cas9 nuclease). The modifications include disruption of the T cell receptor alpha constant (TRAC) and beta-2 microglobulin (B2M) loci, and the simultaneous insertion of an anti-BCMA CAR transgene into the TRAC locus. The CAR is comprised of a humanized scFv specific for BCMA, followed by a CD8 hinge and transmembrane region that is fused to the intracellular signaling domains for CD137 (4-1BB) and CD3. The gene knockouts are intended to reduce the probability of GvHD, redirect the modified T cells towards BCMA-expressing tumor cells, and increase the persistence of the allogeneic cells.


CTX120 is prepared from healthy donor peripheral blood mononuclear cells obtained via a standard leukapheresis procedure. The mononuclear cells are enriched for T cells and activated with anti-CD3/CD28 antibody-coated beads, then electroporated with CRISPR-Cas9 ribonucleoprotein complexes, and transduced with a CAR gene-containing recombinant adeno-associated virus (AAV) vector. The modified T cells are expanded in cell culture, purified, formulated into a suspension, and cryopreserved. The product is to be stored onsite and thawed immediately prior to administration.


The specificity and antitumor cytotoxicity of CTX120 was assessed using in vitro and in vivo pharmacology studies. CTX120 cells released effector cytokines when cocultured with BCMA+ tumor cells in vitro and resulted in tumor cell death. CTX120 inhibited tumor growth in vivo in human tumor xenograft mouse models. In vitro and in vivo safety assessments were performed to assess the risk of immune reactivity and oncogenesis. No off-target edits were identified. Safety studies demonstrate that CTX120 does not cause any clinical or histopathological GvHD in mice and confirm that CTX120 cells do not grow in the absence of cytokines after gene editing.


1 Study Objectives

Primary objective, Part A (dose escalation): To assess the safety of escalating doses of CTX120 in combination with various lymphodepleting and immunomodulatory agents in subjects with relapsed or refractory multiple myeloma to determine the maximum tolerated dose (MTD) and/or recommended dose and regimen for Part B cohort expansion.


Primary objective, Part B (cohort expansion): To assess the efficacy of CTX120 in subjects with relapsed or refractory multiple myeloma, as measured by ORR according to International Myeloma Working Group (IMWG) response criteria (Kumar et al., 2016).


Secondary objectives (Parts A and B): To further characterize the efficacy, safety, and pharmacokinetics of CTX120 and evaluate the changes over time in patient-reported outcomes (PRO) associated with CTX120.


Exploratory objectives (Parts A and B): To identify biomarkers associated with CTX120 that may indicate or predict clinical response, resistance, safety, disease, or pharmacodynamic activity.


2 Subject Eligibility

2.1 Inclusion Criteria

    • 1. Age≥18 years
    • 2. Able to understand and comply with protocol-required study procedures and voluntarily sign a written informed consent document
    • 3. Diagnosis of multiple myeloma with relapsed or refractory disease, as defined by IMWG response criteria (Table 22), and at least 1 of the following:
      • a) Have had at least 2 prior lines of therapy, including an IMiD (e.g., lenalidomide, pomalidomide), PI (e.g., bortezomib, carfilzomib), and a CD38-directed monoclonal antibody (e.g., daratumumab; if approved and available in country/region)
      • b) Multiple myeloma that is double-reftartory or triple-refractory, defined as progression on or within 60 days of treatment with PI, IMiD, and anti-CD38 antibody or PI combination, as part of the same or different regimens
      • c) Multiple myeloma relapsed within 12 months after autologous SCT
      • d) Cohorts 1 and 3 only: At least 1 of the above criteria (3a, b, or c) and previously received a CD38-directed monoclonal antibody
      • e) Cohorts 2 and 3 only: At least 1 of the above criteria (3a, b, c) and previously received lenalidomide
    • 4. Measurable disease, including at least 1 of the following criteria:
      • Serum M-protein≥0.5 g/dL
      • Urine M-protein≥200 mg/24 hours
      • Serum free light chain (FLC) assay: Involved FLC level≥10 mg/dL (100 mg/L),


        provided serum FLC ratio is abnormal
    • 5. Eastern Cooperative Oncology Group (ECOG) performance status 0 or 1 (Table 21)
    • 6. Meets criteria to undergo LD chemotherapy and CAR T cell infusion (all cohorts), daratumumab infusion (Cohorts 1 and 3 only), and lenalidomide administration (Cohorts 2 and 3 only)
    • 7. Adequate organ function:
    • Renal: Estimated glomerular filtration rate>50 mL/min/1.73 m2
    • Liver: Aspartate transaminase or alanine transaminase<3×upper limit of normal (ULN); total bilirubin<2×ULN
    • Cardiac: Hemodynamically stable and left ventricular ejection fraction≥45% by


      echocardiogram
    • Pulmonary: Oxygen saturation level on room air>91% per pulse oximetry
    • 8. Female subjects of childbearing potential (postmenarcheal with an intact uterus and at least 1 ovary, who are less than 1 year postmenopausal) must agree to use acceptable method(s) of contraception from enrollment through at least 12 months after CTX120 infusion.
    • 9. Male subjects must agree to use effective contraception from enrollment through at least 12 months after CTX120 infusion.


2.2 Exclusion Criteria

    • 1. Prior allogeneic SCT
    • 2. Less than 60 days from autologous SCT at time of screening and with unresolved serious complications
    • 3. Plasma cell leukemia (>2.0×109/L circulating plasma cells by standard differential), or nonsecretory multiple myeloma, or Waldenström's macroglobulinemia or POEMS (polyneuropathy, organomegaly, endocrinopathy, monoclonal protein, and skin changes) syndrome, or amyloidosis with end organ involvement and damage
    • 4. Prior treatment with any of the following therapies:
    • Any gene therapy or genetically modified cell therapy, including CAR T cells or natural killer cells
    • Prior treatment with BCMA-directed therapy, including BCMA-directed antibody, bispecific T cell engager, or antibody-drug conjugate
    • Radiation therapy within 14 days of enrollment. Palliative radiation therapy for symptom management is permitted.
    • 5. Known contraindication to daratumumab (Cohorts 1 and 3), lenalidomide (Cohorts 2 and 3), cyclophosphamide, fludarabine, or any of the excipients of CTX120 product
    • 6. Evidence of direct central nervous system (CNS) involvement by multiple myeloma
    • 7. History or presence of clinically relevant CNS pathology such as a seizure disorder, cerebrovascular ischemia/hemorrhage, dementia, cerebellar disease, any autoimmune disease with CNS involvement, or another condition that may increase CAR T cell-related toxicities
    • 8. Unstable angina, clinically significant arrhythmia, or myocardial infarction within 6 months of enrollment
    • 9. Presence of bacterial, viral, or fungal infection that is uncontrolled or requires IV anti-infectives
    • 10. Positive for presence of human immunodeficiency virus (HIV) type 1 or 2, or active hepatitis B virus (HBV) or hepatitis C virus (HCV) infection. Subjects with prior history of HBV or HCV infection who have documented undetectable viral load (by quantitative polymerase chain reaction [PCR] or nucleic acid testing) are permitted. Infectious disease testing (HIV-1, HIV-2, HCV antibody and PCR, HBV surface antigen, HBV surface antibody, HBV core antibody) performed within 30 days of signing the informed consent form (ICF) may be considered for subject eligibility
    • 11. Previous or concurrent malignancy, except basal cell or squamous cell skin carcinoma, adequately resected and in situ carcinoma of cervix, or a previous malignancy that was completely resected and has been in remission for ≥5 years
    • 12. Received live vaccine within 28 days of enrollment
    • 13. Use of systemic antitumor therapy or investigational agent within 14 days prior to enrollment. Use of physiological doses of steroids (e.g., ≤10 mg/day prednisone or equivalent) will be permitted for subjects previously on steroids if clinically indicated
    • 14. Primary immunodeficiency disorder or active autoimmune disease requiring steroids and/or other immunosuppressive therapy
    • 15. Diagnosis of significant psychiatric disorder or other medical condition that could impede the subject's ability to participate in the study
    • 16. Women who are pregnant or breastfeeding


3 Study Design

3.1 Investigational Plan


This is an open-label, multicenter, Phase 1 study evaluating the safety and efficacy of escalating doses of CTX120 in combination with various LD and immunomodulatory agents in subjects with relapsed or refractory multiple myeloma (Table 9). The study is divided into 2 parts: dose escalation and evaluation of different LD regimens (Part A) followed by cohort expansion (Part B).


In Part A, dose escalation begins in adult subjects with 1 of the following: relapsed or refractory multiple myeloma after at least 2 prior lines of therapy, including an IMiD, PI, and CD38-directed monoclonal antibody (where approved/available); progressive multiple myeloma that is double refratrory to IMiD and PI combination or triple-refractory to PI, IMiD, and anti-CD38 antibody, defined as progression on or within 60 days of treatment; or multiple myeloma relapsed within 12 months after autologous SCT. Dose escalation will be performed according to the criteria outlined herein. Based on available data from Part A, a dose level and regimen from 1 or 2 cohorts (Cohorts 1, 2, or 3) is selected for Part B cohort expansion.


In Part B, each expansion cohort is enrolled in 2 stages. In the first stage, up to 27 subjects are enrolled and treated with the recommended dose and regimen of CTX120 for the respective Part B expansion cohort (at or below the MTD determined in Part A). One interim analysis is planned for each expansion cohort when subjects enrolled in the first stage have 3 months of evaluable disease response assessment data.


3.1.1 Study Design


During both dose escalation (Part A) followed by cohort expansion (Part B), the study consists of 3 main stages as follows:


Stage 1: Screening to determine eligibility for treatment (1-2 weeks).


Stage 2: Treatment (Stage 2A and Stage 2B); see Table 9 for treatment by cohort (1-2 weeks)


Stage 3: Follow-up for all cohorts (5 years)


Part A investigates escalating doses of CTX120 in multiple independent cohorts (Cohorts 1, 2, and 3). These cohorts allow preliminary evaluation of the safety and pharmacokinetics of CTX120 when used with different LD and immunomodulatory agents, as summarized in the following Table 9. Subjects may receive an additional dose of CTX120 based on disease response criteria and eligibility, as described herein.









TABLE 9







Part A Dose Cohorts








Cohort
Treatment (Stage 2A + Stage 2B)





1
Stage 2A



Daratumumab administration:



16 mg/kg administered via IV infusion4 within 3 days prior to starting LD



chemotherapy and no more than 14 days prior to CTX120 infusion.



For subjects who achieve stable disease or better on Day 28, up to 5 additional



monthly doses of daratumumab (16 mg/kg IV) continue unless disease



progression or unacceptable toxicity occurs.



LD chemotherapy: Co-administration of fludarabine 30 mg/m2 +



cyclophosphamide 300 mg/m2 IV daily for 3 days.1 Both agents are started or



the same day and administered for 3 consecutive days. LD chemotherapy



must be completed at least 48 hours (but no more than 7 days) prior to



CTX120 infusion.



Stage 2B



Administered at least 48 hours (but no more than 7 days) after completion



of the 3-day course of LD chemotherapy.2,3


2
Stage 2A



LD chemotherapy: Co-administration of fludarabine 30 mg/m2 +



cyclophosphamide 300 mg/m2 IV daily for 3 days.1 Both agents are started



on the same day and administered for 3 consecutive days. LD chemotherapy



must be completed at least 48 hours (but no more than 7 days) prior to



CTX120 infusion.



Lenalidomide administration:



10 mg administered orally once daily for 21 days beginning on the



third day of LD chemotherapy, continuing through CTX120 infusion.



For subjects who achieve stable disease or better on Day 28 post-



CTX120 infusion and have met all criteria described herein, a 28-day cycle (21



days on and 7 days off) of 5 mg lenalidomide administration continues for



up to 5 additional cycles unless disease progression or unacceptable



toxicity occurs.



Stage 2B



Administered at least 48 hours (but no more than 7 days) after completion



of the 3-day course of LD chemotherapy.2,3


3
Stage 2A



Daratumumab administration:



16 mg/kg administered via IV infusion4 within 3 days prior to



starting LD chemotherapy and no more than 14 days prior to CTX120



infusion.



For subjects who achieve stable disease or better on Day 28, up to 5



additional monthly doses of daratumumab (16 mg/kg IV) may continue



unless disease progression or unacceptable toxicity occurs.



LD chemotherapy: Co-administration of fludarabine 30 mg/m2 +



cyclophosphamide 300 mg/m2 IV daily for 3 days.1 Both agents are started



on the same day and administered for 3 consecutive days.



LD chemotherapy must be completed



at least 48 hours (but no more than 7 days) prior to CTX120 infusion.



Lenalidomide administration:



10 mg administered orally once daily for 21 days beginning on the



third day of LD chemotherapy, continuing through CTX120 infusion.



For subjects who achieve stable disease or better on Day 28 post-



CTX120 infusion and have met all criteria described herein, a 28-day cycle



(21 days on and 7 days off) of 5 mg lenalidomide administration may



continue for up to 5 additional cycles unless disease progression or



unacceptable toxicity occurs.



Stage 2B



Administered at least 48 hours (but no more than 7 days) after completion



of the 3-day course of LD chemotherapy.2,3





DL1: Dose Level 1; IV: intravenous(ly); LD: lymphodepleting.



1In Cohorts 2-3, cyclophosphamide may be used at a dose of up to 500 mg/m2 IV for LD chemotherapy. Dose escalation rules and staggering would apply.




2An additional planned dose of CTX120 with LD chemotherapy may be administered 4 to 12 weeks after first CTX120 infusion(s) to subjects who achieve stable disease or better response (based on IMWG criteria) at the Day 28 assessment after first CTX120 infusion(s). The additional dose may be administered without LD chemotherapy if the subject is experiencing significant cytopenias.




3In all cohorts, a subject may receive an additional dose of CTX120 with LD chemotherapy after progressed disease if that subject had a prior response (PR or better response based on IMWG criteria).




4Where approved, daratumumab may be administered as a subcutaneous injection (1800 mg/30,000 units of hyaluronidase-fihj) per local prescribing information rather than an IV infusion.



Note:


Subjects should meet the criteria specified herein prior to both the initiation of LD chemotherapy and infusion of CTX120 (all cohorts) and should meet criteria specified herein for redosing prior to receiving any additional doses of CTX120. For Cohorts 1 and 3, criteria for LD chemotherapy should be confirmed prior to infusion of daratumumab.






The treatment regimens for Cohorts 1-3 are illustrated in FIGS. 22-24.


In the dose escalation part of the study, CTX120 infusion may begin at Dose Level 1 (DL1). In some instances, DL3 or DL4 may be used.


During the post-CTX120 infusion period, subjects are monitored for acute toxicities, including CRS, neurotoxicity, GvHD, and other adverse events (AEs). Toxicity management guidelines are provided herein. During Part A (dose escalation), all subjects are hospitalized for observation for the first 7 days following CTX120 infusion. In Parts A and B, the length of hospitalization for observation may be extended where required by local regulation or site practice. In both Parts A and B, subjects must remain within proximity of the investigative site (i.e., 1-hour transit time) for 28 days after CTX120 infusion.


After the acute observation period, subjects are followed for up to 5 years after CTX120 infusion with physical exams, regular laboratory and disease assessments, and AE evaluations. After completion of this study, all subjects are asked to participate in a separate long-term follow-up study for an additional 10 years to assess long-term safety and survival.


Alternative Lymphodepletion Regimens


Part A (dose escalation) seeks to identify an optimal LD regimen for cohort expansion in Part B. The LD regimen refers to both the LD chemotherapy regimen (i.e., fludarabine and cyclophosphamide) and immunomodulatory agents (i.e., daratumumab and lenalidomide) that may be administered to induce an immune environment amenable to allogeneic CAR T cells. The Part A cohorts are designed to explore 2 different dose levels of cyclophosphamide in the LD chemotherapy regimen and also the addition of daratumumab (Cohort 1), lenalidomide (Cohort 2), or both (Cohort 3) to the LD regimen.


Additional subjects may be enrolled into a Part A cohort under an alternative LD regimen. For example, the higher dose of cyclophosphamide may be used for Cohorts 1 and 2. Dose escalation rules (3+3 design and DLT evaluation) and staggering apply for any subjects enrolled into a cohort with a new LD regimen, as described herein.


3.1.2 Study Subjects


Approximately 6 to 78 subjects in total are treated in Part A (dose escalation). Approximately 70 subjects are to be treated in Part B (cohort expansion).


3.1.3 Study Duration


Subjects participate in this study for 5 years. After completion of this study, all subjects are asked to participate in a separate long-term follow-up study for an additional 10 years to assess long-term safety and survival.


3.2 CTX120 Dose Escalation


Dose escalation is performed using a standard 3+3 design in which 3 or 6 subjects are enrolled at each dose level depending on the occurrence of DLT, as defined herein. The DLT evaluation period begins with the first CTX120 infusion and lasts for 28 days.


Table 10 lists the CAR+ T cell doses of CTX120, based on the total number of CAR+ T cells that may be evaluated in this study, beginning with DL1 and escalate when application, for example, to DL3 or DL4.









TABLE 10







Dose Escalation of CTX120










Dose Level
Total CART T Cell Dose







−1 (de-escalation)
2.5 × 107



1
  5 × 107



2
1.5 × 108



3
4.5 × 108



4
  7.5 × 108 *



5
 1.05 × 109 *







CAR: chimeric antigen receptor.



* A lower dose level consisting of 6 × 108 CARP T cells may be used for deescalation from Dose Level 4. Likewise, a dose level of 9 × 108 CARP+ T cells may be used for de-escalation from Dose Level 5.






Dose escalation is performed according to the following rules:

    • If 0 of 3 subjects experience a DLT, escalate to the next dose level.
    • If 1 of 3 subjects experiences a DLT, expand the current dose level to 6 subjects.
      • If 1 of 6 subjects experiences a DLT, escalate to the next dose level.
      • If ≥2 of 6 subjects experience a DLT:
        • If in DL-1, evaluate alternative dosing schema or declare inability to determine recommended dose for Part B cohort expansion.
        • If in DL1, de-escalate to DL-1.
        • If in DL2, DL3, DL4, or DL5 declare previous dose level the MTD.
    • If ≥2 of 3 subjects experience a DLT:
      • If in DL-1, evaluate alternative dosing schema or declare inability to determine the recommended dose for Part B cohort expansion.
      • If in DL1, decrease to DL-1.
      • If in DL2, DL3, DL4, or DL5 declare previous dose level the MTD.
    • No dose escalation beyond highest dose listed in Table 10.


3.2.1 Maximum Tolerated Dose Definition


The MTD is the highest dose for which DLTs are observed in less than 33% of subjects. An MTD may not be determined in this study. A decision to move to the Part B expansion cohort may be made in the absence of an MTD provided the dose is at or below the maximum dose studied in Part A of the study.


3.2.2 DLT Definitions


Toxicities are graded and documented according to National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) Version 5, with the following exceptions:

    • CRS:
      • American Society for Transplantation and Cellular Therapy (ASTCT) criteria (Lee et al., Biol Blood Marrow Transplant 25, 625-638; 2019)
    • Neurotoxicity, Parts A and B:
      • CTCAE v5.0
      • Immune effector cell-associated neurotoxicity syndrome (ICANS) criteria (Lee et al., 2019)
    • GvHD, Parts A and B:
      • Mount Sinai Acute GvHD International Consortium (MAGIC) criteria (Harris et al., Biol Blood Marrow Transplant 22, 4-10; 2016)


AEs that have no evidence to suggest a plausible causal relationship with CTX120 are not considered DLTs.


A DLT is defined as any of the following CTX120-related events occurring during the DLT evaluation period that persists beyond the specified duration (relative to the time of onset):

    • A. Grade 4 CRS
    • B. Grade 3 or 4 neurotoxicity (based on ICANS criteria)
    • C. Grade≥2 GvHD that is steroid-refractory (e.g., progressive disease after 3 days of steroid treatment [e.g., 1 mg/kg/day], or having no response after 7 days of treatment)
    • D. Death during the DLT period (except due to disease progression)
    • E. Any CTX120-related grade≥3 vital organ toxicity (e.g., pulmonary, cardiac) of any duration, except as listed below.


The following are NOT considered as DLTs:

    • 1. Grade 3 CRS that improves to grade≤2 within 72 hours
    • 2. Grade≤3 tumor lysis syndrome lasting <7 days
    • 3. Grade 3 or 4 fever
    • 4. Grade≥3 allergic reaction improving to grade≤2 within 48 hours of instituting supportive care
    • 5. Grade 3 fatigue lasting <7 days
    • 6. Bleeding in the setting of thrombocytopenia (platelet count<50×109/L); documented bacterial infections or fever in the setting of neutropenia (absolute neutrophil count [ANC]<1000/mm3)
    • 7. Hypogammaglobulinemia
    • 8. Grade 3 or 4 liver function studies that improve to grade≤2 within 7 days
    • 9. Grade 3 or 4 renal insufficiency that improves to grade≤2 within 7 days
    • 10. Grade 3 or 4 cardiac arrythmia that improves to grade≤2 within 48 hours
    • 11. Grade 3 pulmonary toxicity that resolves to grade≤2 within 72 hours. Grade 3 events that are isolated, CTX120-related, and not secondary to supportive treatment as part of CRS will be considered DLTs
    • 12. Grade 3 or 4 thrombocytopenia or neutropenia will be assessed retrospectively. After at least 6 subjects are infused, if ≥50% of subjects have prolonged cytopenias (i.e., lasting more than 28 days postinfusion). Grade≥3 cytopenias that were present at the start of LD chemotherapy may not be considered DLTs.


3.3 CTX120 Redosing (Part A+Part B)


As allogeneic CAR T cells may be susceptible to more rapid clearance than autologous CAR T cells upon lymphocyte recovery, it therefore may be necessary to administer more than a single dose to clear any remaining cancerous cells. In order to achieve greater responses and prolonged durability, redosing may be applied to subjects that do not experience significant toxicity following the first infusion.


3.3.1 Redosing with CTX120


Up to 4 doses of CTX120 per subject may be allowed. Redosing may be permitted in 2 scenarios:


1. Planned redosing with or without LD chemotherapy based on disease response criteria


2. Redosing of CTX120 with LD chemotherapy after progressed disease (PD) if the subject has had an initial objective response after the first CTX120 infusion


To be redosed with CTX120, subjects must meet the redosing criteria and repeat screening assessments, as specified herein.


Subjects who are eligible for redosing, as described above, may be redosed with an LD regimen that is different from the LD regimen administered prior to their initial treatment, if the alternative LD regimen has been cleared in a Part A cohort at the CTX120 dose level, and after consultation with the medical monitor.


3.3.2 Planned Redosing (All Cohorts)


Subjects who responded to the initial CTX120 infusion (stable disease [SD] or better response based on IMWG criteria) with evidence of residual myeloma cells (e.g., minimal residual disease [MRD] positivity, PET-avid lesions) at Day 28 may receive an additional planned CTX120 infusion 4 to 12 weeks after the prior CTX120 infusion. In subjects with significant cytopenias (ANC<1000/μL and/or platelets<25,000/μL), redose may be performed without LD chemotherapy.


For planned redosing, subjects must meet the following criteria:

    • No prior DLT during dose escalation (if applicable)
    • No prior grade≥3 CRS without resolution to grade≤2 within 72 hours following CTX120 infusion; no ongoing CRS of any grade
    • No prior GvHD following CTX120 infusion
    • No prior grade≥2 ICANS following CTX120 infusion; no ongoing ICANS of any grade
    • Meet initial study inclusion criteria and all exclusion criteria except prior treatment with CTX120


Additional redosing criteria at the time of LD chemotherapy and prior to additional CTX120 infusion are as follows.

    • ECOG performance status 0 or 1
    • No requirement for supplemental oxygen to maintain a saturation level>91%
    • No new uncontrolled cardiac arrhythmia
    • No hypotension requiring vasopressor support or fluid bolus
    • No active uncontrolled infection (positive blood cultures for bacteria, fungus, or virus not responding to treatment)
    • Renal: Estimated glomerular filtration rate>50 mL/min/1.73 m2
    • Liver: AST or ALT<3×ULN; total bilirubin<1.5×ULN
    • No worsening of clinical status compared to prior CTX120 infusion that places the subject at increased risk of toxicity
    • No new neurological symptoms suggesting CNS disease involvement
    • Women who are pregnant or breastfeeding are not eligible for redosing


Subjects suitable for redosing should also meet additional safety criteria for LD chemotherapy, if applicable, and for CTX120 dosing as disclosed herein. Subjects who are redosed should be followed per the schedule of assessments (Table 19), consistent with the initial dosing with the following considerations:

    • Echocardiogram (unless new cardiac signs or symptoms) is not required within 3 months of initial CTX120 dose
    • The following disease assessments should be performed prior to redosing with CTX120:
      • Monoclonal protein (serum and urine; see relevant disclosures herein) within 1 week prior to redosing
      • Whole body PET/CT (for subjects with extramedullary disease) within 4 weeks prior to redosing
      • Bone marrow aspirate biopsy within 4 weeks prior to redosing
    • In subjects without extramedullary disease, or if whole body PET/CT is not performed, brain MRI should be performed prior to redosing with CTX120 if clinical suspicion or patient history of CNS involvement.


3.3.3 Redosing After Progressive Disease (All Cohorts)


For all cohorts, a subject may be redosed with CTX120 after PD if the subject has had an initial objective response (PR or better based on IMWG) after the first CTX120 infusion. The additional dose may be administered up to 15 months after the previous CTX120 infusion. Redosing with lymphodepleting chemotherapy in subjects with grade 3 or 4 neutropenia or thrombocytopenia who are >8 weeks post previous CTX120 infusion will not be permitted unless the cytopenias can be clearly attributed to PD or other reversible cause. Redosing without LD may be considered, after consultation with the medical monitor.


To be redosed with CTX120, subjects must meet the criteria disclosed in the above section. Subjects who are redosed should be followed per the schedule of assessments (Table 19), consistent with the initial dosing. Subjects who undergo redosing after PD receives a CTX120 dose that is at or below the highest dose cleared in Part A. Subjects who are eligible for redosing, as described above, may be redosed with an LD regimen that is different from the LD regimen administered prior to their initial treatment, if the alternative LD regimen has been cleared in a Part A cohort at the CTX120 dose level.


In subjects who undergo redosing prior to PD, disease response assessments are to be based on the baseline myeloma disease assessment performed during initial screening. For subjects who are redosed after PD, disease response is assessed relative to the most recent myeloma assessment prior to redosing.


4 Study Treatment

4.1. Lymphodepleting Chemotherapy


All subjects receive LD chemotherapy prior to each infusion of CTX120, except for subjects experiencing significant cytopenias prior to redosing with CTX120 in all cohorts.


The LD chemotherapy may consist of:

    • Fludarabine 30 mg/m2 IV daily for 3 doses and
    • Cyclophosphamide 300 mg/m2 IV


Alternatively, the LD chemotherapy may consist of:

    • Fludarabine 30 mg/m2 IV daily for 3 doses and
    • Cyclophosphamide 500 mg/m2 IV daily for 3 doses


Both agents are started on the same day and administered for 3 consecutive days for all cohorts. Subjects should start LD chemotherapy within 7 days of study enrollment. Adult subjects with moderate impairment of renal function (creatinine clearance [CrCl] 30-70 mL/min/1.73 m2) should have a 20% dose reduction of fludarabine and be monitored closely per the applicable prescribing information.


Both LD chemotherapy agents are started on the same day and administered for 3 consecutive days. Subjects should start LD chemotherapy within 7 days of study enrollment. For subjects in all cohorts, LD chemotherapy is delayed if any of the following signs or symptoms are present:

    • Significant worsening of clinical status that increases the potential risk of AEs associated with LD chemotherapy
    • Requirement for supplemental oxygen to maintain a saturation level>91%
    • New uncontrolled cardiac arrhythmia
    • Hypotension requiring vasopressor support
    • Active infection: Positive blood cultures for bacteria, fungus, or virus not responding to treatment
    • Neurotoxicity known to increase risk of ICANS (e.g., seizures, stroke, change in mental status). Neurotoxicity of benign origin (e.g., headache), lasting less than 48 hours and considered reversible will be allowed.


Additional criteria to be met for LD chemotherapy prior to redosing are specified herein.


During Part A (dose escalation), if LD chemotherapy is delayed more than 30 days or the subject starts anticancer therapy, the subject is replaced. During Part B, subjects with a >30 day delay in receiving LD chemotherapy may be replaced. Subjects whose toxicity(ies) are driven by underlying disease and require anticancer therapy must subsequently meet disease eligibility criteria, treatment washout, and end organ function criteria before restarting LD chemotherapy. Additionally, any subject who receives anticancer therapy after enrollment must have disease evaluation performed prior to starting LD chemotherapy (Cohort 2) or daratumumab (Cohorts 1 and 3).


4.2. Administration of CTX120


CTX120 consists of allogeneic T cells modified with CRISPR-Cas9, resuspended in cryopreservative solution (CryoStor CS-5), and supplied in a 6-mL infusion vial. A flat dose of CTX120 (based on number of CAR+ T cells) is administered as a single IV infusion. The total dose may be contained in multiple vials. Infusion should preferably occur through a central venous catheter. A leukocyte filter must not be used.


Prior to the start of CTX120 infusion, the site pharmacy must ensure that 2 doses of tocilizumab and emergency equipment are available for each specific subject treated. Subjects should be premedicated per the site standard of practice with acetaminophen PO (i.e., paracetamol or its equivalent per site formulary) and diphenhydramine hydrochloride IV or PO (or another H1-antihistamine per site formulary) approximately 30-60 minutes prior to CTX120 infusion. Prophylactic systemic corticosteroids should not be administered, as they may interfere with the activity of CTX120.


There is a dose limit of 7×104 TCR+ cells/kg imposed for all dose levels. Based on the percentage of CAR+ T cells in the CTX120 lot to be administered, enrollment at higher dose levels (e.g., DL4 or DL5) may be restricted to subjects with a minimum weight to ensure the TCR cell limit is not exceeded. Medications that may be discontinued are provided herein. For all cohorts, each CTX120 infusion is delayed if any of the following signs or symptoms are present:

    • New active uncontrolled infection
    • Worsening of clinical status compared to prior to start of LD chemotherapy that places the subject at increased risk of toxicity
    • Neurotoxicity known to increase risk of ICANS (e.g., seizures, stroke, change in mental status). Neurotoxicity of benign origin (e.g., headache) lasting less than 48 hours and considered reversible is allowed.


Each CTX120 infusion is administered at least 48 hours (but no more than 7 days) after the completion of LD chemotherapy. If CTX120 infusion is delayed by more than 10 days, LD chemotherapy must be repeated.


4.2.1 CTX120 Post-Infusion Monitoring


Following CTX120 infusion, subjects' vital signs should be monitored every 30 minutes for 2 hours after infusion or until resolution of any potential clinical symptoms. Subjects in Part A are hospitalized for observation for a minimum of 7 days after CTX120 infusion. Postinfusion hospitalization in Part B is considered based on the safety information obtained during dose escalation and may be performed. In Part B, hospitalization for observation can be considered. In Parts A and B, the length of hospitalization for observation may be extended where required by local regulation or site practice. In both Parts A and B, subjects must remain in proximity of the investigative site (i.e., 1-hour transit time) for at least 28 days after CTX120 infusion. Management of acute CTX120-related toxicities should occur at the study site.


Subjects are monitored for signs of CRS, tumor lysis syndrome (TLS), neurotoxicity, GvHD, and other AEs according to the schedule of assessments (Table 19 and Table 20). Guidelines for the management of CAR T cell-related toxicities are described herein. Subjects should remain hospitalized until CTX120-related nonhematologic toxicities (e.g., fever, hypotension, hypoxia, ongoing neurological toxicity) return to grade 1.


4.3 Daratumumab Administration


Subjects in Cohorts 1 and 3 receive 1 dose of daratumumab (an anti-CD38 monoclonal antibody) 16 mg/kg by IV infusion within 3 days prior to starting LD chemotherapy and within 14 days of CTX120 infusion. For subjects who achieve SD or better on Day 28, up to 5 additional monthly doses of daratumumab (16 mg/kg IV) continues unless disease progression or unacceptable toxicity occurs. Daratumumab administration (including pre- and postinfusion medications, preparation, infusion rates, and postinfusion monitoring) is performed according to the local prescribing information. To facilitate administration, the first 16 mg/kg IV dose may be split to 8 mg/kg over 2 consecutive days.


Disease response is assessed in accordance with IMWG response criteria (Kumar et al., 2016) before repeat dosing with daratumumab. For the first 6 subjects who receive daratumumab at Month 2 and Month 3, the subjects should be monitored for signs of CRS and HLH in the first 7 to 10 days (e.g., every 48 to 72 hours) following each infusion. Daratumumab infusion should be delayed, and discussed with the medical monitor prior to proceeding, if platelets are <25,000/μL (unless transfusion support is planned), as well as for rising ferritin, lactate dehydrogenase, and C-reactive protein (CRP) levels that may be concerning for signs of CRS or HLH. If a subject experiences severe AEs related to daratumumab, redosing with daratumumab is not permitted.


Where approved and available, daratumumab may be administered as a subcutaneous injection (1800 mg/30,000 units of hyaluronidase-fihj), per local prescribing information, rather than as an intravenous infusion.


4.3.1 Daratumumab Infusion Reactions


To reduce the risk of infusion reactions with daratumumab IV, 1 to 3 hours prior to infusion subjects are premedicated with corticosteroids (e.g., IV methylprednisolone 100 mg or equivalent); following the second infusion, the dose of corticosteroid may be reduced [oral or IV methylprednisolone 60 mg], antipyretics (e.g., oral acetaminophen [paracetamol] 650-1000 mg, or equivalent), and antihistamines (e.g., oral or IV diphenhydramine hydrochloride [or another Hi-antihistamine] 25-50 mg, or equivalent).


Subjects are monitored frequently during the entire infusion. For infusion reactions of any grade/severity, infusion is interrupted immediately, and symptoms managed. Permanent discontinuation of therapy if an anaphylactic reaction or life-threatening (grade 4) reaction occurs, and institution of appropriate emergency care. For subjects with grade 1, 2, or 3 reactions, after symptom resolution, the infusion rate is reduced when restarting the infusion, as described in the approved prescribing information or per site practice.


To reduce the risk of delayed infusion reactions, oral corticosteroids (20 mg methylprednisolone or equivalent dose of an intermediate-acting or long-acting corticosteroid in accordance with local standards) are administered to subjects following infusion, per local prescribing information.


For subjects who receive additional monthly doses of daratumumab, only intermediate-acting corticosteroids (e.g., prednisone, methylprednisolone) should be used to reduce the risk of interference with CTX120.


If a subject has an unresolved event of infusion reaction after daratumumab treatment, LD chemotherapy should be delayed and discussed with the medical monitor prior to proceeding.


4.3.2 Additional Considerations


Daratumumab has been associated with herpes zoster (2%) and hepatitis B (1%) reactivation in patients with multiple myeloma. To prevent herpes zoster reactivation, initiate antiviral prophylaxis within 1 week after infusion and continue for 3 months following treatment as per local guidelines. For subjects with latent hepatitis B, consider hepatitis B prophylaxis prior to initiation of daratumumab and for 3 months following treatment (King et al., 2018).


Daratumumab binds to CD38 on red blood cells and results in a positive indirect antiglobulin test (indirect Coombs test). Typing and screening of blood occurs per the approved prescribing information to prevent interference with blood compatibility testing.


Estimated daratumumab plasma concentration after a single dose or 3 consecutive doses is shown in FIG. 25.


4.4. Lenalidomide Therapy


Subjects in Cohorts 2 and 3 receive lenalidomide 10 mg daily for 21 days beginning on the third day of LD chemotherapy (Cycle 1). Lenalidomide should be stopped if a subject develops grade≥3 CRS, grade≥2 ICANS, acute kidney injury (CrCl<30 mL/min), or any other toxicity thought to be related to lenalidomide and is unacceptable.


At Day 28 post-CTX120 infusion, subjects who achieve stable disease or better should restart lenalidomide at 5 mg (21 days on and 7 days off), if ANC≥1000/μL and platelets≥30,000/μL, and continue for 5 more cycles unless disease progression or unacceptable toxicity occurs. If at Day 28, counts are below the threshold for restarting lenalidomide therapy, complete blood count (CBC) is repeated weekly until the threshold to restart is met. If despite supportive care (e.g., granulocyte colony-stimulating factor [G-CSF]), subject's CBC does not reach the count threshold to restart lenalidomide by 6 weeks post-CTX120 infusion, maintenance may start at a later time point. Lenalidomide may be increased to 10 mg for the maintenance cycles if the subject tolerates it.


Subjects should take lenalidomide orally at about the same time each day, with or without food. Refer to lenalidomide local prescribing information for additional guidance and for general risks associated with lenalidomide.


4.4.1 Monitoring for Cytopenia


As per prescribing information, CBC should be checked weekly during the second cycle of lenalidomide, then performed at greater intervals as per local practice. Specifically, for subjects in Cohorts 2 and 3 with SD or better at Day 28, samples are collected weekly to monitor cytopenia resolution (platelet count≥30,000/μL, ANC≥1000/μL) prior to starting additional cycles of lenalidomide, and continue CBC monitoring after restarting lenalidomide per the prescribing information or local practice (e.g., every 7 days [weekly] for Cycle 2; on Days 1 and 15 of Cycle 3; and every 28 days [4 weeks] thereafter). Thromboprophylaxis is recommended for subjects with platelet count≥50,000/μL and a history of prior thromboembolic event.


4.5. Daratumumab Infusion and Lenalidomide Therapy


Subjects in Cohort 3 receive daratumumab (as in Cohort 1), as 1 dose of 16 mg/kg by IV infusion within 3 days prior to the start of LD chemotherapy and within 14 days of CTX120 infusion. Lenalidomide is administered (as in Cohort 2), as 10 mg daily for 21 days beginning on the third day of LD chemotherapy (Cycle 1). The 5 additional monthly doses of daratumumab (16 mg/kg IV) and 5 additional monthly cycles of lenalidomide at 5 mg (21 days on and 7 days off) for subjects who achieve SD or better on Day 28 may or may not be administered in Cohort 3 based on emerging clinical and pharmacokinetics data.


The goal of administering daratumumab and lenalidomide both in Cohort 3 is to deepen and prolong the immunosuppressive and/or immunomodulatory effects achieved with LD chemotherapy alone or LD chemotherapy with either daratumumab or lenalidomide alone. As described above, daratumumab is a monoclonal antibody that suppresses specific T, B, myeloid-derived suppressor, and NK cell subpopulations while lenalidomide is an immunomodulatory drug that potentiates T cell functionality and alters the suppressive microenvironment. Administration of both agents could induce an immune environment even more amenable to expansion, persistence, and function of allogeneic CAR T cells than either agent alone.


The safety profile of co-administered daratumumab and lenalidomide has been shown to be consistent with the safety profile of each agent administered separately (Bahlis et al., Leukemia 34, 1875-1884; 2020; Dimopoulos et al., New England Journal of Medicine 375, 1319-1331; 2016; Facon et al., New England Journal of Medicine 380, 2104-2115; 2019). Assessments and procedures related to daratumumab and lenalidomide safety will be performed in Cohort 3 as described for Cohorts 1 and 2 (Table 19).


4.6 Prior and Concomitant Medications


4.6.1 Allowed Medications


Necessary supportive measures for optimal medical care are given throughout the study, including IV antibiotics to treat infections, growth factors, blood components, and bone-directed therapies (including zoledronic acid or denosumab), except for prohibited medications listed herein.


All concurrent therapies, including prescription and nonprescription medication, and medical procedures must be recorded from the date of signed informed consent through 3 months after CTX120 infusion. Beginning 3 months post-CTX120 infusion, only the following selected concomitant medications will be collected: IV immunoglobulins, vaccinations, anticancer treatments (e.g., chemotherapy, radiation, immunotherapy), immunosuppressants (including steroids), bone-directed therapies, and any investigational agents.


4.6.2 Prohibited Medications


The following medications are prohibited during certain periods of the study as specified below:

    • Corticosteroid therapy at a pharmacologic dose (>10 mg/day of prednisone or equivalent doses of other corticosteroids) and other immunosuppressive drugs should be avoided after CTX120 administration unless medically indicated to treat new toxicity or as part of management of CRS or neurotoxicity associated with CTX120. Use of corticosteroids before and after daratumumab infusion is permitted to prevent infusion reactions.
    • Granulocyte-macrophage colony-stimulating factor (GM-CSF) following CTX120 infusion due to the potential to worsen symptoms of CRS.
    • Care should be taken with administration of G-CSF following CTX120, and requires discussion with medical monitor during dose escalation.
    • Live vaccine within 28 days of enrollment to 3 months following CTX120 infusion.
    • Any anticancer therapy (e.g., chemotherapy, immunotherapy, targeted therapy, radiation, or other investigational agents) other than daratumumab (Cohorts 1 and 3), lenalidomide (Cohorts 2 and 3), or LD chemotherapy (all cohorts) prior to disease progression. Palliative radiation therapy for symptom management is permitted depending on extent, dose, and site(s). Site(s), dose, and extent should be defined and reported to the medical monitor for determination.


5. Toxicity Management

5.1. General Guidance


Prior to LD chemotherapy, infection prophylaxis (e.g., antiviral, antibacterial, antifungal agents) should be initiated according to institutional standard of care for multiple myeloma patients in an immunocompromised setting.


Subjects must be closely monitored for at least 28 days after CTX120 infusion. Significant toxicities have been reported with autologous CAR T cell therapies. Although this is a first-in-human study and the clinical safety profile of CTX120 has not been described, the following general recommendations are provided based on prior experience with autologous CD19 and BCMA CAR T cell therapies:

    • Fever is the most common early manifestation of CRS; however, subjects may also experience weakness, hypotension, or confusion as first presentation.
    • Diagnosis of CRS should be based on clinical symptoms and NOT laboratory values.
    • In subjects who do not respond to CRS-specific management, always consider sepsis and resistant infections. Subjects should be continually evaluated for resistant or emergent bacterial infections, as well as fungal or viral infections.
    • CRS, HLH, and TLS may occur at the same time following CAR T cell infusion. Subjects should be consistently monitored for signs and symptoms of all the conditions and managed appropriately.
    • Neurotoxicity may occur at the time of CRS, during CRS resolution, or following resolution of CRS. Grading and management of neurotoxicity will be performed separately from CRS.
    • Tocilizumab must be administered within 2 hours from the time of order.


In addition to toxicities observed with autologous CAR T cells, signs of GvHD are monitored closely due to the allogeneic nature of CTX120.


The safety profile of CTX120 is continually assessed throughout the study. For Cohorts 1-3, refer to local prescribing information for other general risks associated with daratumumab and lenalidomide.


5.2. Toxicity-Specific Guidance


5.2.1. Infusion Reactions


Infusion reactions have been reported in autologous CAR T cell trials, including transient fever, chills, and/or nausea. If an infusion reaction occurs, acetaminophen (paracetamol) and diphenhydramine hydrochloride (or another H1-antihistamine) may be repeated every 6 hours after CTX120 infusion.


Nonsteroidal anti-inflammatory medications may be prescribed as needed if the subject continues to have fever not relieved by acetaminophen. Systemic steroids should NOT be administered except in cases of life-threatening emergency, as this intervention may have a deleterious effect on CAR T cells. Infusion reactions have also been reported for daratumumab.


5.2.2. Febrile Reaction and Infection Prophylaxis


Infection prophylaxis should occur according to the institutional standard of care for multiple myeloma patients in an immunocompromised setting. In the event of febrile reaction, an evaluation for infection should be initiated and the subject managed appropriately with antibiotics, fluids, and other supportive care as medically indicated and determined by the treating physician. Viral and fungal infections should be considered throughout a subject's medical management if fever persists. If a subject develops sepsis or systemic bacteremia following CTX120 infusion, appropriate cultures and medical management should be initiated. Additionally, consideration of CRS should be given in any instances of fever following CTX120 infusion within 30 days postinfusion.


5.2.3. Tumor Lysis Syndrome


Subjects receiving CAR T cell therapy may be at increased risk of TLS. Subjects should be closely monitored for TLS via laboratory assessments and symptoms from the start of LD chemotherapy until 28 days following CTX120 infusion.


Subjects at increased risk of TLS should receive prophylactic allopurinol (or a non-allopurinol alternative such as febuxostat) and increased oral/IV hydration during screening and before initiation of LD chemotherapy. Prophylaxis can be stopped after 28 days following CTX120 infusion or once the risk of TLS passes.


Sites should monitor and treat TLS as per their institutional standard of care, or according to published guidelines (Cairo et al., Br J Haematol 127, 3-11; 2004). TLS management, including administration of rasburicase, should be instituted promptly when clinically indicated.


5.2.4. Cytokine Release Syndrome


CRS is a major toxicity reported with autologous CAR T cell therapy and has also been observed in early phase studies with allogeneic CAR T cell therapy (Benjamin et al., American Society of Hematology Annual Meeting (San Diego, Calif.); 2018). CRS is due to hyperactivation of the immune system in response to CAR engagement of the target antigen, resulting in multi-cytokine elevation from rapid T cell stimulation and proliferation (Frey et al., Blood 124, 2296; 2014; Maude et al., Cancer J 20, 119-122; 2014). When cytokines are released, a variety of clinical signs and symptoms associated with CRS may occur, including cardiac, gastrointestinal (GI), neurological, respiratory (dyspnea, hypoxia), skin, cardiovascular (hypotension, tachycardia), and constitutional (fever, rigors, sweating, anorexia, headaches, malaise, fatigue, arthralgia, nausea, and vomiting) symptoms, and laboratory (coagulation, renal, and hepatic) abnormalities.


The goal of CRS management is to prevent life-threatening sequelae while preserving the potential for the anticancer effects of CTX120. Symptoms usually occur 1 to 14 days after autologous CAR T cell therapy, but the timing of symptom onset has not been fully defined for allogeneic BCMA CAR T cells.


CRS should be identified and treated based on clinical presentation and not laboratory cytokine measurements. If CRS is suspected, grading should be applied according to the 2019 ASTCT (formerly known as American Society for Blood and Marrow Transplantation) consensus recommendations (Table 11) (Lee et al., Biol Blood Marrow Transplant 25, 625-638; 2019), and management should be performed according to the recommendations in Table 12, which are adapted from published guidelines (Lee et al., Blood 124, 188-195; 2014; Lee et al., 2019).


At the time of the original protocol version (V1.0), the established 2014 Lee criteria for CRS grading were applied (Lee et al., 2014). However, this has been updated to the ASTCT criteria (Lee et al., 2019), which have become the worldwide standard for CRS grading, initially for Part B only (protocol Version 3.0) and later for both parts of the study (protocol Version 5.0).


Neurotoxicity is graded and managed as described herein. End organ toxicity in the context of CRS management (Lee et al., 2019) refers only to hepatic and renal systems (as in the Penn Grading criteria) (Porter et al., J Hematol Oncol 11, 35; 2018).









TABLE 11







ASTCT Cytokine Release Syndrome Grading Criteria











CRS






Parameter
Grade 1
Grade 2
Grade 3
Grade 4





Fever 1
Temperature
Temperature
Temperature
Temperature



≥38° C.
≥38° C.
≥38° C.
≥38° C.


With
None
Not
Requiring a
Requiring


hypotension

requiring
vasopressor
multiple




vasopressors
with
vasopressors





or without
(excluding





vasopressin
vasopressin)


And/or
None
Requiring
Requiring high-
Requiring


hypoxia 2

low-
flow nasal
positive




flow nasal
cannula,3
pressure (e.g.,




cannula3 or
facemask,
CPAP, BiPAP,




blow-by
nonrebreather
intubation,





mask, or
and





Venturi
mechanical





mask
ventilation)





ASTCT: American Society for Transplantation and Cellular Therapy;


BiPAP: bilevel positive airway pressure;


C: Celsius;


CPAP: continuous positive airway pressure;


CRS: cytokine release syndrome;


CTCAE: Common Terminology Criteria for Adverse Events.


Note:


Organ toxicities associated with CRS may be graded according to CTCAE v5.0 but do not influenceCRS grading.



1 Fever is defined as temperature ≥38° C. not attributable to any other cause. In subjects who have CRS then receive antipyretics or anticytokine therapy such as tocilizumab or steroids, fever is no longer required to grade subsequent CRS severity. In this case, CRS grading is driven by hypotension and/or hypoxia.




2 CRS grade is determined by the more severe event: hypotension or hypoxia not attributable to any other cause. For example, a subject with temperature of 39.5° C., hypotension requiring 1 vasopressor, and hypoxia requiring low-flow nasal cannula is classified as grade 3 CRS.




3Low-flow nasal cannula is defined as oxygen delivered at ≤6 L/minute. Low-flow also includes blow-by oxygen delivery, sometimes used in pediatrics. High-flow nasal cannula is defined as oxygen delivered at >6 L/minute.














TABLE 12







Cytokine Release Syndrome Grading and Management Guidance









CRS Severity 1
Tocilizumab
Corticosteroids





Grade 1
Tocilizumab 2
N/A



may be considered




following routine practice



Grade 2
Administer
If no improvement



tocilizumab 8 mg/kg
within 24 hours



IV over 1 hour
after starting



(not to exceed 800
tocilizumab, administer



mg).2
methylprednisolone



Repeat tocilizumab
1 mg/kg IV twice



every 8 hours as
daily.



needed if not responsive
Continue corticosteroid



to IV fluids
use until the



or increasing
event is



supplemental oxygen.
grade ≤1, then taper



Limit to ≤3 doses in a 24-hour
over 3 days.



period; maximum total of 4




doses.



Grade 3
Per grade 2.
Per grade 2.


Grade 4
Per grade 2.
Per grade 2.



If no response to




multiple doses of




tocilizumab and




steroids, consider




using other




anticytokine therapies




(e.g., siltuximab, anakinra).





CRS: cytokine release syndrome;


IV: intravenously;


N/A: not applicable.



1 See (Lee et al., 2019).




2 Refer to tocilizumab prescribing information.







Throughout the duration of CRS, subjects should be provided with supportive care consisting of antipyretics, IV fluids, and oxygen. Subjects who experience grade≥2 CRS (e.g., hypotension, or hypoxia requiring supplemental oxygenation) should be monitored with continuous cardiac telemetry and pulse oximetry. For subjects experiencing grade 3 CRS, consider performing an echocardiogram to assess cardiac function. For grade 3 or 4 CRS, consider intensive care supportive therapy. Intubation for airway protection due to neurotoxicity (e.g., seizure) and not due to hypoxia should not be captured as grade 4 CRS. Similarly, prolonged intubation due to neurotoxicity without other signs of CRS (e.g., hypoxia) is not considered grade 4 CRS. The potential of an underlying infection may be considered in cases of severe CRS, as the presentation (fever, hypotension, hypoxia) is similar. Resolution of CRS is defined as resolution of fever (temperature≥38° C.), hypoxia, and hypotension (Lee et al., 2019).









TABLE 13







High-dose Vasopressors








Pressor
Dose*












Norepinephrine monotherapy
≥20
μg/min


Dopamine monotherapy
≥10
μg/kg/min


Phenylephrine monotherapy
≥200
μg/min


Epinephrine monotherapy
≥10
μg/min








If on vasopressin
Vasopressin + norepinephrine



equivalent



of ≥10 μg/min**


If on combination
Norepinephrine equivalent


vasopressors (not vasopressin)
of ≥20 μg/min**





*All doses are required for ≥3 hours.


**VASST Trial vasopressor equivalent equation: norepinephrine equivalent dose = [norepinephrine (μg/min)] + [dopamine (μg/min)/2] + [epinephrine (μg/min)] + [phenylephrine (μg/min)/10]






5.2.5. Neurotoxicity


Lumbar puncture is required for any grade≥3 neurotoxicity and is strongly recommended for grade 1 and grade 2 events, if clinically feasible. Lumbar puncture must be performed within 48 hours of symptom onset, unless not clinically feasible.


Viral encephalitis (e.g., HHV-6 encephalitis) must be considered in the differential diagnosis for subjects who experience neurocognitive symptoms after receiving CTX120. Whenever lumbar puncture is performed, in addition to the standard panel performed at site (which should include at least cell count, Gram stain, and Neisseria meningitidis), the following viral panel must be performed: CSF PCR analysis for HSV-1 and -2, enterovirus, varicella zoster virus, cytomegalovirus (CMV), and HHV-6.


Results from the infectious disease panel must be available within 5 business days of the lumbar puncture in order to appropriately manage the subject.


Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS)


Neurotoxicity has been observed with autologous CAR T cell therapies. It may occur at the time of CRS, during the resolution of CRS, or following resolution of CRS, and its pathophysiology is unclear. The ASTCT consensus recommendations further defined neurotoxicity associated with CRS as ICANS, a disorder characterized by a pathologic process involving the CNS following any immune therapy that results in activation or engagement of endogenous or infused T cells and/or other immune effector cells (Lee et al., 2019).


Signs and symptoms can be progressive and may include aphasia, altered level of consciousness, impairment of cognitive skills, motor weakness, seizures, and cerebral edema. ICANS grading (Table 14) was developed based on CAR T cell-therapy-associated TOXicity (CARTOX) working group criteria used previously in autologous CAR T cell trials (Neelapu et al., Nat Rev Clin. Oncol 15, 47-62; 2018). ICANS incorporates assessment of level of consciousness, presence/absence of seizures, motor findings, presence/absence of cerebral edema, and overall assessment of neurologic domains by using a modified tool called the ICE (immune effector cell-associated encephalopathy) assessment tool (Table 15).


Evaluation of any new onset neurotoxicity should include a neurological examination (including ICE assessment tool, Table 15), brain magnetic resonance imaging (MRI), and examination of the CSF (via lumbar puncture) as clinically indicated. Infectious etiology should be ruled out by performing a lumbar puncture whenever possible (especially for subjects with Grade 3 or 4 ICANS). If a brain MRI is not possible, all subjects should receive a noncontrast CT scan to rule out intracerebral hemorrhage. Electroencephalogram should also be considered as clinically indicated. Endotracheal intubation may be needed for airway protection in severe cases.


Nonsedating, antiseizure prophylaxis (e.g., levetiracetam) should be considered, especially in subjects with a history of seizures, for at least 21 days following CTX120 infusion or upon resolution of neurological symptoms (unless the antiseizure medication is considered to be contributing to the detrimental symptoms). Subjects who experience grade≥2 ICANS should be monitored with continuous cardiac telemetry and pulse oximetry. For severe or life-threatening neurologic toxicities, intensive care supportive therapy should be provided. Neurology consultation should always be considered. Monitor platelets and for signs of coagulopathy and transfuse blood products appropriately to diminish risk of intracerebral hemorrhage. Table 14 provides neurotoxicity grading, Table 16 provides management guidance, and Table 15 provides neurocognitive assessment performed using the ICE assessment. In addition to treatment guidelines provided in Table 16, nonsteroidal agents (e.g., anakinra, etc.) may be considered for ICANS management (Neill et al., Pract Neurol doi: 10.1136/practneurol-2020-002550; 2020).


For subjects who receive active steroid management for more than 3 days, antifungal and antiviral prophylaxis is recommended to mitigate a risk of severe infection with prolonged steroid use. Consideration for antimicrobial prophylaxis should also be given.









TABLE 14







ICANS Grading











Neurotoxicity






Domain
Grade 1
Grade 2
Grade 3
Grade 4





ICE score 1
7-9
3-6
0-2
0 (subject is






unarousable and






unable to undergo






ICE assessment)


Depressed
Awakens
Awakens
Awakens
Subject is


level of
spon-
to voice
only to
unarousable or


con-
taneously

tactile
requires vigorous


sciousness 2


stimulus
or repetitive






tactile






stimuli to arise;






stupor or coma


Seizure
N/A
N/A
Any clinical
Life-threatening





seizure,
prolonged seizure





focal or
(>5 min) or





generalized,
repetitive clinical





that resolves
or electrical





rapidly, or
seizures without





nonconvulsive
return to baseline





seizures
in between





on EEG that






resolve with






intervention



Motor
N/A
N/A
N/A
Deep focal


findings 3



motor






weakness such






as hemiparesis






or paraparesis


Elevated
N/A
N/A
Focal/local
Diffuse cerebral


ICP/


edema
edema on


cerebral


on
neuroimaging,


edema


neuro-
decerebrate or





imaging 4
decorticate






posturing, cranial






nerve VI palsy,






papilledema,






or Cushing's






triad





CTCAE: Common Terminology Criteria for Adverse Events;


EEG: electroencephalogram;


ICANS: immune effector cell-associated neurotoxicity syndrome;


ICE: immune effector cell-associated encephalopathy (assessment tool);


ICP: intracranial pressure;


N/A: not applicable.


Note:


ICANS grade is determined by the most severe event (ICE score, level of consciousness, seizure, motor findings, raised ICP/cerebral edema) not attributable to any other cause.



1 A subject with an ICE score of 0 may be classified as grade 3 ICANS if awake with global aphasia, but a subject with an ICE score of 0 may be classified as grade 4 ICANS if unarousable (Table 15 for ICE assessment tool).




2 Depressed level of consciousness should be attributable to no other cause (e.g., sedating medication).




3 Tremors and myoclonus associated with immune effector therapies should be graded according to CTCAE v5.0 but do not influence ICANS grading.




4 Intracranial hemorrhage with or without associated edema is not considered a neurotoxicity feature and is excluded from ICANS grading. It may be graded according to CTCAE v5.0.














TABLE 15







ICE Assessment










Assessment
Maximum


Domain

Score





Orientation
Orientation to year, month, city, hospital
4 points


Naming
Name 3 objects (e.g., point to clock, pen, button)
3 points


Following
Ability to follow commands
1 point 


command
(e.g., “Show me 2 fingers”




or “Close your eyes and stick out your tongue”)



Writing
Ability to write a standard sentence
1 point 



(includes a noun and verb)



Attention
Ability to count backward from 100 by 10
1 point 





ICE score will be reported as the total number of points (0-10) across all assessments.













TABLE 16







ICANS Management Guidance








Severity
Management





Grade 1
Provide supportive care per institutional practice.


Grade 2
Consider administering dexamethasone



10 mg IV every 6 hours (or equivalent



methylprednisolone) unless subject already



on equivalent dose of steroids for



CRS.



Continue dexamethasone use until event



is grade ≤1, then taper over 3 days.


Grade 3
Administer dexamethasone 10 mg IV



every 6 hours, unless subject already on



equivalent dose of steroids for CRS.



Continue dexamethasone use until event



is grade ≤1, then taper over 3 days.


Grade 4
Administer methylprednisolone 1000 mg



IV per day for 3 days; if improves,



then manage as above.





CRS: cytokine release syndrome;


ICANS: immune effector cell-associated neurotoxicity syndrome;


IV: intravenously.






Headache, which may occur in a setting of fever or after chemotherapy, is a nonspecific symptom. Headache alone may not necessarily be a manifestation of ICANS and further evaluation should be performed. Weakness or balance problem resulting from deconditioning and muscle loss are excluded from definition of ICANS. Similarly, intracranial hemorrhage with or without associated edema may occur due to coagulopathies in these subjects and are also excluded from definition of ICANS. These and other neurotoxicities should be captured in accordance with CTCAE v5.0.


Human Herpes Virus 6 Encephalitis


Most humans are exposed to HHV-6 during childhood and seroprevalence can approach 100% in adults. HHV-6 is thought to remain clinically latent in most individuals after primary infections and to reactivate to cause disease in persons with severe immunosuppression (Agut et al., Clin Microbiol Rev 28, 313-335; 2015; Hanson et al., Front Immunol 9, 1454; 2018). Two types of HHV-6 (A and B) have been identified. Although no diseases have clearly been linked to HHV-6A infection, HHV-6B is responsible for the childhood disease exanthem subitem. The virus also exhibits neurotropism and persists in brain tissue in a latent form. HHV-6 encephalitis has been predominantly described in immunocompromised patients following allogeneic HSCT, and has also been described in immunocompromised patients receiving autologous CAR T cell therapies (Bhanushali et al., Neurology 80, 1494-1500; 2013; Hanson et al., 2018; Hill et al., Curr Opin Virol 9, 53-60; 2014). Based on data from allogeneic HSCT, immunocompromised patients who are treated with steroids are at higher risk of developing HHV-6 encephalitis.


Diagnosis of HHV-6 encephalitis should be considered in any immunocompromised subject with neurological symptoms (e.g., confusion, memory loss, seizures) following CTX120 infusion. In addition to brain MRI, the following samples are required for diagnostic tests: lumbar puncture for HHV-6 DNA PCR (should be performed within 48 hours of symptoms if clinically feasible) and blood (plasma preferred) for HHV-6 DNA PCR. Diagnosis of HHV-6 encephalitis should be considered in a subject with elevated CSF HHV-6 DNA detected by PCR, elevated blood (plasma preferred) HHV-6 DNA detected by PCR, and acute mental status findings (encephalopathy), or short-term memory loss, or seizures (Hill and Zerr, 2014). Associated brain MRI abnormalities (typically, but not exclusively, non-enhancing, hyperintense lesions in the medial temporal lobes, especially hippocampus and amygdala) may not be seen initially (Ward et al., Haematologica 104, 2155-2163; 2019). Because brain MRI findings may not be present initially, treatment for HHV-6 encephalitis should be considered in the setting of neurological findings and high HHV-6 CSF viral load. CSF protein and cell count often may be unremarkable, although there may be mild protein elevation and mild pleocytosis. Subjects may also experience fever and/or rash (Ward et al., 2019).


In subjects diagnosed with HHV-6 encephalitis, treatment with ganciclovir or foscarnet should be initiated. Drug selection should be dictated by the drug's side effects, the subject's comorbidities, and the site's clinical practice. The recommended duration of therapy is 3 weeks or as per site clinical practice (Hill and Zerr, 2014; Ward et al., 2019).


Once treatment is initiated, peripheral blood HHV-6 viral load should be checked weekly by PCR. Decrease in blood viral load should be seen within 1 to 2 weeks after initiation of treatment. If viral load does not decrease following 1 to 2 weeks of treatment, switching to another antiviral agent (ganciclovir or foscarnet) should be considered. Antiviral therapy should be continued for at least 3 weeks and until PCR testing demonstrates clearance of HHV-6 DNA in blood. At the end of the therapy, lumbar puncture should be performed to confirm clearance of HHV-6 DNA in CSF. If possible, immunosuppressive medications (including steroids) should be reduced during treatment for HHV-6 encephalitis; however, this needs to be balanced with the subject's need for steroids, especially if ICANS is also suspected.


For subjects in whom HHV-6 encephalitis is suspected, retrospective assessment of HHV-6 IgG, IgM, and HHV-6 DNA by PCR should be performed from blood samples collected prior to CTX120 infusion, if available.


In subjects with consistently elevated HHV-6 DNA viral load (e.g., >10,000 copies/mL), and especially when viral load does not decrease following initiation of antiviral therapy, attempt should be made to distinguish HHV-6 reactivation from chromosomally integrated HHV-6 (CIHHV-6). If the site has capabilities to do so, CIHHV-6 can be confirmed by evidence of 1 copy of viral DNA/cellular genome, or viral DNA in hair follicles/nails, or by fluorescence in situ hybridization demonstrating HHV-6 integrated into a human chromosome. In suspected end-organ disease, if biopsy occurs, tissue from the affected organ should be tested for HHV-6 infection by culture, immunochemistry, in situ hybridization, or reverse transcription PCR for mRNA, if the site is able to perform these.


5.2.6. Hemophagocytic Lymphohistiocytosis


Hemophagocytic lymphohistiocytosis has been reported after treatment with autologous CAR T cells (Barrett et al., Curr Opin Pediatr 26, 43-49; 2014; Maude et al., N Engl J Med 371, 1507-1517; 2014; Maude et al., Blood 125, 4017-4023; 2015; Porter et al., 2015; Teachey et al., Blood 121, 5154-5157; 2013) and is included in the boxed warning of the prescribing information for idecabtagene vicleucel, the BCMA-directed autologous CAR T cell therapy for multiple myeloma (ABECMA USPI, 2021). HLH is a clinical syndrome that is a result of an inflammatory response following infusion of CAR T cells in which cytokine production from activated T cells leads to excessive macrophage activation. HLH may also be associated with malignancy, and has been reported for lymphoma, MM, and other cancers (Jordan et al., Blood 118, 4041-4052; 2011; La Rosée, 2015). Signs and symptoms of HLH may include fevers, cytopenias, hepatosplenomegaly, hepatic dysfunction with hyperbilirubinemia, coagulopathy with significantly decreased fibrinogen, and marked elevations in ferritin and CRP. Neurologic findings have also been observed (Jordan et al., 2011; La Rosée, 2015).


CRS and HLH may possess similar clinical syndromes with overlapping clinical features and pathophysiology. If attributed to CAR T toxicity, signs and symptoms of HLH are not graded separately. HLH will likely occur at the time of CRS or as CRS is resolving. HLH should be considered if there are unexplained elevated liver function tests or cytopenias with or without other evidence of CRS. Monitoring of CRP and ferritin may assist with diagnosis and define the clinical course. If these laboratory values further support a diagnosis of HLH, CD25 blood levels should be determined in conjunction with a bone marrow biopsy and aspirate, if safe to conduct, for further confirmation. Where feasible, excess bone marrow samples should be sent to a central laboratory.


If HLH is suspected:

    • Frequently monitor coagulation parameters, including fibrinogen. These tests may be done more frequently than indicated in the schedule of assessments, and frequency should be driven based on laboratory findings.
    • Fibrinogen should be maintained ≥100 mg/dL to decrease risk of bleeding.
    • Coagulopathy should be corrected with blood products.
    • Check for soluble CD25 and triglycerides.
    • If possible, perform bone marrow biopsy to assess for hemophagocytosis.
    • Given the overlap with CRS, subjects should also be managed per CRS treatment guidance in Table 12. Anakinra or other anti-cytokine therapies (e.g., emapalumab) may also be considered following discussion with the medical monitor.


5.2.7. Cytopenias


Grade 3 neutropenia and thrombocytopenia, at times lasting more than 28 days after CAR T cell infusion, have been reported in subjects treated with autologous CAR T cell products (Kymriah USPI, 2017; Raje et al., N Engl J Med 380, 1726-1737; 2019; Yescarta USPI, 2017). Therefore, subjects receiving CTX120 should be monitored for such toxicities and appropriately supported. Monitor platelets and for signs of coagulopathy and transfuse blood products appropriately to diminish risk of hemorrhage. Consideration should be given to antimicrobial and antifungal prophylaxis for any subject with prolonged neutropenia. For subjects experiencing grade≥3 neutropenia, thrombocytopenia, or anemia that has not resolved within 28 days of CTX120 infusion, a CBC with differential should be performed weekly until resolution to grade≤2.


During dose escalation, G-CSF may be considered in cases of grade 3 or 4 neutropenia post-CTX120 infusion. During dose expansion G-CSF may be administered. Antimicrobial and antifungal prophylaxis should be considered for any subject with prolonged neutropenia or on high doses of steroids.


For Cohorts 1 and 3, daratumumab may increase neutropenia and/or thrombocytopenia induced by background therapy. Complete blood cell counts should be monitored periodically during treatment according to the local prescribing information for background therapies. Subjects with neutropenia should be monitored for signs of infection. Daratumumab dose delay may be required to allow recovery of neutrophils and/or platelets, per local prescribing information. Consider supportive care with growth factors for neutropenia or transfusions for thrombocytopenia.


For Cohorts 2 and 3, lenalidomide can cause significant neutropenia and thrombocytopenia. In the multiple myeloma maintenance therapy trials, grade 3 or 4 neutropenia was reported in up to 59% of lenalidomide-treated subjects, and grade 3 or 4 thrombocytopenia in up to 38% of lenalidomide-treated subjects, as noted in the lenalidomide prescribing information. Subjects with neutropenia should be monitored for signs of infection. Subjects should be advised to observe for bleeding or bruising, especially with use of concomitant medication that may increase risk of bleeding. Subjects taking lenalidomide should have their CBCs assessed periodically, as described in the local prescribing information. Lenalidomide dose delay may be required to allow recovery of neutrophils and/or platelets, as per prescribing information, or otherwise specified in other places herein.


5.2.8. Graft Versus Host Disease


GvHD is seen in the setting of allogeneic SCT and is the result of immunocompetent donor T cells (the graft) recognizing the recipient (the host) as foreign. The subsequent immune response activates donor T cells to attack the recipient to eliminate foreign antigen-bearing cells. GvHD is divided into acute, chronic, and overlap syndromes based on both the time from allogeneic SCT and clinical manifestations. Signs of acute GvHD may include a maculopapular rash; hyperbilirubinemia with jaundice due to damage to the small bile ducts, leading to cholestasis; nausea, vomiting, and anorexia; and watery or bloody diarrhea and cramping abdominal pain (Zeiser et al., N Engl J Med 377, 2167-2179, 2017).


To support the proposed clinical study, a 12-week nonclinical Good Laboratory Practice-compliant GvHD and tolerability study was performed in immunocompromised mice treated with a single IV dose of 4×107 CTX120 cells per mouse (approximately 1.6×109 cells/kg). This dose level exceeds the proposed highest clinical dose by more than 100-fold when normalized for body weight. CTX120 did not induce clinical GvHD in immunocompromised (NSG) mice during the course of the 12-week study.


Further, due to the specificity of CAR insertion at the TRAC locus, it is highly unlikely for a T cell to be both CAR+ and TCR+. Remaining TCR+ cells are removed during the manufacturing process by immunoaffinity chromatography on an anti-TCR antibody column to achieve ≤0.15% TCR+ cells in the final product. A dose limit of 7×104 TCR+ cells/kg will be imposed for all dose levels. This limit is lower than the limit of 1×105 TCR+ cells/kg based on published reports on the number of allogeneic cells capable of causing severe GvHD during SCT with haploidentical donors (Bertaina et al., Blood 124, 822-826; 2014). Through this specific editing, purification, and strict product release criteria, the risk of GvHD following CTX120 should be low, although the true incidence is unknown. Subjects should be monitored closely for signs of acute GvHD following infusion of CTX120. The timing of potential symptoms is unknown. However, given that CAR T cell expansion is antigen-driven and will likely occur only in TCR cells, it is unlikely that the number of TCR+ cells will appreciably increase above the number infused.


Diagnosis and grading of GvHD should be based on the published MAGIC criteria (Harris et al., Biol Blood Marrow Transplant 22, 4-10; 2016), as outlined in Table 17.









TABLE 17







Criteria for Grading Acute GvHD












Skin






(active


Lower GI



erythema
Liver

(stool


Stage
only)
(bilirubin)
Upper GI
output/day)





0
No active
<2
No or
<500 mL/day or



(erythematous)
mg/dL
intermittent
<3 episodes/day



GvHD rash

nausea,






vomiting, or






anorexia



1
Maculopapular
2-3
Persistent
500-999



rash
mg/dL
nausea,
mL/day or



<25% BSA

vomiting, or
3-4 episodes/day





anorexia



2
Maculopapular
3.1-6

1000-1500



rash
mg/dL

mL/day or



25-50% BSA


5-7 episodes/day


3
Maculopapular
6.1-15

>1500 mL/day or



rash
mg/dL

>7 episodes/day



>50% BSA





4
Generalized
>15

Severe abdominal



erythroderma
mg/dL

pain



(>50%


with or without ileus,



BSA) plus


or



bullous


grossly bloody stool



formation


(regardless of stool



and


volume)



desquamation






>5%






BSA





BSA: body surface area;


GI: gastrointestinal;


GvHD: graft versus host disease.






Overall GvHD grade is determined based on most severe target organ involvement.

    • Grade 0: No stage 1-4 of any organ
    • Grade 1: Stage 1-2 skin without liver, upper GI, or lower GI involvement
    • Grade 2: Stage 3 rash and/or stage 1 liver and/or stage 1 upper GI and/or stage 1 lower GI
    • Grade 3: Stage 2-3 liver and/or stage 2-3 lower GI, with stage 0-3 skin and/or stage 0-1 upper GI
    • Grade 4: Stage 4 skin, liver, or lower GI involvement, with stage 0-1 upper GI


Potential confounding factors that may mimic GvHD such as infections and reactions to medications should be ruled out. Skin and/or GI biopsy should be obtained for confirmation before or soon after treatment has been initiated. In instance of liver involvement, liver biopsy should be attempted if clinically feasible. Sample(s) of all biopsies will also be sent to a central laboratory for pathology assessment.


Recommendations for management of acute GvHD are outlined in Table 18.









TABLE 18







Acute GvHD Management








Grade
Management





1
Skin: Topical steroids or immunosuppressants; if stage 2:



methylprednisolone 1 mg/kg (or equivalent dose)


2-4
Initiate methylprednisolone 2 mg/kg daily (or equivalent dose).



IV form of steroid such as methylprednisolone



should be considered if there are



concerns with malabsorption.



Steroid taper may begin after improvement



is seen after ≥3 days of steroids. Taper



should be 50% decrease of total daily steroid dose every 5 days.



GI: In addition to steroids, start anti-diarrheal



agents as per standard practice.





GI: gastrointestinal;


IV: intravenous.






Decisions to initiate second-line therapy should be made sooner for subjects with more severe GvHD. For example, secondary therapy may be indicated after 3 days with progressive manifestations of GvHD, after 1 week with persistent grade 3 GvHD, or after 2 weeks with persistent grade 2 GvHD. Second-line systemic therapy may be indicated earlier in subjects who cannot tolerate high-dose glucocorticoid treatment (Martin et al., Biol Blood Marrow Transplant 18, 1150-1163; 2012).


5.2.9. Hypotension and Renal Insufficiency


Hypotension and renal insufficiency have been reported with CAR T cell therapy and should be treated with IV administration of normal saline boluses according to institutional practice guidelines. Dialysis should be considered when appropriate.


6. Study Procedures

Both the dose escalation and expansion parts of the study consist of 3 distinct stages:

    • (1) screening and eligibility confirmation,
    • (2) treatment with various LD/immunomodulatory agents and CTX120 infusion, and
    • (3) follow-up. During the screening period, subjects are assessed according to the eligibility criteria outlined herein. After enrollment, subjects receive various regimens of LD/immunomodulatory agents, followed by CTX120 infusion. After completing the treatment period, subjects are assessed for multiple myeloma response, disease progression, and survival. Throughout all study periods, subjects are regularly monitored for safety.


A complete schedule of assessments is provided in Table 19 and Table 20. Descriptions of all required study procedures are provided in this section. In addition to protocol-mandated assessments, subjects should be followed per institutional guidelines, and unscheduled assessments should be performed when clinically indicated.


Certain assessments for visits after Day 8 may be performed as in-home or alternate-site visits. Assessments include hospital utilization, changes in health and/or changes in medications, body system assessment, vital signs, weight, PRO questionnaire distribution, and blood sample collections for local and central laboratory assessments.


Missed evaluations should be rescheduled and performed as close to the originally scheduled date as possible. An exception is made when rescheduling becomes medically unnecessary or unsafe because it is too close in time to the next scheduled evaluation. In that case, the missed evaluation should be abandoned.


For the purposes of this protocol, there is no Day 0. All visit dates and windows are to be calculated using Day 1 as the date of first CTX120 infusion.









TABLE 19







Schedule of Assessments: Screening, Treatment, and Primary Follow-up (Screening to Month 24)


















Treatment



















(Stage 2)















D-14







to
D-5

Follow-up (Stage 3)































Screening 1
D-6
to
D1

D3 +
D5 ±
D8 ±
D10 ±
D14 ±
D21 ±
D28 ±
M2 ±
M3 ±
M4 ±
M5 ±
M6 ±
M9 ±
M12 ±
M15 ±
M18 ±
M21 ±
M24 ±


Study Stage Day
(Stage 1)
(2)
D-3
(3)
D2
1 d
2 d
2 d
2 d
2 d
2 d
4 d
7 d
7 d
7 d
7 d
14 d
14 d
14 d
14 d
14 d
14 d
21 d





Informed consent
X
























Medical history 4
X
























Physical exam
X
X
X
X
X
X
X
X
X
X
X
X5
X5
X5
X5
X5
X
X
X
X
X
X
X


Vital signs 6
X
X
X
X
X
X
X
X
X
X
X
X5
X5
X5
X5
X5
X
X
X
X
X
X
X


Height, weight 7
X
X
X
X



X

X

X
X7
X7
X7
X7









Pregnancy test 8,9
X
X
X






















ECOG status
X


X







X

X











Echocardiogram
X
























12-lead ECG 10
X
X
X
X







X













ICE assessment 11
X


X
X
X
X
X



X













PRO 12
X


X







X

X


X
X
X

X

X









Concomitant meds 13

Continuous


Adverse events 14

Continuous




















Hospital



Continuous











utilization




















Treatment






























Daratumumab 15

X









X
X
X
X
X









LD chemotherapy 16


X






















Lenalidomide 17


X








X17
X17
X17
X17
X17









CTX120 infusion 18



X*



X**






















Multiple Myeloma Disease/Response Assessments (Central)






























Serum/urine
X










X
X
X
X
X
X
X
X
X
X
X
X


biochemistry 19,20

























Whole body
X










X22

X22


X22
X22
X22
X22
X22
X22
X22


PET/CT scan 21

























BM aspirate/biopsy 23
X








X24

X

X


X

X






CBC w/
X
X
X
X
X
X
X
X
X
X
X
X26
X
X
X
X
X
X
X
X
X
X
X


differential 25, 26

























Serum chemistry
X
X
X
X
X
X
X
X
X
X
X
X5
X5
X5
X5
X5
X
X
X
X
X
X
X


Coagulation parameters
X
X
X
X
X
X
X
X
X
X

X













Viral serology 27
X
























Immunoglobulins
X


X







X
X
X
X
X
X
X
X
X
X
X
X


Ferritin, CRP
X


X
X
X
X
X
X
X
X
X


X
X









TBNK panel 28
X
X
X
X

X
X
X
X
X
X
X
X
X
X
X
X
X
X






Serum B2M 29
X
























BM cytogenetics 29
X





























Biomarkers (Blood, Central)






























CTX120 levels 30
X


X3
X
X
X
X
X
X
X
X
X
X
X
X
X

X



X






pre/

























post





















Cytokines 32
X


X
X
X
X
X
X
X
X
X
X












Anti-CTX120 33
X










X

X


X

X



X


Daratumumab
X
X34
X35
X



X

X
X
X34
X34
X34
X34
X34









PK 34

pre/









pre/
pre/
pre/
pre/
pre/











post









post
post
post
post
post









DNA
X
























Cell-free DNA
X










X
X
X


X

X



X


Exploratory
X

X35
X


X
X
X
X
X
X
X
X
X
X
X

X



X


biomarkers 36





Ab: antibody; AE: adverse event; AESI adverse event of special interest; ANC: absolute neutrophil count; B2M: beta-2 microglobulin; BM: bone marrow; CBC: complete blood count; chemo: chemotherapy; CNS: central nervous system; con meds: concomitant medications; CR: complete response; CRP: C-reactive protein; CRS: cytokine release syndrome; CT: computed tomography; D or d: day; DL: Dose Level; ECG:electrocardiogram; ECOG: Eastern Cooperative Oncology Group; EORTC QLQ-C30 and QLQ-MY20: European Organisation for Research and Treatment of Cancer QLQ-C30 and QLQ-MY20 questionnaires; FLC: free light chain; HBV: hepatitis B virus; HCV: hepatitis C virus; HIV: human immunodeficiency virus; ICE: immune effector cell-associated encephalopathy; IMWG: International Myeloma Working Group; IV: intravenously; LD: lymphodepleting; M: month; M-protein: monoclonal protein; MM: multiple myeloma; MRI: magnetic resonance imaging; PCR: polymerase chain reaction; PET: positron emission tomography; PK: pharmacokinetics; PRO: patient-reported outcome; SAE: serious adverse event; SPEP: serum M-protein quantitation by electrophoresis; TBNK: T, B, and natural killer cells; UPEP: urine M-protein quantitation by electrophoresis.


*NOTE:


For both Part A and Part B, this study allows for planned redosing with CTX120 per the redosing criteria described herein. Subjects who are redosed should be followed per the schedule of assessments consistent with the initial dosing. Some or all Stage 1 screening assessments must be repeated.


NOTE:


Certain assessments for visits after the required hospitalization period (if applicable) may be performed as in-home or alternate-site visits in extenuating circumstances.



1 Screening assessmentst o be completed within 14 days of informed consent. Subjects will be allowed a one-time rescreening, which may take place within 3 months of initial consent.




2 Only for Cohorts 1 and 3. All procedures to be performed prior to daratumumab infusion unless otherwise specified.




3 All assessments on Day 1 are to be performed prior to CTX120 infusion unless otherwise specified; refer to laboratory manual for details.




4 Includes completesurgical and cardiac history.




5 Perform for all cohorts. For Cohorts 2 and 3 only: Repeat assessment if performed ≥ 7 days prior to beginning a new cycle of lenalidomide dosing.




6 Includes sitting blood ressure, heart rate, spiratoryrate, pulse oximetry, and temperature.




7 Height at screening only. For Cohorts 1 and 3 only: Weight at Month 2, 3, 4, and 5 visits prior to daratumumab dosing.




8 Assessed at local laboratory. Refer to Table 25 for hematology and serum chemistry assessments.




9 For female subjects of childbearing potential. Serum pregnancy test conducted at screening. Serum or urine pregnancy test conducted within 72 hours before start of daratumumab (Cohorts 1 and 3 only) and LD chemotherapy (all cohorts). For Cohort 2, prior to starting lenalidomide, as well as during and after administration, pregnancy must be excluded in accordance with local prescribing information.




10 Prior to daratumumab (Cohorts 1 and 3 only), prior to LD chemotherapy, and prior to CTX120 infusion.




11 On Day 1 prior to CTX120 administration. If CNS symptoms persist, ICE assessment (Table 15) should continue to be performed approximately every 2 days until symptom resolution to grade 1 or baseline.




12 EORTC QLQ-C30 and QLQ-MY20, and EuroQol EQ-5D-5L questionnaires. PRO surveys should be administered before any visit-specific procedures are performed.




13 All concomitant medications to be collected up to 3 months post-CTX120 infusion, then only select concomitant medications are collected.




14 Collect all AEs from informed consent to 3 months after each CTX120 infusion and collect only SAEs and AESIs from 3 months after last CTX120 infusion through Month 24 visit. After Month 24 to Month 60 or if a subject starts a new anticancer therapy after Month 3 study visit, only CTX120-related SAEs and CTX120-related AESIs, and new malignancies will be reported. See Table 27 for details.




15 Cohorts 1 and 3 only: First dose of daratumumab administered within 3 days prior to starting LD chemotherapy and no more than 14 days prior to CTX120 infusion. Subsequent (D28 and on) dosing requires achieving SD or better and should continue per the indicated schedule unless disease progression or unacceptable toxicity occurs.




16 For the first CTX120 dose, start LD chemotherapy within 7 days of study enrollment (i.e., confirmation of eligibility). After completion of LD chemotherapy, ensure washout period of ≥ 48 hours (but ≤ 7 days) before CTX120 infusion. Physical exam, weight, and coagulation laboratories performed prior to first dose of LD chemotherapy. Vital signs, CBC, clinical chemistry, and AEs/concomitant medications assessed and recorded daily (i.e., 3 times) during LD chemotherapy.




17 Cohorts 2 and 3 only. Refer to Section 5.4 for dosing instruction and additional criteria for administration. Cycle 1 of lenalidomide beginning on third day of LD chemo and continuing for 21 days through CTX120 infusion. If Cycle 2 of lenalidomide is delayed, adjust subsequent cycles accordingly.




18 CTX120 administered 48 hours to 7 days after completion of LD chemotherapy.




19 Central lab testing and review of M-protein measurements in serum and urine: SPEP, serum immunofixation, serum FLC (kappa and lambda), 24-h UPEP, urine immunofixation, and quantitative immunoglobulins.



Note:


Screening to determine eligibility does NOT require central review, but samples for baseline disease assessment should be collected for central lab testing.


Note:


Screening 24-hour urine collection may begin the day before informed consent



20 A baseline assessment for serum (or urine, if serum is non-measurable) should be performed within 72 hours (or on same day) prior to LD chemotherapy (Cohort 2) or prior to daratumumab infusion (Cohorts 1 and 3).




21 Baseline whole body (vertex to toes) PET/CT to be performed at screening (i.e., within 28 days prior to CTX120 infusion) and upon suspected CR. For subjects with evidence of extramedullary disease (e.g., extramedullary plasmacytoma or myelomatous lesion with soft tissue involvement), postinfusion scans will be conducted per the schedule of assessments, per IMWG response criteria (Table 22), and as clinically indicated. In subjects with extramedullary disease, the CT portion of PET/CT should be of diagnostic quality (e.g., CT with IV contrast) sufficient for tumor size measurement. MRI with contrast may be used for the CT portion when CT is clinically contraindicated or as required by local regulation.




22 Only for subjects with extramedullary disease.




23 Additional BM biopsy and aspirate should be performed to confirm CR (by immunohistochemistry, central testing) as part of disease evaluation. BM biopsy and aspirate should be performed at time of disease relapse whenever clinically feasible. For any BM aspirates collected, samples should be sent for CTX120 levels and/or other exploratory analyses. BM sample collection (aspirate and biopsy) at screening should be performed during the 14-day screening period. All other bone marrow sample collection should be performed ± 5 days of visit date. If HLH is suspected, BM biopsy and aspirate to be performed.




24 If performed, serum and urine multiple myeloma response assessments should also be performed for this time point (Table 19).



25 In case of grade 3 or 4 neutropenia and thrombocytopenia, collect samples weekly until resolution to grade ≤2.


26 For Cohort 2, additional CBC monitoring is required.



27 Infectious disease testing (HIV-1, HIV-2, HCV antibody and PCR, HBV surface antigen, HBV surface antibody, HBV core antibody) performed within 30 days of providing informed consent may be considered for subject eligibility.




28 TBNK panel assessment at screening, before start of daratumumab (Cohorts 1 and 3), before first day of LD chemotherapy, before CTX120 infusion, and all listed time points are assessed at local laboratory. To include 6-color TBNK panel, or equivalent for T, B, and natural killer cells.




29 Serum B2M and cytogenetics (bone marrow) at screening only and assessed locally.




30 In subjects experiencing signs or symptoms of CRS, neurotoxicity, or suspected HLH, additional blood samples should be drawn at intervals outlined in the laboratory manual.




31 Two samples are to be collected on Day 1: one before CTX120 infusion and another 20 (±5) min after the end of CTX120 infusion.




32 Additional cytokine samples should be collected daily for the duration of CRS. During neurotoxicity and suspected HLH, additional cytokine samples are collected.




33 Continue sample collection for all listed time points.




34 Daratumumab-related assessments for Cohorts 1 and 3 only. Day 1 sample collected prior to CTX120 infusion. Two samples are to be collected at each daratumumab dosing: 1 before infusion and another 30 (±15) min after the end of infusion.




35 Prior to first day of LD chemotherapy only.




36 Samples for exploratory biomarkers should also be sent from any lumbar puncture, BM sample collection (aspirate/biopsy), or suspected GvHD tissue biopsy performed following CTX120 infusion. If CRS, neurotoxicity, or HLH occur, collect samples for exploratory biomarker assessment.














TABLE 20







Schedule of Assessments: Progressive Disease and Secondary Follow-up (Months 30-60)
















M30
M36
M42
M48
M54
M60
Progressive
Secondary


Assessments
(±21 d)
(±21 d)
(±21 d)
(±21 d)
(±21 d)
(±21 d)
Disease 1
Follow-Up 2





Physical exam
X
X
X
X
X
X
X
X


Vital signs 3
X
X
X
X
X
X
X
X


PRO 4
X
X
X
X
X
X
X



Concomitant
X
X
X
X
X
X
X
X


medications 5










AEs 6
X
X
X
X
X
X
X
X


MM disease/response
X
X
X
X
X
X
X



assessment 7










CBC with differential 8
X
X
X
X
X
X
X
X


Serum chemistry 8
X
X
X
X
X
X
X
X


Immunoglobulins 8, 9
X
X
X
X
X
X
X



CTX120 levels (blood,

X

X

X
X
X


central) 9, 10










Anti-CTX120 (blood,

X

X

X
X



central) 9










TBNK panel 8
X
X
X
X
X
X
X



Exploratory biomarkers

X

X

X
X
X


(blood, central) 11





Ab: antibody; AE: adverse event; BM: bone marrow; CBC: complete blood count; CT: computed tomography; d: days; EORTC QLQ-C30 and QLQ-MY20: European Organisation for Research and Treatment of Cancer QLQ-C30 and QLQ-MY20 questionnaires; IMWG: International Myeloma Working Group; M: month; M-protein: monoclonal protein; MM: multiple myeloma; PD: progressive disease; PET: positron emission tomography; PRO: patient-reported outcome; SAE: serious adverse event; SCT: stem cell transplant; TBNK: T, B, and natural killer cells.


NOTE:


Certain assessments for visits after the required hospitalization period (if applicable) may be performed as in-home or alternate-site visits in extenuating circumstances.



1 Subjects with PD discontinue the normal schedule of assessments, undergo study assessments listed, then secondary follow-up (see footnote 2).




2 Subjects with PD or who partially withdraw consent discontinue the normal schedule of assessments, attend annual study visits, and undergo secondary follow- up consisting of these procedures at a minimum: abbreviated physical exam, CBC with differential, serum chemistry, disease assessment/survival status, CTX120 persistence, select concomitant medications/procedures (anticancer therapy, disease-related surgery, SCT), and select AEs (treatment-related AEs and SAEs, new malignancies, new/worsening autoimmune, immune deficiency, or neurological disorders).




3 Includes temperature, blood pressure, pulse rate, and respiratory rate.




4 EORTC QLQ-C30, QLQ-MY20, and EuroQol EQ-5D-5L questionnaires. PRO surveys should be administered before any visit-specific procedures are performed.




5 Only select concomitant medications are collected.




6 SAEs and AESIs should be reported through the Month 24 study visit. Only CTX120-related AESIs, CTX120-related SAEs, and new



malignancies will be reported after Month 24 to Month 60 or if a subject begins new anticancer therapy after Month 3 study visit. See Table 27 for details.



7 Disease evaluations based on assessments in accordance with IMWG response criteria (Kumar et al., Lancet Oncol 17, e328-e346; 2016) and will include serum and urine M-protein measurements, and, if deemed appropriate, whole body PET/CT and BM aspirate and biopsy as clinically indicated (Table 25).




8 Assessed at local laboratory. To include 6-color TBNK panel, or equivalent for T, B, and NK cells.




9 Continue sample collection for all listed time points.




10 In addition to time points listed, samples for analysis of CTX120 levels and/or exploratory analyses should be sent to the central laboratory from any unscheduled collection of blood, BM aspirate, or biopsy of extramedullary plasmacytoma.




11 Samples for exploratory biomarkers should be sent from any lumbar puncture, BM sample collection (aspirate/biopsy), or suspected GvHD tissue biopsy performed following CTX120 infusion.







6.1. Subject Screening


The screening period begins on the date that the subject signs the ICF and continues through confirmation of eligibility and enrollment into the study. Once informed consent has been obtained, the subject is screened to confirm study eligibility as outlined in the schedule of assessments (Table 19). Screening assessments should be completed within 14 days of a subject signing the informed consent. Subjects are allowed a one-time rescreening, which may take place within 3 months of the initial consent. If rescreening occurs, subject should reconsent prior to reconfirmation of eligibility criteria.


6.2. Study Assessments


Refer to the schedule of assessments (Table 19 and Table 20) for the timing of the required procedures. Demographic data, including age, sex, race, and ethnicity, are collected. Medical history, including a full history of the subject's disease, previous cancer treatments, and response to treatment from date of diagnosis will be obtained. Cardiac, neurological, and surgical history are obtained.


Physical Exam


Physical examination, including examination of major body systems, including general appearance, skin, neck, head, eyes, ears, nose, throat, heart, lungs, abdomen, lymph nodes, extremities, and nervous system, is performed at every study visit and the results documented. Changes noted from the exam performed at screening are recorded as an AE. For subjects in Cohorts 2 and 3, repeat physical exam if performed 7 or more days prior to beginning a new cycle of lenalidomide dosing.


Vital Signs, Including Height and Weight


Vital signs are recorded at every study visit and include sitting blood pressure, heart rate, respiratory rate, pulse oximetry, and temperature. Weight is obtained according to the schedule in Table 19, and height will only be obtained at screening.


For subjects in Cohorts 1 and 3 only, weight is also obtained on Month 2, 3, 4, and 5 visits prior to daratumumab dosing.


For subjects in Cohorts 2 and 3, vital signs assessments are repeated if performed 7 or more days prior to beginning a new cycle of lenalidomide dosing.


Pregnancy Test


Female subjects of reproductive potential (women who have reached menarche or women who have not been postmenopausal for at least 24 consecutive months, i.e., who have had menses within the preceding 24 months, or have not undergone a sterilization procedure [hysterectomy or bilateral oophorectomy]) must have a serum pregnancy test performed at the time of screening, and a serum or urine pregnancy test within 72 hours before start of daratumumab (Cohorts 1 and 3 only) and LD chemotherapy (all cohorts), including the redosing schedule for respective cohorts. For Cohorts 2 and 3, prior to starting lenalidomide, as well as during and after administration, pregnancy must be excluded in accordance with local prescribing information.


ECOG Performance Status


Performance status is assessed at the screening, CTX120 infusion (Day 1, prior to infusion), Day 28, and Month 3 visits using the ECOG scale to determine the subject's general well-being and ability to perform activities of daily life.









TABLE 21







ECOG Performance Status Scale








Grade
Description





0
Fully active, able to carry on all



pre-disease performance without restriction


1
Restricted in physically strenuous



activity but ambulatory and able to carry



out work of a light or sedentary



nature, e.g., light house work, office work


2
Ambulatory and capable of all



self-care but unable to carry out any work



activities; up and about more



than 50% of waking hours


3
Capable of only limited self-care;



confined to bed or chair more than 50% of



waking hours


4
Completely disabled; cannot carry on any



self-care; totally confined to bed or



chair


5
Dead









Developed by the Eastern Cooperative Oncology Group, Robert L. Comis, MD, Group Chair (Oken et al., Am J Clin Oncol 5, 649-655.; 1982).

Echocardiogram


A transthoracic cardiac echocardiogram (for assessment of left ventricular ejection fraction) is performed and read by trained medical personnel at screening to confirm eligibility. Additional cardiac assessments is recommended during grade 3 or 4 CRS for all subjects who require >1 fluid bolus for hypotension, who are transferred to the intensive care unit for hemodynamic management, or who require any dose of vasopressor for hypotension (Brudno et al., Blood 127, 3321-3330; 2016).


Electrocardiogram


Twelve (12)-lead electrocardiograms (ECGs) are obtained during screening, prior to daratumumab (Cohorts 1 and 3 only) on the first day of treatment, prior to LD chemotherapy on the first day of treatment, prior to CTX120 administration on Day 1, and on Day 28. QTc and QRS intervals are determined from ECGs. Additional ECGs may be obtained.


Immune Effector Cell-Associated Encephalopathy Assessment


Neurocognitive assessment is performed using ICE assessment. The ICE assessment tool is a slightly modified version of the CARTOX-10 screening tool, which now includes a test for receptive aphasia (Neelapu et al., 2018). ICE assessment examines various areas of cognitive function: orientation, naming, following commands, writing, and attention (Table 15).


ICE assessment is performed at screening, before administration of CTX120 on Day 1, and on Days 2, 3, 5, 8, and 28. If a subject experiences CNS symptoms, ICE assessment should continue to be performed approximately every 2 days until resolution of symptoms to grade 1 or baseline. To minimize variability, whenever possible the assessment should be performed by the same research staff member who is familiar with or trained in administration of the ICE assessment tool.


Patient-Reported Outcomes


Three PRO surveys, the European Organisation for Research and Treatment of Cancer (EORTC) QLQ-C30, EORTC QLQ-MY20, and the EuroQol EQ-5D-5L questionnaires, will be administered according to the schedule in Table 19 and Table 20. Questionnaires should be completed (self-administered in the language the subject is most familiar) before clinical assessments are performed.


The EORTC QLQ-C30 is a questionnaire designed to measure cancer patients' physical, psychological, and social functions. It is composed of 5 multi-item scales (physical, role, social, emotional, and cognitive function) and 9 single items (pain, fatigue, financial impact, appetite loss, nausea/vomiting, diarrhea, constipation, sleep disturbance, and quality of life). The EORTC QLQ-C30 is validated and has been widely used among cancer patients, including in multiple myeloma patients (Wisloff et al., Br J. Haematol 92, 604-613; 1996; Wisloff et al., Nordic Myeloma Study Group. Br J. Haematol 97, 29-37; 1997).


The QLQ-MY20 questionnaire is the myeloma-specific module of EORTC QLQ-C30, designed for patients with multiple myeloma to assess the symptoms and side effects of treatment and their impact on everyday life. The module comprises 20 questions addressing 4 domains of quality of life important in myeloma: pain, treatment side effects, social support and future perspective, disease-specific symptoms and their impact on everyday life, treatment side effects, social support, and future perspective (Cocks et al., Eur J Cancer 43, 1670-1678; 2007). The EQ-5D-5L is a generic measure of health status and contains a questionnaire that assesses 5 domains, including mobility, self-care, usual activities, pain/discomfort, and anxiety/depression, plus a visual analog scale. EQ-5D-5L has been used in conjunction with QLQ-C30 and QLQ-MY20 in multiple myeloma (Moreau et al., Leukemia, 33, 2934-2946; 2019).


Multiple Myeloma Disease and Response Assessments


Disease evaluations are based on assessments in accordance with the IMWG criteria for response and MRD assessment in multiple myeloma (Tables 22-23) (Kumar et al., 2016). Determination of study eligibility and decisions regarding subject management and disease progression is made. For efficacy analyses, disease outcome is graded using IMWG response criteria. Multiple myeloma disease and response evaluation should be conducted per the schedule in Table 19 and Table 20, and includes the assessments described below. All response categories (including progression) require 2 consecutive assessments made at any time before the institution of any new therapy.









TABLE 22







Standard IMWG Response Criteria








Response 1
Description





Stringent Complete
Complete response as defined below


Response
plus normal FLC ratio2 and absence


(sCR)
of clonal cells in BM biopsy by



immunohistochemistry (κ/λ ratio ≤4:1 or



≥1:2 for κ and λ patients, respectively,



after counting ≥100 plasma cells).3


Complete
Negative immunofixation on the serum


Response (CR)
and urine and disappearance of



any soft



tissue plasmacytomas and <5%



plasma cells in BM aspirates.4


Very Good
Serum and urine M-protein detectable


Partial Response
by immunofixation but not on


(VGPR)
electrophoresis or ≥90% reduction in



serum M-protein plus urine M-



protein level



<100 mg per 24 h.


Partial Response
≥50% reduction of serum M-protein plus


(PR)
reduction in 24 h urinary M-protein by



≥90% or to <200 mg per 24 h;



If the serum and urine M-protein



are unmeasurable, ≥50% decrease in the



difference between involved and



uninvolved FLC levels is required in



place of the M-protein criteria;



If serum and urine M-protein are



unmeasurable, and serum-free light



assay is also unmeasurable, ≥50%



reduction in plasma cells is required in



place of M-protein, provided baseline



BM plasma-cell percentage was



≥30%. In addition to these criteria,



if present at baseline, ≥50% reduction



in the size (SPD)5 of soft tissue



plasmacytomas is required.


Minimal
≥25% but ≤49% reduction of serum


Response (MR)
M-protein and reduction in 24-h



urine M-protein by 50-89%. In



addition to the above listed criteria, if



present at baseline,



≥50% reduction in the size (SPD)5



of soft tissue plasmacytomas is also



required.


Stable Disease
Not recommended for use as an


(SD)
indicator of response; stability of disease



is best described by providing the



time-to-progression estimates. Not



meeting criteria for CR, VGPR,



PR, MR, or PD.


Progressive
Any one or more of the following criteria:


Disease (PD) 6,7
Increase of 25% from lowest



confirmed response value in ≥1 of the



following criteria:



Serum M-protein (absolute increase



must be ≥0.5 g/dL);



Serum M-protein increase ≥1 g/dL, if



the lowest M component was ≥5



g/dL;



Urine M-protein (absolute increase



must be ≥200 mg/24 h);



In patients without measurable serum



and urine M-protein levels, the



difference between involved and



uninvolved FLC levels (absolute



increase must be >10 mg/dL);



In patients without measurable



serum and urine M-protein levels and



without measurable involved FLC



levels, BM plasma-cell percentage



irrespective of baseline status



(absolute increase must be ≥10%);



Appearance of a new lesion(s), ≥50%



increase from nadir in SPD5 of >1



lesion, or



≥50% increase in the longest diameter



of a previous lesion >1 cm in short



axis;



≥50% increase in circulating plasma cells



(≥200 cells/μL) if this is the only



measure of disease.


Clinical Relapse
Clinical relapse requires ≥1



of the following criteria:



Direct indicators of increasing



disease and/or end organ dysfunction



(CRAB features) related to the



underlying clonal plasma-cell



proliferative disorder. It is not used



in calculation of time to progression



or PFS but is listed as something that



can be reported optionally or for



use in clinical practice;



Development of new soft tissue



plasmacytomas or bone lesions



(osteoporotic fractures do not



constitute progression);



Definite increase in the size of



existing plasmacytomas or bone lesions.



Definite increase is defined as a



50% (and ≥1 cm) increase as measured



serially by the SPD5



of the measurable lesion;



Hypercalcemia (>11 mg/dL);



Decrease in hemoglobin of ≥2 g/dL



not related to therapy or other non-



myeloma—related conditions;



Rise in serum creatinine by ≥2 mg/dL



from the start of the therapy and



attributable to myeloma;



Hyperviscosity related to



serum paraprotein.


Relapse from CR
Any one or more of the following criteria:


(to be used only
Reappearance of serum or urine


if endpoint is
M-protein by immunofixation or


disease-free survival)
electrophoresis;



Development of ≥5% plasma



cells in the BM;



Appearance of any other sign of



progression (i.e., new plasmacytoma,



lytic bone lesion, or



hypercalcemia; see above).


Relapse from MRD-
Any one or more of the following criteria:


Negative
Loss of MRD-negative state (evidence


(to be used only
of clonal plasma cells on NGF or


if endpoint is
NGS, or positive imaging study


disease-free
for recurrence of myeloma);


survival)
Reappearance of serum or urine



M-protein by immunofixation or



electrophoresis;



Development of ≥5% clonal plasma



cells in the BM; Appearance of any



other sign of progression (i.e., new



plasmacytoma, lytic bone lesion, or



hypercalcemia).





BM: bone marrow;


CR: complete response;


CRAB: calcium elevation, renal failure, anemia, lytic bone lesions;


CT: computed tomography;


FLC: free light chain;


h: hour;


IMWG: International Myeloma Working Group;


M-protein: monoclonal protein;


MR: minimal response;


MRD: minimal residual disease;


MRI: magnetic resonance imaging;


NGF: next-generation flow;


NGS: next-generation sequencing;


PD: progressive disease;


PET: positron emission tomography;


PFS: progression-free survival;


PR: partial response;


sCR: stringent complete response;


SD: stable disease;


SPD: sum of products of maximal perpendicular diameters of measured lesions;


VGPR: very good partial response.



1 Derived from international uniform response criteria for multiple myeloma (Durie et al., Leukemia 20, 1467-1473; 2006). Minor response definition and clarifications are disclosed in Rajkumar et al., Blood 117, 4691-4695; 2011). When the only method to measure disease is by serum FLC levels: CR can be defined as a normal FLC ratio of 0.26 to 1.65 in addition to the CR criteria listed previously. VGPR in such patients requires ≥90% decrease in difference between involved and uninvolved FLC levels. All response categories require 2 consecutive assessments made at any time before institution of any new therapy; all categories also require no known evidence of progressive or new bone lesions or extramedullary plasmacytomas if radiographic studies were performed. Radiographic studies are not required to satisfy these response requirements. BM assessments do not need to be confirmed. Each category, except for SD, will be considered unconfirmed until the confirmatory test is performed. Date of initial test is considered as date of response for evaluation of time-dependent outcomes such as duration of response.




2All recommendations regarding clinical uses relating to serum FLC levels or FLC ratio are based on results obtained with the validated Freelite test (Binding Site, Birmingham, UK).




3Presence/absence of clonal cells on immunohistochemistry is based on the κ/λ ratio. An abnormal κ/λ ratio by immunohistochemistry requires ≥100 plasma cells for analysis. An abnormal ratio reflecting presence of an abnormal clone is κ/λ of >4:1 or <1:2.




4Special attention should be given to the emergence of a different monoclonal protein following treatment, especially in the setting of patients having achieved a conventional CR, often related to oligoclonal reconstitution of the immune system. These bands typically disappear over time and in some studies have been associated with a better outcome. Also, appearance of monoclonal IgG κ in patients receiving monoclonal antibodies should be differentiated from the therapeutic antibody.




5Plasmacytoma measurements should be taken from the CT portion of the PET/CT, or MRI scans, or dedicated CT scans where applicable. For patients with only skin involvement, skin lesions should be measured with a ruler.



Measurement of tumor size will be determined by SPD.



6 Positive immunofixation alone in a patient previously classified as achieving CR will not be considered progression. For purposes of calculating time to progression and PFS, patients who have achieved CR and are MRD-negative should be evaluated using criteria listed for PD. Criteria for relapse from CR or relapse from MRD should be used only when calculating disease-free survival.




7 In the case in which a value is felt to be a spurious result per physician discretion (e.g., possible laboratory error), that value will not be considered when determining the lowest value. MRD requires complete response, as defined in Table 22.














TABLE 23







IMWG Minimal Residual Disease Criteria








MRD Status1
Description





Sustained
MRD negativity in BM (NGF and/or


MRD-Negative
NGS) and by imaging as defined below,



confirmed ≥1 year apart. Subsequent



evaluations can be used to further



specify the duration of negativity



(e.g., MRD-negative at 5 years).2


Flow
Absence of phenotypically aberrant


MRD-Negative
clonal plasma cells by NGF3 on BM



aspirates using the EuroFlow standard



operation procedure for MRD



detection in multiple myeloma (or



validated equivalent method) with



sensitivity of ≥1 in 105 nucleated cells.


Sequencing MRD-
Absence of clonal plasma cells by NGS


Negative
on BM aspirate in which presence of



a clone is defined as <2 identical



sequencing reads obtained after DNA



sequencing of BM aspirates using the



LymphoSIGHT platform (or validated



equivalent method) with sensitivity



of ≥1 in 105 nucleated cells.4


Imaging +
MRD negativity, as defined by NGF or


MRD-Negative
NGS plus disappearance of every



area of increased tracer uptake found



at baseline or a preceding PET/CT or



decrease to less mediastinal blood pool



SUV or decrease to less than that of



surrounding normal tissue.5





ASCT: autologous stem cell transplant;


BM: bone marrow;


CT: computed tomography;


FDG: 18F-fluorodeoxyglucose;


IMWG: International Myeloma Working Group;


MFC: multiparameter flow cytometry;


MRD: minimal residual disease;


NGF: next-generation flow;


NGS: next-generation sequencing;


PET: positron emission tomography;


SUV: standard update value;


SUVmax: maximum standardized uptake value.


Note:


For MRD assessment, first BM aspirate should be sent to MRD (not for morphology) and this sample should be taken in 1 draw with a volume of ≥2 mL (to obtain sufficient cells), but maximally 4-5 mL to avoid hemodilution.



1For MRD there is no need for 2 consecutive assessments. MRD tests should be initiated only at the time of suspected complete response. All categories of MRD require no known evidence of progressive or new bone lesions if radiographic studies were performed. However, radiographic studies are not required to satisfy these response requirements except for the requirement of FDG PET if imaging MRD-negative status is reported.




2Sustained MRD negativity when reported should also annotate method used (e.g., sustained flow MRD-negative, sustained sequencing MRD-negative).




3Bone marrow MFC should follow NGF guidelines (Paiva et al., Blood 119, 687-691; 2012). The reference NGF method is an 8-color 2-tube approach that has been extensively validated 5 million cells should be assessed. The flow cytometry method employed should have a sensitivity of detection of ≥1 in 105 plasma cells.




4DNA sequencing assay on BM aspirate should use a validated assay such as LymphoSIGHT (Sequenta).




5Criteria disclosed in Zamagni et al., Clin Cancer Res 21, 4384-4390; 2015), and expert panel (IMPetUs; Italian Myeloma criteria for PET Use) (Nanni et al., Eur J Nucl Med Mol Imaging 43, 414-421; 2016; Usmani et al., Blood 121, 1819-1823; 2013). Baseline positive lesions were identified by presence of focal areas of increased uptake within bones, with or without any underlying lesion identified by CT and present on ≥2 consecutive slices. Alternatively, an SUVmax = 2.5 within osteolytic CT areas >1 cm in size, or SUVmax = 1.5 within osteolytic CT areas ≤1 cm in size were considered positive. Imaging should be performed once MRD negativity is determined by MFC or NGS.














TABLE 24







Required Baseline and Follow-up Tests for Response


Assessment Using IMWG Response Criteria














At
At



Every
No
Sus-
Sus-



Response
Measurable
pected
pected


Test
Assessment
Protein 1
CR
DP 2





SPEP (serum
X

X
X


M-spike ≥1 g/dL) 3






Serum immunofixation

X
X
X


(any)






UPEP (urine M-spike
X

X
X


≥200 mg/24 h)






Urine

X
X



immunofixation (any)






Serum FLC
X

X
X


Serum M-spike






<1 g/dL, urine M-spike






<200 mg/24 h, but






involved Ig FLC ≥10






mg/dL






Any


X
X


Bone marrow
X 4

X



aspirate/biopsy






Serum M-spike,






urine M-spike, or






involved Ig FLC






not meeting






above criteria






but BM plasma cell






percentage ≥30%






Any


X



Plasmacytoma
X 4

X



(PET imaging)






Serum M-spike,






urine M-spike, involved






Ig FLC or BM






not meeting above






criteria, but ≥1






lesion with single






diameter of ≥2 cm






Any


X



Hemoglobin, serum
X


X


calcium, creatinine






(any)





BM: bone marrow;


CR: complete response;


DP: disease progression


FLC: free light chain;


h: hour;


Ig: immunoglobulin;


IMWG: International Myeloma Working Group;


M-spike: spike in monoclonal protein;


PET: positron emission tomography;


SPEP: serum protein electrophoresis;


UPEP: urine protein electrophoresis.



1By electrophoresis.




2Clinical or biochemical.




3Baseline M-spike of ≥0.5 g/dL acceptable if very good partial response or higher is the response endpoint to be measured, and if progression-free survival or time to progression are endpoints of interest.




4To be done at assessment time point or complete response, or as clinically indicated, and then at suspected progression.







6.3. Multiple Myeloma Disease and Response Assessments


Monoclonal Protein Measurements in Serum and Urine


Blood and 24-hour urine samples for M-protein measurements are sent to and analyzed by a central laboratory and reviewed for efficacy analyses per the schedule in Table 22 and Table 23, and as clinically indicated. Serum and 24-hour urine samples are collected for each time point and the following tests performed by a central laboratory:

    • Serum M-protein quantitation by electrophoresis (SPEP)
    • Serum immunofixation
    • Serum free light chain assay (FLC, kappa and lambda)
    • 24-hour urine M-protein quantitation by electrophoresis (UPEP). Note: For screening, 24-hour urine collection may begin the day before informed consent
    • Urine immunofixation
    • Quantitative immunoglobulins (Ig), if needed (e.g., IgA or IgD myeloma)


In addition to central lab testing, serum and urine M-protein assessments may be performed locally and used for determination of study eligibility and clinical decisions regarding patient care. For screening, prior laboratory values (multiple myeloma serum and urine results) obtained locally within 2 weeks of informed consent may be used provided that they were not associated with prior anticancer treatment (at least 2 weeks from last dose of anticancer therapy or at time of disease progression while on therapy).


Whole Body PET/CT Radiographic Disease Assessment


Baseline whole body (vertex to toes) PET/CT is performed at screening (i.e., within 28 days prior to CTX120 infusion) and upon suspected CR. If extramedullary lesions are identified during screening, a CT of diagnostic quality (e.g., with IV contrast or similar) should be performed for targeted region(s). MRI with contrast may be used for the CT portion when CT is clinically contraindicated or as required by local regulation. Unless clinically indicated, postinfusion scans are conducted per the schedule of assessments in Table 19 and Table 20, per IMWG response criteria (Table 21) only for subjects with evidence of extramedullary disease (e.g., extramedullary plasmacytoma or myelomatous lesion with soft tissue involvement). PET/CT (with IV contrast) may be obtained as part of standard of care within 4 weeks prior to subject enrollment may be used to satisfy screening requirements.


Bone Marrow Aspirate and Biopsy


Bone marrow aspirate and biopsy is performed according to the schedule of assessments in Table 19 and Table 20, and as clinically indicated. Bone marrow aspirate/biopsy on Day 14 is optional and requires specific consent. Bone marrow sample collection (aspirate and biopsy) at screening should be performed during the 14-day screening period. Bone marrow biopsy obtained as part of standard of care within 4 weeks prior to subject enrollment may be used to satisfy screening requirements. All other bone marrow sample collection should be performed ±5 days of visit date. Standard institutional guidelines for the bone marrow biopsy should be followed.


Percentage of plasma cells is assessed on bone marrow aspirate and biopsy samples by a central laboratory and reviewed as part of disease response evaluation per IMWG response criteria. For subjects who achieve suspected CR, a bone marrow biopsy to confirm response assessment by immunohistochemistry and MRD evaluation (on bone marrow aspirate) is performed by a central laboratory. At any point that bone marrow collection is performed, aspirate samples should also be sent to a central laboratory for measurement of CTX120 and/or other exploratory analyses.


Extramedullary Plasmacytoma Biopsy


At progression, biopsy of extramedullary plasmacytoma, if present, should be collected (if medically feasible) to confirm disease (local testing) and for biomarker analysis (central testing). For subjects with extramedullary disease, tumor biopsy is also encouraged at screening and at least 1 post-CTX120 infusion timepoint. Excess sample (if available) will be stored for exploratory research.


Beta-2 Microglobulin and Cytogenetics


A serum sample to assess B2M level is obtained at screening and sent to a local laboratory for analysis. A bone marrow sample to evaluate cytogenetics should be performed at screening only and assessed locally (Table 19). Cytogenetics evaluation should include fluorescence in situ hybridization for high-risk genetic abnormalities del(17p), t(4;14), t(14; 16), and lq gain at a minimum.


Disease Staging at Study Entry


Disease staging using the Revised International Staging System (R-ISS) for multiple myeloma (based on the Revised International Staging System (R-ISS)) should be performed at study entry based on screening assessments for cytogenetics, serum B2M, albumin, and lactate dehydrogenase. R-ISS at diagnosis (if known) should be recorded based on medical records.


6.4. Laboratory Tests


Laboratory samples are collected and analyzed according to the schedule of assessment (Table 19 and Table 20). Local laboratories meeting Clinical Laboratory Improvement Amendments requirements are utilized to analyze all tests listed in Table 25 according to standard institutional procedures.









TABLE 25





Local Laboratory Tests
















CBC with
Hematocrit, hemoglobin, red blood


differential
cell count, white blood cell count,



neutrophils, lymphocytes, monocytes,



basophils, eosinophils, platelet



count, ANC


TBNK panel
6-color TBNK panel or equivalent



(commonly staining T cells (CD3,



CD4, CD8), B cells (CD19),



and NK cells (CD56, CD16); see



laboratory manual for



additional instructions)


Serum chemistry 1
ALT (SGPT), AST (SGOT),



bilirubin (total and direct), albumin,



alkaline phosphatase, bicarbonate,



blood urea nitrogen, calcium,



chloride, creatinine, eGFR,



glucose, lactate dehydrogenase,



magnesium, phosphorus,



potassium, sodium, total protein


Coagulation
Prothrombin time, activated partial



thromboplastin time, international



normalized ratio, fibrinogen


Viral serology
HIV-1, HIV-2, hepatitis C virus



antibody and PCR, hepatitis B surface



antigen, hepatitis B surface



antibody, hepatitis B core antibody


Immunoglobulins
IgA, IgG, IgM


CRS/HLH monitoring
Ferritin, CRP


Serum or urine pregnancy 2
Human chorionic gonadotropin





ALT: alanine aminotransferase;


ANC: absolute neutrophil count;


AST: aspartate aminotransferase;


CBC: complete blood count;


CRP: C-reactive protein;


CRS: cytokine release syndrome;


eGFR: estimated glomerular filtration rate;


HIV-1/-2: human immunodeficiency virus type 1 or 2;


HLH: hemophagocytic lymphohistiocytosis;


IgA/G/M: immunoglobulin A, G, or M;


LD: lymphodepleting;


PCR: polymerase chain reaction;


NK: natural killer;


SGOT: serum glutamic oxaloacetic transaminase;


SGPT: serum glutamic pyruvic transaminase,


TBNK: T, B, and NK cells.



1 For Cohort 2: Repeat if performed ≥7 days prior to beginning a new cycle of lenalidomide dosing.




2 For females of childbearing potential only. Serum pregnancy test required at screening. Serum or urine pregnancy test within 72 hours before start of daratumumab (Cohorts 1 and 3 only) and LD chemotherapy (all cohorts), including the redosing schedule for respective cohorts.







6.5. Biomarkers


Blood, bone marrow, CSF samples (only in subjects with treatment-emergent neurotoxicity), and, if applicable, tumor biopsy of extramedullary plasmacytoma are collected to identify biomarkers that may be indicative of clinical response, resistance, safety, disease, pharmacodynamic activity, or the mechanism of action of CTX120. Samples are collected and shipped for testing at a central laboratory.


Analysis of CTX120 Levels


Analysis of levels of transduced BCMA-directed CAR+ T cells is performed on blood samples collected according to the schedule described in Table 19 and Table 20. The time course of the disposition of CTX120 in blood is described using a PCR assay that measures copies of CAR construct per μg DNA. Complementary analyses using flow cytometry to confirm the presence of CAR protein on the cellular surface may also be performed. Samples for analysis of CTX120 levels should be sent to the central laboratory from any blood, bone marrow, CSF, or biopsy of extramedullary plasmacytoma performed following CTX120 infusion. If CRS, neurotoxicity, or HLH occur, samples for assessment of CTX120 levels should be collected in intervals. The trafficking of CTX120 in bone marrow, CSF, or extramedullary plasmacytoma tissue may be evaluated in any of these samples collected as per protocol-specific sampling.


Cytokines


Cytokines, including IL-1β, soluble IL-1 receptor alpha (sIL-1Rα), IL-2, sIL-2Rα, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, IL-15, IL-17a, interferon γ, tumor necrosis factor α, and GM-CSF, are analyzed in a central laboratory. Correlational analysis performed in multiple prior CAR T cell clinical studies have identified these cytokines, and others, as potential predictive markers for severe CRS and/or neurotoxicity, as summarized in a recent review (Wang et al., Biomark Res 6, 4; 2018). Blood for cytokines are collected at specified times as described in Table 19 and Table 25. In subjects experiencing signs or symptoms of CRS, neurotoxicity, and HLH, additional samples should be drawn.


Anti-CTX120 Antibody


The CAR construct is composed of humanized scFv. Blood is collected throughout the study to assess for potential immunogenicity, per Table 19 and Table 20.


Daratumumab Pharmacokinetic Analysis (Cohorts 1 and 3)


Pharmacokinetic analysis of daratumumab may be performed on blood samples collected according to the schedule described in Table 19 and Table 20.


The distribution of daratumumab in CSF, bone marrow, or tumor tissues may be evaluated in any of these samples collected as per protocol-specific sampling.


Exploratory Research Biomarkers


Exploratory research may be conducted to identify molecular (genomic, metabolic, and/or proteomic) biomarkers and immunophenotypes that may be indicative or predictive of clinical response, resistance, safety, disease, pharmacodynamic activity, and/or the mechanism of action of treatment. Samples are collected according to the schedule in Table 19. Samples for exploratory biomarkers should also be sent for analysis from any lumbar puncture or BM sample collection (aspirate/biopsy) performed following CTX120 infusion. In the event of CRS, samples for exploratory biomarker assessment are collected every 48 hours between scheduled visits until CRS resolves.


7. Safety, Adverse Events, and Study Oversight

AEs in response to a query, observed by site personnel, or reported spontaneously by the subject are recorded.


7.1. Adverse Events


An AE is any untoward medical occurrence in a patient or clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. An AE can therefore be any unfavorable and unintended sign (including an abnormal laboratory finding, for example), symptom or disease temporally associated with the use of a medicinal (investigational) product whether or not considered related to the medicinal (investigational) product [(GCP) E6(R2)]. In clinical studies, an AE can include an undesirable medical condition occurring at any time, including screening or washout periods, even if no study treatment has been administered.


The following are considered to be AEs:

    • Aggravation of a pre-existing disease or permanent disorder (any clinically significant worsening in the nature, severity, frequency, or duration of a pre-existing condition)
    • Events resulting from protocol-mandated procedures (e.g., complications from invasive procedures)


The following are not considered to be AEs:

    • Medical or surgical procedures including elective or pre-planned such as surgery, endoscopy, tooth extraction, transfusion. These should be recorded in the relevant eCRF.
      • Note: an untoward medical event occurring during the prescheduled elective procedure or routinely scheduled treatment should be recorded as an AE or SAE
    • Pre-existing diseases or conditions that do not worsen during or after administration of the investigational medicinal product
    • Hospitalization planned for study treatment infusion or observation
    • The malignancy under study or signs and symptoms associated with the disease, as well as progression or relapse of the underlying malignancy (see Section 8.2 Disease Progression)


Abnormal laboratory results without clinical significance should not be recorded as AEs.


7.2. Disease Progression


Disease progression and/or signs and symptoms of disease progression should not be reported as an AE with the following exceptions:

    • Atypical or accelerated progression of malignancy under study that in its nature, presentation, or severity differ from the normal course of the disease, with symptoms meeting serious criteria. In this case worsening of underlying condition should be reported as the SAE.
    • Disease progression with outcome of death within 30 days of CTX120 infusion regardless of relationship to CTX120 should be recorded as SAE and reported.


7.3. Serious Adverse Event


An AE of any untoward medical consequence must be classified as a serious adverse event if it meets any of the following criteria:

    • Results in death
    • Is life-threatening (i.e., an AE that places the subject at immediate risk of death)
    • Requires in-patient hospitalization or prolongs an existing hospitalization (hospitalizations for scheduled medical or surgical procedures or to conduct scheduled observation and treatments do not meet these criteria)
    • Results in persistent or significant disability or incapacity
    • Results in a congenital anomaly or birth defect in the newborn
    • Other important/significant medical events. Important medical events that may not result in death, be life-threatening, or require hospitalization may be considered serious when, based upon appropriate medical judgement, they may jeopardize the patient or subject and may require medical or surgical intervention to prevent one of the outcomes listed in this definition.


Hospitalization for study treatment infusions, or planned hospitalizations following CTX120 infusion, are not considered SAEs. Furthermore, hospitalizations for observation or prolongation of hospitalization for observation alone should not be reported as an SAE unless they are associated with a medically significant event that meets other SAE criteria.


7.4. Adverse Events of Special Interest


AESIs must be reported any time after CTX120 infusion and include:

    • CTX120 infusion reactions
    • Grade≥3 opportunistic/invasive infections
    • Grade≥3 tumor lysis syndrome
    • CRS
    • ICANS
    • Hemophagocytic lymphohistiocytosis
    • GvHD
    • Secondary malignancy
    • Uncontrolled T cell proliferation
    • Any new hematological or autoimmune disorder that is determined to be possibly related or related to CTX120


7.5. Adverse Event Severity


AEs are graded according to CTCAE v5.0, with the exception of CRS, neurotoxicity, and GvHD, which are graded according to the criteria provided herein. When a CTCAE grade or protocol-specified criteria are not available, the toxicity grading in Table 26 can be used.









TABLE 26





Adverse Event Severity


















Grade 1
Mild; asymptomatic or mild




symptoms; clinical or diagnostic




observations only; intervention not indicated.



Grade 2
Moderate; minimal, local, or




noninvasive intervention indicated;




limiting age- appropriate instrumental ADL.1



Grade 3
Severe or medically significant




but not immediately life-threatening;




hospitalization or prolongation of




hospitalization indicated; disabling;




limiting self-care ADL.2



Grade 4
Life-threatening consequences;




urgent intervention indicated.



Grade 5
Death related to AE.







ADL: Activities of Daily Living;



AE: adverse event.




1Instrumental ADL refer to preparing meals, shopping for groceries or clothes, using the telephone, managing money, etc.





2Self-care ADL refer to bathing, dressing and undressing, feeding self, using the toilet, taking medications, and not bedridden.







7.6. Adverse Event Causality


The assessment of relationship is made based on the following definitions:

    • Related: There is a clear causal relationship between the study treatment or procedure and the AE.
    • Possibly related: There is some evidence to suggest a causal relationship between the study treatment or procedure and the AE, but alternative potential causes also exist.
    • Not related: There is no evidence to suggest a causal relationship between the study treatment or procedure and the AE.


If an SAE is assessed to be not related to any study intervention, an alternative etiology must be provided in the CRF. If the relationship between the AE/SAE and the investigational product is determined to be “possible,” a rationale for the assessment must be provided.


7.7. Outcome


The outcome of an AE or SAE classified and reported as follows:

    • Fatal
    • Not recovered/not resolved
    • Recovered/resolved
    • Recovered/resolved with sequelae
    • Recovering/resolving
    • Unknown


7.8. Adverse Event Collection Period


The safety-related information of all subjects enrolled in this study is recorded from the time of ICF signing until end of study; however, there are different reporting requirements for different time periods in the study. Table 27 describes the AEs that should be recorded and reported at each time period of the study.









TABLE 27







Adverse Event Collection by Study Time Period








Time Period
AE Reporting Requirements





Informed consent to 3 months
All AEs


after each CTX120 infusion



3 months after last CTX120
SAEs AESIs


infusion through Month 24 visit



Month 24 to Month 60 visit
CTX120-related SAEs CTX120-


or after a subject receives a new
related AESIs


anticancer therapy after Month 3 visit
New malignancies





AE: adverse event;


AESI: adverse event of special interest;


SAE: serious adverse event.







If a subject receives a new anticancer therapy within 3 months of a CTX120 infusion, all SAEs and AESIs should be reported until 3 months after the CTX120 infusion. If a subject starts a new anticancer therapy more than 3 months after a CTX120 infusion, only CTX120-related SAEs and CTX120-related AESIs, and new malignancies are reported. If a subject does not receive CTX120 therapy after enrollment, the AE reporting period ends 30 days after last study-related procedure (e.g., biopsy, imaging, LD chemotherapy).


8. Stopping Rules and Study Termination

8.1. Stopping Rules for Trial


The study is paused if 1 or more of the following events occur:

    • Life-threatening (grade 4) toxicity attributable to CTX120 that is unmanageable and unexpected
    • Death related to CTX120 within 30 days of infusion
    • Grade≥3 GvHD
    • After at least 12 subjects are enrolled in cohort expansion and at least 1 of the following occurs:
      • >35% grade 3 or 4 neurotoxicity not resolving within 7 days to grade≤2
      • >20% grade≥2 GvHD that is steroid-refractory
      • >30% grade 4 CRS
      • >50% grade 4 neutropenia not resolving within 28 days (except for subjects with baseline neutropenia)
      • >30% grade 4 infections
    • New malignancy (distinct from recurrence/progression of previously-treated malignancy)
    • Lack of efficacy, defined as 2 or fewer responses (including PR+VGPR+CR+stringent complete response [sCR]) after 15 subjects in cohort expansion have 3 months of post-CTX120 assessment


8.2. Stopping Rules for Individual Subjects


Stopping rules for individual subjects are as follows:

    • Any medical condition that would put the subject at risk during continuing study-related treatments or follow-up
    • If a subject is found not to have met eligibility criteria or has a major protocol deviation before the start of LD chemotherapy (Cohort 2) or before the start of daratumumab infusion (Cohorts 1 and 3)


9. Statistical Analyses

9.1. Study Objectives and Hypotheses


The primary objective of Part A is to assess the safety of escalating doses of CTX120 in combination with various LD and immunomodulatory agents in subjects with relapsed or refractory multiple myeloma to determine the MTD and/or recommended dose and regimen for Part B cohort expansion.


The primary objective of Part B is to assess the efficacy of CTX120 in subjects with relapsed or refractory multiple myeloma, as measured by ORR according to IMWG response criteria.


9.2. Study Endpoints


Primary Endpoints


Part A (Dose Escalation): Incidence of AEs defined as DLTs


Part B (Cohort Expansion): Objective response rate (sCR+CR+VGPR+PR), per IMWG response criteria


Part A and B Secondary Endpoints


Efficacy

    • Percentage of subjects with sCR, per IMWG response criteria (Table 22)
    • Percentage of subjects with CR, per IMWG response criteria (Table 22)
    • Percentage of subjects with VGPR, per IMWG response criteria (Table 22)
    • Duration of response is defined as the time between first objective response of sCR/CR/VGPR/PR and disease progression (by IMWG response criteria) or death due to any cause that followed the same objective response.
    • Cumulative duration of response is calculated as the time between the first response of PR or better and the disease progression or death that followed the last objective response a subject ever achieved.
    • Progression-free survival is defined as the time between CTX120 infusion and disease progression (by IMWG response criteria) or death due to any cause. Subjects who have no disease progression will be censored at their last multiple myeloma disease assessment date.
    • Overall survival is defined as the time between CTX120 infusion and death due to any cause. Subjects who are alive are censored at their last date known to be alive.


Safety


Incidence and severity of AEs and clinically significant laboratory abnormalities are summarized and reported according to CTCAE v5.0, except for CRS, which is graded according to ASTCT criteria (Lee et al., 2019); neurotoxicity, which is graded according to ICANS (Lee et al., 2019) and CTCAE v5.0; and GvHD, which is graded according to MAGIC criteria (Harris et al., 2016).


Pharmacokinetics


The levels of CTX120 in blood and other tissues over time are assessed using a PCR assay that measures copies of CAR construct per μg DNA. Complementary analyses using flow cytometry to identify CTX120 in blood may also be performed.


The trafficking of CTX120 in bone marrow, CSF, or extramedullary plasmacytoma tissues may be evaluated in any of these samples collected as per protocol-specific sampling.


Exploratory Endpoints

    • Levels of cytokines in blood and other tissues
    • Incidence of anti-CTX120 antibodies
    • Impact of anti-cytokine therapy on CTX120 proliferation, CRS, and disease response
    • Time to response, defined as the time between the date of CTX120 infusion until first documented response (sCR/CR/VGPR/PR)
    • Time to CR, defined as the time between the date of CTX120 infusion until first documented CR
    • Time to disease progression, defined as time between the date of CTX120 infusion until first evidence of disease progression
    • Percentage of subjects who are MRD-negative
    • Incidence of autologous or allogeneic SCT following CTX120 infusion
    • Incidence and type of subsequent anticancer therapy
    • Anticancer therapy-free survival, defined as the time between date of CTX120 infusion and date of first subsequent anticancer therapy or death due to any cause
    • Other exploratory endpoints


9.3. Analysis Sets


Part A (Dose Escalation)


The DLT evaluable set (DES) includes all subjects who receive CTX120 and complete the DLT evaluation period or discontinue early after experiencing a DLT. The DES is used for determination of the recommended dose for Part B.


Part A+Part B (Dose Escalation+Cohort Expansion)


The enrolled set includes subjects who sign informed consent, meet eligibility criteria, and enroll in the study. The enrolled set is classified according to the assigned dose level of CTX120 and is used for additional analyses of the primary and secondary endpoints.


The treated set includes all subjects who receive any study treatment. The subjects in the treated set are classified according to the received study treatment.


The full analysis set (FAS) includes all subjects who receive CTX120 infusion and have had the opportunity to be followed for at least 3 months (i.e., completed at least 3 months of follow-up or discontinued prior to data cutoff). The FAS is the primary analysis set for disease response assessment.


The safety analysis set (SAS) includes all subjects who receive CTX120 infusion. The subjects in the SAS are classified according to the received dose level of CTX120. The SAS is the primary analysis set for safety assessment of CTX120.


9.4. Sample Size


The sample size in the dose escalation part of the study is approximately 6 to 78 subjects, depending on the number of dose levels and cohorts evaluated, and the occurrence of DLTs. If the study proceeds to cohort expansion (Part B), an optimal Simon 2-stage design is employed independently for each selected cohort. In the first stage, up to 27 subjects are enrolled and treated with CTX120. If the study proceeds to the second stage after the interim analysis, additional subjects are enrolled to achieve a final sample size of 70. Assuming the true ORR of the selected CTX120 dose and regimen is 50%, the study has 90% power (α=0.05, 2-sided) for a 1-sample test of ORR equal to a historical ORR of 30%. The historical ORR is the approximate ORR for currently approved third-line pomalidomide+dexamethasone combination (Miguel et al., Lancet Oncol 14, 1055-1066; 2013), or fourth-line daratumumab monotherapy (Lonial et al., Lancet 387, 1551-1560; 2016) in patients with multiple myeloma.


9.5. Planned Method of Analyses


Efficacy Analysis


The primary analysis of the primary endpoint of ORR is based on independent central review of multiple myeloma disease assessments in the FAS.


Tabulations are produced for appropriate demographic, baseline, efficacy, and safety parameters. ORR is summarized as a proportion with exact 95% confidence interval, and an exact binomial test will be used to compare the observed response rate to an historical response rate of 30%. For time-to-event variables such as duration of response, cumulative duration of response, progression-free survival, and overall survival, medians with 95% confidence intervals are calculated using Kaplan-Meier methods.


Safety Analysis


All safety analysis are based on the SAS. AEs are graded according to CTCAE v5.0, except for CRS (Lee criteria for Part A, ASTCT criteria for Part B), neurotoxicity (ICANS and CTCAE v5.0), and GvHD (MAGIC criteria). The AEs, SAEs, and AESIs are summarized by dose cohort and reported according to the study time period described in Table 27.


Treatment-emergent AEs are defined as AEs that start or worsen on or after the initial CTX120 infusion.


Frequencies of subjects experiencing at least 1 AE are reported by body system and preferred term according to Medical Dictionary for Regulatory Activities (MedDRA) terminology.


Detailed information collected for each AE include description of the event, duration, whether the AE was serious, intensity, relationship to study drug, action taken, clinical outcome, and whether or not it was a DLT. Emphasis in the analysis is placed on AEs classified as dose-limiting.


Vital signs are summarized using descriptive statistics. Summary tables are prepared to examine the distribution of laboratory measures over time.


Pharmacokinetic and Pharmacodynamic Analyses


Levels of CTX120 CAR+ T cells in blood, incidence of anti-CTX120 antibodies, and levels of cytokines in serum are summarized.


Biomarker Analysis


Investigation of additional biomarkers may include assessment of blood components (serum, plasma, and cells), cells from other tissues, extramedullary plasmacytoma tissue, and other subject-derived tissue. These assessments may evaluate DNA, RNA, proteins, and other biologic molecules derived from those tissues. Such evaluations will inform understanding of factors related to the subjects' disease, response to CTX120, and the mechanism of action of the investigational product.


Patient-Reported Outcomes


Descriptive statistics will be presented for PRO, both as reported and as change from baseline.


Results

A number of eligible human multiple myeloma patients were treated by CTX120 alone at multiple doses (e.g., DL3 and DL4), or treated by the combined therapy of CTX120 and daratumumab or the combined therapy of CTX120 and lenalidomide, following the treatment regimens for Cohorts 1 and 2 disclosed herein. In the combined therapy, the patients were given DL3 or DL4 of CTX120.


Preliminary results from the clinical trial disclosed herein show that, at equivalent dose levels, patients treated with either darabumumab or lenalidomide in combination with CTX120 (in Cohorts 1 and 2) exhibited increased depletion of NK cells and lymphocytes as compared with CTX120 monotherapy. FIGS. 26 and 27. Lenalidomide was also observed to enhance CTX120 expansion in human patients. FIG. 28. In addition, at equivalent dose levels, patients treated with either darabumumab or lenalidomide in combination with CTX120 showed higher levels of circulating CAR-T cells and increased anti-myeloma activity compared with CTX120 monotherapy.


Sequence Tables

The following tables provide details for the various nucleotide and amino acid sequences disclosed herein.









TABLE 1







sgRNA Sequences and Target Gene Sequences













SEQ





ID





NO: 










sgRNA Sequences










TRAC sgRNA
Modified
A*G*A*GCAACAGUGCUGUG
1




GCCguuuuagagcuagaaau





agcaaguuaaaauaaggcua





guccguuaucaacuugaaaa





aguggcaccgagucggugcU





*U*U*U







Unmodified
AGAGCAACAGUGCUGUGGCC
2




guuuuagagcuagaaauagc





aaguuaaaauaaggcuaguc





cguuaucaacuugaaaaagu





ggcaccgagucggugcUUUU






TRAC sgRNA
Modified
A*G*A*GCAACAGUGCUGUG
3


spacer

GCC







Unmodified
AGAGCAACAGUGCUGUGGCC
4





β2M sgRNA
Modified
G*C*U*ACUCUCUCUUUCUG
5




GCCguuuuagagcuagaaau





agcaaguuaaaauaaggcua





guccguuaucaacuugaaaa





aguggcaccgagucggugcU





*U*U*U






TRAC sgRNA
Modified
A*G*A*GCAACAGUGCUGUG
1




GCCguuuuagagcuagaaau





agcaaguuaaaauaaggcua





guccguuaucaacuugaaaa





aguggcaccgagucggugcU





*U*U*U







Unmodified
AGAGCAACAGUGCUGUGGCC
2




guuuuagagcuagaaauagc





aaguuaaaauaaggcuaguc





cguuaucaacuugaaaaagu





ggcaccgagucggugcUUUU




Unmodified
GCUACUCUCUCUUUCUGGCC
6




guuuuagagcuagaaauagc





aaguuaaaauaaggcuaguc





cguuaucaacuugaaaaagu





ggcaccgagucggugcUU U





U






β2M sgRNA
Modified
G*C*U*ACUCUCUCUUUCUGGCC
7


spacer
Unmodified
GCUACUCUCUCUUUCUGGCC
8











Target Sequences (PAM)










TRAC
AGAGCAACAGTGCTGTGGCC(TGG)
9





TRAC
AGAGCAACAGTGCTGTGGCC
10





β2M
GCTACTCTCTCTTTCTGGCC(TGG)
11





β2M
GCTACTCTCTCTTTCTGGCC
12 
















TABLE 2







Edited TRAC Gene Sequence












Sequence (Deletions





indicated by dashes (-);
SEQ




insertions indicated by
ID



Description
bold)
NO:






TRAC gene
AA---------------------G
13



edit
AGCAACAAATCTGACT







TRAC gene
AAGAGCAACAGTGCTGT-GCCTGG
14



edit
AGCAACAAATCTGACT







TRAC gene
AAGAGCAACAGTG-------CTGG
15



edit
AGCAACAAATCTGACT







TRAC gene
AAGAGCAACAGT------GCCTGG
16



edit
AGCAACAAATCTGACT







TRAC gene
AAGAGCAACAGTG-----------
17



edit
----------CTGACT







TRAC gene
AAGAGCAACAGTGCTGTGGGCCTG
18



edit
GAGCAACAAATCTGACT







TRAC gene
AAGAGCAACAGTGC TGGCCTGG
19



edit
AGCAACAAATCTGACT







TRAC gene
AAGAGCAACAGTGCTGTGTGCCT
20



edit
GGAGCAACAAATCTGACT
















TABLE 3







Edited β2M gene-edit Gene Sequence












Sequence (Deletions
SEQ




indicated by dashes (-);
ID




insertions indicated by




Description
bold)
NO:






β2M gene-
CGTGGCCTTAGCTGTGCTCGCGCT
21



edit
ACTCTCTCTTTCT-GCCTGGAGGC





TATCCAGCGTGAGTCTCTCCTACC





CTCCCGCT







β2M gene-
CGTGGCCTTAGCTGTGCTCGCGCT
22



edit
ACTCTCTCTTTC GCCTGGAGGCT





ATCCAGCGTGAGTCTCTCCTACCC





TCCCGCT







β2M gene-
CGTGGCCTTAGCTGTGCTCGCGCT
23



edit
ACTCTCTCTTT-----CTGGAGGC





TATCCAGCGTGAGTCTCTCCTACC





CTCCCGCT







β2M gene-
CGTGGCCTTAGCTGTGCTCGCGCT
24



edit
ACTCTCTCTTTCTGGATAGCCTGG





AGGCTATCCAGCGTGAGTCTCTCC





TACCCTCCCGCT







β2M gene-
CGTGGCCTTAGCTGTGCTCGC---
25



edit
----------------------GC





TATCCAGCGTGAGTCTCTCCTACC





CTCCCGCT







β2M gene-
CGTGGCCTTAGCTGTGCTCGCGCT
26



edit
ACTCTCTCTTTCTGTGGCCTGGAG





GCTATCCAGCGTGAGTCTCTCCTA





CCCTCCCGCT
















TBALE 4







Gene Editing/CAR Construct Components


(Nucleotide Sequences)











Name

SEQ



Description
Nucleotide Sequence
ID NO:






CTX-166b
CCTGCAGGCAGCTGCGCGCTCGCTC
27



rAAV
GCTCACTGAGGCCGCCCGGGCGTCG





GGCGACCTTTGGTCGCCCGGCCTCA





GTGAGCGAGCGAGCGCGCAGAGAGG





GAGTGGCCAACTCCATCACTAGGGG





TTCCTGCGGCCGCACGCGTGAGATG





TAAGGAGCTGCTGTGACTTGCTCAA





GGCCTTATATCGAGTAAACGGTAGT





GCTGGGGCTTAGACGCAGGTGTTCT





GATTTATAGTTCAAAACCTCTATCA





ATGAGAGAGCAATCTCCTGGTAATG





TGATAGATTTCCCAACTTAATGCCA





ACATACCATAAACCTCCCATTCTGC





TAATGCCCAGCCTAAGTTGGGGAGA





CCACTCCAGATTCCAAGATGTACAG





TTTGCTTTGCTGGGCCTTTTTCCCA





TGCCTGCCTTTACTCTGCCAGAGTT





ATATTGCTGGGGTTTTGAAGAAGAT





CCTATTAAATAAAAGAATAAGCAGT





ATTATTAAGTAGCCCTGCATTTCAG





GTTTCCTTGAGTGGCAGGCCAGGCC





TGGCCGTGAACGTTCACTGAAATCA





TGGCCTCTTGGCCAAGATTGATAGC





TTGTGCCTGTCCCTGAGTCCCAGTC





CATCACGAGCAGCTGGTTTCTAAGA





TGCTATTTCCCGTATAAAGCATGAG





ACCGTGACTTGCCAGCCCCACAGAG





CCCCGCCCTTGTCCATCACTGGCAT





CTGGACTCCAGCCTGGGTTGGGGCA





AAGAGGGAAATGAGATCATGTCCTA





ACCCTGATCCTCTTGTCCCACAGAT





ATCCAGAACCCTGACCCTGCCGTGT





ACCAGCTGAGAGACTCTAAATCCAG





TGACAAGTCTGTCTGCCTATTCACC





GATTTTGATTCTCAAACAAATGTGT





CACAAAGTAAGGATTCTGATGTGTA





TATCACAGACAAAACTGTGCTAGAC





ATGAGGTCTATGGACTTCAGGCTCC





GGTGCCCGTCAGTGGGCAGAGCGCA





CATCGCCCACAGTCCCCGAGAAGTT





GGGGGGAGGGGTCGGCAATTGAACC





GGTGCCTAGAGAAGGTGGCGCGGGG





TAAACTGGGAAAGTGATGTCGTGTA





CTGGCTCCGCCTTTTTCCCGAGGGT





GGGGGAGAACCGTATATAAGTGCAG





TAGTCGCCGTGAACGTTCTTTTTCG





CAACGGGTTTGCCGCCAGAACACAG





GTAAGTGCCGTGTGTGGTTCCCGCG





GGCCTGGCCTCTTTACGGGTTATGG





CCCTTGCGTGCCTTGAATTACTTCC





ACTGGCTGCAGTACGTGATTCTTGA





TCCCGAGCTTCGGGTTGGAAGTGGG





TGGGAGAGTTCGAGGCCTTGCGCTT





AAGGAGCCCCTTCGCCTCGTGCTTG





AGTTGAGGCCTGGCCTGGGCGCTGG





GGCCGCCGCGTGCGAATCTGGTGGC





ACCTTCGCGCCTGTCTCGCTGCTTT





CGATAAGTCTCTAGCCATTTAAAAT





TTTTGATGACCTGCTGCGACGCTTT





TTTTCTGGCAAGATAGTCTTGTAAA





TGCGGGCCAAGATCTGCACACTGGT





ATTTCGGTTTTTGGGGCCGCGGGCG





GCGACGGGGCCCGTGCGTCCCAGCG





CACATGTTCGGCGAGGCGGGGCCTG





CGAGCGCGGCCACCGAGAATCGGAC





GGGGGTAGTCTCAAGCTGGCCGGCC





TGCTCTGGTGCCTGGCCTCGCGCCG





CCGTGTATCGCCCCGCCCTGGGCGG





CAAGGCTGGCCCGGTCGGCACCAGT





TGCGTGAGCGGAAAGATGGCCGCTT





CCCGGCCCTGCTGCAGGGAGCTCAA





AATGGAGGACGCGGCGCTCGGGAGA





GCGGGCGGGTGAGTCACCCACACAA





AGGAAAAGGGCCTTTCCGTCCTCAG





CCGTCGCTTCATGTGACTCCACGGA





GTACCGGGCGCCGTCCAGGCACCTC





GATTAGTTCTCGAGCTTTTGGAGTA





CGTCGTCTTTAGGTTGGGGGGAGGG





GTTTTATGCGATGGAGTTTCCCCAC





ACTGAGTGGGTGGAGACTGAAGTTA





GGCCAGCTTGGCACTTGATGTAATT





CTCCTTGGAATTTGCCCTTTTTGAG





TTTGGATCTTGGTTCATTCTCAAGC





CTCAGACAGTGGTTCAAAGTTTTTT





TCTTCCATTTCAGGTGTCGTGA





CCACCATGGCGCTTCCGGTGACAGC





ACTGCTCCTCCCCTTGGCGCTGTTG





CTCCACGCAGCAAGGCCGCAGGTGC





AGCTGGTGCAGAGCGGAGCCGAGCT





CAAGAAGCCCGGAGCCTCCGTGAAG





GTGAGCTGCAAGGCCAGCGGCAACA





CCCTGACCAACTACGTGATCCACTG





GGTGAGACAAGCCCCCGGCCAAAGG





CTGGAGTGGATGGGCTACATCCTGC





CCTACAACGACCTGACCAAGTACAG





CCAGAAGTTCCAGGGCAGGGTGACC





ATCACCAGGGATAAGAGCGCCTCCA





CCGCCTATATGGAGCTGAGCAGCCT





GAGGAGCGAGGACACCGCTGTGTAC





TACTGTACAAGGTGGGACTGGGACG





GCTTCTTTGACCCCTGGGGCCAGGG





CACAACAGTGACCGTCAGCAGCGGC





GGCGGAGGCAGCGGCGGCGGCGGCA





GCGGCGGAGGCGGAAGCGAAATCGT





GATGACCCAGAGCCCCGCCACACTG





AGCGTGAGCCCTGGCGAGAGGGCCA





GCATCTCCTGCAGGGCTAGCCAAAG





CCTGGTGCACAGCAACGGCAACACC





CACCTGCACTGGTACCAGCAGAGAC





CCGGACAGGCTCCCAGGCTGCTGAT





CTACAGCGTGAGCAACAGGTTCTCC





GAGGTGCCTGCCAGGTTTAGCGGCA





GCGGAAGCGGCACCGACTTTACCCT





GACCATCAGCAGCGTGGAGTCCGAG





GACTTCGCCGTGTATTACTGCAGCC





AGACCAGCCACATCCCTTACACCTT





CGGCGGCGGCACCAAGCTGGAGATC





AAAAGTGCTGCTGCCTTTGTCCCGG





TATTTCTCCCAGCCAAACCGACCAC





GACTCCCGCCCCGCGCCCTCCGACA





CCCGCTCCCACCATCGCCTCTCAAC





CTCTTAGTCTTCGCCCCGAGGCATG





CCGACCCGCCGCCGGGGGTGCTGTT





CATACGAGGGGCTTGGACTTCGCTT





GTGATATTTACATTTGGGCTCCGTT





GGCGGGTACGTGCGGCGTCCTTTTG





TTGTCACTCGTTATTACTTTGTATT





GTAATCACAGGAATCGCAAACGGGG





CAGAAAGAAACTCCTGTATATATTC





AAACAACCATTTATGAGACCAGTAC





AAACTACTCAAGAGGAAGATGGCTG





TAGCTGCCGATTTCCAGAAGAAGAA





GAAGGAGGATGTGAACTGCGAGTGA





AGTTTTCCCGAAGCGCAGACGCTCC





GGCATATCAGCAAGGACAGAATCAG





CTGTATAACGAACTGAATTTGGGAC





GCCGCGAGGAGTATGACGTGCTTGA





TAAACGCCGGGGGAGAGACCCGGAA





ATGGGGGGTAAACCCCGAAGAAAGA





ATCCCCAAGAAGGACTCTACAATGA





ACTCCAGAAGGATAAGATGGCGGAG





GCCTACTCAGAAATAGGTATGAAGG





GCGAACGACGACGGGGAAAAGGTCA





CGATGGCCTCTACCAAGGGTTGAGT





ACGGCAACCAAAGATACGTACGATG





CACTGCATATGCAGGCCCTGCCTCC





CAGATAATAATAAAATCGCTATCCA





TCGAAGATGGATGTGTGTTGGTTTT





TTGTGTGTGGAGCAACAAATCTGAC





TTTGCATGTGCAAACGCCTTCAACA





ACAGCATTATTCCAGAAGACACCTT





CTTCCCCAGCCCAGGTAAGGGCAGC





TTTGGTGCCTTCGCAGGCTGTTTCC





TTGCTTCAGGAATGGCCAGGTTCTG





CCCAGAGCTCTGGTCAATGATGTCT





AAAACTCCTCTGATTGGTGGTCTCG





GCCTTATCCATTGCCACCAAAACCC





TCTTTTTACTAAGAAACAGTGAGCC





TTGTTCTGGCAGTCCAGAGAATGAC





ACGGGAAAAAAGCAGATGAAGAGAA





GGTGGCAGGAGAGGGCACGTGGCCC





AGCCTCAGTCTCTCCAACTGAGTTC





CTGCCTGCCTGCCTTTGCTCAGACT





GTTTGCCCCTTACTGCTCTTCTAGG





CCTCATTCTAAGCCCCTTCTCCAAG





TTGCCTCTCCTTATTTCTCCCTGTC





TGCCAAAAAATCTTTCCCAGCTCAC





TAAGTCAGTCTCACGCAGTCACTCA





TTAACCCACCAATCACTGATTGTGC





CGGCACATGAATGCACCAGGTGTTG





AAGTGGAGGAATTAAAAAGTCAGAT





GAGGGGTGTGCCCAGAGGAAGCACC





ATTCTAGTTGGGGGAGCCCATCTGT





CAGCTGGGAAAAGTCCAAATAACTT





CAGATTGGAATGTGTTTTAACTCAG





GGTTGAGAAAACAGCTACCTTCAGG





ACAAAAGTCAGGGAAGGGCTCTCTG





AAGAAATGCTACTTGAAGATACCAG





CCCTACCAAGGGCAGGGAGAGGACC





CTATAGAGGCCTGGGACAGGAGCTC





AATGAGAAAGGTAACCACGTGCGGA





CCGAGGCTGCAGCGTCGTCCTCCCT





AGGAACCCCTAGTGATGGAGTTGGC





CACTCCCTCTCTGCGCGCTCGCTCG





CTCACTGAGGCCGGGCGACCAAAGG





TCGCCCGACGCCCGGGCTTTGCCCG





GGCGGCCTCAGTGAGCGAGCGAGCG





CGCAGCTGCCTGCAGG







5′ ITR
CCTGCAGGCAGCTGCGCGCTCGCTC
28




GCTCACTGAGGCCGCCCGGGCGTCG





GGCGACCTTTGGTCGCCCGGCCTCA





GTGAGCGAGCGAGCGCGCAGAGAGG





GAGTGGCCAACTCCATCACTAGGGG





TTCCT







3′ ITR
AGGAACCCCTAGTGATGGAGTTGGC
29




CACTCCCTCTCTGCGCGCTCGCTCG





CTCACTGAGGCCGGGCGACCAAAGG





TCGCCCGACGCCCGGGCTTTGCCCG





GGCGGCCTCAGTGAGCGAGCGAGCG





CGCAGCTGCCTGCAGG







LHA to
GAGATGTAAGGAGCTGCTGTGACTT
30



RHA
GCTCAAGGCCTTATATCGAGTAAAC




(CTX-166b)
GGTAGTGCTGGGGCTTAGACGCAGG





TGTTCTGATTTATAGTTCAAAACCT





CTATCAATGAGAGAGCAATCTCCTG





GTAATGTGATAGATTTCCCAACTTA





ATGCCAACATACCATAAACCTCCCA





TTCTGCTAATGCCCAGCCTAAGTTG





GGGAGACCACTCCAGATTCCAAGAT





GTACAGTTTGCTTTGCTGGGCCTTT





TTCCCATGCCTGCCTTTACTCTGCC





AGAGTTATATTGCTGGGGTTTTGAA





GAAGATCCTATTAAATAAAAGAATA





AGCAGTATTATTAAGTAGCCCTGCA





TTTCAGGTTTCCTTGAGTGGCAGGC





CAGGCCTGGCCGTGAACGTTCACTG





AAATCATGGCCTCTTGGCCAAGATT





GATAGCTTGTGCCTGTCCCTGAGTC





CCAGTCCATCACGAGCAGCTGGTTT





CTAAGATGCTATTTCCCGTATAAAG





CATGAGACCGTGACTTGCCAGCCCC





ACAGAGCCCCGCCCTTGTCCATCAC





TGGCATCTGGACTCCAGCCTGGGTT





GGGGCAAAGAGGGAAATGAGATCAT





GTCCTAACCCTGATCCTCTTGTCCC





ACAGATATCCAGAACCCTGACCCTG





CCGTGTACCAGCTGAGAGACTCTAA





ATCCAGTGACAAGTCTGTCTGCCTA





TTCACCGATTTTGATTCTCAAACAA





ATGTGTCACAAAGTAAGGATTCTGA





TGTGTATATCACAGACAAAACTGTG





CTAGACATGAGGTCTATGGACTTCA





GGCTCCGGTGCCCGTCAGTGGGCAG





AGCGCACATCGCCCACAGTCCCCGA





GAAGTTGGGGGGAGGGGTCGGCAAT





TGAACCGGTGCCTAGAGAAGGTGGC





GCGGGGTAAACTGGGAAAGTGATGT





CGTGTACTGGCTCCGCCTTTTTCCC





GAGGGTGGGGGAGAACCGTATATAA





GTGCAGTAGTCGCCGTGAACGTTCT





TTTTCGCAACGGGTTTGCCGCCAGA





ACACAGGTAAGTGCCGTGTGTGGTT





CCCGCGGGCCTGGCCTCTTTACGGG





TTATGGCCCTTGCGTGCCTTGAATT





ACTTCCACTGGCTGCAGTACGTGAT





TCTTGATCCCGAGCTTCGGGTTGGA





AGTGGGTGGGAGAGTTCGAGGCCTT





GCGCTTAAGGAGCCCCTTCGCCTCG





TGCTTGAGTTGAGGCCTGGCCTGGG





CGCTGGGGCCGCCGCGTGCGAATCT





GGTGGCACCTTCGCGCCTGTCTCGC





TGCTTTCGATAAGTCTCTAGCCATT





TAAAATTTTTGATGACCTGCTGCGA





CGCTTTTTTTCTGGCAAGATAGTCT





TGTAAATGCGGGCCAAGATCTGCAC





ACTGGTATTTCGGTTTTTGGGGCCG





CGGGCGGCGACGGGGCCCGTGCGTC





CCAGCGCACATGTTCGGCGAGGCGG





GGCCTGCGAGCGCGGCCACCGAGAA





TCGGACGGGGGTAGTCTCAAGCTGG





CCGGCCTGCTCTGGTGCCTGGCCTC





GCGCCGCCGTGTATCGCCCCGCCCT





GGGCGGCAAGGCTGGCCCGGTCGGC





ACCAGTTGCGTGAGCGGAAAGATGG





CCGCTTCCCGGCCCTGCTGCAGGGA





GCTCAAAATGGAGGACGCGGCGCTC





GGGAGAGCGGGCGGGTGAGTCACCC





ACACAAAGGAAAAGGGCCTTTCCGT





CCTCAGCCGTCGCTTCATGTGACTC





CACGGAGTACCGGGCGCCGTCCAGG





CACCTCGATTAGTTCTCGAGCTTTT





GGAGTACGTCGTCTTTAGGTTGGGG





GGAGGGGTTTTATGCGATGGAGTTT





CCCCACACTGAGTGGGTGGAGACTG





AAGTTAGGCCAGCTTGGCACTTGAT





GTAATTCTCCTTGGAATTTGCCCTT





TTTGAGTTTGGATCTTGGTTCATTC





TCAAGCCTCAGACAGTGGTTCAAAG





TTTTTTTCTTCCATTTCAGGTGTCG





TGACCACCATGGCGCTTCCGGTGAC





AGCACTGCTCCTCCCCTTGGCGCTG





TTGCTCCACGCAGCAAGGCCGCAGG





TGCAGCTGGTGCAGAGCGGAGCCGA





GCTCAAGAAGCCCGGAGCCTCCGTG





AAGGTGAGCTGCAAGGCCAGCGGCA





ACACCCTGACCAACTACGTGATCCA





CTGGGTGAGACAAGCCCCCGGCCAA





AGGCTGGAGTGGATGGGCTACATCC





TGCCCTACAACGACCTGACCAAGTA





CAGCCAGAAGTTCCAGGGCAGGGTG





ACCATCACCAGGGATAAGAGCGCCT





CCACCGCCTATATGGAGCTGAGCAG





CCTGAGGAGCGAGGACACCGCTGTG





TACTACTGTACAAGGTGGGACTGGG





ACGGCTTGTTGACCCCTGGGGCCAG





GGCACAACAGTGACCGTCAGCAGCG





GCGGCGGAGGCAGCGGCGGCGGCGG





CAGCGGCGGAGGCGGAAGCGAAATC





GTGATGACCCAGAGCCCCGCCACAC





TGAGCGTGAGCCCTGGCGAGAGGGC





CAGCATCTCCTGCAGGGCTAGCCAA





AGCCTGGTGCACAGCAACGGCAACA





CCCACCTGCACTGGTACCAGCAGAG





ACCCGGACAGGCTCCCAGGCTGCTG





ATCTACAGCGTGAGCAACAGGTTCT





CCGAGGTGCCTGCCAGGTTTAGCGG





CAGCGGAAGCGGCACCGACTTTACC





CTGACCATCAGCAGCGTGGAGTCCG





AGGACTTCGCCGTGTATTACTGCAG





CCAGACCAGCCACATCCCTTACACC





TTCGGCGGCGGCACCAAGCTGGAGA





TCAAAAGTGCTGCTGCCTTTGTCCC





GGTATTTCTCCCAGCCAAACCGACC





ACGACTCCCGCCCCGCGCCCTCCGA





CACCCGCTCCCACCATCGCCTCTCA





ACCTCTTAGTCTTCGCCCCGAGGCA





TGCCGACCCGCCGCCGGGGGTGCTG





TTCATACGAGGGGCTTGGACTTCGC





T





TGTTGATATTTACATTTGTGGCTCC





GTTGGCGGGTACGTGCGGCGTCCTT





TTGTTGTCACTCGTTATTACTTTGT





ATTGTAATCACAGGAATCGCAAACG





GGGCAGAAAGAAACTCCTGTATATA





TTCAAACAACCATTTATGAGACCAG





TACAAACTACTCAAGAGGAAGATGG





CTGTAGCTGCCGATTTCCAGAAGAA





GAAGAAGGAGGATGTGAACTGCGAG





TGAAGTTTTCCCGAAGCGCAGACGC





TCCGGCATATCAGCAAGGACAGAAT





CAGCTGTATAACGAACTGAATTTGG





GACGCCGCGAGGTAGTATGACGTGC





TTGATAAACGCCGGGGGAGAGACCC





GGAAATGGGGGGTAAACCCCGAAGA





AAGAATCCCCAAGAAGGACTCTACA





ATGAACTCCAGAAGGATAAGATGGC





GGAGGCCTACTCAGAAATAGGTATG





AAGGGCGAACGACGAGGGGGAAAAG





GTCACGATGGCCTGTACCAAGGGTT





GAGTACGGCAACCAAAGATACGTAC





GATGCACTGCATATGCAGGCCCTGC





CTCCCAGATAATAATAAAATCGCTA





TCCATCGAAGATGGATGTGTGTTGG





TTTTTTGTGTGTGGAGCAACAAATC





TGACTTTGCATGTGCAAACGCCTTC





AACAACAGCATTATTCCAGAAGACA





CCTTCTTCCCCAGCCCAGGTAAGGG





CAGCTTTGGTGCCTTCGGAGGCTGT





TTGCTTGGTTGAGGAATGGCCAGGT





TCTGCCCAGAGCTCTGGTCAATGAT





GTCTAAAACTCCTCTGATTGGTGGT





CTCGGCCTTATCCATTGCCACCAAA





ACCCTCTTTTTACTAAGAAACAGTG





AGCCTTGTTCTGGCAGTCCAGAGAA





TGACAGGGGAAAAAAGCAGATGAAG





AGAAGGTGGCAGGAGAGGGCAGGTG





GCGCAGCCTCAGTCTCTCCAACTGA





GTTCCTGCCTGCCTGCCTTTGCTCA





GACTGTTTGCCCCTTACTGCTCTTC





TAGGCCTCATTCTAAGCCCCTTCTC





CAAGTTGCCTCTCCTTATTTCTCCC





TGTCTGCCAAAAAATCTTTCCCAGC





TCACTAAGTCAGTCTCACGCAGTCA





CTCATTAACCCACCAATCACTGATT





GTGCCGGCACATGAATGCACCAGGT





GTTGAAGTGGAGGAATTAAAAAGTC





AGATGAGGGGTGTGCCCAGAGGAAG





CACCATTCTAGTTGGGGGAGCCCAT





CTGTCAGCTGGGAAAAGTCCAAATA





ACTTCAGATTGGAATGTGTTTTAAC





TCAGGGTTGAGAAAACAGCTACCTT





CAGGACAAAAGTCAGGGAAGGGCTC





TCTGAAGAAATGCTACTTGAAGATA





GCAGCCCTACGAAGGGGAGGGAGAG





GACCCTATAGAGGGCTGGGAGAGGA





GCTGAATGAGAAAGG







TRAC-LHA
GAGATGTAAGGAGCTGCTGTGACTT
31



(800 bp)
GCTCAAGGCCTTATATCGAGTAAAC





GGTAGTGCTGGGGCTTAGACGCAGG





TGTTCTGATTTATAGTTCAAAACCT





CTATCAATGAGAGAGCAATCTCCTG





GTAATGTGATAGATTTCCCAACTTA





ATGCCAACATACCATAAACCTCCCA





TTCTGCTAATGCCCAGCCTAAGTTG





GGGAGACCACTCCAGATTCCAAGAT





GTACAGTTTGCTTTGCTGGGGCTTT





TCCCATGCCTGCCTTTACTCTGCCA





GAGTTATATTGCTGGGGTTTTGAAG





AAGATCCTATTAAATAAAAGAATAA





GCAGTATTATTAAGTAGCCCTGCAT





TTCAGGTTTCCTTGAGTGGCAGGCC





AGGCCTGGCCGTGAACGTTCACTGA





AATCATGGCCTCTTGGCCAAGATTG





ATAGCTTGTGCCTGTCCCTGAGTCC





CAGTCCATCACGAGCAGCTGGTTTC





TAAGATGCTATTTCCCGTATAAAGC





ATGAGACCGTGACTTGCCAGCCCCA





CAGAGCCCCGCCCTTGTCCATCACT





GGCATCTGGACTCCAGCCTGGGTTG





GGGCAAAGAGGGAAATGAGATCATG





TCCTGGCATCTGGACTCCAGCCTGG





GTTGGGGCAAAGAGGGAAATGAGAT





CATGTCCTAACCCTGATCCTCTTGT





CCCACAGATATCCAGAACCCTGACC





CTGCCGTGTACATTCTCAAACAAAT





GTGTCACAAAGTAAGGATTGTGATG





TGTATATCACAGACAAAACTGTGCT





AGACATGAGGTCTATGGACTTCA







TRAC-RHA
TGGAGCAACAAATCTGACTTTGCAT
32



(800 bp)
GTGCAAACGCCTTCAACAACAGCAT





TATTCCAGAAGACACCTTCTTCCCC





AGCCCAGGTAAGGGCAGCTTTGGTG





CCTTCGCAGGCTGTTTCCTTGCTTC





AGGAATGGCCAGGTTCTGCCCAGAG





CTCTGGTCAATGATGTCTAAAACTC





CTCTGATTGGTGGTCTCGGCCTTAT





CCATTGCCACCAAAACCCTCTTTTT





ACTAAGAAACAGTGAGCCTTGTTCT





GGCAGTCCAGAGAATGACACGGGAA





AAAAGCAGATGAAGAGAAGGTGGCA





GGAGAGGGCACGTGGCCCAGCCTCA





GTCTCTCCAACTGAGTTCCTGCCTG





CCTGCCTTTGCTCAGACTGTTTGCC





CCTTACTGCTCTTCTAGGCCTCATT





CTAAGCCCCTTCTCCAAGTTGCCTC





TCCTTATTTCTCCCTGTCTGCCAAA





AAATCTTTCCCAGCTCACTAAGTCA





GTCTCACGGAGTCACTCATTAACCC





ACCAATCACTGATTGTGCCGGCACA





TGAATGCACCAGGTGTTGAAGTGGA





GGAATTAAAAAGTCAGATGAGGGGT





GTGCCCAGAGGAAGCACCATTCTAG





GGGAGCCCATCTGTCAGCTGGGAAA





AGTCCAAATAACTTCAGATTGGAAT





GTGTTTTAACTCAGGGTTGAGAAAA





CAGCTACCTTCAGGACAAAAGTCAG





GGAAGGGCTCTCTGAAGAAATGCTA





CTTGAAGATACCAGCCCTACCAAGG





GCAGGGAGAGGACCCTATAGAGGCC





TGGGACAGGAGCTCAATGAGAAAGG





TTGG







Anti-
ATGGCGCTTCCGGTGACAGCACTGC
33



BCMA
TCCTCCCCTTGGCGCTGTTGCTCCA




CAR
CGCAGCAAGGCCGCAGGTGCAGCTG




(CTX-166b)
GTGCAGAGCGGAGCCGAGCTCAAGA





AGCCCGGAGCCTCCGTGAAGGTGAG





CTGCAAGGCCAGCGGCAACACCCTG





ACCAACTACGTGATCCACTGGGTGA





GACAAGCCCCCGGCCAAAGGCTGGA





GTGGATGGGCTACATCCTGCCCTAC





AACGACCTGACCAAGTACAGCCAGA





AGTTCCAGGGCAGGGTGACCATCAC





CAGGGATAAGAGCGCCTCCACCGCC





TATATGGAGCTGAGCAGCCTGAGGA





GCGAGGACACCGCTGTGTACTACTG





TACAAGGTGGGACTGGGACGGCTTC





TTTGACCCCTGGGGCCAGGGCACAA





CAGTGACCGTCAGCAGCGGCGGCGG





AGGCAGCGGCGGCGGCGGCAGCGGC





GGAGGCGGAAGCGAAATCGTGATGA





CCCAGAGCCCCGCCACACTGAGCGT





GAGCCCTGGCGAGAGGGCCAGCATC





TCCTGCAGGGCTAGCCAAAGCCTGG





TGCACAGCAACGGCAACACCCACCT





GCACTGGTACCAGCAGAGACCCGGA





CAGGCTCCCAGGCTGCTGATCTACA





GCGTGAGCAACAGGTTCTCCGAGGT





GCCTGCCAGGTTTAGCGGCAGCGGA





AGCGGCACCGACTTTACCCTGACCA





TCAGCAGCGTGGAGTCCGAGGACTT





CGCCGTGTATTACTGCAGCCAGACC





AGCCACATCCCTTACACCTTCGGCG





GCGGCACCAAGCTGGAGATCAAAAG





TGCTGCTGCCTTTGTCCCGGTATTT





CTCCCAGCCAAACCGACCACGACTC





CCGCCCCGCGCCCTCCGACACCCGC





TCCCACCATCGCCTCTCAACCTCTT





AGTCTTCGCCCCGAGGCATGCCGAC





CCGCCGCCGGGGGTGCTGTTCATAC





GAGGGGCTTGGACTTCGCTTGTGAT





ATTTACATTTGGGCTCCGTTGGCGG





GTACGTGCGGCGTCCTTTTGTTGTC





ACTCGTTATTACTTTGTATTGTAAT





CACAGGAATCGCAAACGGGGCAGAA





AGAAACTCCTGTATATATTCAAACA





ACCATTTATGAGACCAGTACAAACT





ACTCAAGAGGAAGATGGCTGTAGCT





GCCGATTTCCAGAAGAAGAAGAAGG





AGGATGTGAACTGCGAGTGAAGTTT





TCCCGAAGCGCAGACGCTCCGGCAT





ATCAGCAAGGACAGAATCAGCTGTA





TAACGAACTGAATTTGGGACGCCGC





GAGGAGTATGACGTGCTTGATAAAC





GCCGGGGGAGAGACCCGGAAATGGG





GGGTAAACCCCGAAGAAAGAATCCC





CAAGAAGGACTCTACAATGAACTCC





AGAAGGATAAGATGGCGGAGGCCTA





CTCAGAAATAGGTATGAAGGGCGAA





CGACGACGGGGAAAAGGTCACGATG





GCCTCTACCAAGGGTTGAGTACGGC





AACCAAAGATACGTACGATGCACTG





CATATGCAGGCCCTGCCTCCCAGA







Anti-
CAGGTGCAGCTGGTGCAGAGCGGAG
34



BCMA
CCGAGCTCAAGAAGCCCGGAGCCTC




scFv
CGTGAAGGTGAGCTGCAAGGCCAGC




(CTX-166
GGCAACACCCTGACCAACTACGTGA




& CTX-
TCCACTGGGTGAGACAAGCCCCCGG




166b)
CCAAAGGCTGGAGTGGATGGGCTAC





ATCCTGCCCTACAACGACCTGACCA





AGTACAGCCAGAAGTTCCAGGGCAG





GGTGACCATCACCAGGGATAAGAGC





GCCTCCACCGCCTATATGGAGCTGA





GCAGCCTGAGGAGCGAGGACACCGC





TGTGTACTACTGTACAAGGTGGGAC





TGGGACGGCTTCTTTGACCCCTGGG





GCCAGGGCACAACAGTGACCGTCAG





CAGCGGCGGCGGAGGCAGCGGCGGC





GGCGGCAGCGGCGGAGGCGGAAGCG





AAATCGTGATGACCCAGAGCCCCGC





CACACTGAGCGTGAGCCCTGGCGAG





AGGGCCAGCATCTCCTGCAGGGCTA





GCCAAAGCCTGGTGCACAGCAACGG





CAACACCCACCTGCACTGGTACCAG





CAGAGACCCGGACAGGCTCCCAGGC





TGCTGATCTACAGCGTGAGCAACAG





GTTCTCCGAGGTGCCTGCCAGGITT





AGCGGCAGCGGAAGCGGCACCGACT





TTACCCTGACCATCAGCAGCGTGGA





GTCCGAGGACTTCGCCGTGTATTAC





TGCAGCCAGACCAGCCACATCCCTT





ACACCTTCGGCGGCGGCACCAAGCT





GGAGATCAAA







4-1BB
AAACGGGGCAGAAAGAAACTCCTGT
35




ATATATTCAAACAACCATTTATGAG





ACCAGTACAAACTACTCAAGAGGAA





GATGGCTGTAGCTGCCGATTTCCAG





AAGAAGAAGAAGGAGGATGTGAACT





G







CD28
TCAAAGCGGAGTAGGTTGTTGCATT
36




CCGATTACATGAATATGACTCCTCG





CCGGCCTGGGCCGACAAGAAAACAT





TACCAACCCTATGCCCCCCCACGAG





ACTTCGCTGCGTACAGGTCC







CD3-zeta
CGAGTGAAGTTTTCCCGAAGCGCAG
37




ACGCTCCGGCATATCAGCAAGGACA





GAATCAGCTGTATAACGAACTGAAT





TTGGGACGCCGCGAGGAGTATGACG





TGCTTGATAAACGCCGGGGGAGAGA





CCCGGAAATGGGGGGTAAACCCCGA





AGAAAGAATCCCCAAGAAGGACTCT





ACAATGAACTCCAGAAGGATAAGAT





GGCGGAGGCCTACTCAGAAATAGGT





ATGAAGGGCGAACGACGACGGGGAA





AAGGTCACGATGGCCTCTACCAAGG





GTTGAGTACGGCAACCAAAGATACG





TACGATGCACTGCATATGCAGGCCC





TGCCTCCCAGA







EF-1α
GGCTCCGGTGCCCGTCAGTGGGCAG
38



promoter
AGCGCACATCGCCCACAGTCCCCGA





GAAGTTGGGGGGAGGGGTCGGCAAT





TGAACCGGTGCCTAGAGAAGGTGGC





GCGGGGTAAACTGGGAAAGTGATGT





CGTGTACTGGCTCCGCCTTTTTCCC





GAGGGTGGGGGAGAACCGTATATAA





GTGCAGTAGTCGCCGTGAACGTTCT





TTTTCGCAACGGGTTTGCCGCCAGA





ACACAGGTAAGTGCCGTGTGTGGTT





CCCGCGGGCCTGGCCTCTTTACGGG





TTATGGCCCTTGCGTGCCTTGAATT





ACTTCCACTGGCTGCAGTACGTGAT





TCTTGATCCCGAGCTTCGGGTTGGA





AGTGGGTGGGAGAGTTCGAGGCCTT





GCGCTTAAGGAGCCCCTTCGCCTCG





TGCTTGAGTTGAGGCCTGGCCTGGG





CGCTGGGGCCGCCGCGTGCGAATCT





GGTGGCACCTTCGCGCCTGTCTCGC





TGCTTTCGATAAGTCTCTAGCCATT





TAAAATTTTTGATGACCTGCTGCGA





CGCTTTTTTTCTGGCAAGATAGTCT





TGTAAATGCGGGCCAAGATCTGCAC





ACTGGTATTTCGGTTTTTGGGGCCG





CGGGCGGCGACGGGGCCCGTGCGTC





CCAGCGCACATGTTCGGCGAGGCGG





GGCCTGCGAGCGCGGCCACCGAGAA





TCGGACGGGGGTAGTCTCAAGCTGG





CCGGCCTGCTCTGGTGCCTGGCCTC





GCGCCGCCGTGTATCGCCCCGCCCT





GGGCGGCAAGGCTGGCCCGGTCGGC





ACCAGTTGCGTGAGCGGAAAGATGG





CCGCTTCCCGGCCCTGCTGCAGGGA





GCTCAAAATGGAGGACGCGGCGCTC





GGGAGAGCGGGCGGGTGAGTCACCC





ACACAAAGGAAAAGGGCCTTTCCGT





CCTCAGCCGTCGCTTCATGTGACTC





CACGGAGTACCGGGCGCCGTCCAGG





CACCTCGATTAGTTCTCGAGCTTTT





GGAGTACGTCGTCTTTAGGTTGGGG





GGAGGGGTTTTATGCGATGGAGTTT





CCCCACACTGAGTGGGTGGAGACTG





AAGTTAGGCCAGCTTGGCACTTGAT





GTAATTCTCCTTGGAATTTGCCCTT





TTTGAGTTTGGATCTTGGTTCATTC





TCAAGCCTCAGACAGTGGTTCAAAG





TTTTTTTCTTCCATTTCAGGTGTCG





TGA







3′ poly A
AATAAAATCGCTATCCATCGAAGAT
39




GGATGTGTGTTCiGTTTTTTGTGTG
















TABLE 5







Anti-BCMA CAR Construct Components


(Amino acid sequences)









Name

SEQ ID


Description
Amino Acid Sequence
NO:





CAR (CTX-166b)
MALPVTALLLPLALLLHAARPQVQL
40



VQSGAELKKPGASVKVSCKASGNTL




TNYVIHWVRQAPGQRLEWMGYILPY




NDLTKYSQKFQGRVTITRDKSASTA




YMELSSLRSEDTAVYYCTRWDWDGF




FDPWGQGTTVTVSSGGGGSGGGGSG




GGGSEIVMTQSPATLSVSPGERASI




SCRASQSLVHSNGNTHLHWYQQRPG




QAPRLLIYSVSNRFSEVPARFSGSG




SGTDFTLTISSVESEDFAVYYCSQT




SHIPYTFGGGTKLEIKSAAAFVPVF




LPAKPTTTPAPRPPTPAPTIASQPL




SLRPEACRPAAGGAVHTRGLDFACD




IYIWAPLAGTCGVLLLSLVITLYCN




HRNRKRGRKKLLYIFKQPFMRPVQT




TQEEDGCSCRFPEEEEGGCELRVKF




SRSADAPAYQQGQNQLYNELNLGRR




EEYDVLDKRRGRDPEMGGKPRRKNP




QEGLYNELQKDKMAEAYSEIGMKGE




RRRGKGHDGLYQGLSTATKDTYDAL




HMQALPPR






scFv
QVQLVQSGAELKKPGASVKVSCKAS
41


(CTX-166 (BCMA-
GNTLTNYVIHWVRQAPGQRLEWMGY



11, & CTX-166b)
ILPYNDLTKYSQKFQGRVTITRDKS




ASTAYMELSSLRSEDTAVYYCTRWD




WDGFFDPWGQGTTVTVSSGGGGSGG




GGSGGGGSEIVMTQSPATLSVSPGE




RASISCRASQSLVHSNGNTHLHWYQ




QRPGQAPRLLIYSVSNRFSEVPARF




SGSGSGTDFTLTISSVESEDFAVYY




CSQTSHIPYTFGGGTKLEIK






VH (CTX-166)
QVQLVQSGAELKKPGASVKVSCKAS
42



GNTLTNYVIHWVRQAPGQRLEWMGY




ILPYNDLTKYSQKFQGRVTITRDKS




ASTAYMELSSLRSEDTAVYYCTRWD




WDGFFDPWGQGTTVTVSS






VL (CTX-166)
EIVMTQSPATLSVSPGERASISCRA
43



SQSLVHSNGNTHLHWYQQRPGQAPR




LLIYSVSNRFSEVPARFSGSGSGTD




FTLTISSVESEDFAVYYCSQTSHIP




YTFGGGTKLEIK











VL CDR1
(Kabat or
RASQSLVHSNGNTHLH
44



Chothia)







VL CDR2

SVSNR
45





VL CDR3

SQTSHIPYT
46





VH CDR1
(Kabat)
NYVIH
47





VH CDR2

YILPYNDLTRYSQRFQG
48





VH CDR3

WDWDGFFDP
49





VH CDR1
(Chothia)
GNTLTNY
50





VH CDR2

LPYNDL
51





VH CDR3

WDWDGFFDP
52












linker
GGGGSGGGGSGGGGS
53





Signal peptide-1
MLLLVTSLLLCELPHPAFLLIP
54





CD8 signal peptide
MALPVTALLLPLALLLHAARP
55





CD8a transmembrane
IYIWAPLAGTCGVLLLSLVITLY
56


domain







4-1BB
RRGRRRLLYIFRQPFMRPVQTTQEE
57



DGCSCRFPEEEEGGCEL






CD28
SKRSRLLHSDYMNMTPRRPGPTRKH
58



YQPYAPPRDFAAYRS






CD3-zeta
RVRFSRSADAPAYQQGQNQLYNELN
59



LGRREEYDVLDRRRGRDPEMGGRPR




RRNPQEGLYNELQRDRMAEAYSEIG




MRGERRRGRGHDGLYQGLSTATRDT




YDALHMQALPPR






CD8a
FVPVFLPARPTTTPAPRPPTPAPTI
60


transmembrane
ASQPLSLRPEACRPAAGGAVHTRGL



domain
DFACDIYIWAPLAGTCGVLLLSLVI




TLYCNHRNR

















TABLE 6







Amino Acid Sequences of Daratumumab and CD38











SEQ


Name

ID


Description
Amino Acid Sequences
NO





CD38
MANCEFSPVSGDKPCCRLSRRAQLC
62



LGVSILVLILVVVLAVVVPRWRQQW




SGPGTTKRFPETVLARCVKYTEIHP




EMRHVDCQSVWDAFKGAFISKHPCN




ITEEDYQPLMKLGTQTVPCNKILLW




SRIKDLAHQFTQVQRDMFTLEDTLL




GYLADDLTWCGEFNTSKINYQSCPD




WRKDCSNNPVSVFWKTVSRRFAEAA




CDVVHVMLNGSRSKIFDKNSTFGSV




EVHNLQPEKVQTLEAWVIHGGREDS




RDLCQDPTIKELESIISKRNIQFSC




KNIYRPDKFLQCVKNPEDSSCTSEI






Daratumumab
EVQLLESGGGLVQPGGSLRLSCAVS
63


heavy chain
GFTFNSFAMSWVRQAPGKGLEWVSA



full sequence
ISGSGGGTYYADSVKGRFTISRDNS




KNTLY LQMNSLRAEDTAVYFCAKD




KILWFGEPVFDYWGQGTLVTVSSAS




TKGPSVFPLAPSSKSTSGGTAALGC




LVKDYFPEPVTVSWNSGALTSGVHT




FPAVLQSSGLYSLSSV VTVPSSSL




GTQTYICNVNHKPSNTKVDKRVEPK




SCDKTHTCPPCP




APELLGGPSVFLFPPKPKDTLMISR




TPEVTCVVVDVSHEDPEVKFNWYVD




GVEVHNAKTKPREEQYNSTYRVVSV




LTVLHQDWLNGKEYKCKVSNKALPA




PIEKTISKAKGQPREPQVYTLPPSR




EEMTKNQVSLTCLVKGFYPSDIAVE




WESNGQPENNYKTTPPVLDSDGSFF




LYSKLTVDKSRWQQGNVESC SVMH




EALHNHYTOKSLSLSP GK






Daratumumab
EVQLLESGGGL VQPGGSLRLSCA
64


heavy chain
VSGFTFNSFAMSWVRQAPGKGLEWV



variable
SAISGSGGGTYY ADSVKGRFTISR



region
DNSKNTLYLQMNSLRAEDTAVYFCA




KDKILW FGEPVFDYWGQGTLVTVS




SAS






Daratumumab
EIVLTQSPAT LSLSPGERAT LSC
65


light chain
RASQSVS SYLAWYQQKPQAPRLLI



full
YDASNRATGIPARFSGSGSGTDFTL



sequence
TISSLEPEDFAVYYCQQRSNWPPTF




GQGTKVEIKRTVAAPSVFIFPPSDE




QLKSGTASVVCLLNNFYPREAKVQW




KVDNALQSGNSQESVTEQDSKDSTY




SLSSTLTLSKADYEKHKVYACEVTH




QGLSSPVTKSFNRGEC






Daratumumab
EIVLTQSPATLSLSPGERATLSCRA
66


light chain
SQSVSSYLAWYQQKPGQAPRLLIYD



variable
ASNRATGIPARFSGSGSGTDFTLTI



region
SSLEPEDFAVYYCQQRSNWPPTFGQ




GTKVEIK






Daratumumab
SEAMS
67


heavy chain




CDRl







Daratumumab
AISGSGGGTY YADSVKG
68


heavy chain




CDR2







Daratumumab
DKILWFGEPV FDY
69


heavy chain




CDR3







Daratumumab
RASQSVSSYLA
70


light chain




CDRl







Daratumumab
DASNRAT
71


light chain




CDR2







Daratumumab
QQRSNWPPT
72


light chain




CDR3









Other Embodiments

All of the features disclosed in this specification may be combined in any combination. Each feature disclosed in this specification may be replaced by an alternative feature serving the same, equivalent, or similar purpose. Thus, unless expressly stated otherwise, each feature disclosed is only an example of a generic series of equivalent or similar features.


From the above description, one skilled in the art can easily ascertain the essential characteristics of the present invention, and without departing from the spirit and scope thereof, can make various changes and modifications of the invention to adapt it to various usages and conditions. Thus, other embodiments are also within the claims.


Equivalents

While several inventive embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the function and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the inventive embodiments described herein. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the inventive teachings is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific inventive embodiments described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed. Inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the inventive scope of the present disclosure.


All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.


All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.


The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”


The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.


As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of” or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e. “one or the other but not both”) when preceded by terms of exclusivity, such as “either,” “one of,” “only one of,” or “exactly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.


The term “about” as used herein means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within an acceptable standard deviation, per the practice in the art. Alternatively, “about” can mean a range of up to ±20%, preferably up to ±10%, more preferably up to ±5%, and more preferably still up to ±1% of a given value. Where particular values are described in the application and claims, unless otherwise stated, the term “about” is implicit and in this context means within an acceptable error range for the particular value.


As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.


It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.

Claims
  • 1. A method for treating multiple myeloma (MM), the method comprising: (i) administering to a subject in need thereof an effective amount of one or more lymphodepleting chemotherapeutic agents;(ii) administering to the subject a first dose of a population of genetically engineered T cells after step (i); and(iii) administering to the subject an effective amount of lenalidomide, an effective amount of daratumumab, or a combination thereof;wherein the population of genetically engineered T cells comprise T cells, which comprise a nucleic acid comprising a nucleotide sequence encoding a chimeric antigen receptor (CAR) that binds B-cell maturation antigen (BCMA), a disrupted TRAC gene, and a disrupted β2M gene; and wherein the nucleic acid encoding the CAR is inserted into the disrupted TRAC gene.
  • 2. The method of claim 1, wherein step (i) comprises co-administering to the subject fludarabine at about 30 mg/m2 and cyclophosphamide at about 300 mg/m2 to about 500 mg/m2, optionally at about 300 mg/m2, intravenously per day for three days.
  • 3. The method of claim 1, wherein step (ii) is performed 2-7 days after step (i).
  • 4. The method of claim 1, wherein the first dose of the population of genetically engineered T cells in step (ii) ranges from about 5.0×107 to about 1.05×109 CAR+ T cells, optionally about 5.0×107 to about 7.5×108 CAR+ T cells.
  • 5. The method of claim 4, wherein the first dose of the population of genetically engineered T cells is about 5×107 CAR+ T cells, about 1.5×108 CAR+ T cells, about 4.5×108 CAR+ T cells, about 6×108 CAR+ T cells, about 7.5×108 CAR+ T cells, or about 9×108 CAR+ T cells.
  • 6. The method of claim 4, wherein the first dose of the population of genetically engineered T cells in step (ii) ranges from about 5.0×107 to about 1.5×108 CAR+ T cells, about 1.5×108 to about 4.5×108 CAR+ T cells, about 4.5×108 to about 6.0×108 CAR+ T cells, about 6.0×108 to about 7.5×108 CAR+ T cells, about 7.5×108 to about 9×108 CAR+ T cells, or about 9×108 to about 1.05×109 CAR+ T cells.
  • 7. The method of claim 1, wherein in step (iii), an effective amount of lenalidomide is administered to the subject.
  • 8. The method of claim 7, wherein step (iii) comprises administering to the subject about 10 mg lenalidomide orally per day for 21 days.
  • 9. The method of claim 7, wherein the first dose of lenalidomide in step (iii) starts on the third day of the administration of the lymphodepleting chemotherapeutic agents.
  • 10. The method of claim 7, wherein the method further comprises performing one or more cycles of treatment comprising lenalidomide to the subject after step (ii).
  • 11. The method of claim 10, wherein the first cycle starts 28 days after step (ii), optionally when the subject exhibits stable disease or better.
  • 12. The method of claim 10, wherein the one or more cycles of treatment comprising lenalidomide are up to five cycles, each of which comprises a daily dose of lenalidomide for 21 days, followed by a 7-day resting period; optionally wherein the daily dose of lenalidomide is 5 mg.
  • 13. The method of claim 10, the method further comprising terminating the one or more cycles of the treatment comprising lenalidomide when the subject exhibits disease progression and/or unacceptable toxicity.
  • 14. The method of claim 1, wherein in step (iii), an effective amount of daratumumab is administered to the subject.
  • 15. The method of claim 14, wherein about 16 mg/kg daratumumab is administered to the subject by intravenous infusion within 3 days prior to step (ii), and optionally wherein the dose of about 16 mg/kg of daratumumab is split to 8 mg/kg over two consecutive days.
  • 16. The method of claim 14, wherein about 1800 mg of daratumumab is administered to the subject by subcutaneous injection, and optionally wherein the daratumumab is injected together with hyaluronidase, which optionally is in an amount of about 30,000units.
  • 17. The method of claim 15, wherein the daratumumab is administered to the subject no more than 14 days prior to step (ii).
  • 18. The method of claim 14, wherein the subject is administered multiple doses of daratumumab once per month, optionally up to 5 monthly doses, when the subject exhibits stable disease or better.
  • 19. The method of claim 18, wherein treatment of the daratumumab is terminated when the subject exhibits disease progression and/or unacceptable toxicity.
  • 20. The method of claim 14, wherein prior to the administration of the daratumumab, the subject is administered corticosteroid, antipyretic, antihistamine, or a combination thereof, optionally wherein the subject is administered methylprednisolone at about 100 mg by intravenous infusion or about 60 mg by intravenous infusion or by oral administration, acetaminophen at about 650-1000 mg by oral administration, and diphenhydramine hydrochloride at about 20-50 mg by intravenous infusion or oral administration.
  • 21. The method of claim 1, wherein in step (iii), an effective amount of lenalidomide and an effective amount of daratumumab are administered to the subject.
  • 22. The method of claim 21, wherein about 10 mg lenalidomide is administered to the subject orally per day for 21 days; optionally wherein the first dose of lenalidomide starts on the third day of the administration of the lymphodepleting chemotherapeutic agents.
  • 23. The method of claim 21, wherein the method further comprises performing one or more cycles of treatment comprising lenalidomide to the subject after step (ii).
  • 24. The method of claim 23, wherein the first cycle starts 28 days after step (ii), optionally when the subject exhibits stable disease or better.
  • 25. The method of claim 23, wherein the one or more cycles of treatment comprising lenalidomide are up to five cycles, each of which comprises a daily dose of lenalidomide for 21 days, followed by a 7-day resting period; optionally wherein the daily dose of lenalidomide is 5 mg.
  • 26. The method of claim 22, the method further comprising terminating the one or more cycles of the treatment comprising lenalidomide when the subject exhibits disease progression and/or unacceptable toxicity.
  • 27. The method of claim 21, wherein about 16 mg/kg daratumumab is administered to the subject by intravenous infusion within 3 days prior to step (ii), and optionally wherein the dose of about 16 mg/kg of daratumumab is split to 8 mg/kg over two consecutive days.
  • 28. The method of claim 21, wherein about 1800 mg of daratumumab is administered to the subject by subcutaneous injection, and optionally wherein the daratumumab is injected together with hyaluronidase, which optionally is in an amount of about 30,000units.
  • 29. The method of claim 27, wherein the daratumumab is administered to the subject no more than 14 days prior to step (ii).
  • 30. The method of claim 27, wherein the subject is administered multiple doses of daratumumab once per month, optionally up to 5 monthly doses, when the subject exhibits stable disease or better.
  • 31. The method of claim 30, wherein treatment of the daratumumab is terminated when the subject exhibits disease progression and/or unacceptable toxicity.
  • 32. The method of claim 27, wherein prior to the administration of the daratumumab, the subject is administered corticosteroid, antipyretic, antihistamine, or a combination thereof, optionally wherein the subject is administered methylprednisolone at about 100 mg by intravenous infusion or about 60 mg by intravenous infusion or by oral administration, acetaminophen at about 650-1000 mg by oral administration, and diphenhydramine hydrochloride at about 20-50 mg by intravenous infusion or oral administration.
  • 33. The method of claim 1, wherein the CAR that binds BCMA comprises: (i) an ectodomain comprising an anti-BCMA single chain variable fragment (scFv);(ii) a CD8a transmembrane domain; and(iii) an endodomain comprising a 4-1BB co-stimulatory domain and a CD3ζ signaling domain.
  • 34. The method of claim 33, wherein the anti-BCMA scFv comprises a heavy chain variable domain (VH) comprising SEQ ID NO: 42 and a light chain variable domain (VL) comprising SEQ ID NO: 43.
  • 35. The method of claim 34, wherein the anti-BCMA scFv comprises SEQ ID NO: 41.
  • 36. The method of claims 33, wherein the CAR that binds BCMA comprises the amino acid sequence of SEQ ID NO: 40.
  • 37. The method of claim 37, wherein the nucleic acid encoding the anti-BCMA CAR comprises the nucleotide sequence of SEQ ID NO: 33.
  • 38. The method of claim 1, wherein the disrupted TRAC gene is produced by a CRISPR/Cas9 gene editing system, which comprises a guide RNA comprising a spacer sequence of SEQ ID NO: 4.
  • 39. The method of claim 1, wherein the disrupted TRAC gene has a deletion comprising the SEQ ID NO: 10, optionally wherein the disrupted TRAC gene comprises the nucleotide sequence of SEQ ID NO:30, which substitutes for the deletion comprising SEQ ID NO:10.
  • 40. The method of claim 1, wherein the disrupted β2M gene is produced by a CRISPR/Cas9 gene editing system, which comprises a guide RNA comprising a spacer sequence of SEQ ID NO: 8.
  • 41. The method of claim 1, the disrupted β2M gene comprises at least one of SEQ ID NOs: 21-26.
  • 42. The method of claim 1, wherein in the population of genetically engineered T cells, ≥30% of the genetically engineered T cells are CAR+, ≤0.4% of the genetically engineered T cells are TCR+, and/or ≤30% of the genetically engineered T cells are B2M+.
  • 43. The method of claim 1, wherein the population of genetically engineered T cells is derived from one or more healthy human donors.
  • 44. The method of claim 1, wherein the population of genetically engineered T cells is suspended in a cryopreservation solution.
  • 45. The method of claim 1, wherein the population of genetically engineered T cells is administered by intravenous infusion.
  • 46. The method of claim 1, wherein the method further comprising (iv) monitoring the human patient for development of acute toxicity after step (ii).
  • 47. The method of claim 46, wherein the acute toxicity comprises infusion reactions, febrile reactions, cytokine release syndrome (CRS), immune effector cell-associated neurotoxicity syndrome (ICANS), tumor lysis syndrome, hemophagocytic lymphohistiocytosis (HLH), Cytopenias, GvHD, hypotention, renal insufficiency, viral encephalitis, neutropenia, thrombocytopenia, or a combination thereof.
  • 48. The method of claim 46, wherein the subject is subject to toxicity management if development of toxicity is observed.
  • 49. The method of claim 1, wherein the subject is a human patient, who optionally is 18 years of age or older.
  • 50. The method of claim 1, wherein the subject has relapsed and/or refractory MM.
  • 51. The method of claim 1, wherein the subject has undergone at least two prior therapies for MM, which optionally comprise an immunomodulatory agent, a proteasome inhibitor, an anti-CD38 antibody, or a combination thereof.
  • 52. The method of claim 51, wherein the subject is refractory to one or more prior therapies comprising an immunomodulatory agent, a proteasome inhibitor, and/or an anti-CD38 antibody.
  • 53. The method of claim 52, wherein the subject is double-refractory to prior therapies comprising an immunomodulatory agent and a proteasome inhibitor, or wherein the subject is triple-refractory to prior therapies comprising an immunomodulatory agent, a proteasome inhibitor, and an anti-CD38 antibody.
  • 54. The method of claim 1, wherein the subject relapsed after an autologous stem cell transplant (SCT), and wherein optionally the relapse occurs within 12 months after the SCT.
  • 55. The method of claim 1, wherein the subject has received prior lenalidomide treatment.
  • 56. The method of claim 1, wherein the subject is a human patient having one or more of the following features: (a) measurable disease,(b) Eastern Cooperative Oncology Group performance status 0 or 1,(c) adequate organ function,(d) free of a prior allogeneic stem cell transplantation (SCT),(e) free of autologous SCT within 60 days prior to step (i),(f) free of plasma cell leukemia, non-secretory MM, Waldenstrom's macroglobulinemia, POEM syndrome, and/or amyloidosis with end organ involvement and damage,(g) free of prior gene therapy, anti-BCMA therapy, and non-palliative radiation therapy within 14 days prior to step (i),(h) free of contraindication to lenalidomide, daratumumab, cyclophosphamide, and/or fludarabine,(i) free of central nervous system involvement by MM,(j) free of history or presence of clinically relevant CNS pathology, cerebrovascular ischemia and/or hemorrhage, dementia, a cerebellar disease, an autoimmune disease with CNS involvement,(k) free of unstable angina, arrhythmia, and/or myocardial infarction within 6 month prior to step (i),(l) free of uncontrolled infections, optionally wherein the infection is caused by HIV, HBV, or HCV,(m) free of previous or concurrent malignancy, provided that the malignancy is not basal cell or squamous cell skin carcinoma, adequately resected and in situ carcinoma of cervix, or a previous malignancy that was completely resected and has been in remission for ≥5 years,(n) free of live vaccine administration within 28 days prior to step (i),(o) free of systemic anti-tumor therapy within 14 days prior to step (i), and(p) free of primary immunodeficiency disorders or autoimmune disorders that require immunosuppressive therapy.
  • 57. The method of claim 1, wherein the effective amount of the population of genetically engineered T cells is sufficient to achieve one or more of the following: (a) decrease soft tissue plasmacytomas sizes (SPD) by at least 50% in the subject; (b) decrease serum M-protein levels by at least 25%, optionally by 50% in the subject;(c) decrease 24-hour urine M-protein levels by at least 50%, optionally by 90% in the subject;(d) decrease differences between involved and uninvolved free light chain (FLC) levels by at least 50% in the subject;(e) decrease plasma cell counts by at least 50% in the subject, optionally wherein baseline BM plasma-cell percentage is ≥30%,(f) decrease kappa-to-lambda light chain ratios (κ/λ ratios) to 4:1 or lower in the subject, who has myeloma cells that produce kappa light chains; and(g) increase kappa-to-lambda light chain ratios (κ/λ ratios) to 1:2 or higher in the subject, who has myeloma cells that produce lamda light chains.
  • 58. The method of claim 1, wherein the effective amount of the population of genetically engineered T cells is sufficient to decrease serum M-protein levels by at least 90% and 24-hour urine M-protein levels to less than 100 mg in the subject, and/or wherein the effective amount of the population of genetically engineered T cells is sufficient to decrease serum M-proteins, urine M-proteins, and soft tissue plasmacytomas to undetectable levels, and plasma cell counts to less than 5% of bone marrow (BM) aspirates in the subject.
  • 59. The method of claim 1, wherein the effective amount of the population of genetically engineered T cells is sufficient to achieve Stringent Complete Response (sCR), Complete Response (CR), Very Good Partial Response (VGPR), Partial Response (PR), Minimal Response (MR), or Stable Disease (SD).
  • 60. The method of claim 1, wherein prior to step (i), the human patient does not show one or more of the following features: (a) significant worsening of clinical status,(b) requirement for supplemental oxygen to maintain a saturation level of greater than about 91%,(c) uncontrolled cardiac arrhythmia,(d) hypotension requiring vasopressor support,(e) active infection, and(f) neurological toxicity that increases risk of immune effector cell-associated neurotoxicity syndrome (ICANS).
  • 61. The method of claim 1, wherein prior to step (ii) and after step (i), the human patient does not show one or more of the following features: (a) active uncontrolled infection,(b) worsening of clinical status compared to the clinical status prior to step (i), and(c) neurological toxicity that increases risk of immune effector cell-associated neurotoxicity syndrome (ICANS).
  • 62. The method of claim 1, wherein the method further comprising administering to the subject a second dose of the population of genetically engineered T cells about 4 to 12 weeks after the first dose of the population of genetically engineered T cells, wherein the subject achieve stable disease or better response after the first dose, optionally assessed on Day 28 after the first dose.
  • 63. The method of claim 62, wherein the subject is treated by the lymphodepleting chemotherapeutic agents 2-7 days prior to the second dose of the population of genetically engineered T cells; optionally wherein the subject is administered fludarabine at about 30 mg/m2 and cyclophosphamide at about 300 mg/m2 to about 500 mg/m2, optionally at about 300 mg/m2, intravenously per day for three days.
  • 64. The method of claim 62, wherein the second dose of the population of genetically engineered T cells is not accompanied with lymphodepleting therapy when the subject is experiencing significant cytopenias.
CROSS REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of the filing dates of U.S. Provisional Application No. 63/129,969, filed Dec. 23, 2020, and U.S. Provisional Application No. 63/129,972, filed Dec. 23, 2020, the entire contents of each of which are incorporated by reference herein.

Provisional Applications (2)
Number Date Country
63129969 Dec 2020 US
63129972 Dec 2020 US