The invention provides compositions and methods for treating diseases associated with expression of cd20 or CD22. The invention also relates to chimeric antigen receptor (CAR) specific to cd20 or CD22, vectors encoding the same, and recombinant T or natural killer (NK) cells comprising the cd20 CAR or CD22 CAR. The invention also includes methods of administering a genetically modified T cell or NK cell expressing a CAR that comprises a cd20 or CD22 binding domain.
|
1. An isolated nucleic acid molecule encoding a chimeric antigen receptor (CAR), wherein the CAR comprises a cd20 binding domain, a transmembrane domain, and an intracellular signaling domain, wherein said cd20 binding domain comprises a light chain complementarity determining region 1 (LCDR1), light chain complementarity determining region 2 (LCDR2), light chain complementarity determining region 3 (LCDR3) heavy chain complementarity determining region 1 (HCDR1), heavy chain complementarity determining region 2 (HCDR2), and heavy chain complementarity determining region 3 (HCDR3), wherein the LCDR1, LCDR2, LCDR3, HCDR1, HCDR2, and HCDR3 comprise:
(i) SEQ ID NOs: 147, 148, 149, 136, 137, and 138, respectively;
(ii) SEQ ID NOs: 150, 151, 152, 139, 140, and 141, respectively;
(iii) SEQ ID NOs: 153, 154, 155, 142, 143, and 144, respectively; or
(iv) SEQ ID NOs: 929, 930, 931, 926, 927, and 928, respectively.
2. The isolated nucleic acid molecule of
(i) the VL comprises the amino acid sequence of SEQ ID NO: 156, 129, or 439, or an amino acid sequence with at least 95% identity thereto;
(ii) the VH comprises the amino acid sequence of SEQ ID NO: 145, 118, or 437, or an amino acid sequence with at least 95% identity thereto; or
(iii) the VL and VH comprise:
(a) SEQ ID NOs: 156 and 145, respectively;
(b) SEQ ID NOs: 129 and 118, respectively; or
(c) SEQ ID NOs: 439 and 437, respectively.
3. The isolated nucleic acid molecule of
(i) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 157, 130, or 440, or a nucleotide sequence with at least 95% identity thereto;
(ii) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 146, 119, or 438, or a nucleotide sequence with at least 95% identity thereto; or
(iii) the nucleic acid molecule comprises:
(a) SEQ ID NOs: 157 and 146;
(b) SEQ ID NOs: 130 and 119; or
(c) SEQ ID NOs: 440 and 438.
4. The isolated nucleic acid molecule of
5. The isolated nucleic acid molecule of
6. The isolated nucleic acid molecule of
7. The isolated nucleic acid molecule of
(i) the transmembrane domain comprises the amino acid sequence of SEQ ID NO: 801, or an amino acid sequence with at least 95% identity thereto; or
(ii) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 802 or 1072, or a nucleotide sequence with at least 95% identity thereto.
8. The isolated nucleic acid molecule of
9. The isolated nucleic acid molecule of
(i) the hinge region comprises the amino acid sequence of SEQ ID NO: 799 or SEQ ID NO: 814, or an amino acid sequence with at least 95% identity thereto; or
(ii) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 800 or SEQ ID NO: 815, or a nucleotide sequence with at least 95% identity thereto.
10. The isolated nucleic acid molecule of
11. The isolated nucleic acid molecule of
(i) the costimulatory domain comprises the amino acid sequence of SEQ ID NO: 803, or an amino acid sequence with at least 95% identity thereto; or
(ii) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 804, or a nucleotide sequence with at least 95% identity thereto.
12. The isolated nucleic acid molecule of
13. The isolated nucleic acid molecule of
(i) the primary signaling domain comprises the amino acid sequence of SEQ ID NO: 807 or SEQ ID NO: 805, or an amino acid sequence with at least 95% identity thereto; or
(ii) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 893, 808, 806, or 1074, or a nucleotide sequence with at least 95% identity thereto.
14. The isolated nucleic acid molecule of
15. The isolated nucleic acid molecule of
(i) the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 803, or an amino acid sequence with at least 95% identity thereto, and the amino acid sequence of SEQ ID NO: 807 or SEQ ID NO: 805, or an amino acid sequence with at least 95% identity thereto;
(ii) the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 803 and the amino acid sequence of SEQ ID NO: 807 or SEQ ID NO: 805;
(iii) the intracellular signaling domain comprises the amino acid sequence of SEQ ID NO: 803 and the amino acid sequence of SEQ ID NO: 807 or SEQ ID NO: 805, wherein the sequences comprising the intracellular signaling domain are expressed in the same frame and as a single polypeptide chain, or
(iv) the nucleic acid molecule comprises the nucleotide sequence of SEQ ID NO: 804, or a nucleotide sequence with at least 95% identity thereto, and/or the nucleotide sequence of SEQ ID NO: 893, 808, 806, or 1074, or a nucleotide sequence with at least 95% identity thereto.
16. The isolated nucleic acid molecule of
17. The isolated nucleic acid molecule of
18. The isolated nucleic acid molecule of
19. A nucleic acid comprising:
(i) a first nucleic acid comprising the nucleic acid molecule of
(ii) a second nucleic acid encoding a CAR molecule that binds a B-cell antigen.
20. The nucleic acid of
21. The nucleic acid of
(i) the first and the second nucleic acids are disposed on a single nucleic acid molecule, or
(ii) the first and the second nucleic acids are disposed on separate nucleic acid molecules.
23. The nucleic acid of
(i) a HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 comprising SEQ ID NOs: 773, 774, 775, 776, 777, and 778, respectively;
(ii) a scFv comprising the amino acid sequence of SEQ ID NO: 765; or
(iii) a CAR comprising the amino acid sequence of SEQ ID NO: 764, wherein the CAR molecule comprises or does not comprise a signal peptide comprising the amino acid sequence of SEQ ID NO: 797.
24. The nucleic acid of
(i) a HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 comprising SEQ ID NOs: 785, 786, 787, 788, 789, and 790, respectively;
(ii) a scFv comprising the amino acid sequence of SEQ ID NO: 1047; or
(iii) a CAR comprising the amino acid sequence of SEQ ID NO: 1049, wherein the CAR molecule comprises or does not comprise a signal peptide comprising the amino acid sequence of SEQ ID NO: 797.
25. The nucleic acid of
26. The nucleic acid of
(i) the first nucleic acid and the second nucleic acid comprise a single promoter, wherein the single promoter is an EF-1α promoter; or
(ii) the first nucleic acid comprises a first promoter and the second nucleic acid comprises a second promoter.
28. A method of making a cell, comprising transducing a T cell or an NK cell with the nucleic acid molecule of
29. A method of generating a population of RNA-engineered cells comprising introducing an in vitro transcribed RNA or synthetic RNA into a cell, wherein the RNA comprises the nucleic acid molecule of
31. The isolated cell of
(i) an inhibitory molecule that comprises a first polypeptide that comprises at least a portion of an inhibitory molecule, associated with a second polypeptide that comprises a costimulatory domain and primary signaling domain, or
(ii) an inhibitory molecule that comprises a first polypeptide that comprises at least a portion of Programmed Death 1 (PD1) and a second polypeptide comprising a costimulatory domain and primary signaling domain.
32. A population of immune effector cells, comprising:
(i) a first cell population comprising the nucleic acid molecule of
(ii) a second cell population comprising a nucleic acid encoding a CAR that binds a B-cell antigen.
33. The population of
(i) the B-cell antigen is chosen from CD19, CD22, CD10, CD34, CD123, FLT-3, ROR-1, CD79b, CD79a, or CD179b, or
(ii) the B-cell antigen is CD19 or CD22.
0. 34. The nucleic acid of claim 19, wherein the B cell antigen is CD22, and wherein the CAR molecule that binds the B-cell antigen comprises:
(i) a HCDR1, HCDR2, HCDR3, LCDR1, LCDR2, and LCDR3 comprising:
(a) SEQ ID NOs: 719, 720, 721, 730, 731, and 732, respectively;
(b) SEQ ID NOs: 722, 723, 724, 733, 734, and 735, respectively;
(c) SEQ ID NOs: 725, 726, 727, 736, 737, and 738, respectively; or
(d) SEQ ID NOs: 1036, 1037, 1038, 1039, 1040, and 1041, respectively;
(ii) a light chain variable region (VL) and a heavy chain variable region (VH), wherein:
(a) the VL comprises the amino acid sequence of SEQ ID NO: 840 or 739;
(b) the VH comprises the amino acid sequence of SEQ ID NO: 839 or 728; or
(c) the VL and VH comprise:
(a) SEQ ID NOs: 840 and 839, respectively; or
(b) SEQ ID NOs: 739 and 728, respectively;
(iii) a scFv comprising the amino acid sequence of SEQ ID NO: 742, 835, 836, or 837; or
(iv) a CAR comprising the amino acid sequence of SEQ ID NO: 744, wherein the CAR molecule comprises or does not comprise a signal peptide comprising the amino acid sequence of SEQ ID NO: 797.
0. 35. The nucleic acid of claim 34, wherein:
(i) the VH and VL sequences are connected directly without a linker;
(ii) the VH and VL sequences are connected via a (Gly4-Ser)n linker, wherein n is 0, 1, 2, 3, 4, 5, or 6;
(iii) the VH and VL sequences are connected via a linker, wherein the linker comprises the amino acid sequence of SEQ ID NO: 834; or
(iv) the VH and VL sequences are connected via a linker, wherein the linker comprises the amino acid sequence of SEQ ID NO: 741.
|
This Lymphoidlymphoid
Within 9 days the tumor volume measurement was 200 mm3 and untreated tumors reach endpoint measurement (>1200 m3) by 15-18 days. Anti-tumor activities of therapeutic agents are often tested once tumors are fully engrafted. Thus, there is a suitable window with this model during which the anti-tumor activity of CAR T cells can be observed.
CAR T Cell Dosing:
Mice were dosed 9 days after tumor implantation, with 3×106 CAR T cells. Cells were partially thawed in a 37° C. water bath and then completely thawed by the addition of 1 ml of warmed growth media. The thawed cells were transferred to a 50 ml falcon tube and adjusted to a final volume of 12 ml with growth media. The cells were washed twice and spun at 300 g for 10 minutes and then counted by hemocytometer. T cells were then resuspended at respective concentrations in cold PBS and kept on ice until the mice were dosed. The CARTs were injected intravenously via the tail vein in 200 al, for a dose of 3×106 CART cells. 5 mice per group were either treated with 200 μl of PBS alone (PBS), EGFRvIII-specific, mock CAR T cells as well as CD20-C3H2, CD20-C5H1, CD20-3H5k3, CD20-Ofa, and CD20-8aBBZ. All cells were prepared from the same donor in parallel.
Animal Monitoring:
The health status of the mice was monitored daily, including twice weekly body weight measurements. The percent change in body weight was calculated as (BWcurrent—BWinitial)/(BWinitial)×100%.
The anti-tumor activity of CD20 CAR T cells was assessed in a DLBCL leukemia xenograft model (
The PBS treatment group, which did not receive any T cells, demonstrates baseline TMD8 tumor growth kinetics. The EGFRvIII treatment group received mock CAR-transduced T cells and served as a T cell control to show the non-specific response of human donor T cells in this model. Both the PBS and EGFRvIII treatment groups demonstrated continuous tumor progression throughout this study. CD20-Ofa and CD20-3H5k3 showed similar growth kinetics, suggesting no anti-tumor efficacy by these CARTs. CD20-C3H2, CD20-C5H1, and the control CD20-8aBBZ CAR T cells all showed significantly slower tumor growth, with the strongest and fastest regression seen for CD20-C3H2, followed by CD22-C5H1.
This study demonstrated that the CD20-specific CAR T cells CD22-C3H2 and CD22-C5H1 are capable of leading to the regression of TMD8 tumors. The efficacy was superior to CD20-8aBBZ, the published benchmark CAR.
Genes encoding for single chain variable fragments for anti-CD22 antibodies (CD22-65, CD22-65s, positive control CD22 CAR m971 (m971), and m971s) were cloned into lentiviral CAR expression vectors with the CD3zeta chain and 4-1BB stimulatory molecules: The CD3zeta chain was either wildtype (Zwt) or carried a Q65K mutation (Zmut). The constructs were ranked based on the effector T cell responses of these CD22 CAR-transduced T cells (“CD22 CART” or “CD22 CAR T cells”) in response to CD22 expressing (“CD22+”) targets. Effector T cell responses include, but are not limited to, cellular expansion, proliferation, doubling, cytokine production and target cell killing or cytolytic activity (degranulation).
Generation of CD22 CAR T Cells:
Human scFv encoding lentiviral transfer vectors were used to produce the genomic material packaged into the VSVg pseudotyped lentiviral particles. Lentiviral transfer vector DNA encoding the CAR was mixed with the three packaging components VSVg, gag/pol and rev in combination with lipofectamine reagent to transfect Lenti-X 293T cells (Clontech), followed by medium replacement 12-18 h later. 30 hours after medium change, the media is collected, filtered and stored at −80° C.
CD22 CAR T cells were generated by starting with blood from healthy apheresed donors whose T cells were enriched by negative selection of T cells, CD4+ and CD8+ lymphocytes (Pan T cell isolation, Miltenyi). T cells were activated by the addition of CD3/CD28 beads (DYNABEADS® Human T-Expander CD3/CD28, ThermoFisher Scientific) at a ratio of 1:3 (T cell to bead) in T cell medium (RPMI1640, 10% heat-inactivated fetal calf serum (FCS), 2 mM L-glutamine, 1× Penicillin/Streptomycin, 100 μM non-essential amino acids, 1 mM Sodium Pyruvate, 10 mM Hepes, and 55 μM 2-mercaptoethanol) at 37° C., 5% CO2. T cells were cultured at 0.5×106 T cells in 1 mL medium per well of a 24-well plate. After 24 hours, when T cells were blasting, non-concentrated or concentrated viral supernatant was added; T cells were transduced at a multiplicity of infection (MOI) of 5. T cells began to proliferate, which is monitored by measuring the cell concentration (as counts per mL), and
T cells are diluted in fresh T cell medium every two days. As the T cells began to rest down after approximately 10 days, the logarithmic growth wanes. The combination of slowing growth rate and reduced T cell size (approaching 350 fL) determines the state for T cells to be cryopreserved for later analysis. All CD22 CAR T cells were produced under research grade (i.e., not clinical grade) manufacturing conditions.
Before cryopreserving, the percentage of cells transduced (expressing the CD22-specific CAR on the cell surface) were determined by flow cytometric analysis on a FACS Fortessa (BD) (
Evaluating Potency of CD22 CAR-Redirected T Cells:
To evaluate the functional abilities of CD22 CAR T cells, the cells, generated as described above, were thawed, counted and co-cultured with cancer cells to read out their killing capabilities, secretion of cytokine as well as proliferation. Human scFv bearing CARs CD22-65_Zmut, CD22-65_Zwt, CD22-65s_Zwt, m971_Zmut, and m971s_Zmut were used and compared to a CD19 CAR as well as non-transduced T cells (UTD), which were used as non-targeting T cells control.
T cell killing was directed towards the acute lymphoblastic leukemia (ALL) lines Nalm6 (RRID: CVCL_0092) and
SEM (RRID: CVCL_0095); K562 (RRID: CVCL_0004), a chronic myelogenous leukemia (CML) cell line served as CD22-negative/low control. All cell lines were transduced to express luciferase as a reporter for cell viability/killing. The cytolytic activities of CD22 CARTs were measured at a titration of effector:target cell ratios (E:T) of 10:1, 5:1, 2.5:1, 1.25:1 0.63:1 and 0.31:1. Assays were initiated by mixing the respective number of T cells with a constant number of targets cells (25,000 cells per well of a 96-well plate). After 20 hours, remaining cells in the wells were lysed by addition of Bright-Glo™ Luciferase Assay System (Promega) reagent, to quantify the remaining Luc-expressing cancer cells in each well. “% Killing” was calculated in relation to wells containing target cells alone (0%, maximal Luc signal). The data show that transduction with the CD22 CART encoding lentiviruses transfers andi-CD22 killing activityy to T cells in Nalm6 (
To measure cytokine production of CD22 CAR T cells in response to CD22-expressing target cells, CAR T cells were co-cultured with the same ALL lines as above plus K562, serving as CD22-negative/low control. Cells were cultured at an effector:target ratio of 1:1 and 25,000 cells per well of a 96-well plate for 24 h, after which the media was removed for cytokine analysis using the V-PLEX Human IFN-γ Kit (Meso Scale Diagnostics). These data show that all CD22 CARTs as well as the CD19 CARTs produced IFN-γ when cultured with Nalm6 or SEM (
In the last T cell efficacy assay, the proliferative capacity of the CD22 CAR T cells was assessed. Again, thawed CAR T cells we co-cultured with the ALL lines Nalm6 and SEM. CARTs were stained with CellTracer Violet dye (Thermo-Fisher Scientific) and target cell lines were irradiated prior to co-culture to prevent overgrowth in the wells. They were then cultured at an effector:target ratio of 1:1 and 30,000 cells per well in a 96-well plate for 4 days. On day 4, cells were stained with anti-CD3 antibody as well as soluble CD22-Fc to detect CAR expression. From the intensity of the violet dye of all CD3+ T cells; the lower the fluorescence, the stronger the proliferation, as each cell division of the T cells leads to retention of half the fluorescence in each daughter cell (
All CD22-specific CARs in this experiment were expressed on the cell surface of primary human T cells similarly well: CD22-65_Zmut, CD22-65_Zwt, CD22-65s_Zwt, m971_Zmut, and m971s_Zmut. While all CD22 CARTs showed efficacy in T cell functional assays, m971s_Zmut, CD22-65s_Zwt and CD22-65_Zwt were the top 3 CARTs in all 3 assays. Overall, the transfer of CD22 CARs induced anti-CD22 reactivity but no off-target function was detected.
Anti-tumor activity of a set of CD22 CAR T cells was assessed in vivo in a NALM6 and a SEM xenograft model. CAR T cells with CAR constructs CD22-65_Zmut, CD22-65_Zwt, CD22-65s_Zwt, m971_Zmut, and m971s_Zmut were evaluated.
Cell Lines:
Both Nalm6 (RRID: CVCL_0092) and SEM (RRID: CVCL_0095) are human acute lymphoblastic leukemia (ALL) cell lines. Cells were grown in RMPI medium containing 10% fetal bovine serum and both grow in suspension. Both cell lines persist and expand in mice when implanted intravenously. Cells have been modified to express luciferase, so that that tumor cell growth can also be monitored by imaging the mice after they have been injected with the substrate Luciferin.
Mice:
6 week old NSG (NOD.Cg-PrkdcscidIl2rgtml Wjl/SzJ) mice were received from the Jackson Laboratory (stock number 005557). Animals were allowed to acclimate in the Novartis NIBR animal facility for at least 3 days prior to experimentation. Animals were handled in accordance with Novartis ACUC regulations and guidelines. Electronic transponders for animal identification were implanted on the left flank one day prior to tumor implantation.
Tumor Implantation:
Cells in logarithmic growth phase were harvested and washed in 50 ml falcon tubes at 1200 rpm for 5 minutes, once in growth media and then twice in cold sterile PBS. Cells were resuspended in PBS at a concentration of 5×106 per ml, placed on ice, and injected in mice. Cancer cells were injected intravenously in 200 μl through the caudal vein. Both, the Nalm6 and SEM models endogenously express CD22 and thus, can be used to test the efficacy of CD22-directed CAR T cells in vivo. These models grow well when implanted intravenously in mice, which can be imaged for tumor burden measurements. Upon injection of 1×106 cancer cells, the tumors establish and can be accurately measured within 3 days. Baseline measurements are 4-6×105 photons/second (p/s). Within 7 days the mean bioluminescence measurement is 2-4×106 p/s and untreated tumors reach endpoint measurement (2-3×109) by 21-26 days. Anti-tumor activities of therapeutic agents are often tested once tumors are fully engrafted. Thus, there is a large window with these models during which the anti-tumor activity of CAR T cells can be observed.
CAR T Cell Dosing:
Mice were dosed 7 days after tumor implantation, with 1×106 CART cells for the treatment of Nalm6 and 3×106 CAR T cells for the treatment of SEM. Cells were partially thawed in a 37° C. water bath and then completely thawed by the addition of 1 ml of warmed growth media. The thawed cells were transferred to a 50 ml falcon tube and adjusted to a final volume of 12 ml with growth media. The cells were washed twice and spun at 300 g for 10 minutes and then counted by hemocytometer. T cells were then resuspended at respective concentrations in cold PBS and kept on ice until the mice were dosed. The CARTs were injected intravenously via the tail vein in 200 al, for a dose of 1 or 3×106 CAR T cells for Nalm6 and SEM bearing mice, respectively. 5 mice per group were either treated with 200 μl of PBS alone (PBS), non-transduced T cells (UTD), CD19 CAR T cells, as well as the novel CD22-65_Zmut, CD22-65_Zwt, CD22-65s_Zwt, m971_Zmut, or m971s_Zmut CAR T cells. All cells were prepared from the same donor in parallel.
Animal Monitoring:
The health status of the mice was monitored daily, including twice weekly body weight measurements. The percent change in body weight was calculated as (BWcurrent-BWinitial)/(BWinitial)×100%. Tumors were monitored 2 times weekly by imaging the mice.
The anti-tumor activity of CD22 CAR T cells was assessed in two B-cell acute lymphoblastic leukemia xenograft models. Following tumor cell implantation on day 0, tumor bearing mice were randomized into treatment groups and CAR T cells were administered intravenously via the lateral tail vein on day 7 after tumor implantation. Tumor growth and animal health were monitored until animals achieved endpoint.
In the Nalm6 model, mice which received PBS or UTDT cells were euthanized on day 22, when tumors were causing decreased hind leg mobility. All other groups were euthanized on day 40. The PBS treatment group, which did not receive any T cells, demonstrates baseline Nalm6 tumor growth kinetics in intravenously injected NSG mice. The UTD treatment group received non-transduced T cells and served as a T cell control to show the non-specific response of human donor T cells in this model. Both the PBS and UTD treatment groups demonstrated continuous tumor progression throughout this study. CD22-65_Zmut, CD22-65_Zwt, CD22-65s_Zwt, m971_Zmut, and m971s_Zmut CAR T cells all showed significantly slower tumor growth. CD22-65s_Zwt and m971_Zmut showed complete tumor regression as indicated from mean bioluminescence (
In the SEM model, mice which received PBS or UTD T cells were euthanized on day 27, when tumors were causing decreased hind leg mobility. All other groups were euthanized on day 45. The PBS treatment group, which did not receive any T cells, demonstrates baseline SEM tumor growth kinetics in intravenously injected NSG mice. The UTD treatment group received non-transduced T cells and served as a T cell control to show the non-specific response of human donor T cells in this model. Both the PBS and UTD treatment groups demonstrated continuous tumor progression throughout this study.
CD22-65_Zmut, CD22-65_Zwt, CD22-65s_Zwt, m971_Zmut, m971s_Zmut, and CD19 CAR T cells all showed significantly slower tumor growth. CD19 CARTs showed fastest tumor regression, followed by CD22-65s_Zwt and m971s_Zmut (
Patients and Methods:
An anti-CD22 CAR (“CD22 CAR”) including a longer linker (4×(GGGGS) (SEQ ID NO: 1086); LL) compared to anti-CD22 CAR including a short linker (1×(GGGGS) (SEQ ID NO: 1083); SL) between the light and heavy chains of the scFv was generated (
TABLE 16
Patient characteristics of patients infused with CART22LL T cells.
Pediatric ALL (n = 6)
Characteristic
Value (range, %)
Adult ALL (n = 3)
Age median (range)
14 years (4-25)
47 years (28-64)
Gender (%)
3 M (50%)/3 F (50%)
2 M (66.6%)/1 F (33.3%)
Race
5 caucasian/1 asian
3 caucasian
Prior allogeneic
⅚ (83.3%)
⅓ (33.3%)
transplantation (%)
Prior blinatumomab or
⅚ (83.3%) CART19
⅓ (33.3%) CART19
CART19 (%)
⅙ (16.6%) blinatumomab
3/3 (100%) blinatumomab
BM blast pre CART22
77.5% (0.6-95)
95% (0-97)
% (range)
⅚ (83.3%) CD19− relapse
⅓ (33.3%) CD19− relapse
⅙ (16.6%) CD19+ relapse
⅔ (66.6%) CD19+ relapse
CART22 dose
3.55 × 108 (3.96 × 107-5 × 108)
2 × 108 (5 × 107-5 × 108)
median (range)
CAR expression
36.4% (15-49.7)
25.8% (25-30)
For the adult trial, 5 patients were screened, 4 enrolled (1 patient withdrew consent) and 3 infused (1 manufacturing failure). For the pediatric trial, 9 patients were screened, 8 enrolled (1 screen failure) and 6 infused (two patients were not infused for disease progression).
For the preclinical studies, CART22LL and CART22SL were generated and tested in vivo using xenograft models. NSG mice were engrafted with either a luciferase+ standard B-ALL cell line (NALM6) or primary B-ALL cells obtained from a patient relapsing after CART19 (CHP110R). Additionally, 2-photon imaging was used to study the in vivo behavior and immune synapse formation and flow cytometry to asses T cell activation.
CART22 cells were successfully manufactured for 10 out of 12 patients. In the adult cohort, 3/3 patients developed CRS (gr.1-3) and no neurotoxicity was observed; in the pediatric cohort out of 5 evaluable patients (1 discontinued for lineage switch to AML on pre-infusion marrow), 3/5 developed cytokine-release syndrome (CRS) (all grade 2) and 1 patient had encephalopathy (gr.1). CART22 cells were expanded in the PB with median peak of 1977 (18-40314) copies/ug DNA at day 11-18. In an adult patient who had previously received CART19, a second CART19 re-expansion was observed following CART22 expansion (
A direct comparison of the two different CAR22 constructs (CART22S and CART22SL) was then performed. In xenograft models, CART22SL significantly outperformed CART22LL (
Although feasible and with manageable toxicity, CART22LL led to modest clinical responses for patients with r/r B-ALL. Preclinical evaluation allowed us to conclude that shortening the linker by 15 amino acids significantly increases the anti-leukemia activity of CART22, possibly by leading to more effective interactions between T cells and their targets. Finally, with the caveats of cross-trial comparison, these data suggest that xenograft models can predict the clinical efficacy of CART products and validate the use of In vivo models for lead candidate selection.
Genes encoding for single chain variable fragments for anti-CD22 antibodies were cloned into lentiviral CAR expression vectors with the CD3zeta chain and 4-1BB stimulatory molecules. The following anti-CD22 CAR constructs were evaluated: CD22-65_Zwt, CD22-65s_Zwt (short 1×(GGGGS) linker (SEQ ID NO: 1083); SEQ ID NO: 835), CD22-65ss_Zwt (no linker between the VH and VL regions; SEQ ID NO: 836), CD22-65sLH_Zwt (short 1×(GGGGS) linker (SEQ ID NO: 1083) with the VL region oriented at the N-terminus and the VH region oriented at the C-terminus), CD22-65sKD_Zwt (short 1×(GGGGS) linker (SEQ ID NO: 1083) and mutations in the FR regions of the VH and VL regions; SEQ ID NO: 837), and CD22m971s_Zmut (control) were evaluated. Except for CD22-65sLH_Zwt, all anti-CD22 CAR constructs have the VH region oriented at the N-terminus. The CD3zeta chain was either wildtype (Zwt) or carried a Q65K mutation (Zmut). The constructs were ranked based on the effector T cell responses of these CD22 CAR-transduced T cells (“CD22 CART” or “CD22 CART cells”) in response to CD22 expressing (“CD22+”) targets. Effector T cell responses include, but are not limited to, cellular expansion, proliferation, doubling, cytokine production and target cell killing or cytolytic activity (degranulation).
Generation of CD22 CAR T Cells:
Human scFv encoding lentiviral transfer vectors were used to produce the genomic material packaged into the VSVg pseudotyped lentiviral particles. Lentiviral transfer vector DNA encoding the CAR was mixed with the three packaging components VSVg, gag/pol and rev in combination with lipofectamine reagent to transfect Lenti-X 293T cells (Clontech), followed by medium replacement 12-18h later. 30 hours after medium change, the media is collected, filtered and stored at −80° C. CD22 CAR T cells were generated by starting with blood from healthy apheresed donors whose T cells were enriched by negative selection of T cells, CD4+ and CD8+ lymphocytes (Pan T cell isolation, Miltenyi). T cells were activated by the addition of CD3/CD28 beads (DYNABEADS® Human T-Expander CD3/CD28, ThermoFisher Scientific) at a ratio of 1:3 (T cell to bead) in T cell medium (RPMI1640, 10% heat-inactivated fetal calf serum (FCS), 2 mM L-glutamine, 1× Penicillin/Streptomycin, 100 μM non-essential amino acids, 1 mM Sodium Pyruvate, 10 mM Hepes, and 55 μM 2-mercaptoethanol) at 37° C., 5% CO2. T cells were cultured at 0.5×106 T cells in 1 mL medium per well of a 24-well plate. After 24 hours, when T cells were blasting, non-concentrated or concentrated viral supernatant was added; T cells were transduced at a multiplicity of infection (MOI) of 5. T cells began to proliferate, which is monitored by measuring the cell concentration (as counts per mL), and T cells are diluted in fresh T cell medium every two days. As the T cells began to rest down after approximately 10 days, the logarithmic growth wanes. The combination of slowing growth rate and reduced T cell size (approaching 350 fL) determines the state for T cells to be cryopreserved for later analysis. All CD22 CAR T cells were produced under research grade (i.e., not clinical grade) manufacturing conditions.
Before cryopreserving, the percentage of cells transduced (expressing the CD22-specific CAR on the cell surface) were determined by flow cytometric analysis on a FACS Fortessa (BD) (
Evaluating Potency of CD22 CAR-Redirected T Cells:
To evaluate the functional abilities of CD22 CAR T cells, the cells, generated as described above, were thawed, counted and co-cultured with cancer cells to read out their killing capabilities and secretion of cytokine. Human scFv bearing CARs CD22-65_Zwt, CD22-65s_Zwt, CD22-65ss_Zwt, CD22-65sLH_Zwt, CD22-65sKD_Zwt, and CD22-m971s_Zmut were compared and EGFRvIII CAR T cells as well as non-transduced T cells (UTD) were used as non-targeting T cells control. T cell killing was directed towards the acute lymphoblastic leukemia (ALL) lines Nalm6 (RRID: CVCL_0092) and SEM (RRID: CVCL_0095). Both cell lines were transduced to express luciferase as a reporter for cell viability/killing. The cytolytic activities of CD22 CARTs were measured at a titration of effector:target cell ratios (E:T) of 10:1, 5:1, 2.5:1, 1.25:1 0.63:1 and 0.31:1. Assays were initiated by mixing the respective number of T cells with a constant number of targets cells (25,000 cells per well of a 96-well plate). After 20 hours, remaining cells in the wells were lysed by addition of Bright-Glo™ Luciferase Assay System (Promega) reagent, to quantify the remaining Luc-expressing cancer cells in each well. “% Killing” was calculated in relation to wells containing target cells alone (0%, maximal Luc signal). The data show that transduction with the CD22 CART encoding lentiviruses transfers anti-CD22 killing activity to T cells (Nalm6 and SEM (
To measure cytokine production of CD22 CAR T cells in reponse to CD22-expressing target cells, CAR T cells were co-cultured with the same ALL lines as above. Cells were cultured at an effector:target ratio of 1:1 and 25,000 cells per well of a 96-well plate for 24 h, after which the media was removed for cytokine analysis using the V-PLEX Human IFN-γ Kit (Meso Scale Diagnostics). These data show that all CD22 CARTs, except CD22-65sLH_Zwt, produced IFN-γ when cultured Nalm6 or SEM (
All CD22-specific CARs in this experiment were expressed on the cell surface of primary human T cells similarly well: CD22-65_Zwt, CD22-65s_Zwt, CD22-65ss_Zwt, CD22-65sLH_Zwt, CD22-65sKD_Zwt, and CD22-m971s_Zmut. Additionally, all CD22 CARTs showed efficacy in T cell functional assays, except for CD22-65sLH_Zwt. clp Example 12: In Vivo Activity of CARTs Bearing Human Anti-CD22 scFv with Linker Variants
Anti-tumor activity of a set of CD22 CAR T cells was assessed in vivo in a NALM6 xenograft model. CAR T cells with CAR constructs CD22-65_Zwt, CD22-65s_Zwt, CD22-65ss_Zwt, CD22-65sLH_Zwt, CD22-65sKD_Zwt, and CD22-m971s_Zmut (control) were evaluated.
Cell Line:
Nalm6 (RRID: CVCL_0092) is a human acute lymphoblastic leukemia (ALL) cell line. Cells were grown in RMPI medium containing 10% fetal bovine serum and both grow in suspension. Cells persist and expand in mice when implanted intravenously. Cells have been modified to express luciferase, so that that tumor cell growth can also be monitored by imaging the mice after they have been injected with the substrate Luciferin.
Mice: 6 week old NSG (NOD.Cg-PrkdecscidI12rgtm1Wjl/SzJ) mice were received from the Jackson Laboratory (stock number 005557). Animals were allowed to acclimate in the Novartis NIBR animal facility for at least 3 days prior to experimentation. Animals were handled in accordance with Novartis ACUC regulations and guidelines. Electronic transponders for animal identification were implanted on the left flank one day prior to tumor implantation.
Tumor Implantation:
Cells in logarithmic growth phase were harvested and washed in 50 ml falcon tubes at 1200 rpm for 5 minutes, once in growth media and then twice in cold sterile PBS. Cells were resuspended in PBS at a concentration of 5×106 per ml, placed on ice, and injected inl through the caudal vein. Nalm6 cells endogenously express CD22 and thus, can be used to test the efficacy of CD22-directed CAR T cells in vivo. This model grows well when implanted intravenously in mice, which can be imaged for tumor burden measurements. Upon injection of 1×106 cancer cells, the tumors establish and can be accurately measured within 3 days. Baseline measurements are 4-6×105 photons/second (p/s). Within 7 days the mean bioluminescence measurement is 2-4×106 p/s and untreated tumors reach endpoint measurement (2-3×109) by 20-30 days. Anti-tumor activities of therapeutic agents are often tested once tumors are fully engrafted. Thus, there is a large window with these models during which the anti-tumor activity of CAR T cells can be observed.
CAR T Cell Dosing:
Mice were dosed 7 days after tumor implantation, with 1×106 CART cells for the treatment of Nalm6. Cells were partially thawed in a 37° C. water bath and then completely thawed by the addition of 1 ml of warmed growth media. The thawed cells were transferred to a 50 ml falcon tube and adjusted to a final volume of 12 ml with growth media. The cells were washed twice and spun at 300 g for 10 minutes and then counted by hemocytometer. T cells were then resuspended at respective concentrations in cold PBS and kept on ice until the mice were dosed. The CARTs were injected intravenously via the tail vein in 200 al, for a dose of 1×106 CART cells. 5 mice per group were either treated with 200 μl of PBS alone (PBS), EGFRvIII-transduced T cells, as well as the novel CD22-65_Zwt, CD22-65s_Zwt, CD22-65ss_Zwt, CD22-65sLH_Zwt, CD22-65sKD_Zwt, and CD22-m971s_Zmut CAR T cells. All cells were prepared from the same donor in parallel.
Animal Monitoring:
The health status of the mice was monitored daily, including twice weekly body weight measurements. The percent change in body weight was calculated as (BWcurrent−BWinitial)/(BWinitial)×100%. Tumors were monitored 2 times weekly by imaging the mice.
The anti-tumor activity of CD22 CAR T cells was assessed in a B-cell acute lymphoblastic leukemia xenograft model. Following tumor cell implantation on day 0, tumor bearing mice were randomized into treatment groups and CAR T cells were administered intravenously via the lateral tail vein on day 7 after tumor implantation. Tumor growth and animal health were monitored until animals achieved endpoint.
In this Nalm6 model, mice which received PBS or EGFRvIII T cells were euthanized on day 23, when tumors were causing decreased hind leg mobility. All other groups were euthanized on day 37. The mean bioluminescence for all treatment groups was then determined (
These data demonstrate that the CD22-specific CAR T cells CD22-65s_Zwt and CD22-65ss_Zwt are capable of strongly inhibiting the growth of NALM6 cancer at a low dose of 1×106 CART cells. The efficacy was superior to the control m971 CAR.
To test whether a single CART cell functionalized with two distinct scFVs can be activated by either of the antigens that the CART recognizes, CARTs comprising two distinct scFVs linked together were generated with scFVs targeting CD19 and CD22 (Tables 15 and 17 and
The generated constructs explore two different linkers connecting the two scFVs: LAEAAAK (SEQ ID NO: 1091) and GGGGS (SEQ ID NO: 1083). The LAEAAAK (SEQ ID NO: 1091) is a more rigid linker, while the GGGGS linker (SEQ ID NO: 1083) is a more flexible linker. The impact of the orientation of the light (L) and heavy (H) chains within the anti-CD22 scFV activation was also investigated. The anti-CD19 scFV was oriented as L/H (in a N- to C-terminus orientation) in all of the constructs. Two linkers connecting the H and L chains within the anti-CD22 scFV: GGGGSGGGGSGGGGS (SEQ ID NO: 1084) and GGGGS (SEQ ID NO: 1083) were also explored (annotated as “sh” in Tables 17 and 18). Control constructs engineered with the individual scFVs (anti-CD19 or anti-CD22) were also generated (Table 18).
TABLE 17
Constructs generated in the context of a single CART targeting CD22
and CD19. (Table discloses SEQ ID NOS 1091, 1083, 1091, 1083,
1091, 1083, 1091 and 1083, respectively, in order of appearance)
Distal
Linker
Proximal
H/L
L/H
αCD19
LAEA3K
αCD22
CG#c171
CG#c177
αCD19
G4S
αCD22
CG#c172
CG#c178
αCD19
LAEA3K
αCD22sh
CG#c173
CG#c179
αCD19
G4S
αCD22sh
CG#c174
CG#c180
αCD22
LAEA3K
αCD19
CG#c181
CG#c185
αCD22
G4S
αCD19
CG#c182
CG#c186
αCD22sh
LAEA3K
αCD19
CG#c183
CG#c187
αCD22sh
G4S
αCD19
CG#c184
CG#c188
TABLE 18
CAR construct controls generated to target CD19 or CD20.
H/L
L/H
αCD22
CAR22-65
CG#c175
αCD22sh
CG#c170
CG#c176
αCD19
CAR19
Evaluation of CAR Expression:
The sequences encoding the constructs listed in Tables 17 and 18 were cloned into a lentiviral backbone vector. All of the constructs comprised the leader sequence of the human CD8alpha at their N-terminus, which is expected to be cleaved co-translationally and excluded in the final protein product. Transgene expression was driven by the EF1alpha promoter. The resulting DNAs were used to transfect HEK-293 cells for viral production. Exemplary viral titers are shown in Table 19. Viral titers were determined based on surface expression of the various constructs in SupT1 cells, by FACS using two distinct staining reagents. An anti-idiotype antibody recognizing the scFv directed to CD19 and CD22-FC for staining the scFV directed to CD22 were used. Staining was performed individually.
TABLE 19
viral titers obtained for some constructs tested in the context of
CD19- and CD22- targeting
Sample
CD19 anti-ID Titer (TU/ml)
CD22-Fc (TU/ml)
c171
2.18E+06
2.51E+06
c172
3.29E+05
9.43E+05
c173
2.78E+05
7.34E+05
c174
8.82E+05
1.95E+06
c181
1.99E+06
2.54E+06
c182
1.01E+06
2.53E+06
c183
2.26E+06
1.85E+06
c184
2.24E+06
2.81E+06
c185
1.80E+07
1.99E+07
c186
9.49E+06
1.92E+07
c187
1.64E+07
1.76E+07
c188
3.44E+07
4.84E+07
c170
0.00E+00
6.18E+07
c175
0.00E+00
2.73E+06
c176
0.00E+00
7.84E+06
CAR22-65 Long
0.00E+00
2.60E+07
CAR19
8.16E+07
0.00E+00
For each construct, the viral titers obtained with the two staining reagents were averaged and the viruses were evaluated for their ability to transduce JNL cells. The Jurkat cells were previously transduced with a NFAT-Luciferase reporter construct. A multiplicity of infection of 3 was used. The percent cells positive for CAR in JNLs following transduction with the indicated constructs was determined (
These data show that the scFVs across the different constructs were detected at the cell surface, indicating expression and trafficking to the cell surface for the constructs targeting both CD19 and CD22. Use of a staining reagent that is the antigen itself (CD22-Fc) indicates that the scFV directed to CD22 acquired a correctly folded structure, which is compatible with recognition of the antigen within the CD22 protein. This result was observed whether the anti-CD22 scFV arm was engineered upstream or downstream of the antiCD19 scFV arm.
Evaluation of CAR Activation:
To evaluate whether a CAR comprising two distinct scFVs can be activated by either of the targeted antigens, we co-cultured the untransduced (UTD) and transduced JNLs with target cell lines expressing either CD19 (K562-CD19) or CD22 (K562-CD22). When the CAR recognized the CD19 or CD22 antigen, CAR engagement resulted in downstream NFAT activation. The NFAT-luciferase reporter in the JNL cells provided a measure to read out CAR activation. The expression levels of CD19 and CD22 in the different target cell lines is shown (
The level of NFAT-induced luciferase as a measure of CAR activity is shown (
The disclosures of each and every patent, patent application, and publication cited herein are hereby incorporated herein by reference in their entirety. While this invention has been disclosed with reference to specific aspects, it is apparent that other aspects and variations of this invention may be devised by others skilled in the art without departing from the true spitir and scope of the invention. The appended claims are intended to be construed to include allsuch aspect and equivalent variations.
The patent contains a lengthy “Sequence Listing” section. A copy of the “Sequence Listing” is available in electronic form from the USPTO web site (http://seqdata.uspto.gov/?pageRequest=docDetail&DocID=US10525083B2). An electronic copy of the “Sequence Listing” will also be available from the USPTO upon request and payment of the fee set forth in 37 CFR 1.19(b)(3).
Li, Na, Brogdon, Jennifer, Gill, Saar, Lei, Ming, Huang, Lu, Zhang, Jimin, Guimaraes, Carla, Ruella, Marco, Engels, Boris, Brannetti, Barbara, Granda, Brian, Young, Regina M.
Patent | Priority | Assignee | Title |
Executed on | Assignor | Assignee | Conveyance | Frame | Reel | Doc |
Sep 21 2017 | ZHANG, JIMIN | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Sep 21 2017 | GRANDA, BRIAN | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Sep 21 2017 | BRANNETTI, BARBARA | Novartis Pharma AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066082 | /0281 | |
Sep 22 2017 | BROGDON, JENNIFER | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Sep 22 2017 | HUANG, LU | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Sep 23 2017 | LEI, MING | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Sep 25 2017 | ENGELS, BORIS | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Sep 25 2017 | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | Novartis AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066083 | /0205 | |
Sep 25 2017 | Novartis Pharma AG | Novartis AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066083 | /0106 | |
Sep 25 2017 | LI, NA | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066260 | /0217 | |
Aug 13 2018 | GUIMARAES, CARLA | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066082 | /0540 | |
Sep 14 2018 | NOVARTIS INSTITUTES FOR BIOMEDICAL RESEARCH, INC | Novartis AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066083 | /0148 | |
Oct 15 2019 | The Trustees of the University of Pennsylvania | Novartis AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066164 | /0219 | |
Oct 15 2019 | The Trustees of the University of Pennsylvania | The Trustees of the University of Pennsylvania | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066164 | /0219 | |
Apr 16 2021 | GILL, SAAR | The Trustees of the University of Pennsylvania | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066083 | /0001 | |
Apr 30 2021 | RUELLA, MARCO | The Trustees of the University of Pennsylvania | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066083 | /0001 | |
Jan 05 2022 | Novartis AG | (assignment on the face of the patent) | / | |||
Jan 05 2022 | The Trustees of the University of Pennsylvania | (assignment on the face of the patent) | / | |||
Aug 04 2023 | YOUNG, REGINA M | The Trustees of the University of Pennsylvania | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066083 | /0001 | |
Aug 15 2023 | The Trustees of the University of Pennsylvania | The Trustees of the University of Pennsylvania | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066164 | /0357 | |
Aug 15 2023 | The Trustees of the University of Pennsylvania | Novartis AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066164 | /0357 | |
Jan 11 2024 | Novartis AG | Novartis AG | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066164 | /0555 | |
Jan 11 2024 | Novartis AG | The Trustees of the University of Pennsylvania | ASSIGNMENT OF ASSIGNORS INTEREST SEE DOCUMENT FOR DETAILS | 066164 | /0555 |
Date | Maintenance Fee Events |
Jan 05 2022 | BIG: Entity status set to Undiscounted (note the period is included in the code). |
Date | Maintenance Schedule |
Feb 27 2027 | 4 years fee payment window open |
Aug 27 2027 | 6 months grace period start (w surcharge) |
Feb 27 2028 | patent expiry (for year 4) |
Feb 27 2030 | 2 years to revive unintentionally abandoned end. (for year 4) |
Feb 27 2031 | 8 years fee payment window open |
Aug 27 2031 | 6 months grace period start (w surcharge) |
Feb 27 2032 | patent expiry (for year 8) |
Feb 27 2034 | 2 years to revive unintentionally abandoned end. (for year 8) |
Feb 27 2035 | 12 years fee payment window open |
Aug 27 2035 | 6 months grace period start (w surcharge) |
Feb 27 2036 | patent expiry (for year 12) |
Feb 27 2038 | 2 years to revive unintentionally abandoned end. (for year 12) |