Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma

Eugenia Zah, Eunwoo Nam, Vinya Bhuvan, Uyen Tran, Brenda Y Ji, Stanley B Gosliner, Xiuli Wang, Christine E Brown, Yvonne Y Chen, Eugenia Zah, Eunwoo Nam, Vinya Bhuvan, Uyen Tran, Brenda Y Ji, Stanley B Gosliner, Xiuli Wang, Christine E Brown, Yvonne Y Chen

Abstract

Chimeric antigen receptor (CAR)-T cell therapy has shown remarkable clinical efficacy against B-cell malignancies, yet marked vulnerability to antigen escape and tumor relapse exists. Here we report the rational design and optimization of bispecific CAR-T cells with robust activity against heterogeneous multiple myeloma (MM) that is resistant to conventional CAR-T cell therapy targeting B-cell maturation antigen (BCMA). We demonstrate that BCMA/CS1 bispecific CAR-T cells exhibit superior CAR expression and function compared to T cells that co-express individual BCMA and CS1 CARs. Combination therapy with anti-PD-1 antibody further accelerates the rate of initial tumor clearance in vivo, while CAR-T cell treatment alone achieves durable tumor-free survival even upon tumor re-challenge. Taken together, the BCMA/CS1 bispecific CAR presents a promising treatment approach to prevent antigen escape in CAR-T cell therapy against MM, and the vertically integrated optimization process can be used to develop robust cell-based therapy against novel disease targets.

Conflict of interest statement

Y.Y.C. and E.Z. declare competing financial interest in the form of a patent application whose value may be affected by the publication of this work. The other authors declare no competing interests.

Figures

Fig. 1. High-throughput generation and screening of…
Fig. 1. High-throughput generation and screening of BCMA/CS1 OR-gate CARs.
a A vertically integrated optimization process for CAR-T cell therapy development. b Schematic of a single-chain bispecific (OR-gate) CAR, which contains two ligand-binding domains connected in tandem. A panel of CAR variants was constructed from two CS1-binding scFvs and three BCMA-specific binding domains. Both murine and humanized versions of BCMA-binding scFvs (c11D5.3 and J22.9-xi) were evaluated. The CS1-binding huLuc63 and Luc90 scFvs were fixed at the membrane-distal and membrane-proximal positions, respectively, based on binding-epitope analysis. c Methodology for producing and screening bispecific CARs. CAR-T cells were generated in a 14-day cycle. Starting on day 9 post-stimulation, CAR-T cells were characterized for antitumor function in various assays, which could last up to 2 weeks.
Fig. 2. OR-gate CAR panel exhibits range…
Fig. 2. OR-gate CAR panel exhibits range of efficacy against BCMA+ and CS1+ targets.
a Cell-lysis activity of single-input and bispecific CD8+ CAR-T cells against K562 targets engineered to express either BCMA or CS1. Cells were seeded at an effector-to-target (E:T) ratio of 2:1, where effector-cell seeding was based on CAR+ T-cell count. The fraction of viable K562 cells left after a 20-h coincubation was quantified by fluorescence imaging of target cells using IncuCyte. All bispecific CARs in this panel contained a short extracellular spacer. b Proliferation of single-input and bispecific BCMA/CS1 CD8 + CAR-T cells upon antigen stimulation. CAR-T cells were stained with CellTrace Violet (CTV) dye. CTV median fluorescence intensity (MFI) was quantified by flow cytometry after a 5-day coincubation with parental (BCMA−/CS1−), BCMA+, or CS1+  K562 target cells at a 2:1 E:T ratio. CARs containing huLuc63 paired with dAPRIL, J22.9-xi, and huJ22.9-xi were subsequently eliminated from the panel based on poor cytotoxicity and/or T-cell proliferation. c Cytotoxicity of reduced bispecific CAR-T-cell panel upon repeated antigen challenge. CD8+ CAR-T cells were coincubated with K562 target cells at a 1:1 E:T ratio and rechallenged every 2 days with fresh target cells. Viable target-cell count was quantified by flow cytometry 2 days after each target-cell addition. ‘C#’ denotes the challenge number and ‘D#’ denotes the number of days post challenge. d Characterization of the remaining K562 target-cell populations after four challenges from c reveals differing antigen preference among the panel of bispecific CARs. Values shown are the means of technical triplicate samples, with error bars indicating +1 standard deviation (SD). P-values were calculated by unpaired two-tailed Student’s t-test; n.s. not statistically significant (p > 0.05); *p < 0.05; **p < 0.01, with Bonferroni correction for multiple comparisons applied. P-values in c were calculated for the final time point for each construct, relative to the top-performing CAR, c11D5.3-Luc90. Source data are provided as a Source Data File. P-values in a and b can be referenced in the Source Data File.
Fig. 3. OR-gate CAR-T-cells outperform T cells…
Fig. 3. OR-gate CAR-T-cells outperform T cells co-expressing two separate CARs.
a Single-input and bispecific CARs were tagged with an N-terminal FLAG tag. In DualCAR constructs, the CS1 CAR was N-terminally tagged with a FLAG tag while the BCMA CAR was N-terminally tagged with a HA tag. b CAR surface expression levels were quantified by surface antibody staining of FLAG and HA tags followed by flow cytometry. Each single-input and single-chain bispecific CAR was tagged with an N-terminal FLAG. The first CAR in each DualCAR construct was tagged with the FLAG while the second CAR was tagged with HA. See Supplementary Fig. 4b for construct schematics. c Transduction efficiency as measured by % CAR+ (FLAG+, HA+, or FLAG+HA+) Data shown represent the mean value from three different donors, with error bars indicating +1 SD. d Median fluorescence intensity (MFI) of FLAG antibody staining for T cells expressing either OR-gate or DualCAR constructs. e NM CAR-T-cell proliferation following a 5-day coincubation with MM.1 S target cells. Values shown are the means of technical triplicate samples from the same donor, with error bars indicating +1 SD. Data shown are representative of results from two independent experiments performed with cells from two different healthy donors. P-values for all panels were calculated by unpaired two-tailed Student’s t-test; n.s. not statistically significant (p > 0.05); *p < 0.05; **p < 0.01, with Bonferroni correction for multiple comparisons applied. P-values in c were calculated for the OR-gate CAR constructs, relative to their corresponding DualCAR constructs. Source data are provided as a Source Data File. P-values in e can be referenced in the Source Data File.
Fig. 4. BCMA/CS1 OR-gate CAR-T cells prevent…
Fig. 4. BCMA/CS1 OR-gate CAR-T cells prevent antigen escape in vivo.
a Evaluation of single-input and bispecific CAR-T cells in vivo. Mice were engrafted with a mixture of 1.5 × 106 MM.1 S cells containing a 1:1:1 ratio of BCMA+/CS1−, BCMA−/CS1+, and BCMA+/CS1+ cells. Tumor-bearing animals were treated with 1.5 × 106 EGFRt- or CAR-expressing T cells on day 5 (5 days after tumor injection) and day 13. Six mice were included in each initial treatment group but only five mice in the huLuc63-c11D5.3 group were redosed due to limited T-cell availability. Tumor progression was monitored by bioluminescence imaging. b Survival of mice shown in a. Statistical difference (depicted) between survival of huLuc63-c11D5.3-treatment group compared with other treatment groups was determined using log-rank analysis, applying a Chi-square distribution with one degree of freedom; n.s. not statistically significant (p > 0.05); *p < 0.05; **p < 0.01. P-values for the different treatment groups are as follows: Untreated, p = 0.024; EGFRt, p = 0.024; Luc90, p = 0.011; huLuc63, p = 0.015; c11D5.3, p = 0.008; huc11D5.3-Luc90, p = 0.114. c BCMA/CS1 antigen expression on tumors harvested from mice treated with CS1 single-input Luc90 Short, huLuc63 Long, or BCMA single-input c11D5.3 Long CAR-T cells. Values shown are the means of n = 4 technical replicates for cells stained prior to injection. Thirteen to 18 tumor samples were collected from different locations in the mice (six mice per test group) at the time of sacrifice, with error bars indicating +1 standard deviation (SD), and each sample corresponding to 100 or more events as detected by flow cytometry. d Antigen expression pattern on tumor cells recovered at the time of animal sacrifice. Each data point d corresponds to an individual tumor sample recovered that included more than 100 tumor cells as detected by flow cytometry; each mouse generally contained multiple tumors at the time of sacrifice. Source data are provided as a Source Data File.
Fig. 5. Combination therapy with anti-PD-1 increases…
Fig. 5. Combination therapy with anti-PD-1 increases initial antitumor efficacy but not durability of response in vivo.
a Mice were engrafted with 1.5 × 106 WT MM.1 S cells. Tumor-bearing animals were treated with 1.5 × 106 EGFRt- or CAR-expressing T cells on day 8 (8 days after tumor injection) and day 16. Tumor progression was monitored by bioluminescence imaging and shown for individual animals in each test group (n = 6). On day 133, animals that had been tumor-free for at least 7 weeks (3 each in the huLuc63-c11D5.3 Short groups with and without anti-PD-1) were rechallenged with 1.5 × 106 WT MM.1 S cells. Black arrows indicate time of cell injections; red arrows indicate instances of CAR-T cells eradicating palpable tumor nodules. The endpoint for each animal is marked with an “X.” b CD38 and PD-1 expression on T cells persisting in mice at time of animal sacrifice. Each data point in b corresponds to an individual tumor mass, tissue (e.g., brain and spleen), or cardiac blood sample recovered. For PD-1 expression: 23 samples were collected from six mice in the group not treated with anti-PD-1, and 26 samples were collected from six mice in the group treated with anti-PD-1. For CD38 expression: seven samples were collected from two mice in the group not treated with anti-PD-1, and nine samples were collected from three mice in the group treated with anti-PD-1. Only samples that contained at least 10 human CD45+ cells as detected by flow cytometry were included for analysis. P-values were calculated by unpaired two-tailed Student’s t-test. Source data are provided as a Source Data File.

References

    1. Cancer Facts and Figures 2019, American Cancer Society (2019). .
    1. Cowan AJ, et al. Global burden of multiple myeloma. JAMA Oncol. 2018;98121:1221–1227. doi: 10.1001/jamaoncol.2018.2128.
    1. Moreau P, Attal M, Facon T. Frontline therapy of multiple myeloma. Blood. 2015;125:3076–3085. doi: 10.1182/blood-2014-09-568915.
    1. Ali SA, et al. T cells expressing an anti-B cell maturation antigen chimeric antigen receptor cause remissions of multiple myeloma. Blood. 2016;128:1688–1701. doi: 10.1182/blood-2016-04-711903.
    1. Brudno J, et al. T cells genetically modified to express an anti-B cell maturation antigen chimeric antigen receptor cause remissions of poor-prognosis relapsed multiple myeloma. J. Clin. Oncol. 2018;36:2267–2280. doi: 10.1200/JCO.2018.77.8084.
    1. Berdeja JG, et al. Durable clinical responses in heavily pretreated patients with relapsed/refractory multiple myeloma: Updated results from a multicenter study of bb2121 anti-Bcma CAR T cell therapy. Blood. 2017;130:740.
    1. Cohen AD, et al. Safety and efficacy of B cell maturation antigen (BCMA)-specific chimeric antigen receptor T cells (CART-BCMA) with cyclophosphamide conditioning for refractory multiple myeloma (MM) Blood. 2017;130:505. doi: 10.1182/blood-2017-08-803551.
    1. Smith EL, et al. Development and evaluation of a human single chain variable fragment (scFv) derived Bcma rargeted CAR T cell vector leads to a high objective response rate in patients with advanced MM. Blood. 2017;130:742. doi: 10.1182/blood-2017-02-769869.
    1. Grada Z, et al. TanCAR: A novel bispecific chimeric antigen receptor for cancer immunotherapy. Mol. Ther. Nucleic Acids. 2013;2:e105. doi: 10.1038/mtna.2013.32.
    1. Zah, E., Lin, M.-Y., Silva-Benedict, A., Jensen, M. C. & Chen, Y. Y. T cells expressing CD19/CD20 bi-specific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol. Res. 10.1158/2326-6066.CIR-15-0231 (2016).
    1. Hegde M, et al. Tandem CAR T cells targeting HER2 and IL13R α 2 mitigate tumor antigen escape. J. Clin. Invest. 2016;126:3036–3052. doi: 10.1172/JCI83416.
    1. Qin H, et al. Novel CD19/CD22 Bicistronic chimeric antigen receptors outperform single or bivalent CARs in eradicating CD19+CD22+, CD19−, and CD22− Pre-B Leukemia. Am. Soc. Hematol. 2017;130:810.
    1. Lee L, et al. An APRIL-based chimeric antigen receptor for dual targeting of BCMA and TACI in multiple myeloma. Blood. 2018;131:746–758. doi: 10.1182/blood-2017-05-781351.
    1. Novak AJ, et al. Expression of BCMA, TACI, and BAFF-R in multiple myeloma: a mechanism for growth and survival. Blood. 2004;103:689–694. doi: 10.1182/blood-2003-06-2043.
    1. Moreaux J, et al. The level of TACI gene expression in myeloma cells is associated with a signature of microenvironment dependence versus a plasmablastic signature. Blood. 2005;106:1021–1030. doi: 10.1182/blood-2004-11-4512.
    1. Lee L, et al. Evaluation of B cell maturation antigen as a target for antibody drug conjugate mediated cytotoxicity in multiple myeloma. Br. J. Haematol. 2016;174:911–922. doi: 10.1111/bjh.14145.
    1. Hsi ED, et al. CS1, a potential new therapeutic antibody target for the treatment of multiple myeloma. Clin. Cancer Res. 2008;14:2775–2784. doi: 10.1158/1078-0432.CCR-07-4246.
    1. Tai Y-T, et al. Anti-CS1 humanized monoclonal antibody HuLuc63 inhibits myeloma cell adhesion and induces aritibody-dependent cellular cytotoxicity in the bone marrow mitieu. Blood. 2008;112:1329–1337. doi: 10.1182/blood-2007-08-107292.
    1. Wang X, et al. Lenalidomide enhances the function of CS1 chimeric antigen receptor-redirected T cells against multiple myeloma. Clin. Cancer Res. 2018;24:106–119. doi: 10.1158/1078-0432.CCR-17-0344.
    1. Hudecek M, et al. Receptor affinity and extracellular domain modifications affect tumor recognition by ROR1-specific chimeric antigen receptor T cells. Clin. Cancer Res. 2013;19:3153–3164. doi: 10.1158/1078-0432.CCR-13-0330.
    1. Hudecek M, et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol. Res. 2015;3:125–135. doi: 10.1158/2326-6066.CIR-14-0127.
    1. James JR, Vale RD. Biophysical mechanism of T-cell receptor triggering in a reconstituted system. Nature. 2012;487:64–69. doi: 10.1038/nature11220.
    1. Williams, M., Tso, Y., Landolfi, N. F., Powers, D. B. & Liu, G. Therapeutic use of anti-CS1 antibodies US (US Patents, 2010). .
    1. Chen KH, et al. A compound chimeric antigen receptor strategy for targeting multiple myeloma. Leukemia. 2018;32:402–412. doi: 10.1038/leu.2017.302.
    1. Bouchon A, Cella M, Grierson HL, Cohen JI, Colonna M. Cutting edge: activation of NK cell-mediated cytotoxicity by a SAP-independent receptor of the CD2 family. J. Immunol. 2001;167:5517–5521. doi: 10.4049/jimmunol.167.10.5517.
    1. Turtle CJ, et al. CD19 CAR-T cells of defined CD4+: CD8+ composition in adult B cell ALL patients. J. Clin. Invest. 2016;1:1–16.
    1. Berger C, et al. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J. Clin. Invest. 2008;118:294–305. doi: 10.1172/JCI32103.
    1. Sommermeyer D, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016;30:492–500. doi: 10.1038/leu.2015.247.
    1. Xu Y, et al. Closely-related T-memory stem cells correlate with in-vivo expansion of CAR.CD19-T cells in patients and are preserved by IL-7 and IL-15. Blood. 2014;123:3750–3759. doi: 10.1182/blood-2014-01-552174.
    1. Frigault MJ, et al. Identification of chimeric antigen receptors that mediate constitutive or inducible proliferation of T cells. Cancer Immunol. Res. 2015;3:356–367. doi: 10.1158/2326-6066.CIR-14-0186.
    1. Eyquem J, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature. 2017;543:113–117. doi: 10.1038/nature21405.
    1. Verma V, et al. PD-1 blockade in subprimed CD8 cells induces dysfunctional PD-1+CD38hi cells and anti-PD-1 resistance. Nat. Immunol. 2019;20:1231–1243. doi: 10.1038/s41590-019-0441-y.
    1. Zelba H, et al. Accurate quantification of T-cells expressing PD-1 in patients on anti-PD-1 immunotherapy. Cancer Immunol. Immunother. 2018;67:1845–1851. doi: 10.1007/s00262-018-2244-7.
    1. Rosenzweig M, et al. Preclinical data support leveraging CS1 chimeric antigen receptor T-cell therapy for systemic light chain amyloidosis. Cytotherapy. 2017;19:861–866. doi: 10.1016/j.jcyt.2017.03.077.
    1. Lonial S, et al. Elotuzumab therapy for relapsed or refractory multiple myeloma. N. Engl. J. Med. 2015;373:621–631. doi: 10.1056/NEJMoa1505654.
    1. Magen H, Muchtar E. Elotuzumab: the first approved monoclonal antibody for multiple myeloma treatment. Ther. Adv. Hematol. 2016;7:187–195. doi: 10.1177/2040620716652862.
    1. Zonder JA, et al. A phase 1, multicenter, open-label, dose escalation study of elotuzumab in patients with advanced multiple myeloma. Blood. 2013;120:552–559. doi: 10.1182/blood-2011-06-360552.
    1. Kumar M, Keller B, Makalou N, Sutton RE. Systematic determination of the packaging limit of lentiviral vectors. Hum. Gene Ther. 2001;12:1893–1905. doi: 10.1089/104303401753153947.
    1. Bos TJ, De Bruyne E, Van Lint S, Heirman C, Vanderkerken K. Large double copy vectors are functional but show a size-dependent decline in transduction efficiency. J. Biotechnol. 2010;150:37–40. doi: 10.1016/j.jbiotec.2010.07.010.
    1. Qin H, Haso W, Nguyen SM, Fry TJ. Preclinical development of bispecific chimeric antigen receptor targeting both CD19 and CD22. Blood. 2015;126:4427. doi: 10.1182/blood.V126.23.4427.4427.
    1. Gibson DG, et al. Enzymatic assembly of DNA molecules up to several hundred kilobases. Nat. Methods. 2009;6:343–345. doi: 10.1038/nmeth.1318.
    1. Brogdon, J. et al. WO2016014565_Treatment of cancer using humanized anti-BCMA CAR.pdf. (US Patents, 2016). .
    1. Carpenter RO, et al. B cell maturation antigen is a promising target for adoptive T-cell therapy of multiple myeloma. Clin. Cancer Res. 2013;19:2048–2060. doi: 10.1158/1078-0432.CCR-12-2422.
    1. Oden F, et al. Potent anti-tumor response by targeting. Mol. Oncol. 2015;9:1348–1358. doi: 10.1016/j.molonc.2015.03.010.
    1. Lee LSH, et al. An APRIL based chimeric antigen receptor to simultaneously target BCMA and TACI in multiple myeloma (MM) has potent activity in vitro and in vivo. Blood. 2016;128:379. doi: 10.1182/blood.V128.22.379.379.
    1. Chu J, et al. Genetic modification of T cells redirected toward CS1 enhances eradication of myeloma cells. Clin. Cancer Res. 2014;20:3989–4000. doi: 10.1158/1078-0432.CCR-13-2510.
    1. Yu, J., Hofmeister, C. & Chu, J. WO2014179759_CS1 CAR.pdf. (US Patents, 2014). .
    1. Wang X, et al. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood. 2011;118:1255–1263. doi: 10.1182/blood-2011-02-337360.

Source: PubMed

3
订阅