CAR T Update Part 1: Learning and Opportunities in Hematologic Cancers

Share
T cell attacking cancer cells

Rapid advances in chimeric antigen receptor (CAR) T cell therapy technologies are in development and under investigation in a range of preclinical and clinical research around the globe.

In our paper, published recently in the journal Expert Review of Hematology, we review the main challenges with current CD19-targeted CAR, obstacles to adopting CAR T cell therapy in solid tumors, and various strategies scientists are pursuing to overcome these issues. (1)

This CAR T Update Part 1 provides an overview of antigen escape, T cell persistence, and safety when using CARs in hematologic malignancies within and beyond the setting of B-cell acute lymphoblastic leukemia (B-ALL). We also provide an overview of the range of different strategies under investigation to address these issues.

Refer to CAR T Update Part 2 to learn about how scientists are building on lessons in the setting of hematologic cancers to find ways to achieve efficacy in solid tumor settings. In Part 2, we also discuss issues related to scaling production and financial accessibility.

Scientists at Memorial Sloan Kettering Cancer Center (MSK) pioneered CAR T cell therapy as a treatment for cancer and were the first to demonstrate that CD19 was a robust target. We continue to explore new ways to advance CAR T cells as a treatment for other hematologic cancers, including lymphoma and multiple myeloma, as well as extending their use to solid tumors, including breast cancer, ovarian cancer, lung cancer, and mesothelioma.

The Evolution of CAR T Cell Therapy

CARs are genetically engineered receptors on T cells. They include a human leukocyte antigen-independent antigen recognition domain, usually in the form of a single chain variable fragment (scFv) from an antibody, combined with a T cell activation domain. (2) Once activated, CAR T cells become cytotoxic against the target antigen.

First-, second-, and third-generation CAR receptors have none, one, or two costimulatory domains, respectively. Newer generations, known as armored CARs, also deliver cytokines or PD1-blocking scFvs, which enhance anti-cancer activity.

Despite the high response rates of 80 to 90 percent achieved with CD19-targeted second-generation CAR T cells in treating B-ALL, (3)(4)(5)(6)(7)(8) they have not had comparable responses in other CD19-positive hematologic cancers, or solid tumors.

Antigen Escape

An epitope is the specific segment of a protein that an antibody binds to. There is evidence of the target epitope loss in B-ALL patients who lose durability of remission after treatment with CD19-directed CARs. (9) Other research suggests that epitope loss may be secondary to deletions, mutations, and alternative splicing of the CD19 antigen recognition domain, allowing tumor cells to escape recognition. (10)

CD19 epitope loss has also led to the emergence of a myeloid clone after treatment for mixed- lineage leukemia (MLL)-rearranged B-ALL in disease recurrence. (11) Interestingly, antigen loss has also been observed in lymphoma (12) and chronic lymphocytic leukemia (CLL) (13) with CAR T cell therapy.

Preclinical models have shown that a process called trogocytosis may play a role in antigen escape with both CD28- and 4-1BB-based CARS. Trogocytosis is an active process that tumors use to transfer the target antigen onto T cells so they can escape destruction. It is believed to be reversible but has yet to be demonstrated in humans. (14)

One solution to overcome antigen escape is to combine single-target CARs that address different targets as a pooled product or use dual CARs (15)(16)(17) or tandem CARs. (18) These combination approaches are currently being tested in clinical trials NCT03330691, NCT03241940, and NCT04029038.

Another approach under investigation is low-dose radiation to sensitize antigen-negative tumor cells to CAR T cells and induce their death through the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). (19)

T cell Persistence

So far, there is no consensus on the optimal duration of persistence for maintaining long-term remission with CAR T cell therapy. Comparable response rates have been observed with a median duration of persistence of 14 days (3)and 168 days. (4)Still, relapses have been observed in some cases where CARs were no longer performing immunosurveillance. (8)

T-cell stimulation is a double-edged sword: It increases persistence, but overstimulation can result in exhaustion. (20) Some researchers have demonstrated that longevity can be improved without compromising efficacy by eliminating a signaling redundancy, or by placing immunoreceptor tyrosine-based activation motifs (ITAMS) near the membrane. (21)

A well-studied explanation for decreased persistence of CAR T cells is transgenic immune responses against scFv of murine origin. (22)(23)(24) A fully human scFv-based CAR has shown promising activity against the target antigen in preclinical research. (25) It may be possible to use human scFv-based CARs as a salvage therapy against the same antigen in patients treated with murine-scFv CARs, as shown in a study among B-ALL patients. (26) Clinical trials using humanized (NCT02374333), as well as fully human scFv-based CARs (NCT03684889 and NCT03602612), are in progress.

Several groups have studied central memory (27)(28) and stem cell memory T cells (29)(30)(31)(32) to learn more about persistence mechanisms in adoptive cell therapy (example: CARs). Approaches using defined ratios of CD4:CD8 T cells revealed that the CD4 subset is vital for boosting overall anti-tumor activity. (33)(34) Ongoing clinical trials in this area are NCT01318317, NCT01815749, NCT03389230, and NCT02706405.

Persistence and reduced exhaustion in CAR T cells have been achieved by optimizing the ratio of CD4:CD8 cells through alterations to signaling domains and pathways. (35) Another promising design modification involved diminishing redundant signaling, which improved effector/memory cell ratios and CAR persistence. (21)

An alternative approach to improve persistence involves stimulating the T-cell receptor using antigens from viruses, such as the Epstein-Barr virus (EBV) or cytomegalovirus (CMV), and then using the cytotoxic T cells to transduce CARs.
Dr. Manjusha Namuduri

An alternative approach to improve persistence involves stimulating the T-cell receptor using antigens from viruses, such as the Epstein-Barr virus (EBV) or cytomegalovirus (CMV), and then using the cytotoxic T cells to transduce CARs. Clinical trials underway testing this approach with hematologic and solid tumor antigen-directed CARs are NCT00085930, NCT01109095, NCT03768310, NCT01460901, and NCT01953900.

Finally, tumor vaccines and antigen presenting cells (APCs) may provide an immunological boost to CAR efficacy and persistence. See NCT01953900, NCT02482532, and NCT03186118.

Safety

One of the biggest challenges with CAR T cell therapy is the potential for toxicity, since healthy cells may express the same targets as cancer cells. Cell engineers are incorporating “off” and “on” switches and employing other strategies to mitigate toxicity.

CARs with “off” switches, also called “suicide genes,” can be rapidly destroyed when toxicities arise or after they have eliminated cancer cells. (36)(37)(38)(39) Dasatinib has recently shown it can pause CAR T cell activity temporarily, acting as a reversible switch. (40) Current trials investigating safety “off” switches include NCT02414269, NCT03696784, NCT03500991, NCT02311621, and NCT03016377. Other strategies for inactivating CARs involve checkpoint blockade, (41) titrating activation with an antibody switch, (42) and using self-limiting or transient CARs. (43)(44)(45)(46)

“On” switches activate CARs only in the presence of an activating entity. (47) A platform called GoCAR-T, where a small molecule activates the costimulatory signal only when target antigen- CAR interaction is present, is currently being tested in clinical trials. Recent data from NCT02744287 showed the approach effectively boosted expansion and persistence in four patients. (48) The safety of the GoCAR-T platform may be further strengthened by adding an “off” switch. (49) Another approach is to use synthetic NOTCH receptors that sense environmental cues and function as “smart” CARs. (50)

Toxicities associated with CAR T cell therapy are cytokine release syndrome (CRS) and neurotoxicity, including temporary headache, confusion, and delirium. High-grade CRS, an acute inflammatory response characterized by fever and multiple organ failure and fever, is rare but can occur.

Beyond safety switches, scientists are investigating ways to reduce toxicities without compromising efficacy. For example, one strategy is the prophylactic administration of tocilizumab, an interleukin-6 (IL-6) blocking antibody (see NCT02906371), or an IL-1 antagonist. (51)

Emerging evidence in the setting of CD19-directed CARs for B-ALL shows that the severity of CRS depends on tumor burden. (7)(8)(52) Therefore, strategies to reduce the risk of CRS include optimizing patient selection, changing the conditioning regimen, (53) and using a split-dose approach (54) for CAR T cell infusion.

Advancing CAR T Innovation

At MSK, we are dedicated to finding new ways to improve patient outcomes through groundbreaking preclinical and clinical research. We are currently conducting 15 clinical trials testing CAR T cell therapy in a range of indications, including lymphomas, multiple myeloma, advanced breast cancer, mesothelioma, and lung cancer.

The paper was not funded. Dr. Namuduri is an adhoc consultant for Cellectar Biosciences. Dr. Brentjens receives royalties and grant support from JUNO Therapeutics and is a consultant for JUNO/Celgene. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Refer a Patient
Call our dedicated clinician access number at 646-677-7440 or click the link below, and one of our care advisors will assist you with your referral needs.
  1. Namuduri M, Brentjens RJ. Enhancing CAR T cell efficacy: the next step toward a clinical revolution? Expert Rev Hematol. 2020;13(5):533–543.
  2. Krause A, Guo H-F, Latouche J-B, et al. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med. 1998;188(4):619–626.
  3. Park JH, Riviere I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378(5):449–459.
  4. **Most recent data of the two largest CD19 clinical trial datasets.
  5. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–448.
  6. Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra25.
  7. Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509–1518.
  8. Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5(177):177ra38.
  9. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517.
  10. Grupp SA, Maude SL, Shaw P, et al. T cells engineered with a chimeric antigen receptor (CAR) targeting CD19 (CTL019) have long term persistence and induce durable remissions in children with relapsed, refractory ALL. Am Soc Hematol. 2014;124(21):380.
  11. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov. 2015;5(12):1282–1295.
  12. Gardner R, Wu D, Cherian S, et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood. 2016;127(20):2406–2410.
  13. Brudno JN, Shi V, Stroncek D, et al. T cells expressing a novel fully-human anti-CD19 chimeric antigen receptor induce remissions of advanced lymphoma in a first-in-humans clinical trial. Am Soc Hematol. 2016;128(22):999.
  14. Porter DL, Frey NV, Melenhorst JJ, et al. Randomized, phase II dose optimization study of chimeric antigen receptor modified T cells directed against CD19 (CTL019) in patients with relapsed, refractory CLL. Am Soc Hematol. 2014;124(21):1982
  15. Hamieh M, Dobrin A, Cabriolu A, et al. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature. 2019;568(7750):112–116.
  16. Qin H, Ramakrishna S, Nguyen S, et al. Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol Ther Oncolytics. 2018;11:127–137.
  17. Ruella M, Barrett DM, Kenderian SS, et al. Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J Clin Invest. 2016;126(10):3814–3826.
  18. Hegde M, Corder A, Chow KK, et al. Combinational targeting offsets antigen escape and enhances effector functions of adoptively transferred T cells in glioblastoma. Mol Ther. 2013;21(11):2087–2101.
  19. Hegde M, Mukherjee M, Grada Z, et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest. 2016;126(8):3036–3052.
  20. DeSelm C, Palomba ML, Yahalom J, et al. Low-dose radiation conditioning enables CAR T cells to mitigate antigen escape. Mol Ther. 2018;26(11):2542–2552.
  21. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol. 2015;15(8):486–499.
  22. Feucht J, Sun J, Eyquem J, et al. Calibration of CAR activation potential directs alternative T cell fates and therapeutic potency. Nat Med. 2019;25(1):82–88.
  23. Maus MV, Haas AR, Beatty GL, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1(1):26–31.
  24. Turtle CJ, Hanafi L-A, Berger C, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med. 2016;8(355):355ra116.
  25. Turtle CJ, Hanafi L-A, Berger C, et al. CD19 CAR-T cells of defined CD4+: CD8+composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–2138.
  26. Brudno JN, Hartman SD, Pittaluga S, et al. Clinical anti-lymphoma activity and toxicity of T cells expressing a novel anti-CD19 chimeric antigen receptor with fully-human variable regions. J Clin Oncol. 2018;36(15_suppl):3052.
  27. Maude SL, Barrett DM, Rheingold SR, et al. Efficacy of humanized CD19-targeted chimeric antigen receptor (CAR)-modified T cells in children and young adults with relapsed/refractory acute lymphoblastic leukemia. Am Soc Hematol. 2016;128(22):217.
  28. Wang X, Popplewell LL, Wagner JR, et al. Phase 1 studies of central memory-derived CD19 CAR T-cell therapy following autologous HSCT in patients with B-cell NHL. Blood. 2016;127 (24):2980–2990.
  29. Yang S, Ji Y, Gattinoni L, et al. Modulating the differentiation status of ex vivo-cultured anti-tumor T cells using cytokine cocktails. Cancer Immunol Immunother. 2013;62(4):727–736.
  30. Gattinoni L, Lugli E, Ji Y, et al. A human memory T cell subset with stem cell-like properties. Nat Med. 2011;17(10):1290–1297.
  31. Blaeschke F, Stenger D, Kaeuferle T, et al. Induction of a central memory and stem cell memory phenotype in functionally active CD4(+) and CD8(+) CAR T cells produced in an automated good manufacturing practice system for the treatment of CD19(+) acute lymphoblastic leukemia. Cancer Immunol Immunother. 2018;67(7):1053–1066.
  32. Singh N, Perazzelli J, Grupp SA, et al. Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Sci Transl Med. 2016;8(320):320ra3.
  33. Sabatino M, Hu J, Sommariva M, et al. Generation of clinical-grade CD19-specific CAR-modified CD8+ memory stem cells for the treatment of human B-cell malignancies. Blood. 2016;128(4):519–528.
  34. Wang D, Aguilar B, Starr R, et al. Glioblastoma-targeted CD4+ CAR T cells mediate superior antitumor activity. JCI Insight. 2018;3(10).
  35. Sommermeyer D, Hudecek M, Kosasih PL, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia. 2016;30(2):492–500.
  36. Zhao Z, Condomines M, van der Stegen SJ, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28(4):415–428.
  37. Lupo-Stanghellini MT, Provasi E, Bondanza A, et al. Clinical impact of suicide gene therapy in allogeneic hematopoietic stem cell transplantation. Hum Gene Ther. 2010;21(3):241–250.
  38. Di Stasi A, Tey S-K, Dotti G, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med. 2011;365 (18):1673–1683.
  39. Wang X, Chang W-C, Wong CW, et al. A transgene-encoded cell surface polypeptide for selection, in vivo tracking, and ablation of engineered cells. Blood. 2011;118(5):1255–1263.
  40. Vogler I, Newrzela S, Hartmann S, et al. An improved bicistronic CD20/tCD34 vector for efficient purification and in vivo depletion of gene-modified T cells for adoptive immunotherapy. Mol Ther. 2010;18(7):1330–1338.
  41. Mestermann K, Giavridis T, Weber J, et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci Transl Med. 2019;11(499):eaau5907.
  42. Fedorov VD, Themeli M, Sadelain M. PD-1- and CTLA-4-based inhibitory chimeric antigen receptors (iCARs) divert off-target immunotherapy responses. Sci Transl Med. 2013;5(215):215ra172.
  43. Cao Y, Rodgers DT, Du J, et al. Design of switchable chimeric antigen receptor T cells targeting breast cancer. Angew Chem Int Ed Engl. 2016;55(26):7520–7524.
  44. Zhao Y, Zheng Z, Cohen CJ, et al. High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation. Mol Ther. 2006;13(1):151–159.
  45. Yoon SH, Lee JM, Cho HI, et al. Adoptive immunotherapy using human peripheral blood lymphocytes transferred with RNA encoding Her-2/neu-specific chimeric immune receptor in ovarian cancer xenograft model. Cancer Gene Ther. 2009;16(6):489–497.
  46. Beatty GL, Haas AR, Maus MV, et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res. 2014;2 (2):112–120.
  47. Rowley J, Monie A, Hung C-F, et al. Expression of IL-15RA or an IL-15/IL-15RA fusion on CD8+ T cells modifies adoptively trans- ferred T-cell function in cis. Eur J Immunol. 2009;39(2):491–506.
  48. Rodgers DT, Mazagova M, Hampton EN, et al. Switch-mediated activation and retargeting of CAR-T cells for B-cell malignancies. Proc Natl Acad Sci U S A. 2016;113(4):E459–E468.
  49. GoCAR-T. [cited 2020 Feb 12]. Available from: https://www.fiercebiotech.com/biotech/bellicum-reports-first-data-controllable-car-t
  50. Duong MT, Collinson-Pautz MR, Morschl E, et al. Two-dimensional regulation of CAR-T cell therapy with orthogonal switches. Mol Ther Oncolytics. 2019;12:124–137.
  51. Morsut L, Roybal KT, Xiong X, et al. Engineering customized cell sensing and response behaviors using synthetic notch receptors. Cell. 2016;164(4):780–791.
  52. Giavridis T, van der Stegen SJC, Eyquem J, et al. CAR T cell-induced cytokine release syndrome is mediated by macrophages and aba- ted by IL-1 blockade. Nat Med. 2018;24(6):731–738.
  53. Park JH, Riviere I, Wang X, et al. Impact of disease burden on long-term outcome of 19-28z CAR modified T cells in adult patients with relapsed B-ALL. J Clin Oncol. 2016;34(15_suppl):7003.
  54. Park JH, Rivière I, Wang X, et al. Impact of the conditioning chemotherapy on outcomes in adoptive T cell therapy: results from a phase I clinical trial of autologous CD19-targeted T cells for patients with relapsed CLL. Blood. 2012;120(21):1797.
  55. Frey NV, Shaw PA, Hexner EO, et al. Optimizing chimeric antigen receptor (CAR) T cell therapy for adult patients with relapsed or refractory (r/r) acute lymphoblastic leukemia (ALL). J Clin Oncol. 2016;34(15_suppl):7002.