1,769
18
Essay, 8 pages (1900 words)

Editorial: car t-cell therapies in hematologic tumors

Editorial on the Research Topic

Introduction

Chimeric antigen receptors (CARs) are genetically constructed hybrid receptors that consist of a single-chain variable fragment (scFv) of a monoclonal antibody as the antigen-binding extracellular domain, an intracellular CD3ζ chain as the T cell receptor (TCR) signaling domain, and an additional co-signaling domain, such as CD28 or 4-1BB, to deliver co-stimulation ( 1 , 2 ). The cytotoxic activity of CAR T cells is determined by antigen-binding to the scFv, which leads to phosphorylation of CD3ζ and additional signaling cascades by co-stimulating domains ( 3 ). This mechanism recapitulates intracellular signaling following T-cell activation through the TCR complex, although, in an MHC-independent fashion.

Adoptive T-cell therapy was first clinically employed in 1988 by Rosenberg et al. using ex vivo expanded tumor-infiltrating T-cells in melanoma patients ( 4 ). At around the same time, the adoptive transfer of CAR-modified T cells was first proposed as an anti-tumor approach ( 5 , 6 ). Early clinical trials of CAR T cells showed them to be a promising and safe approach, however, therapeutic efficacy was disappointing, likely due to the inability of CAR-modified T cells to expand and persist in vivo ( 7 9 ). The introduction of co-stimulatory molecules into the signaling domain of CARs greatly increased the potency and persistence of CAR T cells in preclinical studies and their subsequent evaluation in clinical settings demonstrated impressive benefits ( 10 12 ). Most successful clinical results have been observed with CD19-directed CAR T cells in B-cell lymphoproliferative disorders ( 13 , 14 ), which led to the recent FDA and EMA approval of two CAR T-cell therapy products for the treatment of non-Hodgkin lymphomas (NHL) and/or acute lymphoblastic leukemia (ALL). However, in other hematological malignancies, several challenges still need to be overcome for the successful application of CAR T-cell therapies, including identifying target antigens and reversing repressive tumor microenvironments that hamper CAR T-cell function ( 15 , 16 ).

This collection is comprised of a series of reviews providing a comprehensive overview of the current roles of CAR T-cell therapies in several hematological malignancies while also highlighting challenges for toxicity management and future development.

review the role of CAR T-cell immunotherapy in ALL. They provide an overview of the promising results obtained in clinical trials that led to the approval of anti-CD19 CAR T cells for the treatment of relapsed/refractory pediatric patients with ALL. The authors highlight safety concerns as well as potential reasons that preclude durable responses in some patients as major obstacles to overcome the widespread use of this treatment. They also debate on the role of allogeneic hematopoietic stem cell transplantation as consolidation after CAR T-cell therapy, and present strategies currently under investigation for limiting resistance mechanisms, reducing systemic toxicities, and mitigating obstacles to manufacturing processes.

In their article, provide an overview of CAR T-cell therapy in the setting of B-cell NHL and chronic lymphocytic leukemia (CLL). They present results from trials investigating the efficacy and safety of two currently approved second-generation anti-CD19 CAR T-cell products (i. e., axicabtagene ciloleucel and tisagenlecleucel) and a third product (lisocabtagene maraleucel), which is expected to enter the market in 2020. The authors also provide a useful overview of real-world data, including results from special patient populations considered ineligible for pivotal clinical trials (e. g., patients with CNS disease and elderly subjects). Lastly, the review describes novel CAR T-cell-based strategies that are currently under evaluation and which aim to improve efficacy and reduce toxicity in the context of mature B-cell lymphoproliferative disorders.

, review the advances made with CAR T-cell therapy for multiple myeloma. They report results obtained with BCMA-targeted CAR T cells in heavily pretreated patients and outline the potential advantages and disadvantages of new candidate antigens, which are currently under investigation. The authors also provide a thorough description of novel strategies to improve persistence, potency, and trafficking of CAR T cells, such as the refinement of the CAR construct, the use of defined T-cell subpopulations, alternative sources for the production of CAR T cells, the combination approaches with immune modulatory agents, and the use of multi-antigen specific CAR T-cell products.

In their article, provide a comprehensive overview of the current state of CAR T-cell therapy in acute myeloid leukemia (AML). They highlight barriers that limit the full therapeutic potential of this treatment strategy, in particular, the absence of a truly leukemia-specific cell-surface antigen. They also discuss other limitations of CAR T-cell therapy in AML, such as challenges in the manufacturing process due to the presence of leukemic blasts that negatively affect T-cell proliferation and the immunosuppressive mechanisms that diminish CAR T-cell efficacy. The authors also describe potential strategies to overcome such limitations to produce CAR T cells with improved efficacy and tolerability.

CAR T-cell therapies are associated with a unique spectrum of toxicities that are not typically seen with traditional anticancer therapies ( 17 ). in their review focus their attention on these clinical aspects particularly highlighting the role of advanced practice providers (APPs) as members of multidisciplinary care teams in the management of serious adverse events related to these adoptive cell therapies. The article also details approaches for the recognition and grading of the most common CAR T-cell therapy-related toxicities and the available decision support tools for APPs.

In summary, this collection of review articles provides a timely update on the progress made in the field of CAR T-cell therapies for hematologic malignancies, both in terms of the optimal strategies for the management of patients as well as novel products in the development to improve safety and efficacy. Despite the many challenges encountered in moving the field forward, great success has been observed in the last 20 years, leading to the commercial approval of CAR T cells for the treatment of B-cell lymphoproliferative disorders. Many investigations are ongoing to gain a better understanding of the biological mechanisms of toxicity and resistance. These studies on CAR T-cell biology will help develop next-generation strategies to further enhance the efficacy and reduce the toxicity of CAR-transduced cellular therapies, and to extend their applications to other hematological malignancies and solid tumors.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Conflict of Interest

MC received Honoraria from Janssen, Gilead, Abbvie, Shire, and research support from Janssen and Karyopharm Therapeutics. BB received Honoraria from Gilead, Pfizer, Thermofisher, Jazz Pharma, Novartis, Janssen, and research support from Janssen. SN served as a Consultant or Advisory Board member for Kite, a Gilead Company, Merck, Bristol-Myers Squibb, Novartis, Celgene, Pfizer, Allogene Therapeutics, Cell Medica/Kuur, Incyte, Precision Biosciences, Legend Biotech, Adicet Bio, Calibr, and Unum Therapeutics; received research support from Kite, a Gilead Company, Bristol-Myers Squibb, Merck, Poseida, Cellectis, Celgene, Karus Therapeutics, Unum Therapeutics, Allogene Therapeutics, Precision Biosciences, and Acerta; received royalties from Takeda Pharmaceuticals, and has intellectual property related to cell therapy.

Acknowledgments

This was work was supported by an MD Anderson Cancer Center Support Grant P30 CA016672 from the National Institutes of Health and the MD Anderson Cancer Center B-cell Lymphoma Moonshot (SSN). MC would like to express her gratitude for the support given by Fondazione Cassa di Risparmio di Torino (CRT) while contributing to this collection of articles.

References

1. June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science . (2018) 359: 1361–5. doi: 10. 1126/science. aar6711

||

2. Sadelain M, Rivière I, Riddell S. Therapeutic T cell engineering. Nature . (2017) 545: 423–31. doi: 10. 1038/nature22395

||

3. Strati P, Neelapu SS. Chimeric Antigen Receptor-Engineered T cell therapy in lymphoma. Curr Oncol Rep . (2019) 21: 38. doi: 10. 1007/s11912-019-0789-z

||

4. Rosenberg SA, Packard BS, Aebersold PM, Solomon D, Topalian SL, Toy ST, et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med . (1988) 319: 1676–80. doi: 10. 1056/NEJM198812223192527

||

5. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci USA . (1989) 86: 10024–8. doi: 10. 1073/pnas. 86. 24. 10024

||

6. Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, et al. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. Biochem Biophys Res Commun . (1987) 149: 960–8. doi: 10. 1016/0006-291X(87)90502-X

||

7. Park JR, Digiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, et al. Adoptive transfer of chimeric antigen receptor re-directed cytolytic T lymphocyte clones in patients with neuroblastoma. Mol Ther . (2007) 15: 825–33. doi: 10. 1038/sj. mt. 6300104

||

8. Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, et al. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20-specific T cells. Blood . (2008) 112: 2261–71. doi: 10. 1182/blood-2007-12-128843

||

9. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, et al. A phase I study on adoptive immunotherapy using gene-modified T cells for ovarian cancer. Clin Cancer Res . (2006) 12(20 Pt 1): 6106–15. doi: 10. 1158/1078-0432. CCR-06-1183

||

10. Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, Sadelain M. Antigen-dependent CD28 signaling selectively enhances survival and proliferation in genetically modified activated human primary T lymphocytes. J Exp Med . (1998) 188: 619–26. doi: 10. 1084/jem. 188. 4. 619

||

11. Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M. Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor. Nat Biotechnol . (2002) 20: 70–5. doi: 10. 1038/nbt0102-70

||

12. Imai C, Mihara K, Andreansky M, Nicholson IC, Pui CH, Geiger TL, Campana D. Chimeric receptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblastic leukemia. Leukemia . (2004) 18: 676–84. doi: 10. 1038/sj. leu. 2403302

||

13. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med . (2017) 377: 2531–44. doi: 10. 1056/NEJMoa1707447

||

14. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med . (2018) 378: 439–48. doi: 10. 1056/NEJMoa1709866

||

15. Coscia M, Vitale C, Cerrano M, Maffini E, Giaccone L, Boccadoro M, et al. Adoptive immunotherapy with CAR modified T cells in cancer: current landscape and future perspectives. Front Biosci . (2019) 24: 1284–315. doi: 10. 2741/4780

||

16. Cerrano M, Ruella M, Perales MA, Vitale C, Faraci DG, Giaccone L, et al. The advent of CAR T-cell therapy for lymphoproliferative neoplasms: integrating research into clinical practice. Front Immunol . (2020) 11: 888. doi: 10. 3389/fimmu. 2020. 00888

||

17. Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, et al. Chimeric antigen receptor T-cell therapy – assessment and management of toxicities. Nat Rev Clin Oncol . (2018) 15: 47–62. doi: 10. 1038/nrclinonc. 2017. 148

||

Thank's for Your Vote!
Editorial: car t-cell therapies in hematologic tumors. Page 1
Editorial: car t-cell therapies in hematologic tumors. Page 2
Editorial: car t-cell therapies in hematologic tumors. Page 3
Editorial: car t-cell therapies in hematologic tumors. Page 4
Editorial: car t-cell therapies in hematologic tumors. Page 5
Editorial: car t-cell therapies in hematologic tumors. Page 6
Editorial: car t-cell therapies in hematologic tumors. Page 7
Editorial: car t-cell therapies in hematologic tumors. Page 8
Editorial: car t-cell therapies in hematologic tumors. Page 9

This work, titled "Editorial: car t-cell therapies in hematologic tumors" was written and willingly shared by a fellow student. This sample can be utilized as a research and reference resource to aid in the writing of your own work. Any use of the work that does not include an appropriate citation is banned.

If you are the owner of this work and don’t want it to be published on AssignBuster, request its removal.

Request Removal
Cite this Essay

References

AssignBuster. (2022) 'Editorial: car t-cell therapies in hematologic tumors'. 31 October.

Reference

AssignBuster. (2022, October 31). Editorial: car t-cell therapies in hematologic tumors. Retrieved from https://assignbuster.com/editorial-car-t-cell-therapies-in-hematologic-tumors/

References

AssignBuster. 2022. "Editorial: car t-cell therapies in hematologic tumors." October 31, 2022. https://assignbuster.com/editorial-car-t-cell-therapies-in-hematologic-tumors/.

1. AssignBuster. "Editorial: car t-cell therapies in hematologic tumors." October 31, 2022. https://assignbuster.com/editorial-car-t-cell-therapies-in-hematologic-tumors/.


Bibliography


AssignBuster. "Editorial: car t-cell therapies in hematologic tumors." October 31, 2022. https://assignbuster.com/editorial-car-t-cell-therapies-in-hematologic-tumors/.

Work Cited

"Editorial: car t-cell therapies in hematologic tumors." AssignBuster, 31 Oct. 2022, assignbuster.com/editorial-car-t-cell-therapies-in-hematologic-tumors/.

Get in Touch

Please, let us know if you have any ideas on improving Editorial: car t-cell therapies in hematologic tumors, or our service. We will be happy to hear what you think: [email protected]