Open access peer-reviewed chapter - ONLINE FIRST

CAR-T Cells: A Breakthrough in Cancer Treatment

Written By

Ankit Sriwastava, Shubha Gupta, Anand Kumar, Sundaram Gupta and Dharmendra Kumar

Submitted: 03 March 2024 Reviewed: 15 March 2024 Published: 25 June 2024

DOI: 10.5772/intechopen.1005110

Biology of T Cells in Health and Disease IntechOpen
Biology of T Cells in Health and Disease Edited by Hilal Arnouk

From the Edited Volume

Biology of T Cells in Health and Disease [Working Title]

Dr. Hilal Arnouk

Chapter metrics overview

12 Chapter Downloads

View Full Metrics

Abstract

Cancer is a significant health problem that demands ongoing innovation in treatment approaches. This chapter examines the evolution of cancer therapies, highlighting the limitations of conventional methods and the need for precise and effective solutions. Chimeric Antigen Receptor T cell therapy (CAR-T) emerges as a promising solution. The chapter delves into the fundamentals of CAR-T cell therapy, explaining the process of engineering these cells and their mechanism of action. It also discusses the clinical applications of CAR-T cell therapy in approved indications for hematologic malignancies. CAR-T cell therapy has expanded its scope to solid tumors and is exploring futuristic possibilities, such as combination therapies. Although there are challenges, ongoing research focuses on enhancing accessibility. The collaborative and interdisciplinary nature of cancer treatments is emphasized.

Keywords

  • CAR-T
  • T-cells
  • B-ALL
  • DLBCL
  • Hematologic malignancies

1. Introduction

Cancer persists as a substantial global health challenge, demanding ongoing innovation in therapeutic strategies. The contemporary framework of cancer treatment reflects a collaborative, multidisciplinary endeavor to confront the intricacies of the disease. While conventional methods like chemotherapy and radiation exhibit partial effectiveness, their utility is hampered by substantial limitations encompassing issues of both efficacy and adverse effects. As the complexities of cancer become increasingly apparent, the imperative for continual advancements in treatment approaches underscores the pressing need for more targeted and refined solutions to enhance overall efficacy and mitigate the challenges associated with traditional modalities [1]. The pressing need for targeted and effective cancer treatments has propelled the investigation of innovative approaches, with CAR-T cell therapy standing out as a promising frontier in this quest. This chapter is dedicated to unraveling the evolutionary trajectory of cancer treatment, elucidating the limitations inherent in conventional methods. By doing so, it sets the stage for a comprehensive exploration of the revolutionary CAR-T cell therapy—a cutting-edge and transformative modality that holds immense potential in overcoming the challenges posed by traditional treatments. Through this exploration, the chapter aims to provide a nuanced understanding of the historical context and pave the way for a deeper appreciation of the advancements in CAR-T cell therapy within the broader landscape of cancer treatment [2]. The evolution of CAR-T cell therapy is intimately entwined with the historical progression of cancer therapies, particularly within the expansive domain of immunotherapy. Immunotherapy, distinguished by its emphasis on leveraging the body’s own immune system to combat cancer, has traversed a transformative journey marked by pivotal milestones and groundbreaking discoveries. These key advancements have not only reshaped the landscape of oncology but have also laid the foundation for the development of novel and targeted therapies such as CAR-T cell therapy. In tracing this historical trajectory, we gain a profound appreciation for the interconnectedness of these therapeutic approaches and the profound impact they collectively wield in advancing the frontiers of cancer treatment [3]. The evolution of cancer therapies leading up to CAR-T cell therapy represents a series of scientific and clinical breakthroughs. To appreciate the significance of CAR-T cell therapy, it is essential to trace the roots of immunotherapy, acknowledging the key moments and discoveries that paved the way for this revolutionary treatment modality.

Advertisement

2. Understanding CAR-T cell therapy

2.1 What are CAR-T cells?

CAR-T cells, or Chimeric Antigen Receptor T cells, represent a cutting-edge form of immunotherapy designed to harness the body’s immune system to combat cancer. At the heart of CAR-T therapy is the Chimeric Antigen Receptor—a synthetic receptor that combines the specificity of an antibody with the potent cytotoxicity of T cells. This receptor is engineered to recognize and bind to specific antigens expressed on the surface of cancer cells [4]. The Chimeric Antigen Receptor is typically composed of three main components: an extracellular domain, a transmembrane domain, and an intracellular signaling domain. The extracellular domain, derived from an antibody, confers the CAR-T cell with the ability to recognize a particular antigen in cancer cells. The transmembrane domain anchors the receptor in the cell membrane, while the intracellular signaling domain triggers the activation of the T cell upon antigen binding [3].

2.1.1 How CAR-T cells are engineered

CAR-T cells are engineered through a complex process that involves isolating T cells from a patient’s blood, modifying them ex vivo, and then reinfusing them into the patient. This process begins by extracting T cells from the patient, typically through apheresis [5]. Once isolated, these T cells are genetically modified to express the Chimeric Antigen Receptor using viral vectors, often derived from lentiviruses or retroviruses [6]. The modified CAR-T cells are then expanded in culture to create a robust population. This personalized army of CAR-T cells, now armed with the synthetic receptor, is infused back into the patient. Once in the body, these engineered cells seek out and selectively destroy cancer cells expressing the targeted antigen (Figure 1).

Figure 1.

CAR-T cells.

2.2 Mechanism of action

Step-by-step process of CAR-T cell therapy:

Extraction of T cells: the process begins with the extraction of T cells from the patient through apheresis, a technique that separates blood components.

Genetic modification: the isolated T cells undergo genetic modification ex vivo. Using viral vectors, typically derived from lentiviruses or retroviruses, the Chimeric Antigen Receptor (CAR) is introduced into the T cells. This step equips the T cells with the ability to recognize and bind to specific antigens on the surface of cancer cells.

Cell expansion: the genetically modified T cells are cultured and expanded in vitro to create a substantial population of CAR-T cells. This ensures a sufficient quantity of engineered cells for therapeutic effectiveness.

Infusion into the patient: the expanded CAR-T cell population is then infused back into the patient. This infusion marks the introduction of the modified T cells, now armed with the CAR, into the patient’s bloodstream.

Homing to tumor sites: once in the body, CAR-T cells navigate the bloodstream and homing mechanisms to reach tumor sites. The CAR enables these T cells to specifically recognize and bind to cancer cells expressing the targeted antigen.

Activation and cytotoxicity: upon binding to cancer cells, the CAR-T cells become activated. This activation triggers the release of cytotoxic substances and the initiation of immune responses that lead to the destruction of the cancer cells.

Persistence and memory: some CAR-T cells persist in the body, forming a “memory” population. This persistence enhances the ability of the immune system to mount prolonged responses against cancer cells, offering potential long-term protection.

2.3 Interaction between CAR-T cells and cancer cells

The interaction between CAR-T cells and cancer cells is highly specific. The Chimeric Antigen Receptor on the surface of CAR-T cells acts as a molecular sensor, recognizing and binding to specific antigens present on the cancer cell surface. This interaction initiates a cascade of signaling events within the CAR-T cell, leading to its activation.

The engineered CAR-T cells unleash potent cytotoxic mechanisms, including the release of perforin and granzymes, inducing apoptosis (cell death) in the cancer cells. Additionally, the activated CAR-T cells can stimulate other components of the immune system, such as macrophages and natural killer cells, further amplifying the anti-cancer immune response (Figure 2) [7].

Figure 2.

Diagrammatic representation of CAR-T cell interaction with cancer cells lead to cell death.

Advertisement

3. Development and advancements

3.1 Early research and clinical trials

The journey of CAR-T cell therapy commenced with pioneering studies and clinical trials that laid the foundational groundwork. In the early stages, researchers focused on understanding the feasibility and safety of genetically modifying T cells to express Chimeric Antigen Receptors (CARs). Notable studies, such as those by Sadelain and Brentjens, marked crucial milestones in demonstrating the concept’s viability and its potential as a transformative cancer treatment [8]. The early phases of CAR-T cell therapy research faced significant challenges, including issues related to the persistence and functionality of engineered T cells, as well as concerns about off-target effects. These challenges prompted researchers to refine the CAR design, optimize the genetic modification process, and enhance the overall therapeutic efficacy. Breakthrough moments emerged as solutions were devised, leading to improved CAR-T cell technologies and addressing initial hurdles [9].

The landmark clinical trial by Maude et al. in 2014 demonstrated unprecedented success in treating pediatric patients with acute lymphoblastic leukemia (ALL), showcasing the therapy’s potential for achieving sustained remissions and transforming the landscape of cancer treatment [10].

3.2 Key developments in CAR-T cell therapy

Over the years, CAR-T cell therapy has undergone significant technological advancements, revolutionizing cancer treatment. One major breakthrough has been the refinement of CAR design. Researchers have developed next-generation CARs with enhanced signaling domains, optimized co-stimulatory molecules, and improved antigen recognition capabilities. These technological advancements have contributed to the development of CAR-T cells with increased potency and persistence in the body, leading to improved treatment outcomes [11].

Another critical development is the evolution of manufacturing processes. Streamlining and optimizing the production of CAR-T cells have become pivotal for scalability and widespread clinical application. Innovative manufacturing technologies, such as automated systems and closed bioreactors, have been introduced to ensure consistent and high-quality production of CAR-T cell therapies [12]. Enhancing the safety profile of CAR-T cell therapy has been a key focus of research and development. Initial concerns regarding cytokine release syndrome (CRS) and neurotoxicity led to the development of strategies to mitigate these adverse events. The introduction of cytokine-blocking agents and the refinement of dosing regimens have contributed to a more favorable safety profile, making CAR-T cell therapy more manageable for patients [13].

Moreover, advancements in target antigen selection and validation have played a crucial role in improving the therapy’s efficacy. Identifying antigens that are highly specific to cancer cells while sparing healthy tissues has enhanced the precision of CAR-T cell targeting, minimizing off-target effects and maximizing therapeutic efficacy [14].

Advertisement

4. Clinical applications

4.1 Approved indications

CAR-T cell therapy has gained approval for treating specific types of cancers, marking a standard shift in oncology. Notably, the U.S. Food and Drug Administration (FDA) has approved CAR-T therapies for certain hematologic malignancies. Among these, CD19-targeted CAR-T cells have shown remarkable success in treating B-cell acute lymphoblastic leukemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL). The approval of these therapies reflects the transformative impact of CAR-T cell therapy in providing effective treatment options for patients with refractory or relapsed hematologic cancers [15, 16]. Success stories in CAR-T cell therapy are exemplified by cases of patients achieving sustained remissions and, in some instances, achieving complete responses. Notable outcomes include patients with B-ALL experiencing long-term remission following CAR-T treatment. These success stories underscore the therapeutic potential of CAR-T cell therapy, especially in cases where traditional treatments have failed [17].

4.2 Ongoing research and potential applications

Ongoing research is expanding the scope of CAR-T cell therapy beyond hematologic malignancies. Investigations are underway to explore the potential application of CAR-T cells in solid tumors, such as glioblastoma and pancreatic cancer. Researchers are adapting CAR designs and exploring new target antigens to overcome the challenges posed by the tumor microenvironment in solid tumors. This expansion into diverse cancer types represents a promising avenue for broadening the impact of CAR-T cell therapy [18, 19]. The field of CAR-T cell therapy continues to evolve with emerging trends and futuristic possibilities. These include the development of “off-the-shelf” CAR-T cells, overcoming the current limitation of personalized manufacturing for each patient. Additionally, researchers are investigating combination therapies, integrating CAR-T cells with other treatment modalities to enhance overall anti-cancer efficacy. The potential for CAR-T cells to serve as a platform for delivering therapeutic payloads or cytokines is another exciting avenue under exploration [20, 21].

Advertisement

5. Challenges and considerations

5.1 Side effects and toxicities

CAR-T cell therapy, while revolutionary, is associated with specific side effects and toxicities. One of the most common adverse events is Cytokine Release Syndrome (CRS), a systemic inflammatory response triggered by the rapid activation and proliferation of CAR-T cells. CRS can range from mild flu-like symptoms to severe manifestations, potentially impacting multiple organs. Neurotoxicity is another concern, characterized by cognitive disturbances and, in extreme cases, seizures. Understanding and managing these side effects are crucial for optimizing patient outcomes [13, 14]. To mitigate side effects, clinicians employ various strategies. Tocilizumab, an interleukin-6 receptor antagonist, is frequently used to counteract CRS. Corticosteroids may be administered to address severe immune reactions. Monitoring patients closely and intervening promptly are essential aspects of complication management. Ongoing research aims to develop predictive biomarkers that can identify individuals at higher risk of severe complications, enabling proactive interventions to minimize the impact of side effects [22, 23].

5.2 Cost and accessibility

CAR-T cell therapy poses economic challenges due to its intricate manufacturing process, personalized nature, and the costs associated with clinical management of side effects. Infrastructure requirements, including specialized facilities for cell processing, contribute to the overall expense. The initial costs of CAR-T therapies can be substantial, impacting healthcare systems, insurers, and patients alike. Economic implications extend beyond treatment costs to considerations of long-term healthcare expenditures associated with managing potential late effects and complications [24, 25].

Recognizing the need for broader accessibility, ongoing efforts are directed toward reducing the economic barriers to CAR-T cell therapy. This involves research into more cost-effective manufacturing methods, increased collaboration between healthcare providers and manufacturers, and negotiations to establish reimbursement models that ensure affordability for both healthcare systems and patients. Furthermore, exploring strategies for “off-the-shelf” CAR-T cells could potentially streamline production processes, enhancing accessibility by reducing the need for personalized manufacturing [26, 27].

Advertisement

6. Future directions and outlook

6.1 Next-generation CAR-T cell therapies

The field of CAR-T cell therapy is rapidly advancing, with ongoing research focused on developing next-generation technologies to enhance its efficacy and safety. Researchers are exploring novel CAR designs with advanced signaling domains, optimized co-stimulatory molecules, and improved antigen recognition capabilities. Additionally, advancements in gene-editing technologies, such as CRISPR-Cas9, are being harnessed to precisely engineer CAR-T cells, allowing for more controlled and predictable modifications. Ongoing efforts aim to address current limitations, such as antigen escape and persistence of CAR-T cells, with a focus on creating more potent and durable therapies [28, 29].

The future of CAR-T cell therapy holds promise for several innovations. These include the development of “universal” or “off-the-shelf” CAR-T cells, allowing for standardized, readily available treatments without the need for personalized manufacturing. Enhanced safety features, such as suicide switches, are under investigation to provide a fail-safe mechanism for controlling CAR-T cell activity and minimizing adverse events. Moreover, advancements in delivery systems and targeting strategies are being explored to improve the precision and efficiency of CAR-T cell therapy across various cancer types [30, 31].

6.2 Integration with other therapies

Combination therapies involving CAR-T cell therapy and other treatment modalities are a burgeoning area of research. Researchers are exploring synergistic effects by combining CAR-T cells with traditional therapies like chemotherapy or radiation. Additionally, combining CAR-T cell therapy with immune checkpoint inhibitors aims to enhance the overall immune response against cancer cells. These synergistic approaches seek to capitalize on the strengths of different treatments, potentially improving response rates and extending the benefits of CAR-T cell therapy to a broader range of patients [32, 33]. Collaboration is becoming increasingly crucial in advancing cancer treatment. Interdisciplinary collaborations among immunologists, oncologists, geneticists, and other experts are fostering a comprehensive understanding of cancer biology and immune response dynamics. This collaborative approach extends beyond individual therapies, emphasizing the integration of diverse treatment modalities into cohesive and personalized cancer care strategies. Such collaborative efforts are essential for optimizing treatment outcomes and navigating the complexities of individual patient responses [34, 35].

Advertisement

7. Conclusion

In conclusion, CAR-T cell therapy stands as a revolutionary and transformative approach in cancer treatment. The significance of CAR-T cell therapy lies in its ability to harness the body’s own immune system to target and eliminate cancer cells with precision. This therapy has demonstrated remarkable success, particularly in the treatment of certain hematologic malignancies, where conventional treatments have shown limitations. The incorporation of Chimeric Antigen Receptors into T cells represents a groundbreaking strategy that has paved the way for personalized and highly effective cancer therapies.

As of now, CAR-T cell therapy has received approvals for specific indications, showcasing its clinical success and the paradigm shift it has brought to the field of oncology. The therapy’s future potential is vast, with ongoing research focusing on refining current technologies, developing next-generation CAR-T cells, and exploring innovative combination therapies. The continuous evolution of CAR-T cell therapy holds promise for extending its application to a broader spectrum of cancer types and improving its accessibility and safety.

Advertisement

Acknowledgments

The author acknowledges the usage of Chat GPT, Quilbolt,(AI tool used) for language polishing of the manuscript.

References

  1. 1. Debela DT, Muzazu SG, Heraro KD, et al. New approaches and procedures for cancer treatment: Current perspectives. SAGE Open Medicine. 2021;9:20503121211034366. DOI: 10.1177/20503121211034366
  2. 2. Mitra A, Barua A, Huang L, Ganguly S, Feng Q , He B. From bench to bedside: The history and progress of CAR T cell therapy. Frontiers in Immunology. 2023;14:1188049. DOI: 10.3389/fimmu.2023.1188049
  3. 3. Gomes-Silva D, Ramos CA. Cancer immunotherapy using CAR-T cells: From the research bench to the assembly line. Biotechnology Journal. 2018;13(2):1-16. DOI: 10.1002/biot.201700097
  4. 4. Pan K, Farrukh H, Chittepu VCSR, Xu H, Pan CX, Zhu Z. CAR race to cancer immunotherapy: From CAR T, CAR NK to CAR macrophage therapy. Journal of Experimental & Clinical Cancer Research. 2022;41(1):119. DOI: 10.1186/s13046-022-02327-z
  5. 5. Zhang C, Liu J, Zhong JF, Zhang X. Engineering CAR-T cells. Biomarker Research. 2017;5:22. DOI: 10.1186/s40364-017-0102-y
  6. 6. Warnock JN, Daigre C, Al-Rubeai M. Introduction to viral vectors. Methods in Molecular Biology. 2011;737:1-25. DOI: 10.1007/978-1-61779-095-9_1
  7. 7. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia [published correction appears in N Engl J Med. 2016 Mar 10;374(10):998]. The New England Journal of Medicine. 2014;371(16):1507-1517. DOI: 10.1056/NEJMoa1407222
  8. 8. Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancer Discovery. 2013;3(4):388-398. DOI: 10.1158/2159-8290.CD-12-0548
  9. 9. Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. The New England Journal of Medicine. 2013;368(16):1509-1518. DOI: 10.1056/NEJMoa1215134
  10. 10. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. The New England Journal of Medicine. 2014;371(16):1507-1517. DOI: 10.1056/NEJMoa1407222
  11. 11. Park JH, Geyer MB, Brentjens RJ. CD19-targeted CAR T-cell therapeutics for hematologic malignancies: Interpreting clinical outcomes to date. Blood. 2016;127(26):3312-3320. DOI: 10.1182/blood-2016-03-643544
  12. 12. Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR T cell therapy. Molecular Therapy—Methods & Clinical Development. 2017;15(4):92-101. DOI: 10.1016/j.omtm.2016.12.006
  13. 13. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy—Assessment and management of toxicities. Nature Reviews. Clinical Oncology. 2018;15(1):47-62. DOI: 10.1038/nrclinonc.2017.148
  14. 14. Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discovery. 2016;6(6):664-679. DOI: 10.1158/2159-8290.CD-16-0040.*
  15. 15. Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. The New England Journal of Medicine. 2018;378(5):439-448. DOI: 10.1056/NEJMoa1709866
  16. 16. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. The New England Journal of Medicine. 2019;380(1):45-56. DOI: 10.1056/NEJMoa1804980
  17. 17. Park JH, Rivière I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. The New England Journal of Medicine. 2018;378(5):449-459. DOI: 10.1056/NEJMoa1709919
  18. 18. Brown CE, Mackall CL. CAR T cell therapy: Inroads to response and resistance. Nature Reviews. Immunology. 2019;19(2):73-74. DOI: 10.1038/s41577-019-0123-7
  19. 19. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nature Reviews. Clinical Oncology. 2020;17(2):147-167. DOI: 10.1038/s41571-019-0278-6
  20. 20. Fraietta JA, Lacey SF, Orlando EJ, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nature Medicine. 2018;24(5):563-571. DOI: 10.1038/s41591-018-0010-1
  21. 21. Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Molecular Therapy—Oncolytics. 2016;31(3):16011. DOI: 10.1038/mto.2016.11.
  22. 22. Lee DW, Santomasso BD, Locke FL, et al. ASTCT consensus grading for cytokine release syndrome and neurologic toxicity associated with immune effector cells. Biology of Blood and Marrow Transplantation. 2019;25(4):625-638. DOI: 10.1016/j.bbmt.2018.12.758
  23. 23. Brudno JN, Kochenderfer JN. Recent advances in CAR T-cell toxicity: Mechanisms, manifestations and management. Blood Reviews. 2019;34:45-55. DOI: 10.1016/j.blre.2018.10.002
  24. 24. Makita S, Yoshimura K, Tobinai K. Economic evaluation of chimeric antigen receptor T-cell therapy. Current Treatment Options in Oncology. 2020;21(12):102. DOI: 10.1007/s11864-020-00784-2
  25. 25. Pearson S, Cordon-Cardo C, Cohen D, et al. Institutional considerations in leveraging CAR T-cell therapy for patients with cancer during the COVID-19 pandemic. JAMA Oncology. 2020;6(8):1131-1134. DOI: 10.1001/jamaoncol.2020.3051
  26. 26. Puthenveetil G, Musto S, Rodriguez-Molina J, et al. Cost-effectiveness and budget impact of chimeric antigen receptor T-cell therapy in relapsed or refractory large B-cell lymphoma. Value in Health. 2021;24(6):826-834. DOI: 10.1016/j.jval.2020.11.021
  27. 27. Bach PB, Giralt S, Saltz L. FDA approval of Tisagenlecleucel: Promise and complexities of a $475 000 cancer drug. JAMA. 2017;318(23):1861-1862. DOI: 10.1001/jama.2017.17127.*
  28. 28. Majzner RG, Mackall CL. Clinical lessons learned from the first leg of the CAR T cell journey. Nature Medicine. 2019;25(9):1341-1355. DOI: 10.1038/s41591-019-0502-y
  29. 29. June CH, Sadelain M. Chimeric antigen receptor therapy. The New England Journal of Medicine. 2018;379(1):64-73. DOI: 10.1056/NEJMra1706169
  30. 30. Gill S, Maus MV, Porter DL. Chimeric antigen receptor T cell therapy: 25 years in the making. Blood Reviews. 2016;30(3):157-167. DOI: 10.1016/j.blre.2015.10.003
  31. 31. Zhao Z, Condomines M, van der Stegen SJC, et al. Structural design of engineered costimulation determines tumor rejection kinetics and persistence of CAR T cells. Cancer Cell. 2015;28(4):415-428. DOI: 10.1016/j.ccell.2015.09.004
  32. 32. Rafiq S, Yeku OO, Jackson HJ, et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nature Biotechnology. 2018;36(9):847-856. DOI: 10.1038/nbt.4195
  33. 33. Adusumilli PS, Zauderer MG, Rusch VW, et al. Regional delivery of mesothelin-targeted CAR T cells for pleural cancers: Safety and preliminary efficacy in combination with anti-PD-1 agent. Journal of Clinical Oncology. 2019;37(Suppl. 15):2511. DOI: 10.1200/JCO.2019.37.15_suppl.2511
  34. 34. Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168(4):707-723. DOI: 10.1016/j.cell.2017.01.017
  35. 35. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350-1355. DOI: 10.1126/science.aar4060

Written By

Ankit Sriwastava, Shubha Gupta, Anand Kumar, Sundaram Gupta and Dharmendra Kumar

Submitted: 03 March 2024 Reviewed: 15 March 2024 Published: 25 June 2024