101
|
Batra SA, Rathi P, Guo L, Courtney AN, Fleurence J, Balzeau J, Shaik RS, Nguyen TP, Wu MF, Bulsara S, Mamonkin M, Metelitsa LS, Heczey A. Glypican-3-Specific CAR T Cells Coexpressing IL15 and IL21 Have Superior Expansion and Antitumor Activity against Hepatocellular Carcinoma. Cancer Immunol Res 2020; 8:309-320. [PMID: 31953246 PMCID: PMC10765595 DOI: 10.1158/2326-6066.cir-19-0293] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 11/08/2019] [Accepted: 01/10/2020] [Indexed: 01/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth most common cause of cancer-related death in the world, and curative systemic therapies are lacking. Chimeric antigen receptor (CAR)-expressing T cells induce robust antitumor responses in patients with hematologic malignancies but have limited efficacy in patients with solid tumors, including HCC. IL15 and IL21 promote T-cell expansion, survival, and function and can improve the antitumor properties of T cells. We explored whether transgenic expression of IL15 and/or IL21 enhanced glypican-3-CAR (GPC3-CAR) T cells' antitumor properties against HCC. We previously optimized the costimulation in GPC3-CARs and selected a second-generation GPC3-CAR incorporating a 4-1BB costimulatory endodomain (GBBz) for development. Here, we generated constructs encoding IL15, IL21, or both with GBBz (15.GBBz, 21.GBBz, and 21.15.GBBz, respectively) and examined the ability of transduced T cells to kill, produce effector cytokines, and expand in an antigen-dependent manner. We performed gene-expression and phenotypic analyses of GPC3-CAR T cells and CRISPR-Cas9 knockout of the TCF7 gene. Finally, we measured GPC3-CAR T-cell antitumor activity in murine xenograft models of GPC3+ tumors. The increased proliferation of 21.15.GBBz T cells was at least in part dependent on the upregulation and maintenance of TCF-1 (encoded by TCF7) and associated with a higher percentage of stem cell memory and central memory populations after manufacturing. T cells expressing 21.15.GBBz had superior in vitro and in vivo expansion and persistence, and the most robust antitumor activity in vivo These results provided preclinical evidence to support the clinical evaluation of 21.15.GPC3-CAR T cells in patients with HCC.
Collapse
Affiliation(s)
- Sai Arun Batra
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Purva Rathi
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Linjie Guo
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Amy N Courtney
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Julien Fleurence
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Julien Balzeau
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Rahamthulla S Shaik
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Thao P Nguyen
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Meng-Fen Wu
- Dan L Duncan Cancer Center Biostatistics Shared Resource, Baylor College of Medicine, Houston, Texas
| | - Shaun Bulsara
- Dan L Duncan Cancer Center Biostatistics Shared Resource, Baylor College of Medicine, Houston, Texas
| | - Maksim Mamonkin
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Leonid S Metelitsa
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Andras Heczey
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital Liver Tumor Center, Houston, Texas
| |
Collapse
|
102
|
Kintz H, Nylen E, Barber A. Inclusion of Dap10 or 4-1BB costimulation domains in the chPD1 receptor enhances anti-tumor efficacy of T cells in murine models of lymphoma and melanoma. Cell Immunol 2020; 351:104069. [PMID: 32106933 DOI: 10.1016/j.cellimm.2020.104069] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/23/2020] [Accepted: 02/19/2020] [Indexed: 12/30/2022]
Abstract
Chimeric antigen receptors (CAR) utilize costimulatory domains to enhance anti-tumor efficacy. However, it is unclear which costimulatory domain is preferable. Therefore, the intracellular domains of CD28, Dap10, 41BB, GITR, ICOS, or OX40 were compared in a murine chimeric PD1 (chPD1) receptor that targets tumor-associated PD1 ligands. Upon antigen restimulation, T cells expressing chPD1-CD28 receptors had reduced lytic capacity. While most of the chPD1 T cell receptors secreted pro-inflammatory (IFNγ, TNFα, IL-2, GM-CSF, IL-17, and IL-21) and anti-inflammatory cytokines (IL-10), chPD1-Dap10 did not secrete IL-10. Furthermore, chPD1-Dap10 and -41BB receptors induced a memory precursor phenotype, had enhanced persistence in vivo, and superior therapeutic efficacy in murine models of T cell lymphoma and melanoma compared to chPD1-CD28 or chPD1-GITR expressing T cells. Therefore, each costimulatory domain induces differential effects in CAR-expressing T cells and inclusion of Dap10 or 4-1BB costimulatory domains may induce a preferential cytokine profile and differentiation for cancer therapy.
Collapse
Affiliation(s)
- Hailey Kintz
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - Emily Nylen
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA
| | - Amorette Barber
- Department of Biological and Environmental Sciences, Longwood University, Farmville, VA, USA.
| |
Collapse
|
103
|
Long B, Qin L, Zhang B, Li Q, Wang L, Jiang X, Ye H, Zhang G, Yu Z, Jiao Z. CAR T‑cell therapy for gastric cancer: Potential and perspective (Review). Int J Oncol 2020; 56:889-899. [PMID: 32319561 DOI: 10.3892/ijo.2020.4982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/13/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed digestive malignancies and is the third leading cause of cancer‑associated death worldwide. Delayed diagnosis and poor prognosis indicate the urgent need for new therapeutic strategies. The success of chimeric antigen receptor (CAR) T‑cell therapy for chemotherapy‑refractory hematological malignancies has inspired the development of a similar strategy for GC treatment. Although using CAR T‑cells against GC is not without difficulty, results from preclinical studies remain encouraging. The current review summarizes relevant preclinical studies and ongoing clinical trials for the use of CAR T‑cells for GC treatment and investigates possible toxicities, as well as current clinical experiences and emerging approaches. With a deeper understanding of the tumor microenvironment, novel target epitopes and scientific‑technical progress, the potential of CAR T‑cell therapy for GC is anticipated in the near future.
Collapse
Affiliation(s)
- Bo Long
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Long Qin
- The Cuiying Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Boya Zhang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Qiong Li
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Long Wang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Xiangyan Jiang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Huili Ye
- The Cuiying Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Genyuan Zhang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Zeyuan Yu
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Zuoyi Jiao
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
104
|
Pacenta HL, Laetsch TW, John S. CD19 CAR T Cells for the Treatment of Pediatric Pre-B Cell Acute Lymphoblastic Leukemia. Paediatr Drugs 2020; 22:1-11. [PMID: 31749131 DOI: 10.1007/s40272-019-00370-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of cluster of differentiation (CD)-19-targeted chimeric antigen receptor (CAR) T cells for the treatment of pre-B-cell acute lymphoblastic leukemia (B-ALL) is an exciting new advancement in the field of pediatric oncology. Tisagenlecleucel and axicabtagene ciloleucel are the first US FDA-approved CD19-targeted CAR T cells. While various different CD19 CAR T cells are in development, tisagenlecleucel is the only CAR T cell approved for pediatric patients. The multicenter phase II trial that led to the approval of tisagenlecleucel demonstrated excellent responses in individuals with highly refractory disease. Other high-risk groups of patients with B-ALL who experience poor outcomes with standard therapy may also benefit from treatment with tisagenlecleucel. After receiving CAR T cells, patients must be closely monitored for unique toxicities, including cytokine release syndrome, neurotoxicity, and B-cell aplasia. The management of patients with relapsed or refractory disease after administration of CD19 CAR T cells can be challenging, and treatment options vary according to the characteristics of the disease present at relapse. In the many patients who experience a complete response, CAR T cells can lead to a durable remission. This review describes the current design and manufacturing of CAR T cells. Data in the selection and management of pediatric patients are highlighted, as are areas where further studies are needed.
Collapse
Affiliation(s)
- Holly L Pacenta
- Division of Pediatric Hematology-Oncology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9063, USA
- The Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, TX, USA
| | - Theodore W Laetsch
- Division of Pediatric Hematology-Oncology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9063, USA.
- The Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, TX, USA.
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9063, USA.
| | - Samuel John
- Division of Pediatric Hematology-Oncology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390-9063, USA
- The Pauline Allen Gill Center for Cancer and Blood Disorders, Children's Health, Dallas, TX, USA
| |
Collapse
|
105
|
Walsh Z, Yang Y, Kohler ME. Immunobiology of chimeric antigen receptor T cells and novel designs. Immunol Rev 2020; 290:100-113. [PMID: 31355496 DOI: 10.1111/imr.12794] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 01/01/2023]
Abstract
Advances in the development of immunotherapies have offered exciting new options for the treatment of malignant diseases that are refractory to conventional cytotoxic chemotherapies. The adoptive transfer of T cells expressing chimeric antigen receptors (CARs) has demonstrated dramatic results in clinical trials and highlights the promise of novel immune-based approaches to the treatment of cancer. As experience with CAR T cells has expanded with longer follow-up and to a broader range of diseases, new obstacles have been identified which limit the potential lifelong benefits of CAR T cell therapy. These obstacles highlight not only the gaps in knowledge of the optimal clinical application of this "living drug", but also gaps in our understanding of the fundamental biology of CAR T cells themselves. In this review, we discuss the obstacles facing CAR T cell therapy, how these relate to our current understanding of CAR T cell biology and approaches to enhance the clinical efficacy of this therapy.
Collapse
Affiliation(s)
- Zachary Walsh
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yinmeng Yang
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - M Eric Kohler
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.,Division of Blood and Marrow Transplantation and Cellular Therapeutics, Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
106
|
Stern LA, Jonsson VD, Priceman SJ. CAR T Cell Therapy Progress and Challenges for Solid Tumors. Cancer Treat Res 2020; 180:297-326. [PMID: 32215875 DOI: 10.1007/978-3-030-38862-1_11] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The past two decades have marked the beginning of an unprecedented success story for cancer therapy through redirecting antitumor immunity [1]. While the mechanisms that control the initial and ongoing immune responses against tumors remain a strong research focus, the clinical development of technologies that engage the immune system to target and kill cancer cells has become a translational research priority. Early attempts documented in the late 1800s aimed at sparking immunity with cancer vaccines were difficult to interpret but demonstrated an opportunity that more than 100 years later has blossomed into the current field of cancer immunotherapy. Perhaps the most recent and greatest illustration of this is the widespread appreciation that tumors actively shut down antitumor immunity, which has led to the emergence of checkpoint pathway inhibitors that re-invigorate the body's own immune system to target cancer [2, 3]. This class of drugs, with first FDA approvals in 2011, has demonstrated impressive durable clinical responses in several cancer types, including melanoma, lung cancer, Hodgkin's lymphoma, and renal cell carcinoma, with the ongoing investigation in others. The biology and ultimate therapeutic successes of these drugs led to the 2018 Nobel Prize in Physiology or Medicine, awarded to Dr. James Allison and Dr. Tasuku Honjo for their contributions to cancer therapy [4]. In parallel to the emerging science that aided in unleashing the body's own antitumor immunity with checkpoint pathway inhibitors, researchers were also identifying ways to re-engineer antitumor immunity through adoptive cellular immunotherapy approaches. Chimeric antigen receptor (CAR)-based T cell therapy has achieved an early head start in the field, with two recent FDA approvals in 2017 for the treatment of B-cell malignancies [5]. There is an explosion of preclinical and clinical efforts to expand the therapeutic indications for CAR T cell therapies, with a specific focus on improving their clinical utility, particularly for the treatment of solid tumors. In this chapter, we will highlight the recent progress, challenges, and future perspectives surrounding the development of CAR T cell therapies for solid tumors.
Collapse
Affiliation(s)
- Lawrence A Stern
- Department of Hematology and Hematopoietic Cell Transplantation, Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Vanessa D Jonsson
- Department of Hematology and Hematopoietic Cell Transplantation, Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Saul J Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
107
|
Genetically Modified T-Cell Therapy for Osteosarcoma: Into the Roaring 2020s. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:109-131. [PMID: 32483735 DOI: 10.1007/978-3-030-43032-0_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell immunotherapy may offer an approach to improve outcomes for patients with osteosarcoma who fail current therapies. In addition, it has the potential to reduce treatment-related complications for all patients. Generating tumor-specific T cells with conventional antigen-presenting cells ex vivo is time-consuming and often results in T-cell products with a low frequency of tumor-specific T cells. Furthermore, the generated T cells remain sensitive to the immunosuppressive tumor microenvironment. Genetic modification of T cells is one strategy to overcome these limitations. For example, T cells can be genetically modified to render them antigen specific, resistant to inhibitory factors, or increase their ability to home to tumor sites. Most genetic modification strategies have only been evaluated in preclinical models; however, early clinical phase trials are in progress. In this chapter, we will review the current status of gene-modified T-cell therapy with special focus on osteosarcoma, highlighting potential antigenic targets, preclinical and clinical studies, and strategies to improve current T-cell therapy approaches.
Collapse
|
108
|
Marayati R, Quinn CH, Beierle EA. Immunotherapy in Pediatric Solid Tumors-A Systematic Review. Cancers (Basel) 2019; 11:E2022. [PMID: 31847387 PMCID: PMC6966467 DOI: 10.3390/cancers11122022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 12/07/2019] [Accepted: 12/12/2019] [Indexed: 12/27/2022] Open
Abstract
Despite advances in the treatment of many pediatric solid tumors, children with aggressive and high-risk disease continue to have a dismal prognosis. For those presenting with metastatic or recurrent disease, multiple rounds of intensified chemotherapy and radiation are the typical course of action, but more often than not, this fails to control the progression of the disease. Thus, new therapeutics are desperately needed to improve the outcomes for these children. Recent advances in our understanding of both the immune system's biology and its interaction with tumors have led to the development of novel immunotherapeutics as alternative treatment options for these aggressive malignancies. Immunotherapeutic approaches have shown promising results for pediatric solid tumors in early clinical trials, but challenges remain concerning safety and anti-tumor efficacy. In this review, we aim to discuss and summarize the main classes of immunotherapeutics used to treat pediatric solid tumors.
Collapse
Affiliation(s)
| | | | - Elizabeth A. Beierle
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (R.M.); (C.H.Q.)
| |
Collapse
|
109
|
Stock S, Schmitt M, Sellner L. Optimizing Manufacturing Protocols of Chimeric Antigen Receptor T Cells for Improved Anticancer Immunotherapy. Int J Mol Sci 2019; 20:ijms20246223. [PMID: 31835562 PMCID: PMC6940894 DOI: 10.3390/ijms20246223] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/07/2019] [Accepted: 12/08/2019] [Indexed: 01/08/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy can achieve outstanding response rates in heavily pretreated patients with hematological malignancies. However, relapses occur and they limit the efficacy of this promising treatment approach. The cellular composition and immunophenotype of the administered CART cells play a crucial role for therapeutic success. Less differentiated CART cells are associated with improved expansion, long-term in vivo persistence, and prolonged anti-tumor control. Furthermore, the ratio between CD4+ and CD8+ T cells has an effect on the anti-tumor activity of CART cells. The composition of the final cell product is not only influenced by the CART cell construct, but also by the culturing conditions during ex vivo T cell expansion. This includes different T cell activation strategies, cytokine supplementation, and specific pathway inhibition for the differentiation blockade. The optimal production process is not yet defined. In this review, we will discuss the use of different CART cell production strategies and the molecular background for the generation of improved CART cells in detail.
Collapse
Affiliation(s)
- Sophia Stock
- Department of Internal Medicine V, Heidelberg University Hospital, 69120 Heidelberg, Germany; (S.S.); (M.S.)
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, 69120 Heidelberg, Germany; (S.S.); (M.S.)
- National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital, 69120 Heidelberg, Germany; (S.S.); (M.S.)
- National Center for Tumor Diseases (NCT), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- Oncology Business Unit—Medical Affairs, Takeda Pharma Vertrieb GmbH & Co. KG, 10117 Berlin, Germany
- Correspondence:
| |
Collapse
|
110
|
Tang X, Tang Q, Mao Y, Huang X, Jia L, Zhu J, Feng Z. CD137 Co-Stimulation Improves The Antitumor Effect Of LMP1-Specific Chimeric Antigen Receptor T Cells In Vitro And In Vivo. Onco Targets Ther 2019; 12:9341-9350. [PMID: 31807014 PMCID: PMC6847990 DOI: 10.2147/ott.s221040] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/07/2019] [Indexed: 12/18/2022] Open
Abstract
Purpose In previous research, we have found that LMP1-specific chimeric antigen (HELA/CAR) T cells can specifically recognize and kill LMP1-positive NPC cells. However, the tumor-inhibitory effectiveness of HELA/CART cells needs to be enhanced. Methods We created two CARs that contain the T cell receptor-ζ (TCR-ζ) signal transduction domain with the CD28 and CD137 (4-1BB) or CD134 (OX-40) intracellular domains in tandem (HELA/137CAR or HELA/134CAR). Then, the tumor-inhibitory functions of two new CAR-T cells were investigated, both in vitro and in vivo. Results The results showed that, after short-term expansion, primary human T cells were subjected to lentiviral gene transfer, resulting in large numbers of cells with >80% CAR expression. All CART cells were effective in killing SUNE1-LMP1 and C1R-neo cells, while HELA/137CART cells produced greater quantities of IFN-γ and IL-2 than HELA/CART cells. However, the level of IL-2 not INF-γ secreted by HELA/134CART cells was increased under the stimulation of LMP1 antigen. In an LMP1-positive NPC mouse xenograft model, HELA/137CART cells exhibited better antitumor activity and longer survival time in vivo compared with HELA/CAR T cells. Conclusion The findings suggest that CD137 and CD28 is a better costimulatory signaling domain than CD28 only for optimizing tumor-inhibitory roles.
Collapse
Affiliation(s)
- Xiaojun Tang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
| | - Qi Tang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Yuan Mao
- Department of Haematology and Oncology, Geriatric Hospital of Nanjing Medical University, Nanjing, People's Republic of China
| | - Xiaochen Huang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Lizhou Jia
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jin Zhu
- Huadong Medical Institute of Biotechniques, Nanjing, People's Republic of China
| | - Zhenqing Feng
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, People's Republic of China.,Department of Pathology, Nanjing Medical University, Nanjing, People's Republic of China.,Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
111
|
Wu C, Zhang L, Brockman QR, Zhan F, Chen L. Chimeric antigen receptor T cell therapies for multiple myeloma. J Hematol Oncol 2019; 12:120. [PMID: 31752943 PMCID: PMC6873434 DOI: 10.1186/s13045-019-0823-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/07/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematologic malignancy and remains incurable despite the advent of numerous new drugs such as proteasome inhibitors (PIs), immunomodulatory agents (IMiDs), and monoclonal antibodies. There is an unmet need to develop novel therapies for refractory/relapsed MM. In the past few years, chimeric antigen receptor (CAR)-modified T cell therapy for MM has shown promising efficacy in preclinical and clinical studies. Furthermore, the toxicities of CAR-T cell therapy are manageable. This article summarizes recent developments of CAR-T therapy in MM, focusing on promising targets, new technologies, and new research areas. Additionally, a comprehensive overview of antigen selection is presented along with preliminary results and future directions of CAR-T therapy development.
Collapse
Affiliation(s)
- Chao Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Lina Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Qierra R Brockman
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA.,Molecular Medicine Program, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Fenghuang Zhan
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation and Holden Comprehensive Cancer Center, University of Iowa, 585 Newton Rd., Iowa City, IA, 52242, USA
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China.
| |
Collapse
|
112
|
Pesch T, Bonati L, Kelton W, Parola C, Ehling RA, Csepregi L, Kitamura D, Reddy ST. Molecular Design, Optimization, and Genomic Integration of Chimeric B Cell Receptors in Murine B Cells. Front Immunol 2019; 10:2630. [PMID: 31798579 PMCID: PMC6868064 DOI: 10.3389/fimmu.2019.02630] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/23/2019] [Indexed: 11/13/2022] Open
Abstract
Immune cell therapies based on the integration of synthetic antigen receptors comprise a powerful strategy for the treatment of diverse diseases, most notably T cells engineered to express chimeric antigen receptors (CAR) for targeted cancer therapy. In addition to T lymphocytes, B lymphocytes may also represent valuable immune cells that can be engineered for therapeutic purposes such as protein replacement therapy or recombinant antibody production. In this article, we report a promising concept for the molecular design, optimization, and genomic integration of a novel class of synthetic antigen receptors, chimeric B cell receptors (CBCR). We initially optimized CBCR expression and detection by modifying the extracellular surface tag, the transmembrane regions and intracellular signaling domains. For this purpose, we stably integrated a series of CBCR variants using CRISPR-Cas9 into immortalized B cell hybridomas. Subsequently, we developed a reliable and consistent pipeline to precisely introduce cassettes of several kb size into the genome of primary murine B cells also using CRISPR-Cas9 induced HDR. Finally, we were able to show the robust surface expression and antigen recognition of a synthetic CBCR in primary B cells. We anticipate CBCRs and our approach for engineering primary B cells will be a valuable tool for the advancement of future B cell- based immune cell therapies.
Collapse
Affiliation(s)
- Theresa Pesch
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Lucia Bonati
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - William Kelton
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Cristina Parola
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Life Science Graduate School, Systems Biology, ETH Zürich, University of Zurich, Zurich, Switzerland
| | - Roy A. Ehling
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Lucia Csepregi
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Life Science Graduate School, Microbiology and Immunology, ETH Zürich, University of Zurich, Zurich, Switzerland
| | - Daisuke Kitamura
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Sai T. Reddy
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| |
Collapse
|
113
|
Panagopoulou TI, Rafiq QA. CAR-T immunotherapies: Biotechnological strategies to improve safety, efficacy and clinical outcome through CAR engineering. Biotechnol Adv 2019; 37:107411. [DOI: 10.1016/j.biotechadv.2019.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/23/2019] [Accepted: 06/24/2019] [Indexed: 12/25/2022]
|
114
|
Schacker M, Seimetz D. From fiction to science: clinical potentials and regulatory considerations of gene editing. Clin Transl Med 2019; 8:27. [PMID: 31637541 PMCID: PMC6803602 DOI: 10.1186/s40169-019-0244-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
Gene editing technologies such as CRISPR/Cas9 have emerged as an attractive tool not only for scientific research but also for the development of medicinal products. Their ability to induce precise double strand breaks into DNA enables targeted modifications of the genome including selective knockout of genes, correction of mutations or precise insertion of new genetic material into specific loci. Gene editing-based therapies hold a great potential for the treatment of numerous diseases and the first products are already being tested in clinical trials. The treatment indications include oncological malignancies, HIV, diseases of the hematopoietic system and metabolic disorders. This article reviews ongoing preclinical and clinical studies and discusses how gene editing technologies are altering the gene therapy landscape. In addition, it focusses on the regulatory challenges associated with such therapies and how they can be tackled during the drug development process.
Collapse
Affiliation(s)
- Maria Schacker
- Biopharma Excellence GmbH, Agnes-Pockels-Bogen 1, 80992, Munich, Germany.
| | - Diane Seimetz
- Biopharma Excellence GmbH, Agnes-Pockels-Bogen 1, 80992, Munich, Germany
| |
Collapse
|
115
|
Poorebrahim M, Sadeghi S, Fakhr E, Abazari MF, Poortahmasebi V, Kheirollahi A, Askari H, Rajabzadeh A, Rastegarpanah M, Linē A, Cid-Arregui A. Production of CAR T-cells by GMP-grade lentiviral vectors: latest advances and future prospects. Crit Rev Clin Lab Sci 2019; 56:393-419. [PMID: 31314617 DOI: 10.1080/10408363.2019.1633512] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR) T-cells represent a paradigm shift in cancer immunotherapy and a new milestone in the history of oncology. In 2017, the Food and Drug Administration approved two CD19-targeted CAR T-cell therapies (Kymriah™, Novartis, and Yescarta™, Kite Pharma/Gilead Sciences) that have remarkable efficacy in some B-cell malignancies. The CAR approach is currently being evaluated in multiple pivotal trials designed for the immunotherapy of hematological malignancies as well as solid tumors. To generate CAR T-cells ex vivo, lentiviral vectors (LVs) are particularly appealing due to their ability to stably integrate relatively large DNA inserts, and to efficiently transduce both dividing and nondividing cells. This review discusses the latest advances and challenges in the design and production of CAR T-cells, and the good manufacturing practices (GMP)-grade production process of LVs used as a gene transfer vehicle. New developments in the application of CAR T-cell therapy are also outlined with particular emphasis on next-generation allogeneic CAR T-cells.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Solmaz Sadeghi
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR , Tehran , Iran
| | - Elham Fakhr
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) , Heidelberg , Germany
| | - Mohammad Foad Abazari
- Research Center for Clinical Virology, Tehran University of Medical Sciences , Tehran , Iran
| | - Vahdat Poortahmasebi
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,Infectious and Tropical Disease Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,Faculty of Medicine, Department of Bacteriology and Virology, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Asma Kheirollahi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran , Tehran , Iran
| | - Hassan Askari
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Alireza Rajabzadeh
- Applied Cell Sciences and Tissue Engineering Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Malihe Rastegarpanah
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Aija Linē
- Latvian Biomedical Research and Study Centre , Riga , Latvia
| | - Angel Cid-Arregui
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR , Tehran , Iran.,Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ) , Heidelberg , Germany
| |
Collapse
|
116
|
Strohl WR, Naso M. Bispecific T-Cell Redirection versus Chimeric Antigen Receptor (CAR)-T Cells as Approaches to Kill Cancer Cells. Antibodies (Basel) 2019; 8:E41. [PMID: 31544847 PMCID: PMC6784091 DOI: 10.3390/antib8030041] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022] Open
Abstract
The concepts for T-cell redirecting bispecific antibodies (TRBAs) and chimeric antigen receptor (CAR)-T cells are both at least 30 years old but both platforms are just now coming into age. Two TRBAs and two CAR-T cell products have been approved by major regulatory agencies within the last ten years for the treatment of hematological cancers and an additional 53 TRBAs and 246 CAR cell constructs are in clinical trials today. Two major groups of TRBAs include small, short-half-life bispecific antibodies that include bispecific T-cell engagers (BiTE®s) which require continuous dosing and larger, mostly IgG-like bispecific antibodies with extended pharmacokinetics that can be dosed infrequently. Most CAR-T cells today are autologous, although significant strides are being made to develop off-the-shelf, allogeneic CAR-based products. CAR-Ts form a cytolytic synapse with target cells that is very different from the classical immune synapse both physically and mechanistically, whereas the TRBA-induced synapse is similar to the classic immune synapse. Both TRBAs and CAR-T cells are highly efficacious in clinical trials but both also present safety concerns, particularly with cytokine release syndrome and neurotoxicity. New formats and dosing paradigms for TRBAs and CAR-T cells are being developed in efforts to maximize efficacy and minimize toxicity, as well as to optimize use with both solid and hematologic tumors, both of which present significant challenges such as target heterogeneity and the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- William R Strohl
- BiStro Biotech Consulting, LLC, 1086 Tullo Farm Rd., Bridgewater, NJ 08807, USA.
| | - Michael Naso
- Century Therapeutics, 3675 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
117
|
Zabel M, Tauber PA, Pickl WF. The making and function of CAR cells. Immunol Lett 2019; 212:53-69. [PMID: 31181279 PMCID: PMC7058416 DOI: 10.1016/j.imlet.2019.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/24/2019] [Accepted: 06/06/2019] [Indexed: 12/28/2022]
Abstract
Genetically engineered T cells expressing chimeric antigen receptors (CAR) present a new treatment option for patients with cancer. Recent clinical trials of B cell leukemia have demonstrated a response rate of up to 90%. However, CAR cell therapy is frequently accompanied by severe side effects such as cytokine release syndrome and the development of target cell resistance. Consequently, further optimization of CARs to obtain greater long-term efficacy and increased safety is urgently needed. Here we high-light the various efforts of adjusting the intracellular signaling domains of CARs to these major requirements to eventually obtain high-level target cell cytotoxicity paralleled by the establishment of longevity of the CAR expressing cell types to guarantee for extended tumor surveillance over prolonged periods of time. We are convinced that it will be crucial to identify the molecular pathways and signaling requirements utilized by such ‘efficient CARs’ in order to provide a rational basis for their further hypothesis-based improvement. Furthermore, we here discuss timely attempts of how to: i) control ‘on-tumor off-target’ effects; ii) introduce Signal 3 (cytokine responsiveness of CAR cells) as an important building-block into the CAR concept; iii) most efficiently eliminate CAR cells once full remission has been obtained. We also argue that universal systems for the variable and pharmacokinetically-controlled attachment of extracellular ligand recognition domains of choice along with the establishment of ‘off-the-shelf’ cell preparations with suitability for all patients in need of a highly-potent cellular therapy may become future mainstays of CAR cell therapy. Such therapies would have the attraction to work independent of the patients’ histo-compatibility make-up and the availability of functionally intact patient’s cells. Finally, we summarize the evidence that CAR cells may obtain a prominent place in the treatment of non-malignant and auto-reactive T and B lymphocyte expansions in the near future, e.g., for the alleviation of autoimmune diseases and allergies. After the introduction of red blood cell transfusions, which were made possible by the landmark discoveries of the ABO blood groups by Karl Landsteiner, and the establishment of bone marrow transplantation by E. Donnall Thomas to exchange the entire hematopoietic system of a patient suffering from leukemia, the introduction of patient-tailored cytotoxic cellular populations to eradicate malignant cell populations in vivo pioneered by Carl H. June, represents the third major and broadly applicable milestone in the development of human cellular therapies within the rapidly developing field of applied biomedical research of the last one hundred years.
Collapse
Affiliation(s)
- Maja Zabel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter A Tauber
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Winfried F Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
118
|
Marei HE, Althani A, Caceci T, Arriga R, Sconocchia T, Ottaviani A, Lanzilli G, Roselli M, Caratelli S, Cenciarelli C, Sconocchia G. Recent perspective on CAR and Fcγ-CR T cell immunotherapy for cancers: Preclinical evidence versus clinical outcomes. Biochem Pharmacol 2019; 166:335-346. [PMID: 31176617 DOI: 10.1016/j.bcp.2019.06.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 06/03/2019] [Indexed: 12/16/2022]
Abstract
The chimeric antigen receptor T cell (CAR-T cell) immunotherapy currently represents a hot research trend and it is expected to revolutionize the field of cancer therapy. Promising outcomes have been achieved using CAR-T cell therapy for haematological malignancies. Despite encouraging results, several challenges still pose eminent hurdles before being fully recognized. Directing CAR-T cells to target a single tumour associated antigen (TAA) as the case in haematological malignancies might be much simpler than targeting the extensive inhibitory microenvironments associated with solid tumours. This review focuses on the basic principles involved in development of CAR-T cells, emphasizing the differences between humoral IgG, T-cell receptors, CAR and Fcγ-CR constructs. It also highlights the complex inhibitory network that is usually associated with solid tumours, and tackles recent advances in the clinical studies that have provided great hope for the future use of CAR-T cell immunotherapy. While current Fcγ-CR T cell immunotherapy is in pre-clinical stage, is expected to provide a sound therapeutic approach to add to existing classical chemo- and radio-therapeutic modalities.
Collapse
Affiliation(s)
- Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35116, Egypt.
| | - Asma Althani
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Thomas Caceci
- Biomedical Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, United States
| | - Roberto Arriga
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | | | | | - Mario Roselli
- Department of Systems Medicine, Endocrinology and Medical Oncology, University of Rome "Tor Vergata", Rome, Italy
| | - Sara Caratelli
- Institute of Translational Pharmacology-CNR, Rome, Italy
| | | | | |
Collapse
|
119
|
Stoiber S, Cadilha BL, Benmebarek MR, Lesch S, Endres S, Kobold S. Limitations in the Design of Chimeric Antigen Receptors for Cancer Therapy. Cells 2019; 8:cells8050472. [PMID: 31108883 PMCID: PMC6562702 DOI: 10.3390/cells8050472] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022] Open
Abstract
Cancer therapy has entered a new era, transitioning from unspecific chemotherapeutic agents to increasingly specific immune-based therapeutic strategies. Among these, chimeric antigen receptor (CAR) T cells have shown unparalleled therapeutic potential in treating refractory hematological malignancies. In contrast, solid tumors pose a much greater challenge to CAR T cell therapy, which has yet to be overcome. As this novel therapeutic modality matures, increasing effort is being invested to determine the optimal structure and properties of CARs to facilitate the transition from empirical testing to the rational design of CAR T cells. In this review, we highlight how individual CAR domains contribute to the success and failure of this promising treatment modality and provide an insight into the most notable advances in the field of CAR T cell engineering.
Collapse
Affiliation(s)
- Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
- German Center for Translational Cancer Research (DKTK), 80337 Munich, Germany.
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Member of the German Center for Lung Research (DZL), 80337 Munich, Germany.
- German Center for Translational Cancer Research (DKTK), 80337 Munich, Germany.
| |
Collapse
|
120
|
Weinkove R, George P, Dasyam N, McLellan AD. Selecting costimulatory domains for chimeric antigen receptors: functional and clinical considerations. Clin Transl Immunology 2019; 8:e1049. [PMID: 31110702 PMCID: PMC6511336 DOI: 10.1002/cti2.1049] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 12/11/2022] Open
Abstract
Costimulatory signals are required to achieve robust chimeric antigen receptor (CAR) T cell expansion, function, persistence and antitumor activity. These can be provided by incorporating intracellular signalling domains from one or more T cell costimulatory molecules, such as CD28 or 4-1BB, into the CAR. The selection and positioning of costimulatory domains within a CAR construct influence CAR T cell function and fate, and clinical experience of autologous anti-CD19 CAR T cell therapies suggests that costimulatory domains have differential impacts on CAR T cell kinetics, cytotoxic function and potentially safety profile. The clinical impacts of combining costimulatory domains and of alternative costimulatory domains are not yet clearly established, and may be construct- and disease-specific. The aim of this review is to summarise the function and effect of established and emerging costimulatory domains and their combinations within CAR T cells.
Collapse
Affiliation(s)
- Robert Weinkove
- Cancer Immunotherapy Programme Malaghan Institute of Medical Research Wellington New Zealand.,Wellington Blood & Cancer Centre Capital & Coast District Health Board Wellington New Zealand.,Department of Pathology & Molecular Medicine University of Otago Wellington Wellington New Zealand
| | - Philip George
- Cancer Immunotherapy Programme Malaghan Institute of Medical Research Wellington New Zealand.,Wellington Blood & Cancer Centre Capital & Coast District Health Board Wellington New Zealand
| | - Nathaniel Dasyam
- Cancer Immunotherapy Programme Malaghan Institute of Medical Research Wellington New Zealand
| | - Alexander D McLellan
- Department of Microbiology and Immunology University of Otago Dunedin New Zealand
| |
Collapse
|
121
|
Hull CM, Maher J. Novel approaches to promote CAR T-cell function in solid tumors. Expert Opin Biol Ther 2019; 19:789-799. [DOI: 10.1080/14712598.2019.1614164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Caroline M Hull
- School of Cancer and Pharmaceutical Sciences, King’s College London, Division of Cancer Studies, Guy’s Hospital, London, UK
| | - John Maher
- School of Cancer and Pharmaceutical Sciences, King’s College London, Division of Cancer Studies, Guy’s Hospital, London, UK
- Department of Clinical Immunology and Allergy, King’s College Hospital NHS Foundation Trust, London UK
- Department of Immunology, Eastbourne Hospital, Eastbourne, UK
| |
Collapse
|
122
|
Wang L, Yao R, Zhang L, Fan C, Ma L, Liu J. Chimeric antigen receptor T cell therapy and other therapeutics for malignancies: Combination and opportunity. Int Immunopharmacol 2019; 70:498-503. [PMID: 30875561 DOI: 10.1016/j.intimp.2019.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 02/07/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy provides possibility for the treatment of malignancies since clinical trials have shown that CAR-T therapy has a significant anti-tumor effect. Although many efforts have been made to improve the efficacy and reduce the side effects of CAR-T therapy, there are still many problems to solve. With the rapid development of this field, combination immunotherapy has been proved to improve the efficacy of CAR-T therapy. Studies have shown that radiotherapy, chemotherapy, oncolytic virotherapy, BTK inhibitors and immune checkpoint blockade-based therapy may further enhance the efficacy of CAR-T therapy while CRISPR/Cas9 technology and IL-1 blockade may improve the safety. In this review, we summarized the advantages and the mechanisms of the combination immunotherapy based on CAR-T cell therapy.
Collapse
Affiliation(s)
- Luyao Wang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Ruixue Yao
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Lifa Zhang
- 401 Hospital of the People's Liberation Army, China
| | - Chuanbo Fan
- Department of Hematology, Qingdao Hiser Medical Center, China
| | - Leina Ma
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266061, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
123
|
El-Daly SM, Hussein J. Genetically engineered CAR T-immune cells for cancer therapy: recent clinical developments, challenges, and future directions. J Appl Biomed 2019; 17:11. [PMID: 34907752 DOI: 10.32725/jab.2019.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cancer immunotherapy offers tremendous clinical outcomes in cancer management with the potential to induce sustained remission in patients with refractory disease. One of these immunotherapy modalities is the adoptive transfer of autologous T-cells that are genetically engineered ex vivo to express chimeric antigen receptors (CARs). These receptors can direct T-cells to the surface antigens of tumor cells to initiate an efficient and specific cytotoxic response against tumor cells. This review elucidates the structural features of CAR T-cells and their different generations reaching the recent 4th generation (TRUCK). The step-wise treatment process using CAR T-cell therapy and some of the updated prominent clinical applications of this treatment modality in both hematologic and solid malignancies are also covered in the present review. The success of CAR T-cell therapy is still encountered by several limitations for a widespread clinical application of this treatment modality, these challenges along with the recent innovative strategies that have been developed to overcome such drawbacks, as well as, the approaches and future directions aiming for a commercial low cost CAR T-cell immunotherapy modality, are all covered in the present review.
Collapse
Affiliation(s)
- Sherien M El-Daly
- National Research Centre, Medical Research Division, Department of Medical Biochemistry, Dokki, Cairo, Egypt.,National Research Centre, Centre of Excellence for Advanced Sciences, Cancer Biology and Genetics Laboratory, Dokki, Cairo, Egypt
| | - Jihan Hussein
- National Research Centre, Medical Research Division, Department of Medical Biochemistry, Dokki, Cairo, Egypt
| |
Collapse
|
124
|
Zhang R, Deng Q, Sui SN, Jin X, Zhao MF. [The specific cytotoxicities of chimeric antigen receptor-engineered T cells on different lymphomas]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 38:842-847. [PMID: 29166735 PMCID: PMC7364971 DOI: 10.3760/cma.j.issn.0253-2727.2017.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
目的 探讨共刺激分子为4-1BB的二代CD19嵌合抗原受体T细胞(CAR-T细胞)对不同侵袭性淋巴瘤细胞的体内、体外杀伤活性以及CAR-T细胞在体内生存的时间。 方法 采用慢病毒包装并感染T细胞的方法制备CD19 CAR-T细胞,采用CCK-8、ELISA和乳酸脱氢酶细胞毒性检测法检测CD19 CAR-T细胞的增殖能力、炎症因子释放水平和对Raji(套细胞淋巴瘤细胞株)、Pfeiffer(弥漫大B细胞淋巴瘤细胞株)、EHEB(慢性淋巴细胞白血病细胞株)细胞的杀伤活性,采用流式细胞术分析CD19 CAR-T细胞治疗前后荷瘤裸鼠的肿瘤负荷和CAR-T细胞残留水平。 结果 ①与二代CAR-T细胞比较,三代CAR-T细胞的体外(24、48 h)增殖能力(P值均<0.05)、对瘤细胞的杀伤活性(P值均<0.05)较强,差异有统计学意义;而炎症因子的释放水平差异无统计学意义(P值均>0.05)。②效靶比为4∶1且共培养48 h时,二代CD19 CAR-T细胞对Raji细胞[(72.36±2.98)%]、Pfeiffer细胞[(55.92±4.16)%]、EHEB细胞[(35.53±3.93)%]的杀伤活性从强到弱,差异有统计学意义(P=0.013)。③裸鼠体内试验结果显示二代CAR-T细胞在EHEB荷瘤小鼠体内存活时间长于Raji荷瘤小鼠(P=0.046)。 结论 共刺激分子为4-1BB的二代CD19 CAR-T细胞对惰性淋巴瘤细胞株的杀伤活性稍低于对侵袭性淋巴瘤细胞株,但在惰性淋巴瘤细胞株荷瘤小鼠体内存活时间更长,可能更适合惰性淋巴瘤的治疗。
Collapse
Affiliation(s)
- R Zhang
- Department of Hematology and Oncology, Tianjin First Central Hospital, Tianjin 300192, China
| | | | | | | | | |
Collapse
|
125
|
Rahbarizadeh F, Ahmadvand D, Moghimi S. CAR T-cell bioengineering: Single variable domain of heavy chain antibody targeted CARs. Adv Drug Deliv Rev 2019; 141:41-46. [PMID: 31004624 DOI: 10.1016/j.addr.2019.04.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 10/27/2022]
Abstract
Redirecting the recognition specificity of T lymphocytes to designated tumour cell surface antigens by transferring chimeric antigen receptor (CAR) genes is becoming an effective strategy to combat cancer. Today, CAR T-cell therapy has proven successful in the treatment of haematological malignancies and the first CD19 CAR T-cell products has already entered the market. This success is expanding CAR design for broader malignancies including solid tumours. Nevertheless, CARs such as those built on antigen-specific single chain antibody variable fragment (scFv) may induce some adverse effects. Here, we briefly review CAR T-cell bioengineering and discuss selected important initiatives for improved T-cell reprogramming, function and safety. In this respect, we further elaborate on unconventional CARs structured on single variable domain of heavy chain (VHH) antibodies (single-domain antibodies) as an alternative to scFv, because of their interesting immunological and physicochemical characteristics and unique structure, which shows a high degree of homology with human VH3 gene family.
Collapse
|
126
|
Rydzek J, Nerreter T, Peng H, Jutz S, Leitner J, Steinberger P, Einsele H, Rader C, Hudecek M. Chimeric Antigen Receptor Library Screening Using a Novel NF-κB/NFAT Reporter Cell Platform. Mol Ther 2019; 27:287-299. [PMID: 30573301 PMCID: PMC6369451 DOI: 10.1016/j.ymthe.2018.11.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell immunotherapy is under intense preclinical and clinical investigation, and it involves a rapidly increasing portfolio of novel target antigens and CAR designs. We established a platform that enables rapid and high-throughput CAR-screening campaigns with reporter cells derived from the T cell lymphoma line Jurkat. Reporter cells were equipped with nuclear factor κB (NF-κB) and nuclear factor of activated T cells (NFAT) reporter genes that generate a duplex output of enhanced CFP (ECFP) and EGFP, respectively. As a proof of concept, we modified reporter cells with CD19-specific and ROR1-specific CARs, and we detected high-level reporter signals that allowed distinguishing functional from non-functional CAR constructs. The reporter data were highly reproducible, and the time required for completing each testing campaign was substantially shorter with reporter cells (6 days) compared to primary CAR-T cells (21 days). We challenged the reporter platform to a large-scale screening campaign on a ROR1-CAR library, and we showed that reporter cells retrieved a functional CAR variant that was present with a frequency of only 6 in 1.05 × 106. The data illustrate the potential to implement this reporter platform into the preclinical development path of novel CAR-T cell products and to inform and accelerate the selection of lead CAR candidates for clinical translation.
Collapse
Affiliation(s)
- Julian Rydzek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Haiyong Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, USA
| | - Sabrina Jutz
- Institut für Immunologie, Medizinische Universität Wien, Wien, Austria
| | - Judith Leitner
- Institut für Immunologie, Medizinische Universität Wien, Wien, Austria
| | - Peter Steinberger
- Institut für Immunologie, Medizinische Universität Wien, Wien, Austria
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Christoph Rader
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, USA
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany.
| |
Collapse
|
127
|
Sukari A, Abdallah N, Nagasaka M. Unleash the power of the mighty T cells-basis of adoptive cellular therapy. Crit Rev Oncol Hematol 2019; 136:1-12. [PMID: 30878123 DOI: 10.1016/j.critrevonc.2019.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 02/04/2023] Open
Abstract
Adoptive cellular therapy (ACT) is an immunotherapy which involves the passive transfer of lymphocytes into a lymphodepleted host after ex vivo stimulation and expansion. Tumor-infiltrating lymphocytes (TILs) have shown objective tumor responses mainly restricted to melanoma and rely on a laborious manufacturing process. These limitations led to emergence of engineered cells, where normal peripheral blood lymphocytes are modified to express T cell receptors (TCRs) or chimeric antigen receptors (CARs) specific for tumor-associated antigens (TAAs). To date, CD19-targeted chimeric antigen receptor T (CAR T) cells have been the most extensively studied, showing complete and durable responses in B-cell malignancies. Antitumor responses with engineered T cells have often been accompanied by undesired toxicities in clinical trials including cytokine release syndrome (CRS) and neurotoxicity. In this review, we provide an overview of adoptive cellular strategies, early and ongoing clinical trials, adverse events and strategies to mitigate side effects and overcome limitations.
Collapse
Affiliation(s)
- Ammar Sukari
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Nadine Abdallah
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Misako Nagasaka
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Advanced Medical Innovation, St. Marianna University Graduate School of Medicine, Kawasaki, Kanagawa, Japan
| |
Collapse
|
128
|
Chen Y, Sun C, Landoni E, Metelitsa L, Dotti G, Savoldo B. Eradication of Neuroblastoma by T Cells Redirected with an Optimized GD2-Specific Chimeric Antigen Receptor and Interleukin-15. Clin Cancer Res 2019; 25:2915-2924. [PMID: 30617136 DOI: 10.1158/1078-0432.ccr-18-1811] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/30/2018] [Accepted: 01/04/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE A delay in encountering the cognate antigen while in the circulation, and the suboptimal costimulation received at the tumor site are key reasons for the limited activity of chimeric antigen receptor-redirected T cells (CAR-T) in solid tumors. We have explored the benefits of incorporating the IL15 cytokine within the CAR cassette to provide both a survival signal before antigen encounter, and an additional cytokine signaling at the tumor site using a neuroblastoma tumor model. EXPERIMENTAL DESIGN We optimized the construct for the CAR specific for the NB-antigen GD2 without (GD2.CAR) or with IL15 (GD2.CAR.15). We then compared the expansion, phenotype, and antitumor activity of T cells transduced with these constructs against an array of neuroblastoma cell lines in vitro and in vivo using a xenogeneic metastatic model of neuroblastoma. RESULTS We observed that optimized GD2.CAR.15-Ts have reduced expression of the PD-1 receptor, are enriched in stem cell-like cells, and have superior antitumor activity upon repetitive tumor exposures in vitro and in vivo as compared with GD2.CAR-Ts. Tumor rechallenge experiments in vivo further highlighted the role of IL15 in promoting enhanced CAR-T antitumor activity and survival, both in the peripheral blood and tissues. Finally, the inclusion of the inducible caspase-9 gene (iC9) safety switch warranted effective on demand elimination of the engineered GD2.CAR.15-Ts. CONCLUSIONS Our results guide new therapeutic options for GD2.CAR-Ts in patients with neuroblastoma, and CAR-T development for a broad range of solid tumors.
Collapse
Affiliation(s)
- Yuhui Chen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chuang Sun
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Leonid Metelitsa
- Department of Pediatrics, Texas Children's Hospital, Houston, Texas
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. .,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
129
|
Guedan S, Calderon H, Posey AD, Maus MV. Engineering and Design of Chimeric Antigen Receptors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 12:145-156. [PMID: 30666307 PMCID: PMC6330382 DOI: 10.1016/j.omtm.2018.12.009] [Citation(s) in RCA: 285] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T cells engineered with chimeric antigen receptors (CARs) have emerged as a potent new class of therapeutics for cancer, based on their remarkable potency in blood cancers. Since the first clinical reports of their efficacy emerged 7 years ago, investigators have focused on the mechanisms and properties that make CARs effective or toxic, and their effects on T cell biology. Novel CAR designs coupled with improvements in gene transfer technology, incorporating advances in gene editing, have the potential to increase access to engineered cell therapies, as well as improve their potency in solid tumors.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Hugo Calderon
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Avery D Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
130
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
131
|
Omer B, Castillo PA, Tashiro H, Shum T, Huynh MTA, Cardenas M, Tanaka M, Lewis A, Sauer T, Parihar R, Lapteva N, Schmueck-Henneresse M, Mukherjee M, Gottschalk S, Rooney CM. Chimeric Antigen Receptor Signaling Domains Differentially Regulate Proliferation and Native T Cell Receptor Function in Virus-Specific T Cells. Front Med (Lausanne) 2018; 5:343. [PMID: 30619856 PMCID: PMC6297364 DOI: 10.3389/fmed.2018.00343] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/22/2018] [Indexed: 11/16/2022] Open
Abstract
The efficacy of T cells expressing chimeric antigen receptors (CARs) for solid tumors has been limited by insufficient CAR T cell expansion and persistence. The use of virus-specific T cells (VSTs) as carriers for CARs may overcome this limitation since CAR-VSTs can be boosted by viral vaccines or oncolytic viruses. However, there is limited understanding of the optimal combination of endodomains and their influence on the native T cell receptor (TCR) in VSTs. We therefore compared the function of GD2.CARs expressing the TCR zeta chain (ζ) alone or combined with endodomains from CD28 and 4-1BB in varicella zoster virus-specific (VZV) T cells. VZVSTs expressing GD2-CARs recognized VZV-derived peptides and killed GD2-expressing tumor cells. However, after repeated stimulation through their native TCR, the expansion of GD2-CAR.CD28ζ-VZVSTs was 3.3-fold greater (p < 0.001) than non-transduced VZVSTs, whereas GD2-CARζ- and GD2-CAR.41BBζ inhibited VZVST expansion (p < 0.01). Compared to control VZVSTs, GD2-CAR.ζ VZVSTs showed a greater frequency of apoptotic (p < 0.01) T cells, whereas prolonged downregulation of the native αβ TCR was observed in GD2-CAR.41BBζ VZVSTs (p < 0.001). We confirmed that CD28ζ can best maintain TCR function by expressing GD2.CARs in Epstein-Barr virus-specific T cells and CD19-CARs in VZVSTs. In response to CAR stimulation VSTs with CD28ζ endodomains also showed the greatest expansion (6 fold > GD2-CAR.41BBζ VZVSTs (p < 0.001), however anti-tumor efficacy was superior in GD2-CAR.41BBζ-VZVSTs. These findings demonstrate that CAR signaling domains can enhance or diminish the function of the native TCR and indicate that only CD28ζ may preserve the function of the native TCR in tonically signaling CAR-VSTs.
Collapse
Affiliation(s)
- Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Paul A Castillo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Haruko Tashiro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Thomas Shum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Mai T A Huynh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Mara Cardenas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Miyuki Tanaka
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Andrew Lewis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Tim Sauer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Robin Parihar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Michael Schmueck-Henneresse
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Malini Mukherjee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
132
|
Boyiadzis MM, Dhodapkar MV, Brentjens RJ, Kochenderfer JN, Neelapu SS, Maus MV, Porter DL, Maloney DG, Grupp SA, Mackall CL, June CH, Bishop MR. Chimeric antigen receptor (CAR) T therapies for the treatment of hematologic malignancies: clinical perspective and significance. J Immunother Cancer 2018; 6:137. [PMID: 30514386 PMCID: PMC6278156 DOI: 10.1186/s40425-018-0460-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 11/20/2018] [Indexed: 02/06/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapies - adoptive T cell therapies that have been genetically engineered for a new antigen-specificity - have displayed significant success in treating patients with hematologic malignancies, leading to three recent US Food and Drug Administration approvals. Based on the promise generated from these successes, the field is rapidly evolving to include new disease indications and CAR designs, while simultaneously reviewing and optimizing toxicity and management protocols. As such, this review provides expert perspective on the significance and clinical considerations of CAR T cell therapies in order to provide timely information to clinicians about this revolutionary new therapeutic class.
Collapse
Affiliation(s)
| | | | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, National Cancer Institute, Bethesda, MD, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marcela V Maus
- Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - David L Porter
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - David G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Stephan A Grupp
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Oncology, Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Crystal L Mackall
- Cancer Immunology and Immunotherapy Program, Stanford University, Stanford, CA, USA
| | - Carl H June
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael R Bishop
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA. .,The University of Chicago, 5841 S. Maryland Avenue, MC 2115, Chicago, IL, 60637, USA.
| |
Collapse
|
133
|
Munshi PN, Ujjani C. The acceleration of CAR‐T therapy in non‐Hodgkin lymphoma. Hematol Oncol 2018; 37:233-239. [DOI: 10.1002/hon.2568] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Pashna N. Munshi
- Lombardi Comprehensive Cancer CenterMedStar Georgetown University Hospital Washington DC USA
| | - Chaitra Ujjani
- Seattle Cancer Care AllianceUniversity of Washington/Fred Hutchinson Cancer Research Center Seattle Seattle Washington USA
| |
Collapse
|
134
|
Vairy S, Garcia JL, Teira P, Bittencourt H. CTL019 (tisagenlecleucel): CAR-T therapy for relapsed and refractory B-cell acute lymphoblastic leukemia. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:3885-3898. [PMID: 30518999 PMCID: PMC6237143 DOI: 10.2147/dddt.s138765] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Over the past decades, survival of patients with acute lymphoblastic leukemia (ALL) has dramatically improved, but the subgroup of patients with relapsed/refractory ALL still continues to have dismal prognosis. As an emerging therapeutic approach, chimeric antigen receptor-modified T-cells (CAR-T) represent one of the few practice-changing therapies for this subgroup of patients. Originally conceived and built in Philadelphia (University of Pennsylvania), CTL019 or tisagenlecleucel, the first CAR-T approved by the US Food and Drug Administration, showed impressive results in refractory/relapsed ALL since the publication on two pediatric patients in 2013. It is in this context that we provide a review of this product in terms of manufacturing, pharmacology, toxicity, and efficacy studies. Evaluation and management of toxicities, particularly cytokine release syndrome and neurotoxicity, is recognized as an essential part of the patient treatment with broader use of IL-6 receptor inhibitor. An under-assessed aspect, the quality of life of patients entering CAR-T cells treatment, will also be reviewed. By their unique nature, CAR-T cells such as tisagenlecleucel operate in a different way than typical drugs, but also provide unique hope for B-cell malignancies.
Collapse
Affiliation(s)
- Stephanie Vairy
- Division of Haematology and Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada,
| | - Julia Lopes Garcia
- Division of Haematology and Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada,
| | - Pierre Teira
- Division of Haematology and Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada,
| | - Henrique Bittencourt
- Division of Haematology and Oncology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada,
| |
Collapse
|
135
|
Schmidts A, Maus MV. Making CAR T Cells a Solid Option for Solid Tumors. Front Immunol 2018; 9:2593. [PMID: 30467505 PMCID: PMC6235951 DOI: 10.3389/fimmu.2018.02593] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/22/2018] [Indexed: 01/02/2023] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor (CAR) T cells aims to redirect the patient's own immune system to selectively attack cancer cells. To do so, CAR T cells are endowed with specific antigen recognition moieties fused to signaling and costimulatory domains. While this approach has shown great success for the treatment of B cell malignancies, response rates among patients with solid cancers are less favorable. The major challenges for CAR T cell immunotherapy in solid cancers are the identification of unique tumor target antigens, as well as improving CAR T cell trafficking to and expansion at the tumor site. This review focuses on combinatorial antigen targeting, regional delivery and approaches to improve CAR T cell persistence in the face of a hostile tumor microenvironment.
Collapse
Affiliation(s)
- Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
136
|
Mirzaei HR, Jamali A, Jafarzadeh L, Masoumi E, Alishah K, Fallah Mehrjardi K, Emami SAH, Noorbakhsh F, Till BG, Hadjati J. Construction and functional characterization of a fully human anti‐CD19 chimeric antigen receptor (huCAR)‐expressing primary human T cells. J Cell Physiol 2018; 234:9207-9215. [DOI: 10.1002/jcp.27599] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 09/19/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Hamid Reza Mirzaei
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Arezoo Jamali
- Department of Molecular Medicine Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences Tehran Iran
- Department of Medical Laboratory Sciences School of Allied Medical Sciences, Tehran University of Medical Sciences Tehran Iran
| | - Leila Jafarzadeh
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Elham Masoumi
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Khadijeh Alishah
- Department of Biotechnology College of Science, University of Tehran Tehran Iran
| | - Keyvan Fallah Mehrjardi
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Seyed Amir Hossein Emami
- Division of Oncology, Department of Internal Medicine School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Farshid Noorbakhsh
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Brian G. Till
- Clinical Research Division Fred Hutchinson Cancer Research Center Seattle Washington
| | - Jamshid Hadjati
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
137
|
Saetersmoen ML, Hammer Q, Valamehr B, Kaufman DS, Malmberg KJ. Off-the-shelf cell therapy with induced pluripotent stem cell-derived natural killer cells. Semin Immunopathol 2018; 41:59-68. [PMID: 30361801 DOI: 10.1007/s00281-018-0721-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022]
Abstract
Cell therapy is emerging as a very promising therapeutic modality against cancer, spearheaded by the clinical success of chimeric antigen receptor (CAR) modified T cells for B cell malignancies. Currently, FDA-approved CAR-T cell products are based on engineering of autologous T cells harvested from the patient, typically using a central manufacturing facility for gene editing before the product can be delivered to the clinic and infused to the patients. For a broader implementation of advanced cell therapy and to reduce costs, it would be advantageous to use allogeneic "universal" cell therapy products that can be stored in cell banks and provided upon request, in a manner analogous to biopharmaceutical drug products. In this review, we outline a roadmap for development of off-the-shelf cell therapy based on natural killer (NK) cells derived from induced pluripotent stem cells (iPSCs). We discuss strategies to engineer iPSC-derived NK (iPSC-NK) cells for enhanced functional potential, persistence, and homing.
Collapse
Affiliation(s)
| | - Quirin Hammer
- Department of Medicine, Huddinge, Karolinska Institute, Solna, Sweden
| | | | - Dan S Kaufman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Karl-Johan Malmberg
- The KG Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway. .,Department of Medicine, Huddinge, Karolinska Institute, Solna, Sweden. .,Institute for Cancer research, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
138
|
Richards RM, Sotillo E, Majzner RG. CAR T Cell Therapy for Neuroblastoma. Front Immunol 2018; 9:2380. [PMID: 30459759 PMCID: PMC6232778 DOI: 10.3389/fimmu.2018.02380] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 09/25/2018] [Indexed: 12/11/2022] Open
Abstract
Patients with high risk neuroblastoma have a poor prognosis and survivors are often left with debilitating long term sequelae from treatment. Even after integration of anti-GD2 monoclonal antibody therapy into standard, upftont protocols, 5-year overall survival rates are only about 50%. The success of anti-GD2 therapy has proven that immunotherapy can be effective in neuroblastoma. Adoptive transfer of chimeric antigen receptor (CAR) T cells has the potential to build on this success. In early phase clinical trials, CAR T cell therapy for neuroblastoma has proven safe and feasible, but significant barriers to efficacy remain. These include lack of T cell persistence and potency, difficulty in target identification, and an immunosuppressive tumor microenvironment. With recent advances in CAR T cell engineering, many of these issues are being addressed in the laboratory. In this review, we summarize the clinical trials that have been completed or are underway for CAR T cell therapy in neuroblastoma, discuss the conclusions and open questions derived from these trials, and consider potential strategies to improve CAR T cell therapy for patients with neuroblastoma.
Collapse
Affiliation(s)
- Rebecca M. Richards
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Elena Sotillo
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Robbie G. Majzner
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
139
|
Zolov SN, Rietberg SP, Bonifant CL. Programmed cell death protein 1 activation preferentially inhibits CD28.CAR-T cells. Cytotherapy 2018; 20:1259-1266. [PMID: 30309710 DOI: 10.1016/j.jcyt.2018.07.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/17/2018] [Indexed: 12/14/2022]
Abstract
Targeted adoptive immunotherapy with engineered T cells is a promising treatment for refractory hematologic malignancies. However, many patients achieving early complete remissions ultimately relapse. Immunosuppressive ligands are expressed on tumor and supportive cells in the tumor microenvironment (TME). When activated, T cells express associated "checkpoint" receptors. Binding of co-inhibitory ligands and receptors may directly contribute to T-cell functional exhaustion. It is not known whether all T cells engineered to express chimeric antigen receptors (CARs) are subject to checkpoint-mediated regulation. It is also unknown whether distinct CAR signaling moieties modulate T-cell responsiveness to these inhibitory pathways. We have, therefore, directly compared functional co-inhibition in engineered T cells identically targeted to the tumor-associated antigen CD123, but distinct in their mode of T-cell activation: via the endogenous T-cell receptor (ENG), or downstream of CD28 or 41BB-containing CARs. In all cases, we have observed antigen-independent T-cell activation associated with upregulation of the co-inhibitory receptors programmed cell death protein 1 (PD-1, CD279), Tim-3 and Lag-3. Notably, CD28.CAR T cells were uniquely susceptible to PD-1/PD-L1 mediated checkpoint inhibition. Together, our data indicate that PD-1/PD-L1 checkpoint blocking agents may be considered clinically when CD28.CAR T cells do not perform optimally in human trials.
Collapse
Affiliation(s)
- Sergey N Zolov
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Skyler P Rietberg
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA
| | - Challice L Bonifant
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
140
|
Han X, Wang Y, Han WD. Chimeric antigen receptor modified T-cells for cancer treatment. Chronic Dis Transl Med 2018; 4:225-243. [PMID: 30603741 PMCID: PMC6309024 DOI: 10.1016/j.cdtm.2018.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Indexed: 12/12/2022] Open
Abstract
T cells engineered with the chimeric antigen receptor (CAR) are rapidly emerging as an important immunotherapy for hematologic malignancies. The anti-cluster of differentiation (CD)19 CAR-T cell therapy has been remarkably successful against refractory/relapsed acute lymphoblastic leukemia (ALL), and a complete remission rate as high as 90% was observed, in both children and adults. Although the achievement of clinical efficacy using CAR-T cell therapy for solid tumors has encountered several obstacles that were associated with the multiple mechanisms contributing to an immunosuppressive microenvironment, investigators are exploring more optimized approaches to improve the efficiency of CAR-T in solid tumors. In addition, cytokine release syndrome (CRS) and neurotoxicity following CAR-T cell therapy can be severe or even fatal; therefore, the management of these toxicities is significant. Herein, we briefly review the structure of CAR-T and some novel CAR designs, the clinical application of CAR-T cell therapies, as well as the assessment and management of toxicities.
Collapse
Affiliation(s)
- Xiao Han
- Molecular & Immunological Department, Bio-therapeutic Department, The General Hospital of People's Liberation Army, Beijing 100853, China
| | - Yao Wang
- Molecular & Immunological Department, Bio-therapeutic Department, The General Hospital of People's Liberation Army, Beijing 100853, China
| | - Wei-Dong Han
- Molecular & Immunological Department, Bio-therapeutic Department, The General Hospital of People's Liberation Army, Beijing 100853, China
| |
Collapse
|
141
|
Zhang J, Medeiros LJ, Young KH. Cancer Immunotherapy in Diffuse Large B-Cell Lymphoma. Front Oncol 2018; 8:351. [PMID: 30250823 PMCID: PMC6140403 DOI: 10.3389/fonc.2018.00351] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/09/2018] [Indexed: 12/26/2022] Open
Abstract
Remarkable progress has been made in the field of cancer immunotherapy in the past few years. Immunotherapy has become a standard treatment option for patients with various cancers, including melanoma, lymphoma, and carcinomas of the lungs, kidneys, bladder, and head and neck. Promising immunotherapy approaches, such as chimeric antigen receptor (CAR) T cell therapy and therapeutic blockade of immune checkpoints, in particular cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 pathway (PD-1/PD-L1), have boosted the development of new therapeutic regimens for patients with cancer. Immunotherapeutic strategies for diffuse large B-cell lymphoma (DLBCL) include monoclonal anti-CD20 antibody (rituximab), monoclonal anti-PD-1 antibodies (nivolumab and pembrolizumab), monoclonal anti-PD-L1 antibodies (avelumab, durvalumab, and atezolizumab) and chimeric antigen receptor (CAR) T cell therapy. In this review, we outline the latest highlights and progress in using immunotherapy to treat patients with DLBCL, with a focus on the therapeutic blockade of PD-1/PD-L1 and CAR T cell therapy in DLBCL. We also discuss current clinical trials of PD-1/PD-L1 and CAR T cell therapy and review the challenges and opportunities of using immunotherapy for the treatment of DLBCL.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ken H. Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX, United States
| |
Collapse
|
142
|
Perez-Amill L, Marzal B, Urbano-Ispizua A, Juan M, Martín-Antonio B. CAR-T Cell Therapy: A Door Is Open to Find Innumerable Possibilities of Treatments for Cancer Patients. Turk J Haematol 2018; 35:217-228. [PMID: 30185400 PMCID: PMC6256819 DOI: 10.4274/tjh.2018.0196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Seven years ago a chronic lymphocytic leukemia patient was for the first time successfully treated with chimeric antigen receptor (CAR)-modified T cells (CAR-T cells) to target CD19 overexpression in tumor cells. This was the beginning of the development of a new type of immunotherapy treatment in cancer patients. Since then, identification of novel antigens expressed in tumor cells and optimization of both CAR constructs and protocols of administration have opened up new avenues for the successful treatment of other hematological malignancies. However, research still continues to avoid some problems such as toxicities associated with the treatment and to find strategies to avoid tumor cell immune evasion mechanisms. On the other hand, for solid tumors, CAR-T therapy results are still in an early phase. In contrast to hematological malignancies, the complex tumor heterogeneity of solid tumors has led to the research of novel and challenging strategies to improve CAR-T cell activity. Here, we will review the main clinical results obtained with CAR-T cells in hematological malignancies, specifically focusing on CAR-T-19 and CAR-T against B-cell maturation antigen (CAR-T-BCMA). Moreover, we will mention the main problems that decrease CAR-T cell activity in solid tumors and the strategies to overcome them. Finally, we will present some of the first clinical results obtained for solid tumors.
Collapse
Affiliation(s)
- Lorena Perez-Amill
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Hematology, Barcelona, Spain,Lorena Perez-Amill and Berta Marzal contributed to this article equally
| | - Berta Marzal
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Immunology, Barcelona, Spain,Lorena Perez-Amill and Berta Marzal contributed to this article equally
| | - Alvaro Urbano-Ispizua
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Hematology, Barcelona, Spain,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Manel Juan
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Immunology, Barcelona, Spain
| | - Beatriz Martín-Antonio
- Institut d’Investigacions Biomèdiques August Pi i Sunyer Hospital, Clinic of Hematology, Barcelona, Spain,Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| |
Collapse
|
143
|
Si W, Li C, Wei P. Synthetic immunology: T-cell engineering and adoptive immunotherapy. Synth Syst Biotechnol 2018; 3:179-185. [PMID: 30345403 PMCID: PMC6190530 DOI: 10.1016/j.synbio.2018.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/28/2018] [Accepted: 08/13/2018] [Indexed: 12/24/2022] Open
Abstract
During the past decades, the rapidly-evolving cancer is hard to be thoroughly eliminated even though the radiotherapy and chemotherapy do exhibit efficacy in some degree. However, a breakthrough appeared when the adoptive cancer therapy [1] was developed, especially T cells armed with chimeric antigen receptors (CARs) showed great potential in tumor clinical trials recently. CAR-T cells successfully elevated the efficiency and specificity of cytotoxicity. In this review, we will talk about the design of CAR and CAR-included combinatory therapeutic applications in the principles of systems and synthetic immunology.
Collapse
Affiliation(s)
- Wen Si
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Cheng Li
- The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| | - Ping Wei
- Center for Quantitative Biology and Peking-Tsinghua Joint Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.,The MOE Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
144
|
Hong LK, Chen Y, Smith CC, Montgomery SA, Vincent BG, Dotti G, Savoldo B. CD30-Redirected Chimeric Antigen Receptor T Cells Target CD30 + and CD30 - Embryonal Carcinoma via Antigen-Dependent and Fas/FasL Interactions. Cancer Immunol Res 2018; 6:1274-1287. [PMID: 30087115 DOI: 10.1158/2326-6066.cir-18-0065] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 06/05/2018] [Accepted: 08/03/2018] [Indexed: 01/08/2023]
Abstract
Tumor antigen heterogeneity limits success of chimeric antigen receptor (CAR) T-cell therapies. Embryonal carcinomas (EC) and mixed testicular germ cell tumors (TGCT) containing EC, which are the most aggressive TGCT subtypes, are useful for dissecting this issue as ECs express the CD30 antigen but also contain CD30-/dim cells. We found that CD30-redirected CAR T cells (CD30.CAR T cells) exhibit antitumor activity in vitro against the human EC cell lines Tera-1, Tera-2, and NCCIT and putative EC stem cells identified by Hoechst dye staining. Cytolytic activity of CD30.CAR T cells was complemented by their sustained proliferation and proinflammatory cytokine production. CD30.CAR T cells also demonstrated antitumor activity in an in vivo xenograft NOD/SCID/γcnull (NSG) mouse model of metastatic EC. We observed that CD30.CAR T cells, while targeting CD30+ EC tumor cells through the CAR (i.e., antigen-dependent targeting), also eliminated surrounding CD30- EC cells in an antigen-independent manner, via a cell-cell contact-dependent Fas/FasL interaction. In addition, ectopic Fas (CD95) expression in CD30+ Fas- EC was sufficient to improve CD30.CAR T-cell antitumor activity. Overall, these data suggest that CD30.CAR T cells might be useful as an immunotherapy for ECs. Additionally, Fas/FasL interaction between tumor cells and CAR T cells can be exploited to reduce tumor escape due to heterogeneous antigen expression or to improve CAR T-cell antitumor activity. Cancer Immunol Res; 6(10); 1274-87. ©2018 AACR.
Collapse
Affiliation(s)
- Lee K Hong
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yuhui Chen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christof C Smith
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Gianpietro Dotti
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. .,Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
145
|
Abstract
PURPOSE OF REVIEW This review will discuss the challenges facing adoptive cell techniques in the treatment of solid tumors and examine the therapies that are in development for specifically pediatric solid tumors. RECENT FINDINGS Targeting solid tumors with adoptive cell therapy has been limited by the inhibitory tumor microenvironment and heterogeneous expression of targetable antigens. Many creative strategies to overcome these limitations are being developed but still need to be tested clinically. Early phase clinical trials in neuroblastoma with GD2 CAR T cells are promising but results need to be validated on a larger scale. Most research in other pediatric solid tumors is still in early stages. Adoptive cell therapy represents a useful tool to improve the outcomes of many pediatric solid tumors but significant study is still required. Several clinical trials are ongoing to test therapies that have shown promise in the lab.
Collapse
|
146
|
Knochelmann HM, Smith AS, Dwyer CJ, Wyatt MM, Mehrotra S, Paulos CM. CAR T Cells in Solid Tumors: Blueprints for Building Effective Therapies. Front Immunol 2018; 9:1740. [PMID: 30140266 PMCID: PMC6094980 DOI: 10.3389/fimmu.2018.01740] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/13/2018] [Indexed: 01/06/2023] Open
Abstract
Genetic redirection of T lymphocytes with chimeric antigen receptors (CARs) has soared from treating cancers preclinically to FDA approval for hematologic malignancies and commercial-grade production scale in under 30 years. To date, solid tumors are less susceptible to CAR therapies and instead have been treated more successfully with immune checkpoint blockade or tumor-infiltrating lymphocyte therapy. Here, we discuss the current challenges in treating solid tumors with CAR T cells, and the obstacles within the host and tumor microenvironment hindering their efficacy. We present a novel three-pronged approach for enhancing the efficacy of CAR T cells whereby a single infusion product can synergize the power of an optimal CAR construct, a highly potent T cell subset, and rejuvenate the endogenous immune response to conquer therapeutically-resistant solid tumors.
Collapse
Affiliation(s)
- Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Megan M Wyatt
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Shikhar Mehrotra
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Chrystal M Paulos
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Dermatology and Dermatologic Surgery, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
147
|
Kholodenko IV, Kalinovsky DV, Doronin II, Deyev SM, Kholodenko RV. Neuroblastoma Origin and Therapeutic Targets for Immunotherapy. J Immunol Res 2018; 2018:7394268. [PMID: 30116755 PMCID: PMC6079467 DOI: 10.1155/2018/7394268] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/27/2018] [Indexed: 01/30/2023] Open
Abstract
Neuroblastoma is a pediatric solid cancer of heterogeneous clinical behavior. The unique features of this type of cancer frequently hamper the process of determining clinical presentation and predicting therapy effectiveness. The tumor can spontaneously regress without treatment or actively develop and give rise to metastases despite aggressive multimodal therapy. In recent years, immunotherapy has become one of the most promising approaches to the treatment of neuroblastoma. Still, only one drug for targeted immunotherapy of neuroblastoma, chimeric monoclonal GD2-specific antibodies, is used in the clinic today, and its application has significant limitations. In this regard, the development of effective and safe GD2-targeted immunotherapies and analysis of other potential molecular targets for the treatment of neuroblastoma represents an important and topical task. The review summarizes biological characteristics of the origin and development of neuroblastoma and outlines molecular markers of neuroblastoma and modern immunotherapy approaches directed towards these markers.
Collapse
Affiliation(s)
- Irina V. Kholodenko
- Orekhovich Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow 119121, Russia
| | - Daniel V. Kalinovsky
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Igor I. Doronin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Real Target LLC, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| | - Sergey M. Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Institute of Engineering Physics for Biomedicine (PhysBio), National Research Nuclear University “MEPhI”, Moscow 115409, Russia
| | - Roman V. Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
- Real Target LLC, 16/10 Miklukho-Maklaya St., Moscow 117997, Russia
| |
Collapse
|
148
|
Sahin A, Sanchez C, Bullain S, Waterman P, Weissleder R, Carter BS. Development of third generation anti-EGFRvIII chimeric T cells and EGFRvIII-expressing artificial antigen presenting cells for adoptive cell therapy for glioma. PLoS One 2018; 13:e0199414. [PMID: 29975720 PMCID: PMC6033533 DOI: 10.1371/journal.pone.0199414] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 05/13/2018] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive and deadly form of adult brain cancer. Despite of many attempts to identify potential therapies for this disease, including promising cancer immunotherapy approaches, it remains incurable. To address the need of improved persistence, expansion, and optimal antitumor activity of T-cells in the glioma milieu, we have developed an EGFRvIII-specific third generation (G3-EGFRvIII) chimeric antigen receptor (CAR) that expresses both co-stimulatory factors CD28 and OX40 (MR1-CD8TM-CD28-OX40-CD3ζ). To enhance ex vivo target specific activation and optimize T-cell culturing conditions, we generated artificial antigen presenting cell lines (aAPC) expressing the extracellular and transmembrane domain of EGFRvIII (EGFRVIIIΔ654) with costimulatory molecules including CD32, CD80 and 4-1BBL (EGFRVIIIΔ654 aAPC and CD32-80-137L-EGFRVIIIΔ654 aAPC). We demonstrate that the highest cell growth was achieved when G3-EGFRvIII CAR T-cells were cocultured with both co-stimulatory aAPCs and with exposure to EGFRvIII (CD32-80-137L-EGFRVIIIΔ654 aAPCs) in culturing periods of three to six weeks. G3-EGFRvIII CAR T-cells showed an increased level of IFN-γ when cocultured with CD32-80-137L-EGFRVIIIΔ654 aAPCs. Evaluation of G3-EGFRvIII CAR T-cells in an orthotropic human glioma xenograft model demonstrated a prolonged survival of G3-EGFRvIII CAR treated mice compared to control mice. Importantly, we observed survival of G3-EGFRvIII CAR T-cells within the tumor as long as 90 days after implantation in low-dose and single administration, accompanied by a marked tumor stroma demolition. These findings suggest that G3-EGFRvIII CAR cocultured with CD32-80-137L-EGFRVIIIΔ654 aAPCs warrants itself as a potential anti-tumor therapy strategy for glioblastoma.
Collapse
Affiliation(s)
- Ayguen Sahin
- HMS-MGH Center for Nervous System Repair, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, United States of America
- * E-mail:
| | - Carlos Sanchez
- HMS-MGH Center for Nervous System Repair, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, United States of America
| | - Szofia Bullain
- HMS-MGH Center for Nervous System Repair, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, United States of America
| | - Peter Waterman
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, United States of America
| | - Bob S. Carter
- HMS-MGH Center for Nervous System Repair, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, United States of America
| |
Collapse
|
149
|
Labanieh L, Majzner RG, Mackall CL. Programming CAR-T cells to kill cancer. Nat Biomed Eng 2018; 2:377-391. [PMID: 31011197 DOI: 10.1038/s41551-018-0235-9] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/09/2018] [Indexed: 02/06/2023]
Abstract
T cells engineered to express chimeric antigen receptors (CARs) that are specific for tumour antigens have led to high complete response rates in patients with haematologic malignancies. Despite this early success, major challenges to the broad application of CAR-T cells as cancer therapies remain, including treatment-associated toxicities and cancer relapse with antigen-negative tumours. Targeting solid tumours with CAR-T cells poses additional obstacles because of the paucity of tumour-specific antigens and the immunosuppressive effects of the tumour microenvironment. To overcome these challenges, T cells can be programmed with genetic modules that increase their therapeutic potency and specificity. In this Review Article, we survey major advances in the engineering of next-generation CAR-T therapies for haematologic cancers and solid cancers, with particular emphasis on strategies for the control of CAR specificity and activity and on approaches for improving CAR-T-cell persistence and overcoming immunosuppression. We also lay out a roadmap for the development of off-the-shelf CAR-T cells.
Collapse
Affiliation(s)
- Louai Labanieh
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Robbie G Majzner
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Crystal L Mackall
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA. .,Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
150
|
Bishop DC, Xu N, Tse B, O'Brien TA, Gottlieb DJ, Dolnikov A, Micklethwaite KP. PiggyBac-Engineered T Cells Expressing CD19-Specific CARs that Lack IgG1 Fc Spacers Have Potent Activity against B-ALL Xenografts. Mol Ther 2018; 26:1883-1895. [PMID: 29861327 DOI: 10.1016/j.ymthe.2018.05.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 05/04/2018] [Accepted: 05/08/2018] [Indexed: 10/14/2022] Open
Abstract
Clinical trials of CD19-specific chimeric antigen receptor (CAR19) T cells have demonstrated remarkable efficacy against relapsed and refractory B cell malignancies. The piggyBac transposon system offers a less complex and more economical means for generating CAR19 T cells compared to viral vectors. We have previously optimized a protocol for the generation of CAR19 T cells using the piggyBac system, but we found that CAR19 T cells had poor in vivo efficacy and persistence, probably due to deleterious FcγR interactions with the CAR's IgG1 Fc-containing spacer domain. We therefore designed three CD19-specifc CARs that lacked the IgG1 Fc region, and we incorporated combinations of CD28 or 4-1BB transmembrane and co-stimulatory domains. PiggyBac-generated CAR19 T cells expressing these re-designed constructs all demonstrated reactivity in vitro specifically against CD19+ cell lines. However, those combining CD28 transmembrane and co-stimulatory domains showed CD4 predominance and inferior cytotoxicity. At high doses, CAR19 T cells were effective against B-ALL in a xenograft mouse model, regardless of co-stimulatory domain. At diminishing doses, 4-1BB co-stimulation led to greater potency and persistence of CAR19 T cells, and it provided protection against B-ALL re-challenge. Production of potent CAR T cells using piggyBac is simple and cost-effective, and it may enable wider access to CAR T cell therapy.
Collapse
Affiliation(s)
- David C Bishop
- Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Haematology, Westmead Hospital, Sydney, NSW, Australia; Blood and Bone Marrow Transplant Unit, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Ning Xu
- Blood & Marrow Transplant Facility, Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW, Australia; Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia
| | - Benjamin Tse
- Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Tracey A O'Brien
- Blood & Marrow Transplant Facility, Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW, Australia; Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - David J Gottlieb
- Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Haematology, Westmead Hospital, Sydney, NSW, Australia; Blood and Bone Marrow Transplant Unit, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Sydney Cellular Therapies Laboratory, Westmead Hospital, Sydney, NSW, Australia; Department of Medicine, Westmead Hospital, Sydney, NSW, Australia
| | - Alla Dolnikov
- Blood & Marrow Transplant Facility, Kids Cancer Centre, Sydney Children's Hospital, Sydney, NSW, Australia; Children's Cancer Institute, University of New South Wales, Sydney, NSW, Australia; Faculty of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Kenneth P Micklethwaite
- Westmead Institute for Medical Research, Sydney, NSW, Australia; Department of Haematology, Westmead Hospital, Sydney, NSW, Australia; Blood and Bone Marrow Transplant Unit, Westmead Hospital, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Sydney Cellular Therapies Laboratory, Westmead Hospital, Sydney, NSW, Australia.
| |
Collapse
|