51
|
Globerson Levin A, Rivière I, Eshhar Z, Sadelain M. CAR T cells: Building on the CD19 paradigm. Eur J Immunol 2021; 51:2151-2163. [PMID: 34196410 DOI: 10.1002/eji.202049064] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022]
Abstract
Spearheaded by the therapeutic use of chimeric antigen receptors (CARs) targeting CD19, synthetic immunology has entered the clinical arena. CARs are recombinant receptors for antigen that engage cell surface molecules through the variable region of an antibody and signal through arrayed T-cell activating and costimulatory domains. CARs allow redirection of T-cell cytotoxicity against any antigen of choice, independent of MHC expression. Patient T cells engineered to express CARs specific for CD19 have yielded remarkable outcomes in subjects with relapsed/refractory B- cell malignancies, setting off unprecedented interest in T-cell engineering and cell-based cancer immunotherapy. In this review, we present the challenges to extend the use of CAR T cells to solid tumors and other pathologies. We further highlight progress in CAR design, cell manufacturing, and genome editing, which in aggregate hold the promise of generating safer and more effective genetically instructed immunity. Novel engineered cell types, including innate T-cell types, natural killer (NK) cells, macrophages, and induced pluripotent stem cell-derived immune cells, are on the horizon, as are applications of CAR T cells to treat autoimmunity, severe infections, and senescence-associated pathologies.
Collapse
Affiliation(s)
| | - Isabelle Rivière
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zelig Eshhar
- Immunology Lab, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
52
|
Abstract
A paradigm shift has recently occurred in the field of cancer therapeutics. Traditional anticancer agents, such as chemotherapy, radiotherapy and small-molecule drugs targeting specific signalling pathways, have been joined by cellular immunotherapies based on T cell engineering. The rapid adoption of novel, patient-specific cellular therapies builds on scientific developments in tumour immunology, genetic engineering and cell manufacturing, best illustrated by the curative potential of chimeric antigen receptor (CAR) T cell therapy targeting CD19-expressing malignancies. However, the clinical benefit observed in many patients may come at a cost. In up to one-third of patients, significant toxicities occur that are directly associated with the induction of powerful immune effector responses. The most frequently observed immune-mediated toxicities are cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome. This Review discusses our current understanding of their pathophysiology and clinical features, as well as the development of novel therapeutics for their prevention and/or management. This Review discusses our current understanding of the pathophysiological mechanisms of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome associated with chimeric antigen receptor (CAR) T cell therapies, and how this might be used for the prevention or management of these toxicities.
Collapse
|
53
|
Hawkins ER, D'Souza RR, Klampatsa A. Armored CAR T-Cells: The Next Chapter in T-Cell Cancer Immunotherapy. Biologics 2021; 15:95-105. [PMID: 33883875 PMCID: PMC8053711 DOI: 10.2147/btt.s291768] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy engineers T-cells to express a synthetic receptor which redirects effector function to the tumor, to improve efficacy and reduce toxicities associated with conventional treatments, such as radiotherapy and chemotherapy. This approach has proved effective in treating hematological malignancies; however, the same effects have not been observed in solid tumors. The immunosuppressive tumor microenvironment (TME) creates a significant barrier to solid tumor efficacy and reduces the anti-cancer activity of endogenous tumor-resident immune cells, enabling cancer progression. In recent years, researchers have attempted to enhance CAR T-cell function in the TME by engineering the cells to express various proteins alongside the CAR. Examples of this engineering include inducing CAR T-cells to secrete cytokines or express cytokine receptors to modulate the cytokine milieu of the TME. Alternatively, the CAR T-cell may secrete antibody-like proteins to target a range of tumor antigens. Collectively, these methods are termed armored CAR T-cell therapy, and in this review, we will discuss the range of armored CAR T-cell approaches which have been investigated to date.
Collapse
Affiliation(s)
- Elizabeth R Hawkins
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Reena R D'Souza
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Astero Klampatsa
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| |
Collapse
|
54
|
Kumar J, Kumar R, Kumar Singh A, Tsakem EL, Kathania M, Riese MJ, Theiss AL, Davila ML, Venuprasad K. Deletion of Cbl-b inhibits CD8 + T-cell exhaustion and promotes CAR T-cell function. J Immunother Cancer 2021; 9:jitc-2020-001688. [PMID: 33462140 PMCID: PMC7813298 DOI: 10.1136/jitc-2020-001688] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 12/22/2022] Open
Abstract
Background Chimeric antigen receptor (CAR) T-cell therapy is an emerging option for cancer treatment, but its efficacy is limited, especially in solid tumors. This is partly because the CAR T cells become dysfunctional and exhausted in the tumor microenvironment. However, the key pathways responsible for impaired function of exhausted cells remain unclear, which is essential to overcome CAR T-cell exhaustion. Methods Analysis of RNA-sequencing data from CD8+ tumor-infiltrating lymphocytes (TILs) led to identification of Cbl-b as a potential target. The sequencing data were validated using a syngeneic MC38 colon cancer model. To analyze the in vivo role of Cbl-b in T-cell exhaustion, tumor growth, % PD1+Tim3+ cells, and expression of effector cytokines were analyzed in cbl-b+/+ and cbl-b–/– mice. To evaluate the therapeutic potential of Cbl-b depletion, we generated a new CAR construct, hCEAscFv-CD28-CD3ζ.GFP, that recognizes human carcinoembryonic antigen (CEA). cbl-b+/+ and cbl-b–/– CEA-CAR T cells were generated by retroviral transduction. Rag–/– mice bearing MC38-CEA cells were injected with cbl-b+/+ and cbl-b–/–; CEA-CAR T cells, tumor growth, % PD1+Tim3+ cells and expression of effector cytokines were analyzed. Results Our results show that the E3 ubiquitin ligase Cbl-b is upregulated in exhausted (PD1+Tim3+) CD8+ TILs. CRISPR-Cas9-mediated inhibition of Cbl-b restores the effector function of exhausted CD8+ TILs. Importantly, the reduced growth of syngeneic MC38 tumors in cbl-b–/– mice was associated with a marked reduction of PD1+Tim3+ CD8+ TILs. Depletion of Cbl-b inhibited CAR T-cell exhaustion, resulting in reduced MC38-CEA tumor growth, reduced PD1+Tim3+ cells and increased expression of interferon gamma, tumor necrosis factor alpha, and increased tumor cell killing. Conclusion Our studies demonstrate that deficiency of Cbl-b overcomes endogenous CD8+ T-cell exhaustion, and deletion of Cbl-b in CAR T cells renders them resistant to exhaustion. Our results could facilitate the development of efficient CAR T-cell therapy for solid tumors by targeting Cbl-b.
Collapse
Affiliation(s)
- Jitendra Kumar
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Immunology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ritesh Kumar
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Immunology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Amir Kumar Singh
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Immunology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Elviche L Tsakem
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Immunology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Mahesh Kathania
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA.,Immunology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew J Riese
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Arianne L Theiss
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Marco L Davila
- Blood & Marrow Transplantation and Cellular Immunotherapy, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - K Venuprasad
- Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA .,Immunology, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
55
|
Boucher JC, Li G, Kotani H, Cabral ML, Morrissey D, Lee SB, Spitler K, Beatty NJ, Cervantes EV, Shrestha B, Yu B, Kazi A, Wang X, Sebti SM, Davila ML. CD28 Costimulatory Domain-Targeted Mutations Enhance Chimeric Antigen Receptor T-cell Function. Cancer Immunol Res 2021; 9:62-74. [PMID: 33188139 PMCID: PMC7864379 DOI: 10.1158/2326-6066.cir-20-0253] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 08/23/2020] [Accepted: 11/04/2020] [Indexed: 11/16/2022]
Abstract
An obstacle to the development of chimeric antigen receptor (CAR) T cells is the limited understanding of CAR T-cell biology and the mechanisms behind their antitumor activity. We and others have shown that CARs with a CD28 costimulatory domain drive high T-cell activation, which leads to exhaustion and shortened persistence. This work led us to hypothesize that by incorporating null mutations of CD28 subdomains (YMNM, PRRP, or PYAP), we could optimize CAR T-cell costimulation and enhance function. In vivo, we found that mice given CAR T cells with only a PYAP CD28 endodomain had a significant survival advantage, with 100% of mice alive after 62 days compared with 50% for mice with an unmutated endodomain. We observed that mutant CAR T cells remained more sensitive to antigen after ex vivo antigen and PD-L1 stimulation, as demonstrated by increased cytokine production. The mutant CAR T cells also had a reduction of exhaustion-related transcription factors and genes such as Nfatc1, Nr42a, and Pdcd1 Our results demonstrated that CAR T cells with a mutant CD28 endodomain have better survival and function. This work allows for the development of enhanced CAR T-cell therapies by optimizing CAR T-cell costimulation.
Collapse
Affiliation(s)
- Justin C Boucher
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Gongbo Li
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Hiroshi Kotani
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Maria L Cabral
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida
| | - Dylan Morrissey
- Morsani College of Medicine, University of South Florida Health, Tampa, Florida
| | - Sae Bom Lee
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida
| | - Kristen Spitler
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Nolan J Beatty
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, Florida
| | - Estelle V Cervantes
- Morsani College of Medicine, University of South Florida Health, Tampa, Florida
| | - Bishwas Shrestha
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Bin Yu
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Aslamuzzaman Kazi
- Drug Discovery Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Said M Sebti
- Drug Discovery Program, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Marco L Davila
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, Division of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, Florida.
- Morsani College of Medicine, University of South Florida Health, Tampa, Florida
| |
Collapse
|
56
|
Abstract
PURPOSE OF REVIEW Tremendous advances have been made in the treatment armamentarium for acute lymphoblastic leukemia in recent years, which have substantially improved outcomes for these patients. At the same time, unique toxicities have emerged, and without early intervention, are life-threatening. This article will review the novel therapies in acute leukemias and highlight the clinically relevant supportive care advances. RECENT FINDINGS The American Society for Transplantation and Cellular Therapy (ASTCT) has put forth the most recent recommendations in managing the cytokine release syndrome and neurotoxicity after chimeric antigen receptor T cells (CAR-T) and blinatumomab. The hepatic injury incurred by inotuzumab, and the vascular toxicity of tyrosine kinase inhibitors, other relatively novel agents, require subspecialist intervention and multidisciplinary care. Asparaginase, a long-established and key element of pediatric regimens, has made a comeback in the young adult leukemia population. Updated guidelines have been outlined for management of asparaginase thrombotic complications. Lastly, although there have been few changes in the applications of growth factor, antimicrobial prophylaxis, and management of neuropathy, these encompass exceedingly important aspects of care. While the rapidly changing treatment paradigms for acute lymphoblastic leukemia have transformed leukemia-specific outcomes, treatment emergent toxicities have forced much necessary attention to better definitions of these toxicities and on improving supportive care guidelines in acute lymphoblastic leukemia.
Collapse
|
57
|
Kuhn NF, Lopez AV, Li X, Cai W, Daniyan AF, Brentjens RJ. CD103 + cDC1 and endogenous CD8 + T cells are necessary for improved CD40L-overexpressing CAR T cell antitumor function. Nat Commun 2020; 11:6171. [PMID: 33268774 PMCID: PMC7710757 DOI: 10.1038/s41467-020-19833-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/23/2020] [Indexed: 01/05/2023] Open
Abstract
While effective in specific settings, adoptive chimeric antigen receptor (CAR) T cell therapy for cancer requires further improvement and optimization. Our previous results show that CD40L-overexpressing CAR T cells mobilize endogenous immune effectors, resulting in improved antitumor immunity. However, the cell populations required for this protective effect remain to be identified. Here we show, by analyzing Batf3-/- mice lacking the CD103+ conventional dendritic cell type 1 (cDC1) subpopulation important for antigen cross-presentation, that CD40L-overexpressing CAR T cells elicit an impaired antitumor response in the absence of cDC1s. We further find that CD40L-overexpressing CAR T cells stimulate tumor-resident CD11b-CD103- double-negative (DN) cDCs to proliferate and differentiate into cDC1s in wild-type mice. Finally, re-challenge experiments show that endogenous CD8+ T cells are required for protective antitumor memory in this setting. Our findings thus demonstrate the stimulatory effect of CD40L-overexpressing CAR T cells on innate and adaptive immune cells, and provide a rationale for using CD40L-overexpressing CAR T cells to improve immunotherapy responses.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Antigen Presentation
- Antigens, CD/genetics
- Antigens, CD/immunology
- Basic-Leucine Zipper Transcription Factors/deficiency
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/immunology
- CD11b Antigen/deficiency
- CD11b Antigen/genetics
- CD11b Antigen/immunology
- CD40 Ligand/genetics
- CD40 Ligand/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Female
- Gene Expression
- Immunity, Innate
- Immunophenotyping
- Immunotherapy, Adoptive/methods
- Integrin alpha Chains/deficiency
- Integrin alpha Chains/genetics
- Integrin alpha Chains/immunology
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/pathology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasm Transplantation
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Repressor Proteins/deficiency
- Repressor Proteins/genetics
- Repressor Proteins/immunology
Collapse
Affiliation(s)
- Nicholas F Kuhn
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Andrea V Lopez
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xinghuo Li
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Winson Cai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anthony F Daniyan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
58
|
Parayath NN, Stephan SB, Koehne AL, Nelson PS, Stephan MT. In vitro-transcribed antigen receptor mRNA nanocarriers for transient expression in circulating T cells in vivo. Nat Commun 2020; 11:6080. [PMID: 33247092 PMCID: PMC7695830 DOI: 10.1038/s41467-020-19486-2] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
Engineering chimeric antigen receptors (CAR) or T cell receptors (TCR) helps create disease-specific T cells for targeted therapy, but the cost and rigor associated with manufacturing engineered T cells ex vivo can be prohibitive, so programing T cells in vivo may be a viable alternative. Here we report an injectable nanocarrier that delivers in vitro-transcribed (IVT) CAR or TCR mRNA for transiently reprograming of circulating T cells to recognize disease-relevant antigens. In mouse models of human leukemia, prostate cancer and hepatitis B-induced hepatocellular carcinoma, repeated infusions of these polymer nanocarriers induce sufficient host T cells expressing tumor-specific CARs or virus-specific TCRs to cause disease regression at levels similar to bolus infusions of ex vivo engineered lymphocytes. Given their ease of manufacturing, distribution and administration, these nanocarriers, and the associated platforms, could become a therapeutic for a wide range of diseases. Ex vivo engineering of antigen-specific T cells has shown therapeutic efficacy but can be costly and scarce. Here the authors show that in vitro-transcribed antigen receptor mRNA packaged in nanocarriers can directly induce, in vivo, transient their expression in circulating T cells to provide therapeutic effects in mouse models of cancer or viral infection.
Collapse
Affiliation(s)
- N N Parayath
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - S B Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - A L Koehne
- Translational Pathology, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
| | - P S Nelson
- Division of Human Biology and Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - M T Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA. .,Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, 98195, USA. .,Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
59
|
Lanitis E, Rota G, Kosti P, Ronet C, Spill A, Seijo B, Romero P, Dangaj D, Coukos G, Irving M. Optimized gene engineering of murine CAR-T cells reveals the beneficial effects of IL-15 coexpression. J Exp Med 2020; 218:211522. [PMID: 33156338 PMCID: PMC7653685 DOI: 10.1084/jem.20192203] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 07/24/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Limited clinical benefit has been demonstrated for chimeric antigen receptor (CAR) therapy of solid tumors, but coengineering strategies to generate so-called fourth-generation (4G) CAR-T cells are advancing toward overcoming barriers in the tumor microenvironment (TME) for improved responses. In large part due to technical challenges, there are relatively few preclinical CAR therapy studies in immunocompetent, syngeneic tumor-bearing mice. Here, we describe optimized methods for the efficient retroviral transduction and expansion of murine T lymphocytes of a predominantly central memory T cell (TCM cell) phenotype. We present a bicistronic retroviral vector encoding both a tumor vasculature–targeted CAR and murine interleukin-15 (mIL-15), conferring enhanced effector functions, engraftment, tumor control, and TME reprogramming, including NK cell activation and reduced presence of M2 macrophages. The 4G-CAR-T cells coexpressing mIL-15 were further characterized by up-regulation of the antiapoptotic marker Bcl-2 and lower cell-surface expression of the inhibitory receptor PD-1. Overall, this work introduces robust tools for the development and evaluation of 4G-CAR-T cells in immunocompetent mice, an important step toward the acceleration of effective therapies reaching the clinic.
Collapse
Affiliation(s)
- Evripidis Lanitis
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giorgia Rota
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Paris Kosti
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Catherine Ronet
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Bili Seijo
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pedro Romero
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Denarda Dangaj
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
60
|
Grote S, Mittelstaet J, Baden C, Chan KCH, Seitz C, Schlegel P, Kaiser A, Handgretinger R, Schleicher S. Adapter chimeric antigen receptor (AdCAR)-engineered NK-92 cells: an off-the-shelf cellular therapeutic for universal tumor targeting. Oncoimmunology 2020; 9:1825177. [PMID: 33457105 PMCID: PMC7781805 DOI: 10.1080/2162402x.2020.1825177] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite the recent success of CAR T cells targeting CD19 and CD22 in hematological malignancies, the production of CAR T cells still requires an extensive manufacturing process. The well-established NK-92 cell line provides a promising alternative to produce CAR-modified effector cells in a GMP-compliant, cost-effective way. NK-92 can be redirected against a variety of surface antigens by our adapter CAR (AdCAR) system utilizing biotinylated antibodies (bAb) as adapter molecules. Selected bAb were capable of inducing significant AdCAR NK-92-mediated lysis of non-Hodgkin lymphoma (NHL) and mantle-cell lymphoma (MCL) cell lines as well as primary MCL and chronic lymphocytic leukemia (CLL) cells. AdCAR specificity was proven using a JeKo-1 CD19/CD20 knockout antigen-loss model. Moreover, through combinations of bAb, AdCAR NK-92 cells are capable of combatting tumor antigen evasion mechanisms. In conclusion, we successfully generated the AdCAR NK-92 cell line which can be manufactured as an “off-the-shelf, on-demand” product allowing universal and tunable tumor targeting.
Collapse
Affiliation(s)
- Stefan Grote
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| | | | - Caroline Baden
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| | - Kenneth Chun-Ho Chan
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| | - Christian Seitz
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| | - Patrick Schlegel
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| | | | - Rupert Handgretinger
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| | - Sabine Schleicher
- Department of Hematology and Oncology, University Hospital Tuebingen, Children's Hospital, Tuebingen, Germany
| |
Collapse
|
61
|
Burchett R, Walsh S, Wan Y, Bramson JL. A rational relationship: Oncolytic virus vaccines as functional partners for adoptive T cell therapy. Cytokine Growth Factor Rev 2020; 56:149-159. [PMID: 32665126 DOI: 10.1016/j.cytogfr.2020.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 01/02/2023]
Abstract
Tumours employ a variety of immune-evasion and suppression mechanisms to impair development of functional tumor-specific T cells and subvert T cell-mediated immunity in the tumour microenvironment. Adoptive T cell therapy (ACT) aims to overcome these barriers and overwhelm tumor defenses with a bolus of T cells that were selectively expanded ex vivo. Although this strategy has been effective in liquid tumors and melanomas, many tumors appear to be resistant to ACT. Several factors are thought to play into this resistance, including poor engraftment and persistence of transferred cells, tumour cell heterogeneity and antigen loss, poor immune cell recruitment and infiltration into the tumour, and susceptibility to local immunosuppression in the tumor microenvironment. Oncolytic viruses (OV) have been identified as powerful stimulators of the anti-tumour immune response. As such, OVs are inherently well-positioned to act in synergy with ACT to bolster the anti-tumour T cell response. Further, OV vaccines, wherein tumour-associated antigens are encoded into the viral backbone, have proven to be remarkable in boosting antigen-specific T cell response. Pre-clinical studies have revealed remarkable therapeutic outcomes when OV vaccines are paired with ACT. In this scenario, OV vaccines are thought to function in a "push and pull" manner, where push refers to expanding T cells in the periphery and pull refers to recruiting those cells into the tumour that has been rendered amenable to T cell attack by the actions of the OV. In this review, we discuss barriers that limit eradication of tumors by T cells, highlight attributes of OVs that break down these barriers and present strategies for rational combinations of ACT with OV vaccines.
Collapse
Affiliation(s)
- Rebecca Burchett
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Scott Walsh
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Yonghong Wan
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan L Bramson
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
62
|
Amor C, Feucht J, Leibold J, Ho YJ, Zhu C, Alonso-Curbelo D, Mansilla-Soto J, Boyer JA, Li X, Giavridis T, Kulick A, Houlihan S, Peerschke E, Friedman SL, Ponomarev V, Piersigilli A, Sadelain M, Lowe SW. Senolytic CAR T cells reverse senescence-associated pathologies. Nature 2020; 583:127-132. [PMID: 32555459 PMCID: PMC7583560 DOI: 10.1038/s41586-020-2403-9] [Citation(s) in RCA: 651] [Impact Index Per Article: 130.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 05/06/2020] [Indexed: 12/21/2022]
Abstract
Cellular senescence is characterized by stable cell-cycle arrest and a secretory program that modulates the tissue microenvironment1,2. Physiologically, senescence serves as a tumour-suppressive mechanism that prevents the expansion of premalignant cells3,4 and has a beneficial role in wound-healing responses5,6. Pathologically, the aberrant accumulation of senescent cells generates an inflammatory milieu that leads to chronic tissue damage and contributes to diseases such as liver and lung fibrosis, atherosclerosis, diabetes and osteoarthritis1,7. Accordingly, eliminating senescent cells from damaged tissues in mice ameliorates the symptoms of these pathologies and even promotes longevity1,2,8-10. Here we test the therapeutic concept that chimeric antigen receptor (CAR) T cells that target senescent cells can be effective senolytic agents. We identify the urokinase-type plasminogen activator receptor (uPAR)11 as a cell-surface protein that is broadly induced during senescence and show that uPAR-specific CAR T cells efficiently ablate senescent cells in vitro and in vivo. CAR T cells that target uPAR extend the survival of mice with lung adenocarcinoma that are treated with a senescence-inducing combination of drugs, and restore tissue homeostasis in mice in which liver fibrosis is induced chemically or by diet. These results establish the therapeutic potential of senolytic CAR T cells for senescence-associated diseases.
Collapse
Affiliation(s)
- Corina Amor
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Judith Feucht
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Josef Leibold
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu-Jui Ho
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Changyu Zhu
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Direna Alonso-Curbelo
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge Mansilla-Soto
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jacob A Boyer
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xiang Li
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Theodoros Giavridis
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amanda Kulick
- Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shauna Houlihan
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ellinor Peerschke
- Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vladimir Ponomarev
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alessandra Piersigilli
- Laboratory of Comparative Pathology, Rockefeller University, Weill Cornell Medicine and Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Scott W Lowe
- Department of Cancer Biology and Genetics, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
63
|
Lv C, Chen S, Hu F, Huang D, Wang T, Du J, Wang J, Wu H. Pluripotent stem cell-derived CD19-CAR iT cells effectively eradicate B-cell lymphoma in vivo. Cell Mol Immunol 2020; 18:773-775. [PMID: 32541834 DOI: 10.1038/s41423-020-0429-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 01/24/2023] Open
Affiliation(s)
- Cui Lv
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shoubing Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Fangxiao Hu
- School of Life Sciences, University of Science and Technology of China, Hefei, China.,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Dehao Huang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Tongjie Wang
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Juan Du
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangdong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Jinyong Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, China. .,CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangdong, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| | - Hongling Wu
- CAS Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stem cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China. .,Guangzhou Regenerative Medicine and Health-Guangdong Laboratory (GRMH-GDL), Guangdong, China.
| |
Collapse
|
64
|
Wijewarnasuriya D, Bebernitz C, Lopez AV, Rafiq S, Brentjens RJ. Excessive Costimulation Leads to Dysfunction of Adoptively Transferred T Cells. Cancer Immunol Res 2020; 8:732-742. [PMID: 32213625 PMCID: PMC7269815 DOI: 10.1158/2326-6066.cir-19-0908] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/12/2020] [Accepted: 03/19/2020] [Indexed: 12/21/2022]
Abstract
Although clinical responses with CD19-targeting chimeric antigen receptor (CAR) T-cell treatment have been observed in patients with certain hematologic malignancies, high rates of disease relapse highlight the necessity to understand and improve mechanisms of CAR T-cell failure. Because T-cell dysfunction is thought to contribute to CAR T-cell treatment failure, understanding what mechanisms drive T cells into this dysfunctional state may aid optimal design of efficacious CAR T cells. Dysfunctional CAR T cells have been characterized as having upregulated inhibitory receptors and decreased cytolytic capabilities. Previous studies have identified a role for sustained CAR CD3ζ signaling in CAR T-cell dysfunction. Here, we demonstrate a mechanism that drives dysfunction in CAR T cells through excessive costimulation. Fully activated CD19-targeted CAR T cells were rendered dysfunctional upon stimulation with both endogenous CD28 stimulation and CAR-mediated CD28 costimulation. Costimulation-driven dysfunction of CAR T cells was demonstrated in a syngeneic immunocompetent mouse model, in which CAR T cells were activated with signals 1 (CD3ζ), 2 (CD28), and 3 (IL12). Thus, we show that CAR T-cell dysfunction can be driven through excessive CD28 and 4-1BB costimulation.See related article by Drakes et al., p. 743.
Collapse
Affiliation(s)
- Dinali Wijewarnasuriya
- Weill Cornell Graduate School of Medical Sciences, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina Bebernitz
- Weill Cornell Graduate School of Medical Sciences, New York, New York
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrea V Lopez
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarwish Rafiq
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University School of Medicine, Atlanta, Georgia
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
- Cellular Therapeutics Center, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pharmacology, Weill Cornell Graduate School of Medical Sciences, New York, New York
| |
Collapse
|
65
|
Gavriilaki E, Sakellari I, Gavriilaki M, Anagnostopoulos A. A New Era in Endothelial Injury Syndromes: Toxicity of CAR-T Cells and the Role of Immunity. Int J Mol Sci 2020; 21:E3886. [PMID: 32485958 PMCID: PMC7312228 DOI: 10.3390/ijms21113886] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 12/13/2022] Open
Abstract
Immunotherapy with chimeric antigen receptor T (CAR-T cells) has been recently approved for patients with relapsed/refractory B-lymphoproliferative neoplasms. Along with great efficacy in patients with poor prognosis, CAR-T cells have been also linked with novel toxicities in a significant portion of patients. Cytokine release syndrome (CRS) and neurotoxicity present with unique clinical phenotypes that have not been previously observed. Nevertheless, they share similar characteristics with endothelial injury syndromes developing post hematopoietic cell transplantation (HCT). Evolution in complement therapeutics has attracted renewed interest in these life-threatening syndromes, primarily concerning transplant-associated thrombotic microangiopathy (TA-TMA). The immune system emerges as a key player not only mediating cytokine responses but potentially contributing to endothelial injury in CAR-T cell toxicity. The interplay between complement, endothelial dysfunction, hypercoagulability, and inflammation seems to be a common denominator in these syndromes. As the indications for CAR-T cells and patient populations expand, there in an unmet clinical need of better understanding of the pathophysiology of CAR-T cell toxicity. Therefore, this review aims to provide state-of-the-art knowledge on cellular therapies in clinical practice (indications and toxicities), endothelial injury syndromes and immunity, as well as potential therapeutic targets.
Collapse
Affiliation(s)
- Eleni Gavriilaki
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| | - Ioanna Sakellari
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| | - Maria Gavriilaki
- Laboratory of Clinical Neurophysiology, AHEPA Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Achilles Anagnostopoulos
- Hematology Department—BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece; (I.S.); (A.A.)
| |
Collapse
|
66
|
Alhabbab RY. Targeting Cancer Stem Cells by Genetically Engineered Chimeric Antigen Receptor T Cells. Front Genet 2020; 11:312. [PMID: 32391048 PMCID: PMC7188929 DOI: 10.3389/fgene.2020.00312] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
The term cancer stem cell (CSC) starts 25 years ago with the evidence that CSC is a subpopulation of tumor cells that have renewal ability and can differentiate into several distinct linages. Therefore, CSCs play crucial role in the initiation and the maintenance of cancer. Moreover, it has been proposed throughout several studies that CSCs are behind the failure of the conventional chemo-/radiotherapy as well as cancer recurrence due to their ability to resist the therapy and their ability to re-regenerate. Thus, the need for targeted therapy to eliminate CSCs is crucial; for that reason, chimeric antigen receptor (CAR) T cells has currently been in use with high rate of success in leukemia and, to some degree, in patients with solid tumors. This review outlines the most common CSC populations and their common markers, in particular CD133, CD90, EpCAM, CD44, ALDH, and EGFRVIII, the interaction between CSCs and the immune system, CAR T cell genetic engineering and signaling, CAR T cells in targeting CSCs, and the barriers in using CAR T cells as immunotherapy to treat solid cancers.
Collapse
Affiliation(s)
- Rowa Y. Alhabbab
- Division of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
67
|
Abstract
Adoptive cell therapy with ex vivo expanded tumor infiltrating lymphocytes or gene engineering T cells expressing chimeric antigen receptors (CAR) is a promising treatment for cancer patients. This production utilizes T-cell activation and transduction with activation beads and RetroNectin, respectively. However, the high cost of production is an obstacle for the broad clinical application of novel immunotherapeutic cell products. To facilitate production we refined our approach by using artificial antigen presenting cells (aAPCs) with receptors that ligate CD3, CD28, and the CD137 ligand (CD137L or 41BBL), as well as express the heparin binding domain (HBD), which binds virus for gene-transfer. We have used these aAPC for ex vivo gene engineering and expansion of tumor infiltrating lymphocytes and CAR T cells. We found that aAPCs can support efficacious T-cell expansion and transduction. Moreover, aAPCs expanded T cells exhibit higher production of IFN-γ and lower traits of T-cell exhaustion compared with bead expanded T cells. Our results suggest that aAPC provide a more physiological stimulus for T-cell activation than beads that persistently ligate T cells. The use of a renewable cell line to replace 2 critical reagents (beads and retronectin) for CAR T-cell production can significantly reduce the cost of production and make these therapies more accessible to patients.
Collapse
|
68
|
Ma L, Dichwalkar T, Chang JYH, Cossette B, Garafola D, Zhang AQ, Fichter M, Wang C, Liang S, Silva M, Kumari S, Mehta NK, Abraham W, Thai N, Li N, Wittrup KD, Irvine DJ. Enhanced CAR-T cell activity against solid tumors by vaccine boosting through the chimeric receptor. SCIENCE (NEW YORK, N.Y.) 2020; 365:162-168. [PMID: 31296767 DOI: 10.1126/science.aav8692] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 06/10/2019] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor-T cell (CAR-T) therapy has been effective in the treatment of hematologic malignancies, but it has shown limited efficacy against solid tumors. Here we demonstrate an approach to enhancing CAR-T function in solid tumors by directly vaccine-boosting donor cells through their chimeric receptor in vivo. We designed amphiphile CAR-T ligands (amph-ligands) that, upon injection, trafficked to lymph nodes and decorated the surfaces of antigen-presenting cells, thereby priming CAR-Ts in the native lymph node microenvironment. Amph-ligand boosting triggered massive CAR-T expansion, increased donor cell polyfunctionality, and enhanced antitumor efficacy in multiple immunocompetent mouse tumor models. We demonstrate two approaches to generalizing this strategy to any chimeric antigen receptor, enabling this simple non-human leukocyte antigen-restricted approach to enhanced CAR-T functionality to be applied to existing CAR-T designs.
Collapse
Affiliation(s)
- Leyuan Ma
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Tanmay Dichwalkar
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jason Y H Chang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Benjamin Cossette
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Daniel Garafola
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Angela Q Zhang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael Fichter
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chensu Wang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Simon Liang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Murillo Silva
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Sudha Kumari
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Naveen K Mehta
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Wuhbet Abraham
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nikki Thai
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Na Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - K Dane Wittrup
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Darrell J Irvine
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
69
|
Singh AK, McGuirk JP. CAR T cells: continuation in a revolution of immunotherapy. Lancet Oncol 2020; 21:e168-e178. [PMID: 32135120 DOI: 10.1016/s1470-2045(19)30823-x] [Citation(s) in RCA: 217] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/19/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
The recent clinical successes of immunotherapy, as a result of a broader and more profound understanding of cancer immunobiology, and the leverage of this knowledge to effectively eradicate malignant cells, has revolutionised the field of cancer therapeutics. Immunotherapy is now considered the fifth pillar of cancer care, alongside surgery, chemotherapy, radiotherapy, and targeted therapy. Recently, the success of genetically modified T cells that express chimeric antigen receptors (CAR T cells) has generated considerable excitement. CAR T-cell therapy research and development has built on experience generated by laboratory research and clinical investigation of lymphokine-activated killer cells, tumour-infiltrating lymphocytes, and allogeneic haemopoietic stem-cell transplantation for cancer treatment. This Review aims to provide a background on the field of adoptive T-cell therapy and the development of genetically modified T cells, most notably CAR T-cell therapy. Many challenges exist to optimise efficacy, minimise toxicity, and broaden the application of immunotherapies based on T cells.
Collapse
Affiliation(s)
- Anurag K Singh
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, USA.
| | - Joseph P McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Westwood, KS, USA
| |
Collapse
|
70
|
Kansal R, Richardson N, Neeli I, Khawaja S, Chamberlain D, Ghani M, Ghani QUA, Balazs L, Beranova-Giorgianni S, Giorgianni F, Kochenderfer JN, Marion T, Albritton LM, Radic M. Sustained B cell depletion by CD19-targeted CAR T cells is a highly effective treatment for murine lupus. Sci Transl Med 2020; 11:11/482/eaav1648. [PMID: 30842314 DOI: 10.1126/scitranslmed.aav1648] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/21/2018] [Accepted: 01/30/2019] [Indexed: 12/12/2022]
Abstract
The failure of anti-CD20 antibody (Rituximab) as therapy for lupus may be attributed to the transient and incomplete B cell depletion achieved in clinical trials. Here, using an alternative approach, we report that complete and sustained CD19+ B cell depletion is a highly effective therapy in lupus models. CD8+ T cells expressing CD19-targeted chimeric antigen receptors (CARs) persistently depleted CD19+ B cells, eliminated autoantibody production, reversed disease manifestations in target organs, and extended life spans well beyond normal in the (NZB × NZW) F1 and MRL fas/fas mouse models of lupus. CAR T cells were active for 1 year in vivo and were enriched in the CD44+CD62L+ T cell subset. Adoptively transferred splenic T cells from CAR T cell-treated mice depleted CD19+ B cells and reduced disease in naive autoimmune mice, indicating that disease control was cell-mediated. Sustained B cell depletion with CD19-targeted CAR T cell immunotherapy is a stable and effective strategy to treat murine lupus, and its effectiveness should be explored in clinical trials for lupus.
Collapse
Affiliation(s)
- Rita Kansal
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Noah Richardson
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Saleem Khawaja
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Damian Chamberlain
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Marium Ghani
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Qurat-Ul-Ain Ghani
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Louisa Balazs
- Department of Pathology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Sarka Beranova-Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Francesco Giorgianni
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - James N Kochenderfer
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Tony Marion
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lorraine M Albritton
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
71
|
Abadir E, Gasiorowski RE, Silveira PA, Larsen S, Clark GJ. Is Hematopoietic Stem Cell Transplantation Required to Unleash the Full Potential of Immunotherapy in Acute Myeloid Leukemia? J Clin Med 2020; 9:E554. [PMID: 32085578 PMCID: PMC7073661 DOI: 10.3390/jcm9020554] [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: 01/31/2020] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
From monoclonal antibodies (mAbs) to Chimeric Antigen Receptor (CAR) T cells, immunotherapies have enhanced the efficacy of treatments against B cell malignancies. The same has not been true for Acute Myeloid Leukemia (AML). Hematologic toxicity has limited the potential of modern immunotherapies for AML at preclinical and clinical levels. Gemtuzumab Ozogamicin has demonstrated hematologic toxicity, but the challenge of preserving normal hematopoiesis has become more apparent with the development of increasingly potent immunotherapies. To date, no single surface molecule has been identified that is able to differentiate AML from Hematopoietic Stem and Progenitor Cells (HSPC). Attempts have been made to spare hematopoiesis by targeting molecules expressed only on later myeloid progenitors as well as AML or using toxins that selectively kill AML over HSPC. Other strategies include targeting aberrantly expressed lymphoid molecules or only targeting monocyte-associated proteins in AML with monocytic differentiation. Recently, some groups have accepted that stem cell transplantation is required to access potent AML immunotherapy and envision it as a rescue to avoid severe hematologic toxicity. Whether it will ever be possible to differentiate AML from HSPC using surface molecules is unclear. Unless true specific AML surface targets are discovered, stem cell transplantation could be required to harness the true potential of immunotherapy in AML.
Collapse
Affiliation(s)
- Edward Abadir
- Dendritic Cell Research, ANZAC Research Institute, Concord 2139, NSW, Australia;
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown 2050, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| | - Robin E. Gasiorowski
- The University of Sydney, Camperdown 2039, NSW, Australia;
- Department of Haematology, Concord Repatriation and General Hospital, Concord 2039, NSW, Australia
| | - Pablo A. Silveira
- Dendritic Cell Research, ANZAC Research Institute, Concord 2139, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| | - Stephen Larsen
- Institute of Haematology, Royal Prince Alfred Hospital, Camperdown 2050, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| | - Georgina J. Clark
- Dendritic Cell Research, ANZAC Research Institute, Concord 2139, NSW, Australia;
- The University of Sydney, Camperdown 2039, NSW, Australia;
| |
Collapse
|
72
|
Riedell PA, Bishop MR. Safety and efficacy of axicabtagene ciloleucel in refractory large B-cell lymphomas. Ther Adv Hematol 2020; 11:2040620720902899. [PMID: 32064069 PMCID: PMC6990602 DOI: 10.1177/2040620720902899] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/11/2019] [Indexed: 12/18/2022] Open
Abstract
Aggressive large B-cell lymphomas represent a diverse population of diseases that are typically treated with anti-CD20 based immunochemotherapy. While this treatment is effective for a large proportion of patients, those that become refractory to induction therapy or experience disease relapse suffer an inferior overall prognosis, and novel treatment options are needed. Adoptive T-cell immunotherapy in the form of chimeric antigen receptor (CAR) T-cell therapy is one of the most revolutionary breakthroughs in the past several decades for the treatment of relapsed/refractory aggressive large B-cell lymphomas. Based on data from the pivotal ZUMA-1 study, axicabtagene ciloleucel (axi-cel) became the first-in-class anti-CD19 directed CAR T-cell therapy approved for patients with diffuse large B-cell lymphoma and other aggressive B-cell lymphoma variants. In this review, we provide an overview of CAR T-cell therapy, including its biology, manufacturing, and treatment course. In addition, we highlight the available efficacy data, review pertinent safety concerns, including cytokine release syndrome and neurologic toxicity, as well as provide an overview of emerging therapeutic strategies in the cellular therapy arena.
Collapse
Affiliation(s)
- Peter A. Riedell
- Hematopoietic Cellular Therapy Program, Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Michael R. Bishop
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Ave., MC 2115, Chicago, IL 60637, USA
| |
Collapse
|
73
|
Jacoby E, Shahani SA, Shah NN. Updates on CAR T-cell therapy in B-cell malignancies. Immunol Rev 2020; 290:39-59. [PMID: 31355492 DOI: 10.1111/imr.12774] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 05/09/2019] [Indexed: 12/22/2022]
Abstract
By increasing disease-free survival and offering the potential for long-term cure, chimeric antigen receptor (CAR) T-cell therapy has dramatically expanded therapeutic options among those with high-risk B-cell malignancies. As CAR T-cell utilization evolves however, novel challenges are generated. These include determining how to optimally integrate CAR T cells into standard of care and overcoming mechanisms of resistance to CAR T-cell therapy, such as evolutionary stress induced on cancer cells leading to immunophenotypic changes that allow leukemia to evade this targeted therapy. Compounding these challenges are the limited ability to determine differences between various CAR T-cell constructs, understanding the generalizability of trial outcomes from multiple sites utilizing unique CAR manufacturing strategies, and comparing distinct criteria for toxicity grading while defining optimal management. Additionally, as understanding of CAR behavior in humans has developed, strategies have appropriately evolved to proactively mitigate toxicities. These challenges offer complimentary insights and guide next steps to enhance the efficacy of this novel therapeutic modality. With a focus on B-cell malignancies as the paradigm for effective CAR T-cell therapy, this review describes advances in the field as well as current challenges and future directions.
Collapse
Affiliation(s)
- Elad Jacoby
- Division of Pediatric Hematology, Oncology and BMT, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shilpa A Shahani
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
74
|
Habib R, Nagrial A, Micklethwaite K, Gowrishankar K. Chimeric Antigen Receptors for the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1263:117-143. [PMID: 32588326 DOI: 10.1007/978-3-030-44518-8_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has dramatically revolutionised cancer treatment. The FDA approval of two CAR-T cell products for otherwise incurable refractory B-cell acute lymphoblastic leukaemia (B-ALL) and aggressive B-cell non-Hodgkin lymphoma has established this treatment as an effective immunotherapy option. The race for extending CAR-T therapy for various tumours is well and truly underway. However, response rates in solid organ cancers have been inadequate thus far, partly due to challenges posed by the tumour microenvironment (TME). The TME is a complex structure whose role is to subserve the persistence and proliferation of tumours as well as support their escape from immune surveillance. It presents several obstacles like inhibitory immune checkpoint proteins, immunosuppressive cells, cytokines, chemokines, stromal factors and adverse metabolic pathways. CAR structure and CAR-T therapies have evolved to overcome these obstacles, and we now have several novel CARs with improved anti-tumour activity demonstrated in xenograft models and in some clinical trials. This chapter provides a discussion of the evolution of CAR-T therapies to enable targeting specific aspects of the TME.
Collapse
Affiliation(s)
- Rosemary Habib
- Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
| | - Adnan Nagrial
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
| | - Kenneth Micklethwaite
- Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.,Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia.,Sydney Cellular Therapies Laboratory, Blood and Bone Marrow Transplant Unit, Department of Haematology, Sydney Medical School, Westmead Hospital, Sydney, NSW, Australia
| | - Kavitha Gowrishankar
- Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
75
|
Curran M, Mairesse M, Matas-Céspedes A, Bareham B, Pellegrini G, Liaunardy A, Powell E, Sargeant R, Cuomo E, Stebbings R, Betts CJ, Saeb-Parsy K. Recent Advancements and Applications of Human Immune System Mice in Preclinical Immuno-Oncology. Toxicol Pathol 2019; 48:302-316. [PMID: 31847725 DOI: 10.1177/0192623319886304] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significant advances in immunotherapies have resulted in the increasing need of predictive preclinical models to improve immunotherapeutic drug development, treatment combination, and to prevent or minimize toxicity in clinical trials. Immunodeficient mice reconstituted with human immune system (HIS), termed humanized mice or HIS mice, permit detailed analysis of human immune biology, development, and function. Although this model constitutes a great translational model, some aspects need to be improved as the incomplete engraftment of immune cells, graft versus host disease and the lack of human cytokines and growth factors. In this review, we discuss current HIS platforms, their pathology, and recent advances in their development to improve the quality of human immune cell reconstitution. We also highlight new technologies that can be used to better understand these models and how improved characterization is needed for their application in immuno-oncology safety, efficacy, and new modalities therapy development.
Collapse
Affiliation(s)
- Michelle Curran
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Maelle Mairesse
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Alba Matas-Céspedes
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom.,Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Bethany Bareham
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Giovanni Pellegrini
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ardi Liaunardy
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Edward Powell
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Rebecca Sargeant
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Emanuela Cuomo
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Richard Stebbings
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Catherine J Betts
- Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
76
|
Yao S, Jianlin C, Yarong L, Botao L, Qinghan W, Hongliang F, Lu Z, Hongmei N, Pin W, Hu C, Liangding H, Bin Z. Donor-Derived CD123-Targeted CAR T Cell Serves as a RIC Regimen for Haploidentical Transplantation in a Patient With FUS-ERG+ AML. Front Oncol 2019; 9:1358. [PMID: 31850234 PMCID: PMC6901822 DOI: 10.3389/fonc.2019.01358] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/18/2019] [Indexed: 01/16/2023] Open
Abstract
Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) following chemotherapy is part of standard treatment protocol for patients with acute myeloid leukemia (AML). FUS-ERG+ AML is rare but has an extremely poor prognosis even with allo-HSCT in remission, possibly due to its a leukemia stem cell (LSC)-driven disease resulting in chemotherapy resistance and a novel therapy is urgently required. It has been reported that FUS-ERG-positive AML expresses CD123, a marker of LSC, in some cases. CD123-targeted CAR T cell (CART123) is promising immunotherapy, but how to improve the complete remission (CR) rate and rescue potential hematopoietic toxicity still need to explore. Case Presentation: We used donor-derived CART123 as part of conditioning regimen for haploidentical HSCT (haplo-HSCT) in a patient with FUS-ERG+ AML who relapsed after allogeneic transplantation within 3 months, resists to multi-agent chemotherapy and donor lymphocyte infusion (DLI) and remained non-remission, aiming to reduce these chemotherapy-resistant blasts and rescue potential hematopoietic toxicity. The blasts in BM were reduced within 2 weeks and coincided with CAR copies expansion after CART123 infusion. The patient achieved full donor chimerism, CR with incomplete blood count recovery, and myeloid implantation. Conclusion: Our results hints that CART123 reduces the chemotherapy-resistant AML blasts for FUS-ERG+ AML without affecting the full donor chimerism and myeloid implantation.
Collapse
Affiliation(s)
- Sun Yao
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Chen Jianlin
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Liu Yarong
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Li Botao
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Wang Qinghan
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Fang Hongliang
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Zhang Lu
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China
| | - Ning Hongmei
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Wang Pin
- R&D Department, HRAIN Biotechnology Co., Ltd., Shanghai, China.,Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, United States.,Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, United States.,Department of Pharmaceutical Sciences and Pharmacology, University of Southern California, Los Angeles, CA, United States
| | - Chen Hu
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research, Department of Hematopoietic Stem Cell Transplantation, The Cell and Gene Therapy Center, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Hu Liangding
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| | - Zhang Bin
- Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research, Department of Hematopoietic Stem Cell Transplantation, The Cell and Gene Therapy Center, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), The Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing, China
| |
Collapse
|
77
|
He Y, Schreiber K, Wolf SP, Wen F, Steentoft C, Zerweck J, Steiner M, Sharma P, Shepard HM, Posey A, June CH, Mandel U, Clausen H, Leisegang M, Meredith SC, Kranz DM, Schreiber H. Multiple cancer-specific antigens are targeted by a chimeric antigen receptor on a single cancer cell. JCI Insight 2019; 4:130416. [PMID: 31672936 PMCID: PMC6948763 DOI: 10.1172/jci.insight.130416] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 09/11/2019] [Indexed: 12/22/2022] Open
Abstract
Human cancer cells were eradicated by adoptive transfer of T cells transduced with a chimeric antigen receptor (CAR) made from an antibody (237Ab) that is highly specific for the murine Tn-glycosylated podoplanin (Tn-PDPN). The objectives were to determine the specificity of these CAR-transduced T (CART) cells and the mechanism for the absence of relapse. We show that although the 237Ab bound only to cell lines expressing murine Tn-PDPN, the 237Ab-derived 237CART cells lysed multiple different human and murine cancers not predicted by the 237Ab binding. Nevertheless, the 237CART cell reactivities remained cancer specific because all recognitions were dependent on the Tn glycosylation that resulted from COSMC mutations that were not present in normal tissues. While Tn was required for the recognition by 237CART, Tn alone was not sufficient for 237CART cell activation. Activation of 237CART cells required peptide backbone recognition but tolerated substitutions of up to 5 of the 7 amino acid residues in the motif recognized by 237Ab. Together, these findings demonstrate what we believe is a new principle whereby simultaneous recognition of multiple independent Tn-glycopeptide antigens on a cancer cell makes tumor escape due to antigen loss unlikely.
Collapse
Affiliation(s)
| | - Karin Schreiber
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Steven P. Wolf
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Frank Wen
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Catharina Steentoft
- Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan Zerweck
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Madeline Steiner
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
| | - Preeti Sharma
- Department of Biochemistry, University of Illinois, Urbana, Illinois, USA
| | | | - Avery Posey
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Cellular Therapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Carl H. June
- Center for Cellular Therapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ulla Mandel
- Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Leisegang
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Campus Buch, Berlin, Germany
| | | | - David M. Kranz
- Department of Biochemistry, University of Illinois, Urbana, Illinois, USA
| | - Hans Schreiber
- Committee on Cancer Biology, and
- Department of Pathology, The University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, The University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
78
|
Gorabi AM, Hajighasemi S, Sathyapalan T, Sahebkar A. Cell transfer-based immunotherapies in cancer: A review. IUBMB Life 2019; 72:790-800. [PMID: 31633881 DOI: 10.1002/iub.2180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 09/21/2019] [Indexed: 12/17/2022]
Abstract
In cell transfer therapy (CTT), immune cells such as innate immune-derived natural killer cells and dendritic cells as well as acquired immune-related T lymphocytes such as tumor-infiltrating lymphocytes and cytokine-activated or genetically modified peripheral blood T cells are used in the management of cancer. These therapies are increasingly becoming the most used treatment modality in cancer after tumor resection, chemotherapy, and radiotherapy. In adoptive cell transfer, the lymphocytes isolated from either a donor or the patient are modified ex vivo and reinfused to target malignant cells. Transferring in vitro-manipulated immune cells produces a continuous antitumor immune response. In this review, we evaluate the recent advances in CTT for the management of various malignancies.
Collapse
Affiliation(s)
- Armita M Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeideh Hajighasemi
- Faculty of Paramedicine, Department of Medical Biotechnology, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
79
|
Salman H, Pinz KG, Wada M, Shuai X, Yan LE, Petrov JC, Ma Y. Preclinical Targeting of Human Acute Myeloid Leukemia Using CD4-specific Chimeric Antigen Receptor (CAR) T Cells and NK Cells. J Cancer 2019; 10:4408-4419. [PMID: 31413761 PMCID: PMC6691696 DOI: 10.7150/jca.28952] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 05/12/2019] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy lacking targeted therapy due to shared molecular and transcriptional circuits as well as phenotypic markers with normal hematopoietic stem cells (HSCs). Identifying leukemia specific markers expressed on AML or AML subtypes for therapeutic targeting is of exquisite clinical value. Here we show that CD4, a T lymphocytes membrane glycoprotein that interacts with major histocompatibility complex class II antigens and is also expressed in certain AML subsets but not on HSCs is a proper target for genetically engineered chimeric antigen receptor T cells (CAR-T cells). Treatment with CD4 redirected CAR-T cell (CD4CAR) specifically eliminated CD4-expressing AML cell lines in vitro and exhibited a potent anti-leukemic effect in a systemic AML murine model in vivo. We also utilized natural killers as another vehicle for CAR engineered cells and this strategy similarly and robustly eliminated CD4- expressing AML cells in vitro and had a potent in vivo anti-leukemic effect and was noted to have shorter in vivo persistence. Our data offer a proof of concept for immunotherapeutic targeting of CD4 as a strategy to treat CD4 expressing refractory AML as a bridge to stem cell transplant (SCT) in a first in human clinical trial.
Collapse
Affiliation(s)
- Huda Salman
- Department of Internal Medicine, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Kevin G Pinz
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Masayuki Wada
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Xiao Shuai
- Department of Hematology, West China hospital of Sichuan University, Chengdu, P.R. China
| | - Lulu E Yan
- iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| | - Jessica C Petrov
- Department of Internal Medicine, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA
| | - Yupo Ma
- Department of Internal Medicine, Stony Brook Medicine, Stony Brook University Medical Center, Stony Brook, NY 11794, USA.,iCell Gene Therapeutics LLC, Research & Development Division, Long Island High Technology Incubator, Stony Brook, NY 11790, USA
| |
Collapse
|
80
|
Hu W, Wang G, Huang D, Sui M, Xu Y. Cancer Immunotherapy Based on Natural Killer Cells: Current Progress and New Opportunities. Front Immunol 2019; 10:1205. [PMID: 31214177 PMCID: PMC6554437 DOI: 10.3389/fimmu.2019.01205] [Citation(s) in RCA: 265] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/13/2019] [Indexed: 12/15/2022] Open
Abstract
Cancer immunotherapy has been firmly established as a new milestone for cancer therapy, with the development of multiple immune cells as therapeutic tools. Natural killer (NK) cells are innate immune cells endowed with potent cytolytic activity against tumors, and meanwhile act as regulatory cells for the immune system. The efficacy of NK cell-mediated immunotherapy can be enhanced by immune stimulants such as cytokines and antibodies, and adoptive transfer of activated NK cells expanded ex vivo. In addition, NK cells can arm themselves with chimeric antigen receptors (CARs), which may greatly enhance their anti-tumor activity. Most recently, extracellular vesicles (EVs) derived from NK cells show promising anti-tumor effects in preclinical studies. Herein, we carefully review the current progress in these NK cell-based immunotherapeutic strategies (NK cells combined with stimulants, adoptive transfer of NK cells, CAR-NK cells, and NK EVs) for the treatment of cancers, and discussed the challenges and opportunities for opening a new horizon for cancer immunotherapy.
Collapse
Affiliation(s)
- Weilei Hu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Guosheng Wang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongsheng Huang
- Department of Surgery & Clinical Research Institute of Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Center for Cancer Biology and Innovative Therapeutics, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, China
| | - Meihua Sui
- Department of Surgery & Clinical Research Institute of Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Center for Cancer Biology and Innovative Therapeutics, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, China
| | - Yibing Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
81
|
Yáñez L, Sánchez-Escamilla M, Perales MA. CAR T Cell Toxicity: Current Management and Future Directions. Hemasphere 2019; 3:e186. [PMID: 31723825 PMCID: PMC6746032 DOI: 10.1097/hs9.0000000000000186] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/28/2018] [Accepted: 02/08/2019] [Indexed: 12/17/2022] Open
Abstract
By late 2018, 2 chimeric antigen receptor T (CAR T) cell products have been approved by US and European regulatory authorities. Tisagenlecleucel (Kymriah, Novartis) is indicated in the treatment of patients up to 25 years of age with B-cell acute lymphoblastic leukemia (ALL) that is refractory or in second or later relapse, or adult patients with large B-cell lymphoma relapsed or refractory (r/r) after 2 or more lines of systemic therapy, including diffuse large B-cell lymphoma (DLBCL) not otherwise specified, high grade B-cell lymphoma and DLBCL arising from follicular lymphoma. Axicabtagene ciloleucel (Yescarta, Kite) is indicated for the treatment of adult patients with large B-cell lymphoma relapsed or refractory after 2 or more lines of systemic therapy, including DLBCL not otherwise specified, primary mediastinal large B-cell lymphoma, high grade B-cell lymphoma, and DLBCL arising from follicular lymphoma (ZUMA-1 trial). This review will offer a practical guide for the recognition and management of the most important toxicities related to the use of the current commercial CAR T cells, and also highlight strategies to diminish these side effects in the future.
Collapse
Affiliation(s)
- Lucrecia Yáñez
- Department of Hematology, University Hospital Marqués de Valdecilla, Santander, Spain
- Department of Hematological Malignancies and Stem Cell Transplantation, Research Institute of Marques de Valdecilla (IDIVAL), Santander, Spain
| | - Miriam Sánchez-Escamilla
- Department of Hematological Malignancies and Stem Cell Transplantation, Research Institute of Marques de Valdecilla (IDIVAL), Santander, Spain
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College; New York, NY
| |
Collapse
|
82
|
Kuhn NF, Purdon TJ, van Leeuwen DG, Lopez AV, Curran KJ, Daniyan AF, Brentjens RJ. CD40 Ligand-Modified Chimeric Antigen Receptor T Cells Enhance Antitumor Function by Eliciting an Endogenous Antitumor Response. Cancer Cell 2019; 35:473-488.e6. [PMID: 30889381 PMCID: PMC6428219 DOI: 10.1016/j.ccell.2019.02.006] [Citation(s) in RCA: 176] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/16/2018] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cells provide great efficacy in B cell malignancies. However, improved CAR T cell therapies are still needed. Here, we engineered tumor-targeted CAR T cells to constitutively express the immune-stimulatory molecule CD40 ligand (CD40L) and explored efficacy in different mouse leukemia/lymphoma models. We observed that CD40L+ CAR T cells circumvent tumor immune escape via antigen loss through CD40/CD40L-mediated cytotoxicity and induction of a sustained, endogenous immune response. After adoptive cell transfer, the CD40L+ CAR T cells displayed superior antitumor efficacy, licensed antigen-presenting cells, enhanced recruitment of immune effectors, and mobilized endogenous tumor-recognizing T cells. These effects were absent in Cd40-/- mice and provide a rationale for the use of CD40L+ CAR T cells in cancer treatment.
Collapse
Affiliation(s)
- Nicholas F Kuhn
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Terence J Purdon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dayenne G van Leeuwen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea V Lopez
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anthony F Daniyan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
83
|
Grover NS, Savoldo B. Challenges of driving CD30-directed CAR-T cells to the clinic. BMC Cancer 2019; 19:203. [PMID: 30841880 PMCID: PMC6404322 DOI: 10.1186/s12885-019-5415-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 02/27/2019] [Indexed: 01/24/2023] Open
Abstract
Chimeric antigen receptor T (CAR-T) cells are a promising new treatment for patients with relapsed or refractory hematologic malignancies, including lymphoma. Given the success of CAR-T cells directed against CD19, new targets are being developed and tested, since not all lymphomas express CD19. CD30 is promising target as it is universally expressed in virtually all classical Hodgkin lymphomas, anaplastic large cell lymphomas, and in a proportion of other lymphoma types, including cutaneous T cell lymphomas and diffuse large B cell lymphomas. Preclinical studies with CD30-directed CAR-T cells support the feasibility of this approach. Recently, two clinical trials of CD30-directed CAR-T cells in relapsed/refractory CD30+ lymphomas, including Hodgkin lymphoma, have been reported with minimal toxicities noted and preliminary efficacy seen in a proportion of patients. However, improving the persistence and expansion of CAR-T cells is key to further enhancing the efficacy of this treatment approach. Future directions include optimizing the lymphodepletion regimen, enhancing migration to the tumor site, and combination with other immune regulators. Several ongoing and upcoming clinical trials of CD30-directed CAR-T cells are expected to further enhance this approach to treat patients with relapsed and refractory CD30+ lymphomas.
Collapse
Affiliation(s)
- Natalie S Grover
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA.
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
84
|
Chobrutskiy BI, Zaman S, Diviney A, Mihyu MM, Blanck G. T-cell receptor-α CDR3 domain chemical features correlate with survival rates in bladder cancer. J Cancer Res Clin Oncol 2019; 145:615-623. [PMID: 30539280 DOI: 10.1007/s00432-018-2815-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 12/04/2018] [Indexed: 01/04/2023]
Abstract
PURPOSE In certain cancer settings, a T-cell response to cancer represents a relatively favorable outcome. Thus, the near-future challenges include a better understanding of exactly which T-cell features contribute to a response to which cancer antigen-groups, to maximize the opportunities for tumor-infiltrating lymphocyte (TIL)-based therapies and other immunotherapies. METHODS The immune receptor complementarity determining region-3 (CDR3) is considered to be important for antigen binding, hence, in this report, we evaluated the chemical features of the CDR3 of 846 T-cell receptor-α (TCR-α) coding regions associated with bladder tumor tissue, using bioinformatics databases. RESULTS Results indicated that statistically significantly distinct, low value, CDR3 region isoelectric points associate with a better outcome (log rank p < 0.027, overall survival). Moreover, in samples representing the more favorable isoelectric points, known driver mutations, for example, PIK3CA (E → K) with chemically complementary features overlap the better-outcome, low isoelectric point samples. Further work extended these results, i.e., survival rate-CDR3 associations, to other CDR3 chemical features and other cancers, consistent with the initial isoelectric point-related, bladder cancer findings. CONCLUSIONS A bioinformatics assessment of cancer-associated TCR biochemical features may improve the accuracy of the predictions of which TILs will be best for ex-vivo amplification and which patients will benefit from other immunotherapies.
Collapse
Affiliation(s)
- Boris I Chobrutskiy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Saif Zaman
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Andrea Diviney
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Moody M Mihyu
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, USA.
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, 12901 Bruce B. Downs Bd. MDC7, Tampa, FL, 33612, USA.
| |
Collapse
|
85
|
Kansagra AJ, Frey NV, Bar M, Laetsch TW, Carpenter PA, Savani BN, Heslop HE, Bollard CM, Komanduri KV, Gastineau DA, Chabannon C, Perales MA, Hudecek M, Aljurf M, Andritsos L, Barrett JA, Bachanova V, Bonini C, Ghobadi A, Gill SI, Hill J, Kenderian S, Kebriaei P, Nagler A, Maloney D, Liu HD, Shah NN, Kharfan-Dabaja MA, Shpall EJ, Mufti GJ, Johnston L, Jacoby E, Bazarbachi A, DiPersio JF, Pavletic SZ, Porter DL, Grupp SA, Sadelain M, Litzow MR, Mohty M, Hashmi SK. Clinical Utilization of Chimeric Antigen Receptor T Cells in B Cell Acute Lymphoblastic Leukemia: An Expert Opinion from the European Society for Blood and Marrow Transplantation and the American Society for Blood and Marrow Transplantation. Biol Blood Marrow Transplant 2019; 25:e76-e85. [PMID: 30576834 PMCID: PMC8335749 DOI: 10.1016/j.bbmt.2018.12.068] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 02/06/2023]
Abstract
On August 30, 2017 the US Food and Drug Administration approved tisagenlecleucel (Kymriah; Novartis, Basel, Switzerland), a synthetic bioimmune product of anti-CD19 chimeric antigen receptor T cells (CAR-T), for the treatment of children and young adults with relapsed/refractory B cell acute lymphoblastic leukemia (B-ALL). With this new era of personalized cancer immunotherapy, multiple challenges are present, ranging from implementation of a CAR-T program to safe delivery of the drug, long-term toxicity monitoring, and disease assessments. To address these issues experts representing the American Society for Blood and Marrow Transplant, the European Society for Blood and Marrow Transplantation, the International Society of Cell and Gene Therapy, and the Foundation for the Accreditation of Cellular Therapy formed a global CAR-T task force to identify and address key questions pertinent for hematologists and transplant physicians regarding the clinical use of anti CD19 CAR-T therapy in patients with B-ALL. This article presents an initial roadmap for navigating common clinical practice scenarios that will become more prevalent now that the first commercially available CAR-T product for B-ALL has been approved.
Collapse
Affiliation(s)
- Ankit J Kansagra
- Department of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Noelle V Frey
- Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Merav Bar
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Theodore W Laetsch
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center and Children's Health, Dallas, Texas
| | - Paul A Carpenter
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Bipin N Savani
- Division of Hematology/Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC
| | - Krishna V Komanduri
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, Florida
| | - Dennis A Gastineau
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, Arizona
| | - Christian Chabannon
- Institut Paoli-Calmettes, Centre de Lutte Contre le Cancer; Centre d'Investigations Cliniques en Biothérapie, Université d'Aix-Marseille, Inserm CBT 1409, Marseille, France
| | - Miguel A Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Mahmoud Aljurf
- Oncology Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Leslie Andritsos
- Division of Hematology, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - John A Barrett
- Stem Cell Transplantation and Cellular Therapy Program, GW Cancer Center, George Washington University, Washington, DC
| | - Veronika Bachanova
- Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Chiara Bonini
- Experimental Hematology Unit, University Vita-Salute San Raffaele and Ospedale San Raffaele, Milano, Italy
| | - Armin Ghobadi
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Saar I Gill
- Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joshua Hill
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Saad Kenderian
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota; Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Arnon Nagler
- The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David Maloney
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, Washington
| | - Hien D Liu
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ghulam J Mufti
- Department of Haematological Medicine, King's College, London, United Kingdom
| | - Laura Johnston
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Elad Jacoby
- The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Steven Z Pavletic
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David L Porter
- Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, New York
| | - Mark R Litzow
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Mohamad Mohty
- Hôpital Saint-Antoine, APHP, Sorbonne Universite, INSERM UMRs 938, Paris, France
| | - Shahrukh K Hashmi
- Oncology Centre, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia; Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
86
|
Kansagra AJ, Frey NV, Bar M, Laetsch TW, Carpenter PA, Savani BN, Heslop HE, Bollard CM, Komanduri KV, Gastineau DA, Chabannon C, Perales MA, Hudecek M, Aljurf M, Andritsos L, Barrett JA, Bachanova V, Bonini C, Ghobadi A, Gill SI, Hill JA, Kenderian S, Kebriaei P, Nagler A, Maloney D, Liu HD, Shah NN, Kharfan-Dabaja MA, Shpall EJ, Mufti GJ, Johnston L, Jacoby E, Bazarbachi A, DiPersio JF, Pavletic SZ, Porter DL, Grupp SA, Sadelain M, Litzow MR, Mohty M, Hashmi SK. Clinical utilization of Chimeric Antigen Receptor T-cells (CAR-T) in B-cell acute lymphoblastic leukemia (ALL)-an expert opinion from the European Society for Blood and Marrow Transplantation (EBMT) and the American Society for Blood and Marrow Transplantation (ASBMT). Bone Marrow Transplant 2019; 54:1868-1880. [PMID: 31092900 PMCID: PMC8268756 DOI: 10.1038/s41409-019-0451-2] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/24/2018] [Indexed: 01/05/2023]
Abstract
On August 30, 2017, the U.S. Food and Drug Administration (US-FDA) approved tisagenlecleucel (KYMRIAH, Novartis, Basel, Switzerland), a synthetic bioimmune product of anti-CD19 chimeric antigen receptor-T cells (CAR-T), for the treatment of children and young adults with relapsed/refractory B-cell acute lymphoblastic leukemia (B-ALL). With this new era of personalized cancer immunotherapy, multiple challenges are present ranging from implementation of a CAR-T program to safe delivery of the drug, long-term toxicity monitoring and disease assessments. To address these issues, experts representing the American Society for Blood and Marrow Transplant (ASBMT), the European Group for Blood and Marrow Transplantation (EBMT), the International Society of Cell and Gene Therapy (ISCT), and the Foundation for the Accreditation of Cellular Therapy (FACT), formed a global CAR-T task force to identify and address key questions pertinent for hematologists and transplant physicians regarding the clinical use of anti CD19 CAR-T therapy in patients with B-ALL. This article presents an initial roadmap for navigating common clinical practice scenarios that will become more prevalent now that the first commercially available CAR-T product for B-ALL has been approved.
Collapse
Affiliation(s)
- Ankit J. Kansagra
- 0000 0000 9482 7121grid.267313.2Department of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Noelle V. Frey
- 0000 0004 1936 8972grid.25879.31Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA USA
| | - Merav Bar
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Theodore W. Laetsch
- 0000 0000 9482 7121grid.267313.2Division of Pediatric Hematology-Oncology, Department of Pediatrics, University of Texas Southwestern Medical Center and Children’s Health, Dallas, TX USA
| | - Paul A. Carpenter
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Bipin N. Savani
- 0000 0004 1936 9916grid.412807.8Division of Hematology/Oncology, Department of Internal Medicine, Vanderbilt University Medical Center, Nashville, TN USA
| | - Helen E. Heslop
- 0000 0004 0445 0041grid.63368.38Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children’s Hospital, Houston, TX USA
| | - Catherine M. Bollard
- 0000 0004 0482 1586grid.239560.bCenter for Cancer and Immunology Research, Children’s National Health System, Washington, DC USA
| | - Krishna V. Komanduri
- 0000 0004 0414 313Xgrid.418456.aSylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL USA
| | - Dennis A. Gastineau
- 0000 0000 8875 6339grid.417468.8Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ USA
| | - Christian Chabannon
- 0000 0001 2176 4817grid.5399.6Institut Paoli-Calmettes, Centre de Lutte Contre le Cancer; Centre d’Investigations Cliniques en Biothérapie, Université d’Aix-Marseille, Inserm CBT, 1409 Marseille, France
| | - Miguel A. Perales
- 0000 0001 2171 9952grid.51462.34Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY USA
| | - Michael Hudecek
- 0000 0001 1378 7891grid.411760.5Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Mahmoud Aljurf
- 0000 0001 2191 4301grid.415310.2Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Leslie Andritsos
- 0000 0001 1545 0811grid.412332.5Division of Hematology, Ohio State University Wexner Medical Center, Columbus, OH USA
| | - John A. Barrett
- 0000 0004 1936 9510grid.253615.6Stem Cell Transplantation and Cellular Therapy Program, GW Cancer Center, George Washington University, Washington, DC USA
| | - Veronika Bachanova
- 0000000419368657grid.17635.36Division of Hematology/Oncology/Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN USA
| | - Chiara Bonini
- grid.15496.3f0000 0001 0439 0892Experimental Hematology Unit, University Vita-Salute San Raffaele and Ospedale San Raffaele, Milano, Italy
| | - Armin Ghobadi
- 0000 0001 2355 7002grid.4367.6Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO USA
| | - Saar I. Gill
- 0000 0004 1936 8972grid.25879.31Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA USA
| | - Joshua A. Hill
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Saad Kenderian
- 0000 0004 0459 167Xgrid.66875.3aDivision of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN USA ,0000 0004 0459 167Xgrid.66875.3aDepartment of Immunology, Mayo Clinic, Rochester, MN USA
| | - Partow Kebriaei
- 0000 0001 2291 4776grid.240145.6Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Arnon Nagler
- 0000 0004 1937 0546grid.12136.37The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - David Maloney
- 0000000122986657grid.34477.33Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA USA
| | - Hien D. Liu
- 0000 0000 9891 5233grid.468198.aH. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Nirali N. Shah
- 0000 0001 2297 5165grid.94365.3dPediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | | | - Elizabeth J. Shpall
- 0000 0004 1937 0546grid.12136.37The Chaim Sheba Medical Center, Tel-Hashomer, Affiliated with the Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ghulam J. Mufti
- 0000 0001 2322 6764grid.13097.3cDepartment of Haematological Medicine, King’s College, London, UK
| | - Laura Johnston
- 0000000419368956grid.168010.eDepartment of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Elad Jacoby
- 0000 0000 9891 5233grid.468198.aH. Lee Moffitt Cancer Center and Research Institute, Tampa, FL USA
| | - Ali Bazarbachi
- 0000 0004 0581 3406grid.411654.3Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - John F. DiPersio
- 0000 0004 0459 167Xgrid.66875.3aDivision of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN USA
| | - Steven Z. Pavletic
- 0000 0001 2297 5165grid.94365.3dExperimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD USA
| | - David L. Porter
- 0000 0004 1936 8972grid.25879.31Cell Therapy and Transplant Program, Abramson Cancer Center and the Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA USA
| | - Stephan A. Grupp
- 0000 0004 1936 8972grid.25879.31Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA USA
| | - Michel Sadelain
- 0000 0001 2171 9952grid.51462.34Center for Cell Engineering and Immunology Program, Sloan Kettering Institute, New York, NY USA
| | - Mark R. Litzow
- 0000 0004 0459 167Xgrid.66875.3aDepartment of Immunology, Mayo Clinic, Rochester, MN USA
| | - Mohamad Mohty
- 0000 0001 2308 1657grid.462844.8Hôpital Saint-Antoine, APHP, Sorbonne Universite, INSERM UMRs 938, Paris, France
| | - Shahrukh K. Hashmi
- 0000 0001 1545 0811grid.412332.5Division of Hematology, Ohio State University Wexner Medical Center, Columbus, OH USA ,0000 0004 0459 167Xgrid.66875.3aDepartment of Immunology, Mayo Clinic, Rochester, MN USA
| |
Collapse
|
87
|
Johnson MO, Wolf MM, Madden MZ, Andrejeva G, Sugiura A, Contreras DC, Maseda D, Liberti MV, Paz K, Kishton RJ, Johnson ME, de Cubas AA, Wu P, Li G, Zhang Y, Newcomb DC, Wells AD, Restifo NP, Rathmell WK, Locasale JW, Davila ML, Blazar BR, Rathmell JC. Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism. Cell 2018; 175:1780-1795.e19. [PMID: 30392958 PMCID: PMC6361668 DOI: 10.1016/j.cell.2018.10.001] [Citation(s) in RCA: 486] [Impact Index Per Article: 69.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 08/16/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022]
Abstract
Activated T cells differentiate into functional subsets with distinct metabolic programs. Glutaminase (GLS) converts glutamine to glutamate to support the tricarboxylic acid cycle and redox and epigenetic reactions. Here, we identify a key role for GLS in T cell activation and specification. Though GLS deficiency diminished initial T cell activation and proliferation and impaired differentiation of Th17 cells, loss of GLS also increased Tbet to promote differentiation and effector function of CD4 Th1 and CD8 CTL cells. This was associated with altered chromatin accessibility and gene expression, including decreased PIK3IP1 in Th1 cells that sensitized to IL-2-mediated mTORC1 signaling. In vivo, GLS null T cells failed to drive Th17-inflammatory diseases, and Th1 cells had initially elevated function but exhausted over time. Transient GLS inhibition, however, led to increased Th1 and CTL T cell numbers. Glutamine metabolism thus has distinct roles to promote Th17 but constrain Th1 and CTL effector cell differentiation.
Collapse
Affiliation(s)
- Marc O. Johnson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Melissa M. Wolf
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew Z. Madden
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Gabriela Andrejeva
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ayaka Sugiura
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Diana C. Contreras
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Damian Maseda
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maria V. Liberti
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Katelyn Paz
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Rigel J. Kishton
- Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Matthew E. Johnson
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aguirre A. de Cubas
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Pingsheng Wu
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Gongbo Li
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Yongliang Zhang
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Dawn C. Newcomb
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Andrew D. Wells
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia and the Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas P. Restifo
- Center for Cell-Based Therapy, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - W. Kimryn Rathmell
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jason W. Locasale
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Marco L. Davila
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Bruce R. Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jeffrey C. Rathmell
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA,Vanderbilt Center for Immunobiology, Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University School of Medicine, Nashville, TN 37232, USA,Lead Contact,Correspondence:
| |
Collapse
|
88
|
Disruption of a self-amplifying catecholamine loop reduces cytokine release syndrome. Nature 2018; 564:273-277. [PMID: 30542164 PMCID: PMC6512810 DOI: 10.1038/s41586-018-0774-y] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 10/22/2018] [Indexed: 11/08/2022]
Abstract
Cytokine release syndrome (CRS) is a life-threatening complication of several new immunotherapies used to treat cancers and autoimmune diseases1-5. Here we report that atrial natriuretic peptide can protect mice from CRS induced by such agents by reducing the levels of circulating catecholamines. Catecholamines were found to orchestrate an immunodysregulation resulting from oncolytic bacteria and lipopolysaccharide through a self-amplifying loop in macrophages. Myeloid-specific deletion of tyrosine hydroxylase inhibited this circuit. Cytokine release induced by T-cell-activating therapeutic agents was also accompanied by a catecholamine surge and inhibition of catecholamine synthesis reduced cytokine release in vitro and in mice. Pharmacologic catecholamine blockade with metyrosine protected mice from lethal complications of CRS resulting from infections and various biotherapeutic agents including oncolytic bacteria, T-cell-targeting antibodies and CAR-T cells. Our study identifies catecholamines as an essential component of the cytokine release that can be modulated by specific blockers without impairing the therapeutic response.
Collapse
|
89
|
Switchable control over in vivo CAR T expansion, B cell depletion, and induction of memory. Proc Natl Acad Sci U S A 2018; 115:E10898-E10906. [PMID: 30373813 PMCID: PMC6243241 DOI: 10.1073/pnas.1810060115] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy represents a powerful strategy in immuno-oncology. Nevertheless, associated life-threatening toxicities and chronic B cell aplasia have underscored the need to control engineered T cells in the patient. To address these challenges, we have previously developed a switchable CAR (sCAR) T cell platform that allows dose-titratable control over CAR T cell activity by using antibody-based switches. Here, we demonstrate in a syngeneic murine model that the switchable platform can impart antitumor efficacy while dissociating long-term persistence from chronic B cell aplasia. Further, the functional reversibility of the switchable platform can be leveraged to incorporate “rest” phases through cyclical dosing of the switch to enable the induction of a robust central memory population for in vivo, on-demand expansion of sCAR T cells. Chimeric antigen receptor (CAR) T cells with a long-lived memory phenotype are correlated with durable, complete remissions in patients with leukemia. However, not all CAR T cell products form robust memory populations, and those that do can induce chronic B cell aplasia in patients. To address these challenges, we previously developed a switchable CAR (sCAR) T cell system that allows fully tunable, on/off control over engineered cellular activity. To further evaluate the platform, we generated and assessed different murine sCAR constructs to determine the factors that afford efficacy, persistence, and expansion of sCAR T cells in a competent immune system. We find that sCAR T cells undergo significant in vivo expansion, which is correlated with potent antitumor efficacy. Most importantly, we show that the switch dosing regimen not only allows control over B cell populations through iterative depletion and repopulation, but that the “rest” period between dosing cycles is the key for induction of memory and expansion of sCAR T cells. These findings introduce rest as a paradigm in enhancing memory and improving the efficacy and persistence of engineered T cell products.
Collapse
|
90
|
Li G, Boucher JC, Kotani H, Park K, Zhang Y, Shrestha B, Wang X, Guan L, Beatty N, Abate-Daga D, Davila ML. 4-1BB enhancement of CAR T function requires NF-κB and TRAFs. JCI Insight 2018; 3:121322. [PMID: 30232281 PMCID: PMC6237232 DOI: 10.1172/jci.insight.121322] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/07/2018] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptors (CARs) have an antigen-binding domain fused to transmembrane, costimulatory, and CD3ζ domains. Two CARs with regulatory approval include a CD28 or 4-1BB costimulatory domain. While both CARs achieve similar clinical outcomes, biologic differences have become apparent but not completely understood. Therefore, in this study we aimed to identify mechanistic differences between 4-1BB and CD28 costimulation that contribute to the biologic differences between the 2 CARs and could be exploited to enhance CAR T cell function. Using CD19-targeted CAR T cells with 4-1BB we determined that enhancement of T cell function is driven by NF-κB. Comparison to CAR T cells with CD28 also revealed that 4-1BB is associated with more antiapoptotic proteins and dependence on persistence for B cell killing. While TNF receptor-associated factor 2 (TRAF2) has been presupposed to be required for 4-1BB costimulation in CAR T cells, we determined that TRAF1 and TRAF3 are also critical. We observed that TRAFs impacted CAR T viability and proliferation, as well as cytotoxicity and/or cytokines, in part by regulating NF-κB. Our study demonstrates how 4-1BB costimulation in CAR T cells impacts antitumor eradication and clinical outcomes and has implications for enhanced CAR design.
Collapse
Affiliation(s)
- Gongbo Li
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Justin C. Boucher
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Hiroshi Kotani
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Kyungho Park
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Yongliang Zhang
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Bishwas Shrestha
- Clinical Science Division, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Lawrence Guan
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Nolan Beatty
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Daniel Abate-Daga
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Marco L. Davila
- Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.,Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
91
|
Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies. J Immunol Res 2018; 2018:4054815. [PMID: 30306093 PMCID: PMC6166361 DOI: 10.1155/2018/4054815] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/26/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are powerful immune effectors whose antitumor activity is regulated through a sophisticated network of activating and inhibitory receptors. As effectors of cancer immunotherapy, NK cells are attractive as they do not attack healthy self-tissues nor do they induce T cell-driven inflammatory cytokine storm, enabling their use as allogeneic adoptive cellular therapies. Clinical responses to adoptive NK-based immunotherapy have been thwarted, however, by the profound immunosuppression induced by the tumor microenvironment, particularly severe in the context of solid tumors. In addition, the short postinfusion persistence of NK cells in vivo has limited their clinical efficacy. Enhancing the antitumor immunity of NK cells through genetic engineering has been fueled by the promise that impaired cytotoxic functionality can be restored or augmented with the use of synthetic genetic approaches. Alongside expressing chimeric antigen receptors to overcome immune escape by cancer cells, enhance their recognition, and mediate their killing, NK cells have been genetically modified to enhance their persistence in vivo by the expression of cytokines such as IL-15, avoid functional and metabolic tumor microenvironment suppression, or improve their homing ability, enabling enhanced targeting of solid tumors. However, NK cells are notoriously adverse to endogenous gene uptake, resulting in low gene uptake and transgene expression with many vector systems. Though viral vectors have achieved the highest gene transfer efficiencies with NK cells, nonviral vectors and gene transfer approaches—electroporation, lipofection, nanoparticles, and trogocytosis—are emerging. And while the use of NK cell lines has achieved improved gene transfer efficiencies particularly with viral vectors, challenges with primary NK cells remain. Here, we discuss the genetic engineering of NK cells as they relate to NK immunobiology within the context of cancer immunotherapy, highlighting the most recent breakthroughs in viral vectors and nonviral approaches aimed at genetic reprogramming of NK cells for improved adoptive immunotherapy of cancer, and, finally, address their clinical status.
Collapse
|
92
|
Caruso HG, Heimberger AB, Cooper LJN. Steering CAR T cells to distinguish friend from foe. Oncoimmunology 2018; 8:e1271857. [PMID: 31646067 PMCID: PMC6791456 DOI: 10.1080/2162402x.2016.1271857] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 12/02/2016] [Accepted: 12/07/2016] [Indexed: 02/03/2023] Open
Abstract
CD19-specific chimeric antigen receptor (CAR)+ T cells have demonstrated clinical efficacy and long-lasting remissions, concomitant with tolerable normal B-cell aplasia. However, many tumor-associated antigens (TAAs) are expressed on normal tissues, the destruction of which would lead to intolerable toxicity. Thus, there is a need to engineer CAR+ T cells with improved safety profiles to restrict toxicity against TAA-expressing normal tissues. Bioengineering approaches include: (i) targeting CAR+ T cells to the tumor site, (ii) limiting CAR+ T-cell persistence, and (iii) restricting CAR activation. We review and evaluate strategies to engineer CAR+ T cells to reduce the potential of on-target, off-tissue toxicity.
Collapse
Affiliation(s)
- Hillary G Caruso
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amy B Heimberger
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Laurence J N Cooper
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Ziopharm Oncology, Boston, MA, USA
| |
Collapse
|
93
|
Escobar G, Barbarossa L, Barbiera G, Norelli M, Genua M, Ranghetti A, Plati T, Camisa B, Brombin C, Cittaro D, Annoni A, Bondanza A, Ostuni R, Gentner B, Naldini L. Interferon gene therapy reprograms the leukemia microenvironment inducing protective immunity to multiple tumor antigens. Nat Commun 2018; 9:2896. [PMID: 30042420 PMCID: PMC6057972 DOI: 10.1038/s41467-018-05315-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/14/2018] [Indexed: 11/13/2022] Open
Abstract
Immunotherapy is emerging as a new pillar of cancer treatment with potential to cure. However, many patients still fail to respond to these therapies. Among the underlying factors, an immunosuppressive tumor microenvironment (TME) plays a major role. Here we show that monocyte-mediated gene delivery of IFNα inhibits leukemia in a mouse model. IFN gene therapy counteracts leukemia-induced expansion of immunosuppressive myeloid cells and imposes an immunostimulatory program to the TME, as shown by bulk and single-cell transcriptome analyses. This reprogramming promotes T-cell priming and effector function against multiple surrogate tumor-specific antigens, inhibiting leukemia growth in our experimental model. Durable responses are observed in a fraction of mice and are further increased combining gene therapy with checkpoint blockers. Furthermore, IFN gene therapy strongly enhances anti-tumor activity of adoptively transferred T cells engineered with tumor-specific TCR or CAR, overcoming suppressive signals in the leukemia TME. These findings warrant further investigations on the potential development of our gene therapy strategy towards clinical testing. An immune suppressive tumor microenvironment (TME) is a limitation for immunotherapy. Here the authors show that, in a B cell acute lymphoblastic leukemia mouse model, gene-based delivery of IFNα reprograms the leukemia-induced immunosuppressive TME into immunostimulatory and enhances T-cell responses.
Collapse
Affiliation(s)
- Giulia Escobar
- Vita-Salute San Raffaele University, 20132, Milan, Italy.,Targeted Cancer Gene Therapy Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Luigi Barbarossa
- Targeted Cancer Gene Therapy Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.,San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Giulia Barbiera
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Margherita Norelli
- Vita-Salute San Raffaele University, 20132, Milan, Italy.,Division of Immunology, Transplant and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Marco Genua
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Anna Ranghetti
- Targeted Cancer Gene Therapy Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Tiziana Plati
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Barbara Camisa
- Division of Immunology, Transplant and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Chiara Brombin
- CUSSB-University Center for Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, 20132, Milan, Italy
| | - Davide Cittaro
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy.,Centre for Translational Genomics and Bioinformatics, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Attilio Bondanza
- Vita-Salute San Raffaele University, 20132, Milan, Italy.,Division of Immunology, Transplant and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy
| | - Renato Ostuni
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy
| | - Bernhard Gentner
- San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy. .,Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy.
| | - Luigi Naldini
- Vita-Salute San Raffaele University, 20132, Milan, Italy. .,Targeted Cancer Gene Therapy Unit, IRCCS San Raffaele Scientific Institute, 20132, Milan, Italy. .,San Raffaele Telethon Institute for Gene Therapy, 20132, Milan, Italy.
| |
Collapse
|
94
|
Ruella M, June CH. Predicting Dangerous Rides in CAR T Cells: Bridging the Gap between Mice and Humans. Mol Ther 2018; 26:1401-1403. [PMID: 29784587 PMCID: PMC5986966 DOI: 10.1016/j.ymthe.2018.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA.
| |
Collapse
|
95
|
Magnani CF, Mezzanotte C, Cappuzzello C, Bardini M, Tettamanti S, Fazio G, Cooper LJN, Dastoli G, Cazzaniga G, Biondi A, Biagi E. Preclinical Efficacy and Safety of CD19CAR Cytokine-Induced Killer Cells Transfected with Sleeping Beauty Transposon for the Treatment of Acute Lymphoblastic Leukemia. Hum Gene Ther 2018; 29:602-613. [PMID: 29641322 DOI: 10.1089/hum.2017.207] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Infusion of patient-derived CD19-specific chimeric antigen receptor (CAR) T cells engineered by viral vectors achieved complete remission and durable response in relapsed and refractory (r/r) B-lineage neoplasms. Here, we expand on those findings by providing a preclinical evaluation of allogeneic non-viral cytokine-induced killer (CIK) cells transfected with the Sleeping Beauty (SB) transposon CD19CAR (CARCIK-CD19). Specifically, thanks to a large-scale 18-day manufacturing process, it was possible to achieve stable CD19CAR expression (62.425 ± 6.399%) and efficient T-cell expansion (23.36 ± 3.00-fold). Frozen/thawed CARCIK-CD19 remained fully functional both in vitro and in an established patient-derived xenograft (PDX) of MLL-ENL rearranged acute lymphoblastic leukemia (ALL). CARCIK-CD19 showed a dose-dependent antitumor response and prolonged persistence in a PDX, bearing the feature of a Philadelphia-like ALL with PAX5/AUTS2 translocation, and in a survival model of lymphoma, achieving complete eradication of disseminated tumors. Finally, the infusion of CARCIK-CD19 proved to be safe and well tolerated in a biodistribution and toxicity model. The infused cells persisted in the hematopoietic and post-injection perfused organs until the end of the study and consisted of CD8+, CD56+, and CAR+ T cells. Overall, these findings provide important implications for non-viral technology and the proof-of-concept that donor-derived CARCIK-CD19 are indeed effective against relapsed ALL, a possibility that will be tested in Phase I/II clinical trials after allogeneic hematopoietic stem-cell transplantation.
Collapse
Affiliation(s)
- Chiara F Magnani
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Claudia Mezzanotte
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Claudia Cappuzzello
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Michela Bardini
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Sarah Tettamanti
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Grazia Fazio
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | | | - Giuseppe Dastoli
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Giovanni Cazzaniga
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Andrea Biondi
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| | - Ettore Biagi
- 1 Tettamanti Research Center, Department of Pediatrics, University of Milano-Bicocca , San Gerardo Hospital/Fondazione MBBM, Monza, Italy
| |
Collapse
|
96
|
Siegler EL, Wang P. Preclinical Models in Chimeric Antigen Receptor-Engineered T-Cell Therapy. Hum Gene Ther 2018; 29:534-546. [PMID: 29390873 DOI: 10.1089/hum.2017.243] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy has enormous potential in inducing long-term remission in cancer patients, and chimeric antigen receptor (CAR)-engineered T cells have been largely successful in treating hematological malignancies in the clinic. CAR-T therapy has not been as effective in treating solid tumors, in part due to the immunosuppressive tumor microenvironment. Additionally, CAR-T therapy can cause dangerous side effects, including off-tumor toxicity, cytokine release syndrome, and neurotoxicity. Animal models of CAR-T therapy often fail to predict such adverse events and frequently overestimate the efficacy of the treatment. Nearly all preclinical CAR-T studies have been performed in mice, including syngeneic, xenograft, transgenic, and humanized mouse models. Recently, a few studies have used primate models to mimic clinical side effects better. To date, no single model perfectly recapitulates the human immune system and tumor microenvironment, and some models have revealed CAR-T limitations that were contradicted or missed entirely in other models. Careful model selection based on the primary goals of the study is a crucial step in evaluating CAR-T treatment. Advancements are being made in preclinical models, with the ultimate objective of providing safer, more effective CAR-T therapy to patients.
Collapse
Affiliation(s)
- Elizabeth Louise Siegler
- 1 Department of Biomedical Engineering, University of Southern California , Los Angeles, California
| | - Pin Wang
- 1 Department of Biomedical Engineering, University of Southern California , Los Angeles, California.,2 Department of Pharmacology and Pharmaceutical Sciences, University of Southern California , Los Angeles, California.,3 Mork Family Department of Chemical Engineering and Materials Science, University of Southern California , Los Angeles, California
| |
Collapse
|
97
|
Arai Y, Choi U, Corsino CI, Koontz SM, Tajima M, Sweeney CL, Black MA, Feldman SA, Dinauer MC, Malech HL. Myeloid Conditioning with c-kit-Targeted CAR-T Cells Enables Donor Stem Cell Engraftment. Mol Ther 2018; 26:1181-1197. [PMID: 29622475 DOI: 10.1016/j.ymthe.2018.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 01/04/2023] Open
Abstract
We report a novel approach to bone marrow (BM) conditioning using c-kit-targeted chimeric antigen receptor T (c-kit CAR-T) cells in mice. Previous reports using anti-c-kit or anti-CD45 antibody linked to a toxin such as saporin have been promising. We developed a distinctly different approach using c-kit CAR-T cells. Initial studies demonstrated in vitro killing of hematopoietic stem cells by c-kit CAR-T cells but poor expansion in vivo and poor migration of CAR-T cells into BM. Pre-treatment of recipient mice with low-dose cyclophosphamide (125 mg/kg) together with CXCR4 transduction in the CAR-T cells enhanced trafficking to and expansion in BM (<1%-13.1%). This resulted in significant depletion of the BM c-kit+ population (9.0%-0.1%). Because congenic Thy1.1 CAR-T cells were used in the Thy1.2-recipient mice, anti-Thy1.1 antibody could be used to deplete CAR-T cells in vivo before donor BM transplant. This achieved 20%-40% multilineage engraftment. We applied this conditioning to achieve an average of 28% correction of chronic granulomatous disease mice by wild-type BM transplant. Our findings provide a proof of concept that c-kit CAR-T cells can achieve effective BM conditioning without chemo-/radiotherapy. Our work also demonstrates that co-expression of a trafficking receptor can enhance targeting of CAR-T cells to a designated tissue.
Collapse
Affiliation(s)
- Yasuyuki Arai
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Uimook Choi
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Cristina I Corsino
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Sherry M Koontz
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Masaki Tajima
- Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Colin L Sweeney
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Mary A Black
- Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Steven A Feldman
- Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Mary C Dinauer
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Harry L Malech
- Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
98
|
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been clinically proven to efficiently combat haematological malignancies. However, continuous efforts are required to increase the specificity of CAR T-cells against tumour versus normal tissues, and are essential to improve their antitumour activity in solid tumours. This review summarises the structure of major CAR designs, and strategies to overcome immunosuppressive tumour microenvironment, and reduce toxicities. Along with reviewing currently available techniques that allow the elimination of CAR T-cells after they fulfil their desired functions, using suicide genes, drug elimination strategies are also introduced. A better understanding of the strengths and pitfalls of CAR T-cell therapy will provide fundamental knowledge for the improvement of engineered T-cell therapy in the near future.
Collapse
|
99
|
Kornete M, Marone R, Jeker LT. Highly Efficient and Versatile Plasmid-Based Gene Editing in Primary T Cells. THE JOURNAL OF IMMUNOLOGY 2018; 200:2489-2501. [PMID: 29445007 DOI: 10.4049/jimmunol.1701121] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/22/2018] [Indexed: 12/31/2022]
Abstract
Adoptive cell transfer is an important approach for basic research and emerges as an effective treatment for various diseases, including infections and blood cancers. Direct genetic manipulation of primary immune cells opens up unprecedented research opportunities and could be applied to enhance cellular therapeutic products. In this article, we report highly efficient genome engineering in primary murine T cells using a plasmid-based RNA-guided CRISPR system. We developed a straightforward approach to ablate genes in up to 90% of cells and to introduce precisely targeted single nucleotide polymorphisms in up to 25% of the transfected primary T cells. We used gene editing-mediated allele switching to quantify homology-directed repair, systematically optimize experimental parameters, and map a native B cell epitope in primary T cells. Allele switching of a surrogate cell surface marker can be used to enrich cells, with successful simultaneous editing of a second gene of interest. Finally, we applied the approach to correct two disease-causing mutations in the Foxp3 gene. Repairing the cause of the scurfy syndrome, a 2-bp insertion in Foxp3, and repairing the clinically relevant Foxp3K276X mutation restored Foxp3 expression in primary T cells.
Collapse
Affiliation(s)
- Mara Kornete
- Department of Biomedicine, Basel University Hospital and University of Basel, CH-4031 Basel, Switzerland; and Transplantation Immunology and Nephrology, Basel University Hospital, CH-4031 Basel, Switzerland
| | - Romina Marone
- Department of Biomedicine, Basel University Hospital and University of Basel, CH-4031 Basel, Switzerland; and Transplantation Immunology and Nephrology, Basel University Hospital, CH-4031 Basel, Switzerland
| | - Lukas T Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, CH-4031 Basel, Switzerland; and Transplantation Immunology and Nephrology, Basel University Hospital, CH-4031 Basel, Switzerland
| |
Collapse
|
100
|
Sharpe ME. T-cell Immunotherapies and the Role of Nonclinical Assessment: The Balance between Efficacy and Pathology. Toxicol Pathol 2018; 46:131-146. [PMID: 29471776 PMCID: PMC5843031 DOI: 10.1177/0192623317752101] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gene-engineered T-cell therapies have the potential to revolutionize the treatment of cancer. These therapies have shown exceptional clinical efficacy specifically in the field of B-cell malignancies and the first products (Kymriah™ and Yescarta™) have recently been approved in the United States for specific indications. The power of these treatments is also linked with a distinct set of toxicities both predicted and unpredicted, including off-tumor activity, cytokine release syndromes, and neurotoxicity, occasionally with fatal consequences. As these therapies begin to reach more patients, it is critical to develop the nonclinical tools to adequately determine the mechanisms driving these toxicities, to assess the safety risks of candidate products, and to develop strategies for safety management.
Collapse
Affiliation(s)
- Michaela E. Sharpe
- Cell and Gene Therapy Catapult, Guy’s Hospital, Great Maze Pond, London, United Kingdom
| |
Collapse
|