401
|
Biology and clinical application of CAR T cells for B cell malignancies. Int J Hematol 2016; 104:6-17. [PMID: 27262700 PMCID: PMC5512169 DOI: 10.1007/s12185-016-2039-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/31/2016] [Accepted: 05/31/2016] [Indexed: 10/21/2022]
Abstract
Chimeric antigen receptor (CAR)-modified T cells have generated broad interest in oncology following a series of dramatic clinical successes in patients with chemorefractory B cell malignancies. CAR therapy now appears to be on the cusp of regulatory approval as a cell-based immunotherapy. We review here the T cell biology and cell engineering research that led to the development of second generation CARs, the selection of CD19 as a CAR target, and the preclinical studies in animal models that laid the foundation for clinical trials targeting CD19+ malignancies. We further summarize the status of CD19 CAR clinical therapy for non-Hodgkin lymphoma and B cell acute lymphoblastic leukemia, including their efficacy, toxicities (cytokine release syndrome, neurotoxicity and B cell aplasia) and current management in humans. We conclude with an overview of recent pre-clinical advances in CAR design that argues favorably for the advancement of CAR therapy to tackle other hematological malignancies as well as solid tumors.
Collapse
|
402
|
Sadelain M. Chimeric antigen receptors: driving immunology towards synthetic biology. Curr Opin Immunol 2016; 41:68-76. [PMID: 27372731 DOI: 10.1016/j.coi.2016.06.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/06/2016] [Accepted: 06/08/2016] [Indexed: 12/15/2022]
Abstract
The advent of second generation chimeric antigen receptors and the CD19 paradigm have ushered a new therapeutic modality in oncology. In contrast to earlier forms of adoptive cell therapy, which were based on the isolation and expansion of naturally occurring T cells, CAR therapy is based on the design and manufacture of engineered T cells with optimized properties. A new armamentarium, comprising not only CARs but also chimeric costimulatory receptors, chimeric cytokine receptors, inhibitory receptors and synthetic Notch receptors, expressed in naïve, central memory or stem cell-like memory T cells, is being developed for clinical use in a wide range of cancers. Immunological principles are thus finding a new purpose thanks to advances in genetic engineering, synthetic biology and cell manufacturing sciences.
Collapse
Affiliation(s)
- Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
403
|
Turtle CJ, Riddell SR, Maloney DG. CD19-Targeted chimeric antigen receptor-modified T-cell immunotherapy for B-cell malignancies. Clin Pharmacol Ther 2016; 100:252-8. [PMID: 27170467 DOI: 10.1002/cpt.392] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/04/2016] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptors (CARs) comprise a tumor-targeting moiety, often in the form of a single chain variable fragment derived from a monoclonal antibody, fused to one or more intracellular T-cell signaling sequences. Lymphodepletion chemotherapy followed by infusion of T cells that are genetically modified to express a CD19-specific CAR is a promising therapy for patients with refractory CD19(+) B-cell malignancies, producing rates of complete remission that are remarkably high in acute lymphoblastic leukemia and encouraging in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Responses are often durable, although additional studies are needed to define the role of CAR-T cell immunotherapy in the context of other treatments. CAR-modified T-cell immunotherapy can be complicated by cytokine release syndrome and neurologic toxicity, which in most cases are manageable and reversible. Here we review recent clinical trial data and discuss issues for the field.
Collapse
Affiliation(s)
- C J Turtle
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - S R Riddell
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| | - D G Maloney
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA
| |
Collapse
|
404
|
T cells expressing CD19-specific Engager Molecules for the Immunotherapy of CD19-positive Malignancies. Sci Rep 2016; 6:27130. [PMID: 27255991 PMCID: PMC4891739 DOI: 10.1038/srep27130] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 05/16/2016] [Indexed: 12/31/2022] Open
Abstract
T cells expressing chimeric antigen receptors (CARs) or the infusion of bispecific T-cell engagers (BITEs) have shown antitumor activity in humans for CD19-positive malignancies. While BITEs redirect the large reservoir of resident T cells to tumors, CAR T cells rely on significant in vivo expansion to exert antitumor activity. We have shown that it is feasible to modify T cells to secrete solid tumor antigen-specific BITEs, enabling T cells to redirect resident T cells to tumor cells. To adapt this approach to CD19-positive malignancies we now generated T cells expressing secretable, CD19-specific BITEs (CD19-ENG T cells). CD19-ENG T cells recognized tumor cells in an antigen-dependent manner as judged by cytokine production and tumor killing, and redirected bystander T cells to tumor cells. Infusion of CD19-ENG T cells resulted in regression of leukemia or lymphoma in xenograft models and a survival advantage in comparison to control mice. Genetically modified T cells expressing engager molecules may present a promising addition to current CD19-targeted immunotherapies.
Collapse
|
405
|
Braun M, Ress ML, Yoo YE, Scholz CJ, Eyrich M, Schlegel PG, Wölfl M. IL12-mediated sensitizing of T-cell receptor-dependent and -independent tumor cell killing. Oncoimmunology 2016; 5:e1188245. [PMID: 27622043 DOI: 10.1080/2162402x.2016.1188245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 04/25/2016] [Accepted: 05/05/2016] [Indexed: 10/21/2022] Open
Abstract
Interleukin 12 (IL12) is a key inflammatory cytokine critically influencing Th1/Tc1-T-cell responses at the time of initial antigen encounter. Therefore, it may be exploited for cancer immunotherapy. Here, we investigated how IL12, and other inflammatory cytokines, shape effector functions of human T-cells. Using a defined culture system, we followed the gradual differentiation and function of antigen-specific CD8(+) T cells from their initial activation as naïve T cells through their expansion phase as early memory cells to full differentiation as clonally expanded effector T cells. The addition of IL12 8 days after the initial priming event initiated two mechanistically separate events: First, IL12 sensitized the T-cell receptor (TCR) for antigen-specific activation, leading to an approximately 10-fold increase in peptide sensitivity and, in consequence, enhanced tumor cell killing. Secondly, IL12 enabled TCR/HLA-independent activation and cytotoxicity: this "non-specific" effect was mediated by the NK cell receptor DNAM1 (CD226) and dependent on ligand expression of the target cells. This IL12 regulated, DNAM1-mediated killing is dependent on src-kinases as well as on PTPRC (CD45) activity. Thus, besides enhancing TCR-mediated activation, we here identified for the first time a second IL12 mediated mechanism leading to activation of a receptor-dependent killing pathway via DNAM1.
Collapse
Affiliation(s)
- Matthias Braun
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany; Else-Kröner Forschungskolleg for Interdisciplinary Translational Immunology, School of Medicine, University of Würzburg, Würzburg, Germany
| | - Marie L Ress
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg , Würzburg, Germany
| | - Young-Eun Yoo
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg , Würzburg, Germany
| | - Claus J Scholz
- Core Unit Systems Medicine, University of Würzburg , Würzburg, Germany
| | - Matthias Eyrich
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg , Würzburg, Germany
| | - Paul G Schlegel
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany; Clinical Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Matthias Wölfl
- Children's Hospital, Pediatric Hematology, Oncology and Stem Cell Transplantation, University of Würzburg, Würzburg, Germany; Clinical Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
406
|
CAR models: next-generation CAR modifications for enhanced T-cell function. Mol Ther Oncolytics 2016; 3:16014. [PMID: 27231717 PMCID: PMC4871190 DOI: 10.1038/mto.2016.14] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/04/2016] [Accepted: 04/06/2016] [Indexed: 12/18/2022] Open
Abstract
T cells genetically targeted with a chimeric antigen receptor (CAR) to B-cell malignancies have demonstrated tremendous clinical outcomes. With the proof in principle for CAR T cells as a therapy for B-cell malignancies being established, current and future research is being focused on adapting CAR technology to other cancers, as well as enhancing its efficacy and/or safety. The modular nature of the CAR, extracellular antigen-binding domain fused to a transmembrane domain and intracellular T-cell signaling domains, allows for optimization by replacement of the various components. These modifications are creating a whole new class of therapeutic CARs. In this review, we discuss the recent major advances in CAR design and how these modifications will impact its clinical application.
Collapse
|
407
|
Torikai H, Cooper LJ. Translational Implications for Off-the-shelf Immune Cells Expressing Chimeric Antigen Receptors. Mol Ther 2016; 24:1178-86. [PMID: 27203439 DOI: 10.1038/mt.2016.106] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022] Open
Abstract
Chimeric antigen receptor (CAR) endows specificity to T-cells independent of human leukocyte antigen (HLA). This enables one immunoreceptor to directly target the same surface antigen on different subsets of tumor cells from multiple HLA-disparate recipients. Most approaches manufacture individualized CAR(+)T-cells from the recipient or HLA-compatible donor, which are revealing promising clinical results. This is the impetus to broaden the number of patients eligible to benefit from adoptive immunotherapy such as to infuse third-party donor derived CAR(+)T-cells. This will overcome issues associated with (i) time to manufacture T-cells, (ii) cost to generate one product for one patient, (iii) inability to generate a product from lymphopenic patients or patient's immune cells fail to complete the manufacturing process, and (iv) heterogeneity of T-cell products produced for or from individual recipients. Establishing a biobank of allogeneic genetically modified immune cells from healthy third-party donors, which are cryopreserved and validated in advance of administration, will facilitate the centralizing manufacturing and widespread distribution of CAR(+)T-cells to multiple points-of-care in a timely manner. To achieve this, it is necessary to engineer an effective strategy to avoid deleterious allogeneic immune responses leading to toxicity and rejection. We review the strategies to establish "off-the-shelf" donor-derived biobanks for human application of CAR(+)T-cells as a drug.
Collapse
Affiliation(s)
- Hiroki Torikai
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Laurence Jn Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Ziopharm Oncology Inc., Boston, Massachusetts, USA
| |
Collapse
|
408
|
Zhang H, Ye ZL, Yuan ZG, Luo ZQ, Jin HJ, Qian QJ. New Strategies for the Treatment of Solid Tumors with CAR-T Cells. Int J Biol Sci 2016; 12:718-29. [PMID: 27194949 PMCID: PMC4870715 DOI: 10.7150/ijbs.14405] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 01/16/2016] [Indexed: 01/11/2023] Open
Abstract
Recent years, we have witnessed significant progresses in both basic and clinical studies regarding novel therapeutic strategies with genetically engineered T cells. Modification with chimeric antigen receptors (CARs) endows T cells with tumor specific cytotoxicity and thus induce anti-tumor immunity against malignancies. However, targeting solid tumors is more challenging than targeting B-cell malignancies with CAR-T cells because of the histopathological structure features, specific antigens shortage and strong immunosuppressive environment of solid tumors. Meanwhile, the on-target/off-tumor toxicity caused by relative expression of target on normal tissues is another issue that should be reckoned. Optimization of the design of CAR vectors, exploration of new targets, addition of safe switches and combination with other treatments bring new vitality to the CAR-T cell based immunotherapy against solid tumors. In this review, we focus on the major obstacles limiting the application of CAR-T cell therapy toward solid tumors and summarize the measures to refine this new cancer therapeutic modality.
Collapse
Affiliation(s)
- Hao Zhang
- 1. Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Zhen-Long Ye
- 1. Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Zhen-Gang Yuan
- 1. Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Zheng-Qiang Luo
- 2. Xinyuan Institute of Medicine and Biotechnology College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Hua-Jun Jin
- 1. Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China
| | - Qi-Jun Qian
- 1. Laboratory of Viral and Gene Therapy, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai 200438, China;; 2. Xinyuan Institute of Medicine and Biotechnology College of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China;; 3. Ningbo NO.5 Hospital (Ningbo Cancer Hospital), Ningbo 315201, China
| |
Collapse
|
409
|
Barth MJ, Chu Y, Hanley PJ, Cairo MS. Immunotherapeutic approaches for the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma. Br J Haematol 2016; 173:597-616. [PMID: 27062282 DOI: 10.1111/bjh.14078] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
With the introduction of the anti-CD20 monoclonal antibody rituximab, B-cell non-Hodgkin lymphoma was the first malignancy successfully treated with an immunotherapeutic agent. Since then, numerous advances have expanded the repertoire of immunotherapeutic agents available for the treatment of a variety of malignancies, including many lymphoma subtypes. These include the introduction of monoclonal antibodies targeting a variety of cell surface proteins, including the successful targeting of immunoregulatory checkpoint receptors present on T-cells or tumour cells. Additionally, cellular immunotherapeutic approaches utilize T- or Natural Killer-cells generated with chimeric antigen receptors against cell surface proteins or Epstein-Barr virus-associated latent membrane proteins. The following review describes the current state of immunotherapy for non-Hodgkin lymphoma including a summary of currently available data and promising agents currently in clinical development with future promise in the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Matthew J Barth
- Department of Pediatrics, Roswell Park Cancer Institute, Buffalo, NY, USA.,Division of Pediatric Hematology/Oncology, University at Buffalo, Buffalo, NY, USA
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Patrick J Hanley
- Program for Cell Enhancement and Technologies for Immunotherapy, Division of Blood and Marrow Transplantation, Sheikhz Zayed Institute for Pediatric Surgical Innovation, Washington, D.C., USA.,Center for Cancer and Immunology Research, Children's National Health System, The George Washington University, Washington, D.C., USA
| | - Mitchell S Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA.,Department of Medicine, New York Medical College, Valhalla, NY, USA.,Department of Pathology, New York Medical College, Valhalla, NY, USA.,Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, USA.,Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
410
|
Maus MV, Levine BL. Chimeric Antigen Receptor T-Cell Therapy for the Community Oncologist. Oncologist 2016; 21:608-17. [PMID: 27009942 DOI: 10.1634/theoncologist.2015-0421] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED : The field of cancer immunotherapy has rapidly progressed in the past decade as several therapeutic modalities have entered into the clinic. One such immunotherapy that has shown promise in the treatment of cancer is the use of chimeric antigen receptor (CAR)-modified T lymphocytes. CARs are engineered receptors constructed from antigen recognition regions of antibodies fused to T-cell signaling and costimulatory domains that can be used to reprogram a patient's T cells to specifically target tumor cells. CAR T-cell therapy has demonstrated sustained complete responses for some patients with advanced leukemia, and a number of CAR therapies are being evaluated in clinical studies. CAR T-cell therapy-associated toxicities, including cytokine release syndrome, macrophage activation syndrome, and tumor lysis syndrome, have been observed and effectively managed in the clinic. In patients with significant clinical responses, sustained B-cell aplasia has also been observed and is a marker of CAR T-cell persistence that might provide long-term disease control. Education on CAR T-cell therapy efficacy and safety management is critical for clinicians and patients who are considering this novel type of treatment. In the present report, the current landscape of CAR T-cell therapy, the effective management of patients undergoing treatment, and which patients are the most suitable candidates for current trials are discussed. IMPLICATIONS FOR PRACTICE The present report describes the current status of chimeric antigen receptor (CAR) T lymphocytes as an immunotherapy for patients with relapsed or refractory B-cell malignancies. CAR T cells targeting CD19, a protein expressed on many B-cell malignancies, typically induce high complete response rates in patients with B-cell leukemia or lymphoma who have very limited therapeutic options. Recent clinical trial results of CD19 CAR T-cell therapies and the management of CAR T-cell-associated adverse events are discussed. The present report will therefore inform physicians regarding the efficacy and safety of CAR T cells as a therapy for B-cell malignancies.
Collapse
Affiliation(s)
- Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, Pennsylvania, USA Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
411
|
Abstract
The engineered expression of chimeric antigen receptors (CARs) on the surface of T cells enables the redirection of T-cell specificity. Early clinical trials using CAR T cells for the treatment of patients with cancer showed modest results, but the impressive outcomes of several trials of CD19-targeted CAR T cells in the treatment of patients with B-cell malignancies have generated an increased enthusiasm for this approach. Important lessons have been derived from clinical trials of CD19-specific CAR T cells, and ongoing clinical trials are testing CAR designs directed at novel targets involved in haematological and solid malignancies. In this Review, we discuss these trials and present strategies that can increase the antitumour efficacy and safety of CAR T-cell therapy. Given the fast-moving nature of this field, we only discuss studies with direct translational application currently or soon-to-be tested in the clinical setting.
Collapse
|
412
|
Li S, Yang Z, Shen J, Shan J, Qian C. Adoptive therapy with CAR redirected T cells for hematological malignancies. SCIENCE CHINA-LIFE SCIENCES 2016; 59:370-8. [DOI: 10.1007/s11427-016-5036-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/26/2016] [Indexed: 01/01/2023]
|
413
|
The future of cancer treatment: immunomodulation, CARs and combination immunotherapy. Nat Rev Clin Oncol 2016; 13:273-90. [PMID: 26977780 DOI: 10.1038/nrclinonc.2016.25] [Citation(s) in RCA: 736] [Impact Index Per Article: 92.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the past decade, advances in the use of monoclonal antibodies (mAbs) and adoptive cellular therapy to treat cancer by modulating the immune response have led to unprecedented responses in patients with advanced-stage tumours that would otherwise have been fatal. To date, three immune-checkpoint-blocking mAbs have been approved in the USA for the treatment of patients with several types of cancer, and more patients will benefit from immunomodulatory mAb therapy in the months and years ahead. Concurrently, the adoptive transfer of genetically modified lymphocytes to treat patients with haematological malignancies has yielded dramatic results, and we anticipate that this approach will rapidly become the standard of care for an increasing number of patients. In this Review, we highlight the latest advances in immunotherapy and discuss the role that it will have in the future of cancer treatment, including settings for which testing combination strategies and 'armoured' CAR T cells are recommended.
Collapse
|
414
|
Wang W, Qin DY, Zhang BL, Wei W, Wang YS, Wei YQ. Establishing guidelines for CAR-T cells: challenges and considerations. SCIENCE CHINA-LIFE SCIENCES 2016; 59:333-9. [DOI: 10.1007/s11427-016-5026-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 01/17/2016] [Indexed: 01/08/2023]
|
415
|
Petrozziello E, Sturmheit T, Mondino A. Exploiting cytokines in adoptive T-cell therapy of cancer. Immunotherapy 2016; 7:573-84. [PMID: 26065481 DOI: 10.2217/imt.15.19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Adoptive immunotherapy with tumor-reactive autologous T cells, either expanded from tumor specimens or genetically engineered to express tumor-reactive T-cell receptors and chimeric antigen receptors, is holding promising results in clinical trials. Several critical issues have been identified and results underline the possibility to exploit cytokines to further ameliorate the efficacy of current treatment protocols, also encompassing adoptive T-cell therapy. Here we review latest developments on the use of cytokines to better direct the nature of the T-cell infusion product, T-cell function and persistence in vivo, as well as to modulate the tumor microenvironment.
Collapse
Affiliation(s)
- Elisabetta Petrozziello
- Division of Immunology, Transplantation & Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| | - Tabea Sturmheit
- Division of Immunology, Transplantation & Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy.,Vita-Salute San Raffaele University, San Raffaele Scientific Institute Milan, Italy
| | - Anna Mondino
- Division of Immunology, Transplantation & Infectious Diseases, San Raffaele Scientific Institute, Via Olgettina, 58, 20132 Milan, Italy
| |
Collapse
|
416
|
Pegram HJ, Smith EL, Rafiq S, Brentjens RJ. CAR therapy for hematological cancers: can success seen in the treatment of B-cell acute lymphoblastic leukemia be applied to other hematological malignancies? Immunotherapy 2016; 7:545-61. [PMID: 26065479 DOI: 10.2217/imt.15.6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has recently come into the spotlight due to impressive results in patients with B-cell acute lymphoblastic leukemia. By targeting CD19, a marker expressed most B-cell tumors, as well as normal B cells, CAR T-cell therapy has been investigated as a treatment strategy for B-cell leukemia and lymphoma. This review will discuss the successes of this therapy for the treatment of B-cell acute lymphoblastic leukemia and the challenges to this therapeutic strategy. We will also discuss application of CAR T-cell therapy to chronic lymphocytic leukemia and other B-cell malignancies including a follicular lymphoma, diffuse large B-cell lymphoma, as well as acute and plasma cell malignancies.
Collapse
Affiliation(s)
- Hollie J Pegram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric L Smith
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarwish Rafiq
- 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.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Molecular Pharmacology & Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
417
|
Phase 1 clinical trial demonstrated that MUC1 positive metastatic seminal vesicle cancer can be effectively eradicated by modified Anti-MUC1 chimeric antigen receptor transduced T cells. SCIENCE CHINA-LIFE SCIENCES 2016; 59:386-97. [PMID: 26961900 DOI: 10.1007/s11427-016-5024-7] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Accepted: 01/16/2016] [Indexed: 02/05/2023]
Abstract
Recent progress in chimeric antigen receptor-modified T-cell (CAR-T cell) technology in cancer therapy is extremely promising, especially in the treatment of patients with B-cell acute lymphoblastic leukemia. In contrast, due to the hostile immunosuppressive microenvironment of a solid tumor, CAR T-cell accessibility and survival continue to pose a considerable challenge, which leads to their limited therapeutic efficacy. In this study, we constructed two anti-MUC1 CAR-T cell lines. One set of CAR-T cells contained SM3 single chain variable fragment (scFv) sequence specifically targeting the MUC1 antigen and co-expressing interleukin (IL) 12 (named SM3-CAR). The other CAR-T cell line carried the SM3 scFv sequence modified to improve its binding to MUC1 antigen (named pSM3-CAR) but did not co-express IL-12. When those two types of CAR-T cells were injected intratumorally into two independent metastatic lesions of the same MUC1(+) seminal vesicle cancer patient as part of an interventional treatment strategy, the initial results indicated no side-effects of the MUC1 targeting CAR-T cell approach, and patient serum cytokines responses were positive. Further evaluation showed that pSM3-CAR effectively caused tumor necrosis, providing new options for improved CAR-T therapy in solid tumors.
Collapse
|
418
|
Perales MA, Sauter CS, Armand P. Reprint of: Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2016; 22:S9-S14. [PMID: 26899275 DOI: 10.1016/j.bbmt.2016.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in non-transplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Craig S Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
419
|
Sadelain M, Brentjens R, Rivière I, Park J. CD19 CAR Therapy for Acute Lymphoblastic Leukemia. Am Soc Clin Oncol Educ Book 2016:e360-3. [PMID: 25993197 DOI: 10.14694/edbook_am.2015.35.e360] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chimeric antigen receptor (CAR) therapy is an emerging immunotherapy that shows great promise for cancer, in particular acute lymphoblastic leukemia (ALL). CARs are recombinant receptors for antigen, which, in a single molecule, redirect the specificity and function of T lymphocytes. Following their genetic transfer to patient T cells, the latter acquire the ability to recognize leukemia cells and destroy them. Several years ago, we identified CD19 as an attractive target for CAR therapy for most B cell malignancies, including ALL. We and others have reported remarkable clinical outcomes in adults and children with ALL, achieving a high complete remission rate irrespective of age, prior treatments, or other prognostic markers. Severe cytokine release may develop in patients with high tumor burdens. Several interventions are available to curb the cytokine release syndrome when it occurs. Based on the impressive results obtained with CD19 CAR therapy for ALL, it is realistic to expect that CD19 CARs will become part of the armamentarium for B cell-ALL and other B cell malignancies.
Collapse
Affiliation(s)
- Michel Sadelain
- From the Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Renier Brentjens
- From the Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Isabelle Rivière
- From the Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jae Park
- From the Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
420
|
Dai H, Wang Y, Lu X, Han W. Chimeric Antigen Receptors Modified T-Cells for Cancer Therapy. J Natl Cancer Inst 2016; 108:djv439. [PMID: 26819347 PMCID: PMC4948566 DOI: 10.1093/jnci/djv439] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
The genetic modification and characterization of T-cells with chimeric antigen receptors (CARs) allow functionally distinct T-cell subsets to recognize specific tumor cells. The incorporation of costimulatory molecules or cytokines can enable engineered T-cells to eliminate tumor cells. CARs are generated by fusing the antigen-binding region of a monoclonal antibody (mAb) or other ligand to membrane-spanning and intracellular-signaling domains. They have recently shown clinical benefit in patients treated with CD19-directed autologous T-cells. Recent successes suggest that the modification of T-cells with CARs could be a powerful approach for developing safe and effective cancer therapeutics. Here, we briefly review early studies, consider strategies to improve the therapeutic potential and safety, and discuss the challenges and future prospects for CAR T-cells in cancer therapy.
Collapse
Affiliation(s)
- Hanren Dai
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China
| | - Yao Wang
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China
| | - Xuechun Lu
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China
| | - Weidong Han
- Affiliations of authors: Department of Immunology (HD, YW, WH) and Department of Molecular Biology (WH), Institute of Basic Medicine, School of Life Sciences, Department of Bio-therapeutic (HD, YW, WH), and Department of Hematology (XL), Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
421
|
Hernandez-Alcoceba R, Poutou J, Ballesteros-Briones MC, Smerdou C. Gene therapy approaches against cancer using in vivo and ex vivo gene transfer of interleukin-12. Immunotherapy 2016; 8:179-98. [PMID: 26786809 DOI: 10.2217/imt.15.109] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IL-12 is an immunostimulatory cytokine with strong antitumor properties. Systemic administration of IL-12 in cancer patients led to severe toxic effects, prompting the development of gene therapy vectors able to express this cytokine locally in tumors. Both nonviral and viral vectors have demonstrated a high antitumor efficacy in preclinical tumor models. Some of these vectors, including DNA electroporation, adenovirus and ex vivo transduced dendritic cells, were tested in patients, showing low toxicity and moderate antitumor efficacy. IL-12 activity can be potentiated by molecules with immunostimulatory, antiangiogenic or cytotoxic activity. These combination therapies are of clinical interest because they could lower the threshold for IL-12 efficacy, increasing the therapeutic potential of gene therapy and preventing the toxicity mediated by this cytokine.
Collapse
Affiliation(s)
- Ruben Hernandez-Alcoceba
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - Joanna Poutou
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - María Cristina Ballesteros-Briones
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| | - Cristian Smerdou
- Division of Gene Therapy, CIMA, University of Navarra, Pamplona 31008 Spain.,Instituto de Investigación Sanitaria de Navarra, c/Irunlarrea 3, Pamplona 31008, Spain
| |
Collapse
|
422
|
Tsai AK, Davila E. Producer T cells: Using genetically engineered T cells as vehicles to generate and deliver therapeutics to tumors. Oncoimmunology 2016; 5:e1122158. [PMID: 27467930 PMCID: PMC4910704 DOI: 10.1080/2162402x.2015.1122158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/11/2015] [Accepted: 11/14/2015] [Indexed: 12/27/2022] Open
Abstract
Adoptive cell transfer (ACT) is an emerging anticancer therapy that has shown promise in various malignancies. Redirecting antigen specificity by genetically engineering T cells to stably express receptors has become an effective variant of ACT. A novel extension of this approach is to utilize engineered T cells to produce and deliver anticancer therapeutics that enhance cytotoxic T cell function and simultaneously inhibit immunosuppressive processes. Here, we review the potential of using T cells as therapeutic-secreting vehicles for immunotherapies and present theoretical and established arguments in support of further development of this unique cell-based immunotherapy.
Collapse
Affiliation(s)
- Alexander K Tsai
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland , Baltimore, Baltimore, MD, USA
| | - Eduardo Davila
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland, Baltimore, Baltimore, MD, USA; Department of Microbiology and Immunology, University of Maryland, Baltimore, Baltimore, MD, USA
| |
Collapse
|
423
|
Kasenda B, Kühnl A, Chau I. Beginning of a novel frontier: T-cell-directed immune manipulation in lymphomas. Expert Rev Hematol 2016; 9:123-35. [DOI: 10.1586/17474086.2016.1122513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
424
|
Abstract
The past several years can be considered a renaissance era in the treatment of metastatic melanoma. Following a 30-year stretch in which oncologists barely put a dent in a very grim overall survival (OS) rate for these patients, things have rapidly changed course with the recent approval of three new melanoma drugs by the FDA. Both oncogene-targeted therapy and immune checkpoint blockade approaches have shown remarkable efficacy in a subset of melanoma patients and have clearly been game-changers in terms of clinical impact. However, most patients still succumb to their disease, and thus, there remains an urgent need to improve upon current therapies. Fortunately, innovations in molecular medicine have led to many silent gains that have greatly increased our understanding of the nature of cancer biology as well as the complex interactions between tumors and the immune system. They have also allowed for the first time a detailed understanding of an individual patient's cancer at the genomic and proteomic level. This information is now starting to be employed at all stages of cancer treatment, including diagnosis, choice of drug therapy, treatment monitoring, and analysis of resistance mechanisms upon recurrence. This new era of personalized medicine will foreseeably lead to paradigm shifts in immunotherapeutic treatment approaches such as individualized cancer vaccines and adoptive transfer of genetically modified T cells. Advances in xenograft technology will also allow for the testing of drug combinations using in vivo models, a truly necessary development as the number of new drugs needing to be tested is predicted to skyrocket in the coming years. This chapter will provide an overview of recent technological developments in cancer research, and how they are expected to impact future diagnosis, monitoring, and development of novel treatments for metastatic melanoma.
Collapse
Affiliation(s)
| | | | | | - Patrick Hwu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Gregory Lizée
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
425
|
Ji Y, Hocker JD, Gattinoni L. Enhancing adoptive T cell immunotherapy with microRNA therapeutics. Semin Immunol 2015; 28:45-53. [PMID: 26710685 DOI: 10.1016/j.smim.2015.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 12/15/2022]
Abstract
Adoptive T cell-based immunotherapies can mediate complete and durable regressions in patients with advanced cancer, but current response rates remain inadequate. Maneuvers to improve the fitness and antitumor efficacy of transferred T cells have been under extensive exploration in the field. Small non-coding microRNAs have emerged as critical modulators of immune system homeostasis and T cell immunity. Here, we summarize recent advances in our understanding of the role of microRNAs in regulating T cell activation, differentiation, and function. We also discuss how microRNA therapeutics could be employed to fine-tune T cell receptor signaling and enhance T cell persistence and effector functions, paving the way for the next generation of adoptive immunotherapies.
Collapse
Affiliation(s)
- Yun Ji
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA.
| | - James D Hocker
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA
| | - Luca Gattinoni
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, USA.
| |
Collapse
|
426
|
Ma X, Yan W, Zheng H, Du Q, Zhang L, Ban Y, Li N, Wei F. Regulation of IL-10 and IL-12 production and function in macrophages and dendritic cells. F1000Res 2015; 4. [PMID: 26918147 PMCID: PMC4754024 DOI: 10.12688/f1000research.7010.1] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2015] [Indexed: 12/20/2022] Open
Abstract
Interleukin-10 and Interleukin-12 are produced primarily by pathogen-activated antigen-presenting cells, particularly macrophages and dendritic cells. IL-10 and IL-12 play very important immunoregulatory roles in host defense and immune homeostasis. Being anti- and pro-inflammatory in nature, respectively, their functions are antagonistically opposing. A comprehensive and in-depth understanding of their immunological properties and signaling mechanisms will help develop better clinical intervention strategies in therapy for a wide range of human disorders. Here, we provide an update on some emerging concepts, controversies, unanswered questions, and opinions regarding the immune signaling of IL-10 and IL-12.
Collapse
Affiliation(s)
- Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Wenjun Yan
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Hua Zheng
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Qinglin Du
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Lixing Zhang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Yi Ban
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY, USA
| | - Na Li
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Science and Biotechnology, Shanghai Jiaotong University, Shanghai, USA
| |
Collapse
|
427
|
Lorentzen CL, Straten PT. CD19-Chimeric Antigen Receptor T Cells for Treatment of Chronic Lymphocytic Leukaemia and Acute Lymphoblastic Leukaemia. Scand J Immunol 2015; 82:307-19. [PMID: 26099639 DOI: 10.1111/sji.12331] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 06/14/2015] [Indexed: 02/02/2023]
Abstract
Adoptive cell therapy (ACT) for cancer represents a promising new treatment modality. ACT based on the administration of cytotoxic T cells genetically engineered to express a chimeric antigen receptor (CAR) recognizing CD19 expressed by B cell malignancies has been shown to induce complete lasting responses in patients with chronic lymphocytic leukaemia (CLL) and acute lymphoblastic leukaemia (ALL). So far, eleven clinical trials including 99 CLL and ALL patients treated with CAR T cells targeting CD19 have been published, and the results from these trials are promising with impressive clinical responses in heavily pretreated patients. Thus, CAR T cell therapy has induced complete responses in both CLL and ALL, and surprisingly, current results indicate that patients with ALL are more prone to respond than are CLL patients. Importantly, the majority of CAR cell studies have observed severe therapy-associated toxicities, which needs attention. Herein we review current data and discuss key aspects of this powerful approach to treat and potentially cure B cell malignancies.
Collapse
Affiliation(s)
- C L Lorentzen
- University of Copenhagen, Copenhagen, Denmark.,Center for Cancer Immune Therapy (CCIT), Department of Hematology, 65Q9 Copenhagen University Hospital, Herlev, Denmark
| | - P T Straten
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, 65Q9 Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
428
|
Erker C, Harker-Murray P, Burke MJ. Emerging immunotherapy in pediatric lymphoma. Future Oncol 2015; 12:257-70. [PMID: 26616565 DOI: 10.2217/fon.15.282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hodgkin and non-Hodgkin lymphoma collectively are the third most common cancer diagnosed in children each year. For children who relapse or have refractory disease, outcomes remain poor. Immunotherapy has recently emerged as a novel approach to treat hematologic malignancies. The field has been rapidly expanding over the past few years broadening its armamentarium which now includes monoclonal antibodies, antibody-drug conjugates and cellular therapies including bispecific T-cell engagers and chimeric antigen receptor-engineered T cells. Many of these agents are in their infancy stages and only beginning to make their mark on lymphoma treatment while others have begun to show promising efficacy in relapsed disease. In this review, the authors provide an overview of current and emerging immunotherapies in the field of pediatric lymphoma.
Collapse
Affiliation(s)
- Craig Erker
- Division of Pediatric Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Paul Harker-Murray
- Division of Pediatric Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Michael J Burke
- Division of Pediatric Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
429
|
Whilding LM, Maher J. CAR T-cell immunotherapy: The path from the by-road to the freeway? Mol Oncol 2015; 9:1994-2018. [PMID: 26563646 PMCID: PMC5528729 DOI: 10.1016/j.molonc.2015.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptors are genetically encoded artificial fusion molecules that can re-program the specificity of peripheral blood polyclonal T-cells against a selected cell surface target. Unparallelled clinical efficacy has recently been demonstrated using this approach to treat patients with refractory B-cell malignancy. However, the approach is technically challenging and can elicit severe toxicity in patients. Moreover, solid tumours have largely proven refractory to this approach. In this review, we describe the important structural features of CARs and how this may influence function. Emerging clinical experience is summarized in both solid tumours and haematological malignancies. Finally, we consider the particular challenges imposed by solid tumours to the successful development of CAR T-cell immunotherapy, together with a number of innovative strategies that have been developed in an effort to reverse the balance in favour of therapeutic benefit.
Collapse
Affiliation(s)
- Lynsey M Whilding
- King's College London, King's Health Partners Integrated Cancer Centre, Department of Research Oncology, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK.
| | - John Maher
- King's College London, King's Health Partners Integrated Cancer Centre, Department of Research Oncology, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK; Department of Immunology, Barnet Hospital, Royal Free London NHS Foundation Trust, Barnet, Hertfordshire, EN5 3DJ, UK; Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
430
|
Maude S, Barrett DM. Current status of chimeric antigen receptor therapy for haematological malignancies. Br J Haematol 2015; 172:11-22. [PMID: 26560054 DOI: 10.1111/bjh.13792] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The field of adoptive cell transfer includes chimeric antigen receptor (CAR) engineered T cells, constructs that emerged from basic research into principles of immunology and have transformed into clinically effective therapies for haematological malignancies. T cells engineered to express these artificial receptors hold great promise, but also carry significant risk. While permanent genetic modification of mature T cells appears safe, modulating their in vivo function is difficult, partly because the robust response can trigger other arms of the immune system. Suicide systems and toxicity management with cytokine blockade or signal transduction modulators have emerged as a new frontier in this field, a far cry from early problems getting CAR T cells to work at all. Currently, clinical trials in patients with relapsed or refractory B cell malignancies treated with CD19-specific CAR T cells have induced durable remissions in adults and children. Results from these trials indicate that more work needs to be done to understand biomarkers of efficacy, the role of T cell persistence and how to integrate this care into standard practice. Cell therapy will not be a 'one size fits all' class of medicine, and here we will discuss the development of this therapy and important questions for its future.
Collapse
Affiliation(s)
- Shannon Maude
- Abramson Cancer Center and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David M Barrett
- Abramson Cancer Center and the Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
431
|
Characterization and Functional Analysis of scFv-based Chimeric Antigen Receptors to Redirect T Cells to IL13Rα2-positive Glioma. Mol Ther 2015; 24:354-363. [PMID: 26514825 DOI: 10.1038/mt.2015.199] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 10/23/2015] [Indexed: 12/30/2022] Open
Abstract
Immunotherapy with T cells expressing chimeric antigen receptors (CARs) is an attractive approach to improve outcomes for patients with glioblastoma (GBM). IL13Rα2 is expressed at a high frequency in GBM but not in normal brain, making it a promising CAR T-cell therapy target. IL13Rα2-specific CARs generated up to date contain mutated forms of IL13 as an antigen-binding domain. While these CARs target IL13Rα2, they also recognize IL13Rα1, which is broadly expressed. To overcome this limitation, we constructed a panel of IL13Rα2-specific CARs that contain the IL13Rα2-specific single-chain variable fragment (scFv) 47 as an antigen binding domain, short or long spacer regions, a transmembrane domain, and endodomains derived from costimulatory molecules and CD3.ζ (IL13Rα2-CARs). IL13Rα2-CAR T cells recognized IL13Rα2-positive target cells in coculture and cytotoxicity assays with no cross-reactivity to IL13Rα1. However, only IL13Rα2-CAR T cells with a short spacer region produced IL2 in an antigen-dependent fashion. In vivo, T cells expressing IL13Rα2-CARs with short spacer regions and CD28.ζ, 41BB.ζ, and CD28.OX40.ζ endodomains had potent anti-glioma activity conferring a significant survival advantage in comparison to mice that received control T cells. Thus, IL13Rα2-CAR T cells hold the promise to improve current IL13Rα2-targeted immunotherapy approaches for GBM and other IL13Rα2-positive malignancies.
Collapse
|
432
|
Morello A, Sadelain M, Adusumilli PS. Mesothelin-Targeted CARs: Driving T Cells to Solid Tumors. Cancer Discov 2015; 6:133-46. [PMID: 26503962 DOI: 10.1158/2159-8290.cd-15-0583] [Citation(s) in RCA: 324] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/08/2015] [Indexed: 12/19/2022]
Abstract
UNLABELLED Chimeric antigen receptors (CAR) are synthetic receptors that target T cells to cell-surface antigens and augment T-cell function and persistence. Mesothelin is a cell-surface antigen implicated in tumor invasion, which is highly expressed in mesothelioma and lung, pancreas, breast, ovarian, and other cancers. Its low-level expression in mesothelia, however, commands thoughtful therapeutic interventions. Encouragingly, recent clinical trials evaluating active immunization or immunoconjugates in patients with pancreatic adenocarcinoma or mesothelioma have shown responses without toxicity. Altogether, these findings and preclinical CAR therapy models using either systemic or regional T-cell delivery argue favorably for mesothelin CAR therapy in multiple solid tumors. SIGNIFICANCE Recent success obtained with adoptive transfer of CAR T cells targeting CD19 in patients with refractory hematologic malignancies has generated much enthusiasm for T-cell engineering and raises the prospect of implementing similar strategies for solid tumors. Mesothelin is expressed in a wide range and a high percentage of solid tumors, which we review here in detail. Mesothelin CAR therapy has the potential to treat multiple solid malignancies.
Collapse
Affiliation(s)
- Aurore Morello
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Prasad S Adusumilli
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, New York. Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
433
|
Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2015; 22:17-22. [PMID: 26485445 DOI: 10.1016/j.bbmt.2015.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 01/21/2023]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in nontransplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
|
434
|
Leignadier J, Favre S, Luther SA, Luescher IF. CD8 engineered cytotoxic T cells reprogram melanoma tumor environment. Oncoimmunology 2015; 5:e1086861. [PMID: 27141342 DOI: 10.1080/2162402x.2015.1086861] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/18/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022] Open
Abstract
Cytotoxic T lymphocytes (CTL) from CD8β-deficient mice have powerful FasL-mediated cytotoxicity and IFNγ responses, but ablated Ca2+ and NFAT signaling, which can be restored by transduction with CD8β. Upon infection with lymphocytic choriomeningitis virus (LCMV), these cells yielded GP33-specific CTL (CD8βR) that exhibited high FasL/Fas-mediated cytotoxicity, IFNγ CXCL9 and 10 chemokine responses. Transfer of these cells in B16-GP33 tumor bearing mice resulted in (i) massive T cell tumor infiltration, (ii) strong reduction of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Treg) and IL-17-expressing T helper cells, (iii) maturation of tumor-associated antigen-presenting cells and (iv) production of endogenous, B16 melanoma-specific CTL that eradicated the tumor long after the transferred CD8βR CTL perished. Our study demonstrates that the synergistic combination of strong Fas/FasL mediated cytotoxicity, IFNγ and CXCL9 and 10 responses endows adoptively transferred CTL to reprogram the tumor environment and to thus enable the generation of endogenous, tumoricidal immunity.
Collapse
Affiliation(s)
- Julie Leignadier
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| | - Stephanie Favre
- Department of Biochemistry, University of Lausanne , Epalinges, Switzerland
| | - Sanjiv A Luther
- Department of Biochemistry, University of Lausanne , Epalinges, Switzerland
| | - Immanuel F Luescher
- Ludwig Center for Cancer Research, University of Lausanne , Epalinges, Switzerland
| |
Collapse
|
435
|
Abstract
Second-generation chimeric antigen receptors (CARs) retarget and reprogramme T cells to augment their antitumour efficacy. The combined activating and co-stimulatory domains incorporated in these CARs critically determine the function, differentiation, metabolism and persistence of engineered T cells. CD19-targeted CARs that incorporate CD28 or 4-1BB signalling domains are the best known to date. Both have shown remarkable complete remission rates in patients with refractory B cell malignancies. Recent data indicate that CD28-based CARs direct a brisk proliferative response and boost effector functions, whereas 4-1BB-based CARs induce a more progressive T cell accumulation that may compensate for less immediate potency. These distinct kinetic features can be exploited to further develop CAR-based T cell therapies for a variety of cancers. A new field of immunopharmacology is emerging.
Collapse
|
436
|
Abstract
Cancer immunotherapy was selected as the Breakthrough of the Year 2013 by the editors of Science, in part because of the successful treatment of refractory hematological malignancies with adoptive transfer of chimeric antigen receptor (CAR)-engineered T cells. Effective treatment of B cell leukemia may pave the road to future treatment of solid tumors, using similar approaches. The prostate expresses many unique proteins and, since the prostate gland is a dispensable organ, CAR T cells can potentially be used to target these tissue-specific antigens. However, the location and composition of prostate cancer metastases complicate the task of treating these tumors. It is therefore likely that more sophisticated CAR T cell approaches are going to be required for prostate metastasis than for B cell malignancies. Two main challenges that need to be resolved are how to increase the migration and infiltration of CAR T cells into prostate cancer bone metastases and how to counteract the immunosuppressive microenvironment found in bone lesions. Inclusion of homing (chemokine) receptors in CAR T cells may improve their recruitment to bone metastases, as may antibody-based combination therapies to normalize the tumor vasculature. Optimal activation of CAR T cells through the introduction of multiple costimulatory domains would help to overcome inhibitory signals from the tumor microenvironment. Likewise, combination therapy with checkpoint inhibitors that can reduce tumor immunosuppression may help improve efficacy. Other elegant approaches such as induced expression of immune stimulatory cytokines upon target recognition may also help to recruit other effector immune cells to metastatic sites. Although toxicities are difficult to predict in prostate cancer, severe on-target/off-tumor toxicities have been observed in clinical trials with use of CAR T cells against hematological malignancies; therefore, the choice of the target antigen is going to be crucial. This review focuses on different means of accomplishing maximal effectiveness of CAR T cell therapy for prostate cancer bone metastases while minimizing side effects and CAR T cell-associated toxicities. CAR T cell-based therapies for prostate cancer have the potential to be a therapy model for other solid tumors.
Collapse
|
437
|
Abstract
Twenty-five years after its inception, the genetic engineering of T cells is now a therapeutic modality pursued at an increasing number of medical centers. This immunotherapeutic strategy is predicated on gene transfer technology to instruct T lymphocytes to recognize and reject tumor cells. Chimeric antigen receptors (CARs) are synthetic receptors that mediate antigen recognition, T cell activation, and - in the case of second-generation CARs - costimulation to augment T cell functionality and persistence. We demonstrated over a decade ago that human T cells engineered with a CD19-specific CAR eradicated B cell malignancies in mice. Several phase I clinical trials eventually yielded dramatic results in patients with leukemia or lymphoma, especially acute lymphoblastic leukemia (ALL). This review recounts the milestones of CD19 CAR therapy and summarizes lessons learned from the CD19 paradigm.
Collapse
|
438
|
Pato A, Eisenberg G, Machlenkin A, Margalit A, Cafri G, Frankenburg S, Merims S, Peretz T, Lotem M, Gross G. Messenger RNA encoding constitutively active Toll-like receptor 4 enhances effector functions of human T cells. Clin Exp Immunol 2015. [PMID: 26212048 DOI: 10.1111/cei.12688] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Adoptive T cell therapy of cancer employs a large number of ex-vivo-propagated T cells which recognize their targets either by virtue of their endogenous T cell receptor (TCR) or via genetic reprogramming. However, both cell-extrinsic and intrinsic mechanisms often diminish the in-vivo potency of these therapeutic T cells, limiting their clinical efficacy and broader use. Direct activation of human T cells by Toll-like receptor (TLR) ligands induces T cell survival and proliferation, boosts the production of proinflammatory cytokines and augments resistance to regulatory T cell (Treg) suppression. Removal of the TLR ligand-binding region results in constitutive signalling triggered by the remaining cytosolic Toll/interleukin-1 receptor (TIR) domain. The use of such TIR domains therefore offers an ideal means for equipping anti-tumour T cells with the arsenal of functional attributes required for improving current clinical protocols. Here we show that constitutively active (ca)TLR-4 can be expressed efficiently in human T cells using mRNA electroporation. The mere expression of caTLR-4 mRNA in polyclonal CD8 and CD4 T cells induced the production of interferon (IFN)-γ, triggered the surface expression of CD25, CD69 and 4-1BB and up-regulated a panel of cytokines and chemokines. In tumour-infiltrating lymphocytes prepared from melanoma patients, caTLR-4 induced robust IFN-γ secretion in all samples tested. Furthermore, caTLR-4 enhanced the anti-melanoma cytolytic activity of tumour-infiltrating lymphocytes and augmented the secretion of IFN-γ, tumour necrosis factor (TNF)-α and granulocyte-macrophage colony-stimulating factor (GM-CSF) for at least 4 days post-transfection. Our results demonstrate that caTLR-4 is capable of exerting multiple T cell-enhancing effects and can potentially be used as a genetic adjuvant in adoptive cell therapy.
Collapse
Affiliation(s)
- A Pato
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat, Shmona.,Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - G Eisenberg
- Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - A Machlenkin
- Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - A Margalit
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat, Shmona.,Department of Biotechnology, Tel-Hai College, Upper, Galilee
| | - G Cafri
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat, Shmona.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - S Frankenburg
- Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - S Merims
- Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - T Peretz
- Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - M Lotem
- Sharett Institute of Oncology, Hadassah Hebrew University Hospital, Jerusalem
| | - G Gross
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat, Shmona.,Department of Biotechnology, Tel-Hai College, Upper, Galilee
| |
Collapse
|
439
|
Seyedin SN, Schoenhals JE, Lee DA, Cortez MA, Wang X, Niknam S, Tang C, Hong DS, Naing A, Sharma P, Allison JP, Chang JY, Gomez DR, Heymach JV, Komaki RU, Cooper LJ, Welsh JW. Strategies for combining immunotherapy with radiation for anticancer therapy. Immunotherapy 2015; 7:967-980. [PMID: 26310908 DOI: 10.2217/imt.15.65] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Radiation therapy controls local disease but also prompts the release of tumor-associated antigens and stress-related danger signals that primes T cells to promote tumor regression at unirradiated sites known as the abscopal effect. This may be enhanced by blocking inhibitory immune signals that modulate immune activity through a variety of mechanisms. Indeed, abscopal responses have occurred in patients with lung cancer or melanoma when given anti-CTLA4 antibody and radiation. Other approaches involve expanding and reinfusing T or NK cells or engineered T cells to express receptors that target specific tumor peptides. These approaches may be useful for immunocompromised patients receiving radiation. Preclinical and clinical studies are testing both immune checkpoint-based strategies and adoptive immunotherapies with radiation.
Collapse
Affiliation(s)
- Steven N Seyedin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jonathan E Schoenhals
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, TX, USA
| | - Dean A Lee
- Faculty, Graduate School of Biomedical Sciences, University of Texas Health Sciences Center, Houston, TX, USA
| | - Maria A Cortez
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, TX, USA
| | - Xiaohong Wang
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, TX, USA
| | - Sharareh Niknam
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, TX, USA
| | - Chad Tang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Padmanee Sharma
- Department of Immunology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James P Allison
- Department of Immunology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Daniel R Gomez
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ritsuko U Komaki
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Laurence J Cooper
- Department of Pediatrics, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
440
|
Heiblig M, Elhamri M, Michallet M, Thomas X. Adoptive immunotherapy for acute leukemia: New insights in chimeric antigen receptors. World J Stem Cells 2015; 7:1022-1038. [PMID: 26328018 PMCID: PMC4550626 DOI: 10.4252/wjsc.v7.i7.1022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/28/2014] [Accepted: 06/19/2015] [Indexed: 02/06/2023] Open
Abstract
Relapses remain a major concern in acute leukemia. It is well known that leukemia stem cells (LSCs) hide in hematopoietic niches and escape to the immune system surveillance through the outgrowth of poorly immunogenic tumor-cell variants and the suppression of the active immune response. Despite the introduction of new reagents and new therapeutic approaches, no treatment strategies have been able to definitively eradicate LSCs. However, recent adoptive immunotherapy in cancer is expected to revolutionize our way to fight against this disease, by redirecting the immune system in order to eliminate relapse issues. Initially described at the onset of the 90’s, chimeric antigen receptors (CARs) are recombinant receptors transferred in various T cell subsets, providing specific antigens binding in a non-major histocompatibility complex restricted manner, and effective on a large variety of human leukocyte antigen-divers cell populations. Once transferred, engineered T cells act like an expanding “living drug” specifically targeting the tumor-associated antigen, and ensure long-term anti-tumor memory. Over the last decades, substantial improvements have been made in CARs design. CAR T cells have finally reached the clinical practice and first clinical trials have shown promising results. In acute lymphoblastic leukemia, high rate of complete and prolonged clinical responses have been observed after anti-CD19 CAR T cell therapy, with specific but manageable adverse events. In this review, our goal was to describe CAR structures and functions, and to summarize recent data regarding pre-clinical studies and clinical trials in acute leukemia.
Collapse
|
441
|
Abstract
Immunotherapy is a promising treatment modality for cancer as it can promote specific and durable anti-cancer responses. However, limitations to current approaches remain. Therapeutics administered as soluble injections often require high doses and frequent re-dosing, which can result in systemic toxicities. Soluble bolus-based vaccine formulations typically elicit weak cellular immune responses, limiting their use for cancer. Current methods for ex vivo T cell expansion for adoptive T cell therapies are suboptimal, and achieving high T cell persistence and sustained functionality with limited systemic toxicity following transfer remains challenging. Biomaterials can play important roles in addressing some of these limitations. For example, nanomaterials can be employed as vehicles to deliver immune modulating payloads to specific tissues, cells, and cellular compartments with minimal off-target toxicity, or to co-deliver antigen and danger signal in therapeutic vaccine formulations. Alternatively, micro-to macroscale materials can be employed as devices for controlled molecular and cellular delivery, or as engineered microenvironments for recruiting and programming immune cells in situ. Recent work has demonstrated the potential for combining cancer immunotherapy and biomaterials, and the application of biomaterials to cancer immunotherapy is likely to enable the development of effective next-generation platforms. This review discusses the application of engineered materials for the delivery of immune modulating agents to the tumor microenvironment, therapeutic cancer vaccination, and adoptive T cell therapy.
Collapse
Affiliation(s)
- Alexander S. Cheung
- School of Engineering and Applied Sciences, and Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - David J. Mooney
- School of Engineering and Applied Sciences, and Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| |
Collapse
|
442
|
Barrett DM, Grupp SA, June CH. Chimeric Antigen Receptor- and TCR-Modified T Cells Enter Main Street and Wall Street. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:755-61. [PMID: 26188068 PMCID: PMC4507286 DOI: 10.4049/jimmunol.1500751] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The field of adoptive cell transfer (ACT) is currently comprised of chimeric Ag receptor (CAR)- and TCR-engineered T cells and has emerged from principles of basic immunology to paradigm-shifting clinical immunotherapy. ACT of T cells engineered to express artificial receptors that target cells of choice is an exciting new approach for cancer, and it holds equal promise for chronic infection and autoimmunity. Using principles of synthetic biology, advances in immunology, and genetic engineering have made it possible to generate human T cells that display desired specificities and enhanced functionalities. Clinical trials in patients with advanced B cell leukemias and lymphomas treated with CD19-specific CAR T cells have induced durable remissions in adults and children. The prospects for the widespread availability of engineered T cells have changed dramatically given the recent entry of the pharmaceutical industry to this arena. In this overview, we discuss some of the challenges and opportunities that face the field of ACT.
Collapse
Affiliation(s)
- David M Barrett
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | - Stephan A Grupp
- Division of Oncology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
443
|
Suzuki M, Curran KJ, Cheung NKV. Chimeric antigen receptors and bispecific antibodies to retarget T cells in pediatric oncology. Pediatr Blood Cancer 2015; 62:1326-36. [PMID: 25832831 PMCID: PMC4976492 DOI: 10.1002/pbc.25513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/20/2015] [Indexed: 12/22/2022]
Abstract
Cancer immunotherapy using antigen-specific T cells has broad therapeutic potential. Chimeric antigen receptors and bispecific antibodies can redirect T cells to kill tumors without human leukocyte antigens (HLA) restriction. Key determinants of clinical potential include the choice of target antigen, antibody specificity, antibody affinity, tumor accessibility, T cell persistence, and tumor immune evasion. For pediatric cancers, additional constraints include their propensity for bulky metastatic disease and the concern for late toxicities from treatment. Nonetheless, the recent preclinical and clinical developments of these T cell based therapies are highly encouraging.
Collapse
Affiliation(s)
- Maya Suzuki
- Department of Pediatrics; Memorial Sloan Kettering Cancer Center; 1275 York Avenue, New York 10065 NY
| | - Kevin J. Curran
- Department of Pediatrics; Memorial Sloan Kettering Cancer Center; 1275 York Avenue, New York 10065 NY
| | - Nai-Kong V. Cheung
- Department of Pediatrics; Memorial Sloan Kettering Cancer Center; 1275 York Avenue, New York 10065 NY
| |
Collapse
|
444
|
Wang M, Yin B, Wang HY, Wang RF. Current advances in T-cell-based cancer immunotherapy. Immunotherapy 2015; 6:1265-78. [PMID: 25524383 DOI: 10.2217/imt.14.86] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is a leading cause of death worldwide; due to the lack of ideal cancer biomarkers for early detection or diagnosis, most patients present with late-stage disease at the time of diagnosis, thus limiting the potential for successful treatment. Traditional cancer treatments, including surgery, chemotherapy and radiation therapy, have demonstrated very limited efficacy for patients with late-stage disease. Therefore, innovative and effective cancer treatments are urgently needed for cancer patients with late-stage and refractory disease. Cancer immunotherapy, particularly adoptive cell transfer, has shown great promise in the treatment of patients with late-stage disease, including those who are refractory to standard therapies. In this review, we will highlight recent advances and discuss future directions in adoptive cell transfer based cancer immunotherapy.
Collapse
Affiliation(s)
- Mingjun Wang
- Center for Inflammation & Epigenetics, Houston Methodist Research Institute, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
445
|
Abstract
PURPOSE OF REVIEW Over the last 18 months, substantial progress has been made in demonstrating the clinical efficacy of harnessing the immune system to treat a variety of both solid and hematologic malignancies. This review summarizes and evaluates these seminal studies. RECENT FINDINGS The two treatment modalities most responsible for the success of immune based therapies in cancer are adoptive T-cell therapy and immunoregulatory antibodies. Specifically, immunotherapy is generating responses in malignancies that would otherwise have no traditional curative options such as CD19-targeted chimeric antigen receptors to treat relapsed/refractory acute lymphocytic leukemia and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and PD-1 blockade alone or in combination to treat metastatic melanoma and other solid tumors. SUMMARY We are at a turning point for the field of cancer immunotherapy. The scientific community is now, after decades of research, proving that these treatments have great promise for patients. Ongoing preclinical research and clinical trials over the next few years will determine the extent of impact cancer immunotherapy will have on the treatment of the general population.
Collapse
|
446
|
Jackson SR, Yuan J, Teague RM. Targeting CD8+ T-cell tolerance for cancer immunotherapy. Immunotherapy 2015; 6:833-52. [PMID: 25290416 DOI: 10.2217/imt.14.51] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In the final issue of Science in 2013, the American Association of Science recognized progress in the field of cancer immunotherapy as the 'Breakthrough of the Year.' The achievements were actually twofold, owing to the early success of genetically engineered chimeric antigen receptors (CAR) and to the mounting clinical triumphs achieved with checkpoint blockade antibodies. While fundamentally very different, the common thread of these independent strategies is the ability to prevent or overcome mechanisms of CD8(+) T-cell tolerance for improved tumor immunity. Here we discuss how circumventing T-cell tolerance has provided experimental insights that have guided the field of clinical cancer immunotherapy to a place where real breakthroughs can finally be claimed.
Collapse
Affiliation(s)
- Stephanie R Jackson
- Saint Louis University School of Medicine, Department of Molecular Microbiology & Immunology, 1100 South Grand Blvd, St Louis, MO 63104, USA
| | | | | |
Collapse
|
447
|
Li HW, Andreola G, Carlson AL, Shao S, Lin CP, Zhao G, Sykes M. Rapid Functional Decline of Activated and Memory Graft-versus-Host-Reactive T Cells Encountering Host Antigens in the Absence of Inflammation. THE JOURNAL OF IMMUNOLOGY 2015; 195:1282-92. [PMID: 26085679 DOI: 10.4049/jimmunol.1401511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 05/22/2015] [Indexed: 01/04/2023]
Abstract
Inflammation in the priming host environment has critical effects on the graft-versus-host (GVH) responses mediated by naive donor T cells. However, it is unclear how a quiescent or inflammatory environment impacts the activity of GVH-reactive primed T and memory cells. We show in this article that GVH-reactive primed donor T cells generated in irradiated recipients had diminished ability compared with naive T cells to increase donor chimerism when transferred to quiescent mixed allogeneic chimeras. GVH-reactive primed T cells showed marked loss of cytotoxic function and activation, and delayed but not decreased proliferation or accumulation in lymphoid tissues when transferred to quiescent mixed chimeras compared with freshly irradiated secondary recipients. Primed CD4 and CD8 T cells provided mutual help to sustain these functions in both subsets. CD8 help for CD4 cells was largely IFN-γ dependent. TLR stimulation after transfer of GVH-reactive primed T cells to mixed chimeras restored their cytotoxic effector function and permitted the generation of more effective T cell memory in association with reduced PD-1 expression on CD4 memory cells. Our data indicate that an inflammatory host environment is required for the maintenance of GVH-reactive primed T cell functions and the generation of memory T cells that can rapidly acquire effector functions. These findings have important implications for graft-versus-host disease and T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Hao Wei Li
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Giovanna Andreola
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Alicia L Carlson
- Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114; and Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Steven Shao
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| | - Charles P Lin
- Advanced Microscopy Program, Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114; and Center for Systems Biology, Massachusetts General Hospital, Boston, MA 02114
| | - Guiling Zhao
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129
| | - Megan Sykes
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129;
| |
Collapse
|
448
|
Klinke DJ. Enhancing the discovery and development of immunotherapies for cancer using quantitative and systems pharmacology: Interleukin-12 as a case study. J Immunother Cancer 2015; 3:27. [PMID: 26082838 PMCID: PMC4468964 DOI: 10.1186/s40425-015-0069-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/28/2015] [Indexed: 12/22/2022] Open
Abstract
Recent clinical successes of immune checkpoint modulators have unleashed a wave of enthusiasm associated with cancer immunotherapy. However, this enthusiasm is dampened by persistent translational hurdles associated with cancer immunotherapy that mirror the broader pharmaceutical industry. Specifically, the challenges associated with drug discovery and development stem from an incomplete understanding of the biological mechanisms in humans that are targeted by a potential drug and the financial implications of clinical failures. Sustaining progress in expanding the clinical benefit provided by cancer immunotherapy requires reliably identifying new mechanisms of action. Along these lines, quantitative and systems pharmacology (QSP) has been proposed as a means to invigorate the drug discovery and development process. In this review, I discuss two central themes of QSP as applied in the context of cancer immunotherapy. The first theme focuses on a network-centric view of biology as a contrast to a "one-gene, one-receptor, one-mechanism" paradigm prevalent in contemporary drug discovery and development. This theme has been enabled by the advances in wet-lab capabilities to assay biological systems at increasing breadth and resolution. The second theme focuses on integrating mechanistic modeling and simulation with quantitative wet-lab studies. Drawing from recent QSP examples, large-scale mechanistic models that integrate phenotypic signaling-, cellular-, and tissue-level behaviors have the potential to lower many of the translational hurdles associated with cancer immunotherapy. These include prioritizing immunotherapies, developing mechanistic biomarkers that stratify patient populations and that reflect the underlying strength and dynamics of a protective host immune response, and facilitate explicit sharing of our understanding of the underlying biology using mechanistic models as vehicles for dialogue. However, creating such models require a modular approach that assumes that the biological networks remain similar in health and disease. As oncogenesis is associated with re-wiring of these biological networks, I also describe an approach that combines mechanistic modeling with quantitative wet-lab experiments to identify ways in which malignant cells alter these networks, using Interleukin-12 as an example. Collectively, QSP represents a new holistic approach that may have profound implications for how translational science is performed.
Collapse
Affiliation(s)
- David J Klinke
- Department of Chemical Engineering and Mary Babb Randolph Cancer Center, West Virginia University, Morgantown, WV 25606 USA
| |
Collapse
|
449
|
Haji-Fatahaliha M, Hosseini M, Akbarian A, Sadreddini S, Jadidi-Niaragh F, Yousefi M. CAR-modified T-cell therapy for cancer: an updated review. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1339-49. [PMID: 26068778 DOI: 10.3109/21691401.2015.1052465] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The use of chimeric antigen receptor (CAR)-modified T cells is a promising approach for cancer immunotherapy. These genetically modified receptors contain an antigen-binding moiety, a hinge region, a transmembrane domain, and an intracellular costimulatory domain resulting in T-cell activation subsequent to antigen binding. Optimal tumor removal through CAR-modified T cells requires suitable target antigen selection, co-stimulatory signaling domain, and the ability of CAR T cells to traffic, persist, and retain antitumor function after adoptive transfer. There are several elements which can improve antitumor function of CAR T cells, including signaling, conditioning chemotherapy and irradiation, tumor burden of the disease, T-cell phenotype, and supplementary cytokine usage. This review outlines four generations of CAR. The pre-clinical and clinical studies showed that this technique has a great potential for treatment of solid and hematological malignancies. The main purpose of the current review is to focus on the pre-clinical and clinical developments of CAR-based immunotherapy.
Collapse
Affiliation(s)
- Mostafa Haji-Fatahaliha
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Maryam Hosseini
- b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Asiye Akbarian
- d Department of Microbiology , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Sanam Sadreddini
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Farhad Jadidi-Niaragh
- e Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Yousefi
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran.,c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
450
|
Ji Y, Gattinoni L. miR-155 releases the brakes on antitumor T cells. Oncoimmunology 2015; 4:e1026533. [PMID: 26405586 DOI: 10.1080/2162402x.2015.1026533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022] Open
Abstract
Homeostatic γC cytokines are essential to support the expansion and function of tumor-specific T cells, but their effects are constrained by suppressor of cytokine signaling (SOCS) proteins as well as phosphoinositide and tyrosine-specific phosphatases. The microRNA miR-155 counteracts these inhibitory hurdles to potentiate intracellular cytokine signaling and T cell antitumor immunity.
Collapse
Affiliation(s)
- Yun Ji
- Experimental Transplantation and Immunology Branch; Center for Cancer Research; National Cancer Institute; National Institutes of Health ; Bethesda, MD USA
| | - Luca Gattinoni
- Experimental Transplantation and Immunology Branch; Center for Cancer Research; National Cancer Institute; National Institutes of Health ; Bethesda, MD USA
| |
Collapse
|