301
|
Abramson JS. Anti-CD19 CAR T-Cell Therapy for B-Cell Non-Hodgkin Lymphoma. Transfus Med Rev 2019; 34:29-33. [PMID: 31677848 DOI: 10.1016/j.tmrv.2019.08.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 07/31/2019] [Accepted: 08/09/2019] [Indexed: 01/24/2023]
Abstract
CAR T-cells are autologous T-cells transduced with a chimeric antigen receptor which targets the modified T-cell against a specified cancer antigen. Anti-CD19 CAR T-cells currently represent transformational therapy for relapsed/refractory aggressive B-cell lymphomas where durable remissions can be induced in patients with previously incurable chemotherapy-refractory disease. Three anti-CD19 CAR T-cells are currently Food and Drug Administration and European Medicines Agency approved or in advanced-stage development: axicabtagene ciloleucel, tisagenlecleucel, and lisocabtagene maraleucel. Although all targeting CD19 on the surface of malignant (and healthy) B-cells, these products differ from one another in multiple ways including construct, manufacturing, dose, design of pivotal clinical trials, and toxicity profile. Efficacy and safety data for anti-CD19 CAR T-cell therapy in aggressive B-cell lymphomas will be reviewed, as well as novel CAR T-cell designs and strategies for overcoming treatment resistance.
Collapse
Affiliation(s)
- Jeremy S Abramson
- Massachusetts General Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
302
|
Han C, Kwon BS. Chimeric antigen receptor T-cell therapy for cancer: a basic research-oriented perspective. Immunotherapy 2019; 10:221-234. [PMID: 29370727 DOI: 10.2217/imt-2017-0133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have outstanding therapeutic potential for treating blood cancers. The prospects for this technology have accelerated basic research, clinical translation and Big Pharma's investment in the field of T-cell therapeutics. This interest has led to the discovery of key factors that affect CAR T-cell efficacy and play pivotal roles in T-cell immunology. Herein, we introduce advances in adoptive immunotherapy and the birth of CAR T cells, and review CAR T-cell studies that focus on three important features: CAR constructs, target antigens and T-cell phenotypes. At last, we highlight novel strategies that overcome the tumor microenvironment and circumvent CAR T-cell side effects, and consider the future direction of CAR T-cell development.
Collapse
Affiliation(s)
- Chungyong Han
- Immunotherapeutics Branch, Division of Convergence Technology Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Byoung S Kwon
- Immunotherapeutics Branch, Division of Convergence Technology Research Institute, National Cancer Center, Goyang 10408, Korea.,Eutilex Co., Ltd, Suite #1401, Daeryung Technotown 17, Gasan digital 1-ro 25, Geumcheon-gu, Seoul 08594, Korea.,Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70118, USA
| |
Collapse
|
303
|
Coeshott C, Vang B, Jones M, Nankervis B. Large-scale expansion and characterization of CD3 + T-cells in the Quantum ® Cell Expansion System. J Transl Med 2019; 17:258. [PMID: 31391068 PMCID: PMC6686483 DOI: 10.1186/s12967-019-2001-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/23/2019] [Indexed: 12/27/2022] Open
Abstract
Background The rapid evolution of cell-based immunotherapies such as chimeric antigen receptor T-cells for treatment of hematological cancers has precipitated the need for a platform to expand these cells ex vivo in a safe, efficient, and reproducible manner. In the Quantum® Cell Expansion System (Quantum system) we evaluated the expansion of T-cells from healthy donors in a functionally-closed environment that reduces time and resources needed to produce a therapeutic dose. Methods Mononuclear cells from leukapheresis products from 5 healthy donors were activated with anti-CD3/CD28 Dynabeads® and expanded in the Quantum system for 8–9 days using xeno-free, serum-free medium and IL-2. Harvested cells were phenotyped by flow cytometry and evaluated for cytokine secretion by multiplex assays. Results From starting products of 30 or 85 × 106 mononuclear cells, CD3+ T-cell populations expanded over 500-fold following stimulation to provide yields up to 25 × 109 cells within 8 days. T-cell yields from all donors were similar in terms of harvest numbers, viability and doubling times. Functionality (secretion of IFN-γ, IL-2 and TNF-α) was retained in harvested T-cells upon restimulation in vitro and T-cells displayed therapeutically-relevant less-differentiated phenotypes of naïve and central memory T-cells, with low expression of exhaustion markers LAG-3 and PD-1. Conclusions The Quantum system has been successfully used to produce large quantities of functional T-cells at clinical dosing scale and within a short timeframe. This platform could have wide applicability for autologous and allogeneic cellular immunotherapies for the treatment of cancer. Electronic supplementary material The online version of this article (10.1186/s12967-019-2001-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claire Coeshott
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA.
| | - Boah Vang
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA
| | - Mark Jones
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA
| | - Brian Nankervis
- Terumo BCT, Inc., 10810 West Collins Avenue, Lakewood, CO, 80215, USA
| |
Collapse
|
304
|
Chimeric Antigen Receptor T-Cell Therapy Clinical Results in Pediatric and Young Adult B-ALL. Hemasphere 2019; 3:e279. [PMID: 31723849 PMCID: PMC6745916 DOI: 10.1097/hs9.0000000000000279] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 06/02/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cell therapy has revolutionized the care of patients with relapsed and refractory B-cell acute lymphoblastic leukemia (B-ALL). Results from clinical trials across multiple institutions report remarkable remission rates with CD19-directed CAR-modified T-cell therapy. These remissions are also proving to be durable in many patients with a relapse-free survival (RFS) of approximately 50% to 60% at 1 year across several trials and institutions in this population that has been historically very difficult to treat. In addition, new products are being developed to enhance upon the original CAR T-cell products, which include a humanized CAR, allogeneic CARs, and both CD22 and biallelic CD19 and CD22 constructs. Toxicity after CAR-modified T-cell therapy is characterized by cytokine release syndrome (CRS) and neurotoxicity in the acute post-infusion period and B-cell aplasia as a long-term consequence of treatment. This review will summarize the published data thus far on the use of CAR-modified T-cell therapy in pediatric B-ALL and outline the various CAR products now being developed for this population. Delivery of this therapy and the decision to pursue hematopoietic stem cell transplant (HSCT) after treatment will be discussed.
Collapse
|
305
|
Gao Q, Dong X, Xu Q, Zhu L, Wang F, Hou Y, Chao C. Therapeutic potential of CRISPR/Cas9 gene editing in engineered T-cell therapy. Cancer Med 2019; 8:4254-4264. [PMID: 31199589 PMCID: PMC6675705 DOI: 10.1002/cam4.2257] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 04/19/2019] [Accepted: 05/07/2019] [Indexed: 12/27/2022] Open
Abstract
Cancer patients have been treated with various types of therapies, including conventional strategies like chemo-, radio-, and targeted therapy, as well as immunotherapy like checkpoint inhibitors, vaccine and cell therapy etc. Among the therapeutic alternatives, T-cell therapy like CAR-T (Chimeric Antigen Receptor Engineered T cell) and TCR-T (T Cell Receptor Engineered T cell), has emerged as the most promising therapeutics due to its impressive clinical efficacy. However, there are many challenges and obstacles, such as immunosuppressive tumor microenvironment, manufacturing complexity, and poor infiltration of engrafted cells, etc still, need to be overcome for further treatment with different forms of cancer. Recently, the antitumor activities of CAR-T and TCR-T cells have shown great improvement with the utilization of CRISPR/Cas9 gene editing technology. Thus, the genome editing system could be a powerful genetic tool to use for manipulating T cells and enhancing the efficacy of cell immunotherapy. This review focuses on pros and cons of various gene delivery methods, challenges, and safety issues of CRISPR/Cas9 gene editing application in T-cell-based immunotherapy.
Collapse
Affiliation(s)
- Qianqian Gao
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
| | - Xuan Dong
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
| | - Qumiao Xu
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
| | - Linnan Zhu
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
| | - Fei Wang
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
- BGI Education CenterUniversity of Chinese Academy of Sciences, Beishan Industrial ZoneShenzhenChina
| | - Yong Hou
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
| | - Cheng‐chi Chao
- BGI‐Shenzhen, Beishan Industrial ZoneShenzhenChina
- Shenzhen Key Laboratory of GenomicsBeishan Industrial ZoneShenzhenChina
- Guangdong Enterprise Key Laboratory of Human Disease GenomicsBeishan Industrial ZoneShenzhenChina
- AbVision, IncMilpitasCalifornia
| |
Collapse
|
306
|
Azimi CS, Tang Q, Roybal KT, Bluestone JA. NextGen cell-based immunotherapies in cancer and other immune disorders. Curr Opin Immunol 2019; 59:79-87. [PMID: 31071513 DOI: 10.1016/j.coi.2019.03.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/29/2019] [Indexed: 12/27/2022]
Abstract
T lymphocyte and other cell therapies have the potential to transform how we treat cancers and other diseases that have few therapeutic options. Here, we review the current progress in engineered T cell therapies and look to the future of what will establish cell therapy as the next pillar of medicine. The tools of synthetic biology along with fundamental knowledge in cell biology and immunology have enabled the development of approaches to engineer cells with enhanced capacity to recognize and treat disease safely and effectively. This along with new modes of engineering cells with CRISPR and strategies to make universal 'off-the-shelf' cell therapies will provide more rapid, flexible, and cheaper translation to the clinic.
Collapse
Affiliation(s)
- Camillia S Azimi
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Qizhi Tang
- UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA; Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Kole T Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| | - Jeffrey A Bluestone
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
307
|
Jiang X, Xu J, Liu M, Xing H, Wang Z, Huang L, Mellor AL, Wang W, Wu S. Adoptive CD8 + T cell therapy against cancer:Challenges and opportunities. Cancer Lett 2019; 462:23-32. [PMID: 31356845 DOI: 10.1016/j.canlet.2019.07.017] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/11/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022]
Abstract
Cancer immunotherapy is a new and promising option for cancer treatment. Unlike traditional chemo- and radiotherapy, immunotherapy actives host immune system to attack malignancies, and this potentially offers long-term protection from recurrence with less toxicity in comparison to conventional chemo- and radiation therapy. In adoptive CD8+ T cell therapy (ACT), large numbers of tumor-specific T cells are sourced from patients and expanded in vitro and infused back to patients. T cells can be expanded from naturally-induced tumor-specific CD8+ T cells isolated from tumor infiltrating lymphocytes (TIL) or genetically-modified autologous circulating CD8+ T cells. The engineered T cells expressed tumor-specific antigen receptors including chimeric antigen receptors (CARs) and T cell receptors (TCRs), prepared from cultured B and T cell clones, respectively. The most successful ACT, anti-CD19 chimeric antigen receptor T (CAR-T) cell therapy directed against B cell lymphoma, is already approved for use based on evidence of efficacy. Efficacy of solid tumors is not yet forthcoming. This review summarizes current technology developments using ACT in clinical trials. In this review, differences between various ACT approaches are discussed. Furthermore, resistance factors in the tumor microenvironment are also considered, as are immune related adverse effects, critical clinic monitoring parameters and potential mitigation approaches.
Collapse
Affiliation(s)
- Xiaotao Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of Proteomics, Guangzhou, Guangdong, People's Republic of China.
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, People's Republic of China
| | - Mingfeng Liu
- Department of Breast, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Hui Xing
- Department of Obstetrics and Gynecology, Xiangyang Central Hospital, Xiangyang, Hubei, People's Republic of China.
| | - Zhiming Wang
- Sino-British Research Center for Molecular Oncology, National Center for the International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| | - Lei Huang
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-Upon-Tyne, United Kingdom.
| | - Andrew L Mellor
- Institute of Cellular Medicine, Faculty of Medical Sciences, Framlington Place, Newcastle University, Newcastle-Upon-Tyne, United Kingdom.
| | - Wei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic of China.
| | - Sha Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangdong Provincial Key Laboratory of Proteomics, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
308
|
Quach DH, Becerra-Dominguez L, Rouce RH, Rooney CM. A strategy to protect off-the-shelf cell therapy products using virus-specific T-cells engineered to eliminate alloreactive T-cells. J Transl Med 2019; 17:240. [PMID: 31340822 PMCID: PMC6657103 DOI: 10.1186/s12967-019-1988-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022] Open
Abstract
Background The use of “off-the-shelf” cellular therapy products derived from healthy donors addresses many of the challenges associated with customized cell products. However, the potential of allogeneic cell products to produce graft-versus-host disease (GVHD), and their likely rejection by host alloreactive T-cells are major barriers to their clinical safety and efficacy. We have developed a molecule that when expressed in T-cells, can eliminate alloreactive T-cells and hence can be used to protect cell therapy products from allospecific rejection. Further, expression of this molecule in virus-specific T-cells (VSTs) should virtually eliminate the potential for recipients to develop GVHD. Methods To generate a molecule that can mediate killing of cognate alloreactive T-cells, we fused beta-2 microglobulin (B2M), a universal component of all human leukocyte antigen (HLA) class I molecules, to the cytolytic endodomain of the T cell receptor ζ chain, to create a chimeric HLA accessory receptor (CHAR). To determine if CHAR-modified human VSTs could eliminate alloreactive T-cells, we co-cultured them with allogeneic peripheral blood mononuclear cells (PBMC), and assessed proliferation of PBMC-derived alloreactive T-cells and the survival of CHAR-modified VSTs by flow cytometry. Results The CHAR was able to transport HLA molecules to the cell surface of Daudi cells, that lack HLA class I expression due to defective B2M expression, illustrating its ability to complex with human HLA class I molecules. Furthermore, VSTs expressing CHAR were protected from allospecific elimination in co-cultures with allogeneic PBMCs compared to unmodified VSTs, and mediated killing of alloreactive T-cells. Unexpectedly, CHAR-modified VSTs eliminated not only alloreactive HLA class I restricted CD8 T-cells, but also alloreactive CD4 T-cells. This beneficial effect resulted from non-specific elimination of activated T-cells. Of note, we confirmed that CHAR-modified VSTs did not affect pathogen-specific T-cells which are essential for protective immunity. Conclusions Human T-cells can be genetically modified to eliminate alloreactive T-cells, providing a unique strategy to protect off-the-shelf cell therapy products. Allogeneic cell therapies have already proved effective in treating viral infections in the stem cell transplant setting, and have potential in other fields such as regenerative medicine. A strategy to prevent allograft rejection would greatly increase their efficacy and commercial viability. Electronic supplementary material The online version of this article (10.1186/s12967-019-1988-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David H Quach
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, 1102 Bates Ave, Suite 1770, Houston, TX, 77030, USA
| | - Luis Becerra-Dominguez
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, 1102 Bates Ave, Suite 1770, Houston, TX, 77030, USA
| | - Rayne H Rouce
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, 1102 Bates Ave, Suite 1770, Houston, TX, 77030, USA.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital and Baylor College of Medicine, 1102 Bates Ave, Suite 1770, Houston, TX, 77030, USA. .,Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular Virology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
309
|
Bertaina A, Roncarolo MG. Graft Engineering and Adoptive Immunotherapy: New Approaches to Promote Immune Tolerance After Hematopoietic Stem Cell Transplantation. Front Immunol 2019; 10:1342. [PMID: 31354695 PMCID: PMC6635579 DOI: 10.3389/fimmu.2019.01342] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 05/28/2019] [Indexed: 12/11/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative therapeutic option for a wide range of immune and hematologic malignant and non-malignant disorders. Once transplanted, allogeneic cells have to support myeloid repopulation and immunological reconstitution, but also need to become tolerant to the host via central or peripheral mechanisms to achieve the desired therapeutic effect. Peripheral tolerance after allogeneic HSCT may be achieved by several mechanisms, though blocking alloreactivity to the host human leukocyte antigens while preserving immune responses to pathogens and tumor antigens remains a challenge. Recently uncovered evidence on the mechanisms of post-HSCT immune reconstitution and tolerance in transplanted patients has allowed for the development of novel cell-based therapeutic approaches. These therapies are aimed at inducing long-term peripheral tolerance and reducing the risk of graft-vs-host disease (GvHD), while sparing the graft-vs-leukemia (GvL) effect. Thus, ensuring effective long term remission in hematologic malignancies. Today, haploidentical stem cell transplants have become a widely used treatment for patients with hematological malignancies. A myriad of ex vivo and in vivo T-cell depletion strategies have been adopted, with the goal of preventing GvHD while preserving GvL in the context of immunogenetic disparity. αβ T-cell/CD19 B-cell depletion techniques, in particular, has gained significant momentum, because of the high rate of leukemia-free survival and the low risk of severe GvHD. Despite progress, better treatments are still needed in a portion of patients to further reduce the incidence of relapse and achieve long-term tolerance. Current post-HSCT cell therapy approaches designed to induce tolerance and minimizing GvHD occurrence include the use of (i) γδ T cells, (ii) regulatory Type 1 T (Tr1) cells, and (iii) engineered FOXP3+ regulatory T cells. Future protocols may include post-HSCT infusion of allogeneic effector or regulatory T cells engineered with a chimeric antigen receptor (CAR). In the present review, we describe the most recent advances in graft engineering and post-HSCT adoptive immunotherapy.
Collapse
Affiliation(s)
- Alice Bertaina
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
| | - Maria Grazia Roncarolo
- Division of Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford School of Medicine, Stanford, CA, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, United States
| |
Collapse
|
310
|
Strohl WR, Naso M. Bispecific T-Cell Redirection versus Chimeric Antigen Receptor (CAR)-T Cells as Approaches to Kill Cancer Cells. Antibodies (Basel) 2019; 8:E41. [PMID: 31544847 PMCID: PMC6784091 DOI: 10.3390/antib8030041] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022] Open
Abstract
The concepts for T-cell redirecting bispecific antibodies (TRBAs) and chimeric antigen receptor (CAR)-T cells are both at least 30 years old but both platforms are just now coming into age. Two TRBAs and two CAR-T cell products have been approved by major regulatory agencies within the last ten years for the treatment of hematological cancers and an additional 53 TRBAs and 246 CAR cell constructs are in clinical trials today. Two major groups of TRBAs include small, short-half-life bispecific antibodies that include bispecific T-cell engagers (BiTE®s) which require continuous dosing and larger, mostly IgG-like bispecific antibodies with extended pharmacokinetics that can be dosed infrequently. Most CAR-T cells today are autologous, although significant strides are being made to develop off-the-shelf, allogeneic CAR-based products. CAR-Ts form a cytolytic synapse with target cells that is very different from the classical immune synapse both physically and mechanistically, whereas the TRBA-induced synapse is similar to the classic immune synapse. Both TRBAs and CAR-T cells are highly efficacious in clinical trials but both also present safety concerns, particularly with cytokine release syndrome and neurotoxicity. New formats and dosing paradigms for TRBAs and CAR-T cells are being developed in efforts to maximize efficacy and minimize toxicity, as well as to optimize use with both solid and hematologic tumors, both of which present significant challenges such as target heterogeneity and the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- William R Strohl
- BiStro Biotech Consulting, LLC, 1086 Tullo Farm Rd., Bridgewater, NJ 08807, USA.
| | - Michael Naso
- Century Therapeutics, 3675 Market St., Philadelphia, PA 19104, USA
| |
Collapse
|
311
|
Juillerat A, Tkach D, Busser BW, Temburni S, Valton J, Duclert A, Poirot L, Depil S, Duchateau P. Modulation of chimeric antigen receptor surface expression by a small molecule switch. BMC Biotechnol 2019; 19:44. [PMID: 31269942 PMCID: PMC6610870 DOI: 10.1186/s12896-019-0537-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 06/24/2019] [Indexed: 12/26/2022] Open
Abstract
Background Engineered therapeutic cells have attracted a great deal of interest due to their potential applications in treating a wide range of diseases, including cancer and autoimmunity. Chimeric antigen receptor (CAR) T-cells are designed to detect and kill tumor cells that present a specific, predefined antigen. The rapid expansion of targeted antigen beyond CD19, has highlighted new challenges, such as autoactivation and T-cell fratricide, that could impact the capacity to manufacture engineered CAR T-cells. Therefore, the development of strategies to control CAR expression at the surface of T-cells and their functions is under intense investigations. Results Here, we report the development and evaluation of an off-switch directly embedded within a CAR construct (SWIFF-CAR). The incorporation of a self-cleaving degradation moiety controlled by a protease/protease inhibitor pair allowed the ex vivo tight and reversible control of the CAR surface presentation and the subsequent CAR-induced signaling and cytolytic functions of the engineered T-cells using the cell permeable Asunaprevir (ASN) small molecule. Conclusions The strategy described in this study could, in principle, be broadly adapted to CAR T-cells development to circumvent some of the possible hurdle of CAR T-cell manufacturing. This system essentially creates a CAR T-cell with an integrated functional rheostat. Electronic supplementary material The online version of this article (10.1186/s12896-019-0537-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Diane Tkach
- Cellectis Inc, 430E, 29th street, New York, NY, 10016, USA
| | - Brian W Busser
- Cellectis Inc, 430E, 29th street, New York, NY, 10016, USA
| | - Sonal Temburni
- Cellectis Inc, 430E, 29th street, New York, NY, 10016, USA
| | - Julien Valton
- Cellectis Inc, 430E, 29th street, New York, NY, 10016, USA
| | | | - Laurent Poirot
- Cellectis, 8 rue de la croix Jarry, 75013, Paris, France
| | - Stéphane Depil
- Cellectis, 8 rue de la croix Jarry, 75013, Paris, France
| | | |
Collapse
|
312
|
Hill L, Lulla P, Heslop HE. CAR-T cell Therapy for Non-Hodgkin Lymphomas: A New Treatment Paradigm. ADVANCES IN CELL AND GENE THERAPY 2019; 2:e54. [PMID: 31777773 PMCID: PMC6880960 DOI: 10.1002/acg2.54] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/20/2019] [Indexed: 12/27/2022]
Abstract
The majority of patients with B-cell non-Hodgkin lymphoma (NHL) can be cured with standard chemoimmunotherapy. However, patients who fail first line therapy have dismal outcomes, particularly if they have disease that is resistant to salvage therapy, including chemoimmunotherapy, radiation and/or autologous stem cell transplantation. Indolent B-NHLs, such as follicular lymphoma (FL), although not generally considered curable may be treated over many years with good prognosis. However, a subset of B-NHLs can undergo histologic transformation into more aggressive subtypes with outcomes similar to aggressive B-NHLs. In recent years, T cells genetically modified with chimeric antigen receptors (CARs), have demonstrated a remarkable capacity to induce complete and durable clinical responses in patients with chemotherapy-refractory lymphomas. Indeed, two autologous CD19-directed CAR-modified T cell products have now been FDA-approved for the treatment of patients with relapsed or refractory diffuse large B-cell lymphoma (DLBCL), primary mediastinal B-cell lymphoma (PMBCL) and transformed FL, while a plethora of other CAR-T cell targets are being explored in ongoing clinical trials. The purpose of this review is to summarize the clinical efficacy and unique toxicities of individually developed CAR-T cell products for the treatment of lymphomas, and their evolution from the laboratory bench to commercialization.
Collapse
Affiliation(s)
- LaQuisa Hill
- The Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas
| | - Premal Lulla
- The Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas
| | - Helen E Heslop
- The Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, Texas
| |
Collapse
|
313
|
Puig-Saus C, Ribas A. Gene editing: Towards the third generation of adoptive T-cell transfer therapies. IMMUNO-ONCOLOGY TECHNOLOGY 2019; 1:19-26. [PMID: 35755321 PMCID: PMC9216344 DOI: 10.1016/j.iotech.2019.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
First-generation adoptive T-cell transfer (ACT) administering tumor-infiltrating lymphocytes (TILs), and second-generation ACT using autologous T cells genetically modified to express tumor-specific T-cell receptors (TCRs) or chimeric antigen receptors (CARs) have both shown promise for the treatment of several cancers, including melanoma, leukemia and lymphoma. However, these treatments require labor-intensive manufacturing of the cell product for each patient, frequently utilize lentiviral or retroviral vectors to genetically modify the T cells, and have limited antitumor efficacy in solid tumors. Gene editing is revolutionizing the field of gene therapy, and ACT is at the forefront of this revolution. Gene-editing technologies can be used to re-engineer the phenotype of T cells to increase their antitumor potency, to generate off-the-shelf ACT products, and to replace endogenous TCRs with tumor-specific TCRs or CARs using homology-directed repair (HDR) donor templates. Adeno-associated viral vectors or linear DNA have been used as HDR donor templates. Of note, non-viral delivery substantially reduces the time required to generate clinical-grade reagents for manufacture of T-cell products—a critical step for the translation of personalized T-cell therapies. These technological advances in the field using gene editing open the door to the third generation of ACT therapies. CRISPR-Cas9 allows the generation of tumor-specific T cells for adoptive T-cell transfer (ACT). Gene editing allows generation of off-the-shelf ACT products. Gene editing can tailor T-cell phenotype and increase antitumor potency. Non-viral gene editing is a requirement for personalized ACT. Personalized third-generation ACT: gene-edited neoantigen-specific T cells.
Collapse
Affiliation(s)
- Cristina Puig-Saus
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, USA
| | - Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, USA.,Division of Surgical Oncology, Department of Surgery, UCLA, Los Angeles, USA.,Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, USA
| |
Collapse
|
314
|
Ju A, Lee SW, Lee YE, Han KC, Kim JC, Shin SC, Park HJ, EunKyeong Kim E, Hong S, Jang M. A carrier-free multiplexed gene editing system applicable for suspension cells. Biomaterials 2019; 217:119298. [PMID: 31280073 DOI: 10.1016/j.biomaterials.2019.119298] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/22/2019] [Indexed: 12/26/2022]
Abstract
Genetically engineered cells via CRISPR/Cas9 system can serve as powerful sources for cancer immunotherapeutic applications. Furthermore, multiple genetic alterations are necessary to overcome tumor-induced immune-suppressive mechanisms. However, one of the major obstacles is the technical difficulty with efficient multiple gene manipulation of suspension cells due to the low transfection efficacy. Herein, we established a carrier-free multiplexed gene editing platform in a simplified method, which can enhance the function of cytotoxic CD8+ T cells by modulating suspension cancer cells. Our multiple Cas9 ribonucleoproteins (RNPs) enable simultaneous disruption of two programmed cell death 1 (PD-1) ligands, functioning as negative regulators in the immune system, by accessing engineered Cas9 proteins with abilities of complexation and cellular penetration. In addition, combination with electroporation enhanced multiple gene editing efficacy, compared with that by treatment of multiple Cas9 RNPs alone. This procedure resulted in high gene editing at multiple loci of suspension cells. The treatment of multiple Cas9 RNPs targeting both ligands strongly improved Th1-type cytokine production of cytotoxic CD8+ T cells, resulting in synergistic cytotoxic effects against cancer. Simultaneous suppression of PD-L1 and PD-L2 on cancer cells via our developed editing system allows effective anti-tumor immunity. Furthermore, the treatment of multiple Cas9 RNPs targeting PD-L1, PD-L2, and TIM-3 had approximately 70-90% deletion efficacy. Thus, our multiplexed gene editing strategy endows potential clinical utilities in cancer immunotherapy.
Collapse
Affiliation(s)
- Anna Ju
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sung Won Lee
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Young Eun Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 136-701, Republic of Korea
| | - Ki-Cheol Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jin-Chul Kim
- Natural Constituents of Research Center, Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - Sang Chul Shin
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyun Jung Park
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea
| | - Eunice EunKyeong Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seokmann Hong
- Department of Integrative Bioscience and Biotechnology, Institute of Anticancer Medicine Development, Sejong University, Seoul, 05006, Republic of Korea.
| | - Mihue Jang
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
315
|
Abstract
The successes with chimeric antigen receptor (CAR) T cell therapy in early clinical trials involving patients with pre-B cell acute lymphoblastic leukaemia (ALL) or B cell lymphomas have revolutionized anticancer therapy, providing a potentially curative option for patients who are refractory to standard treatments. These trials resulted in rapid FDA approvals of anti-CD19 CAR T cell products for both ALL and certain types of B cell lymphoma - the first approved gene therapies in the USA. However, growing experience with these agents has revealed that remissions will be brief in a substantial number of patients owing to poor CAR T cell persistence and/or cancer cell resistance resulting from antigen loss or modulation. Furthermore, the initial experience with CAR T cells has highlighted challenges associated with manufacturing a patient-specific therapy. Understanding the limitations of CAR T cell therapy will be critical to realizing the full potential of this novel treatment approach. Herein, we discuss the factors that can preclude durable remissions following CAR T cell therapy, with a primary focus on the resistance mechanisms that underlie disease relapse. We also provide an overview of potential strategies to overcome these obstacles in an effort to more effectively incorporate this unique therapeutic strategy into standard treatment paradigms.
Collapse
Affiliation(s)
- Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Terry J Fry
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
316
|
Hull CM, Maher J. Novel approaches to promote CAR T-cell function in solid tumors. Expert Opin Biol Ther 2019; 19:789-799. [DOI: 10.1080/14712598.2019.1614164] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Caroline M Hull
- School of Cancer and Pharmaceutical Sciences, King’s College London, Division of Cancer Studies, Guy’s Hospital, London, UK
| | - John Maher
- School of Cancer and Pharmaceutical Sciences, King’s College London, Division of Cancer Studies, Guy’s Hospital, London, UK
- Department of Clinical Immunology and Allergy, King’s College Hospital NHS Foundation Trust, London UK
- Department of Immunology, Eastbourne Hospital, Eastbourne, UK
| |
Collapse
|
317
|
Mardiana S, Lai J, House IG, Beavis PA, Darcy PK. Switching on the green light for chimeric antigen receptor T-cell therapy. Clin Transl Immunology 2019; 8:e1046. [PMID: 31073403 PMCID: PMC6500780 DOI: 10.1002/cti2.1046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/18/2022] Open
Abstract
Adoptive cellular therapy involving genetic modification of T cells with chimeric antigen receptor (CAR) transgene offers a promising strategy to broaden the efficacy of this approach for the effective treatment of cancer. Although remarkable antitumor responses have been observed following CAR T‐cell therapy in a subset of B‐cell malignancies, this has yet to be extended in the context of solid cancers. A number of promising strategies involving reprogramming the tumor microenvironment, increasing the specificity and safety of gene‐modified T cells and harnessing the endogenous immune response have been tested in preclinical models that may have a significant impact in patients with solid cancers. This review will discuss these exciting new developments and the challenges that must be overcome to deliver a more sustained and potent therapeutic response.
Collapse
Affiliation(s)
- Sherly Mardiana
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Junyun Lai
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Imran Geoffrey House
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Paul Andrew Beavis
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Phillip Kevin Darcy
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia.,Department of Pathology University of Melbourne Parkville VIC Australia.,Department of Immunology Monash University Clayton VIC Australia
| |
Collapse
|
318
|
Zanders ED, Svensson F, Bailey DS. Therapy for glioblastoma: is it working? Drug Discov Today 2019; 24:1193-1201. [PMID: 30878561 DOI: 10.1016/j.drudis.2019.03.008] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 02/06/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) remains one of the most intransigent of cancers, with a median overall survival of only 15 months after diagnosis. Drug treatments have largely proven ineffective; it is thought that this is related to the heterogeneous nature and plasticity of GBM-initiating stem cell lineages. Although many combination drug therapies are being positioned to address tumour heterogeneity, the most promising therapeutic approaches for GBM to date appear to be those targeting GBM by vaccination or antibody- and cell-based immunotherapy. We review the most recent clinical trials for GBM and discuss the role of adaptive clinical trials in developing personalised treatment strategies to address intra- and inter-tumoral heterogeneity.
Collapse
Affiliation(s)
- Edward D Zanders
- IOTA Pharmaceuticals Ltd, St John's Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK
| | - Fredrik Svensson
- IOTA Pharmaceuticals Ltd, St John's Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK
| | - David S Bailey
- IOTA Pharmaceuticals Ltd, St John's Innovation Centre, Cowley Road, Cambridge CB4 0WS, UK.
| |
Collapse
|
319
|
Qasim W. Allogeneic CAR T cell therapies for leukemia. Am J Hematol 2019; 94:S50-S54. [PMID: 30632623 DOI: 10.1002/ajh.25399] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/27/2022]
Abstract
Allogeneic chimeric antigen receptor T (CAR T) cells can offer advantages over autologous T cell therapies, including the availability of "fit" cells for production, and elimination of risks associated with inadvertent transduction of leukemic blasts. However, allogeneic T cell therapies must address HLA barriers and conventionally rely on the availability of a suitable HLA-matched donor if graft-vs-host-disease and rejection effects are to be avoided. More recently, the incorporation of additional genome editing manipulations, to disrupt T cell receptor expression and address other critical pathways have been explored. Clinical trials are underway investigating non-HLA matched T cells expressing anti-CD19 CARs for the treatment of B cell acute lymphoblastic leukemia (B-ALL) and anti-CD123 CAR for acute myeloid leukemia (AML). Such approaches continue to be refined and improved to widen accessibility and reduce the cost of T cell therapies for a wider range of conditions.
Collapse
Affiliation(s)
- Waseem Qasim
- University College London, Great Ormond Street Institute of Child Health London United Kingdom
| |
Collapse
|
320
|
Kamiya T, Seow SV, Wong D, Robinson M, Campana D. Blocking expression of inhibitory receptor NKG2A overcomes tumor resistance to NK cells. J Clin Invest 2019; 129:2094-2106. [PMID: 30860984 PMCID: PMC6486333 DOI: 10.1172/jci123955] [Citation(s) in RCA: 245] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
A key mechanism of tumor resistance to immune cells is mediated by expression of peptide-loaded HLA-E in tumor cells, which suppresses natural killer (NK) cell activity via ligation of the NK inhibitory receptor CD94/NKG2A. Gene expression data from approximately 10,000 tumor samples showed widespread HLAE expression, with levels correlating with those of KLRC1 (NKG2A) and KLRD1 (CD94). To bypass HLA-E inhibition, we developed a way to generate highly functional NK cells lacking NKG2A. Constructs containing a single-chain variable fragment derived from an anti-NKG2A antibody were linked to endoplasmic reticulum-retention domains. After retroviral transduction in human peripheral blood NK cells, these NKG2A Protein Expression Blockers (PEBLs) abrogated NKG2A expression. The resulting NKG2Anull NK cells had higher cytotoxicity against HLA-E-expressing tumor cells. Transduction of anti-NKG2A PEBL produced more potent cytotoxicity than interference with an anti-NKG2A antibody and prevented de novo NKG2A expression, without affecting NK cell proliferation. In immunodeficient mice, NKG2Anull NK cells were significantly more powerful than NKG2A+ NK cells against HLA-E-expressing tumors. Thus, NKG2A downregulation evades the HLA-E cancer immune-checkpoint, and increases the anti-tumor activity of NK cell infusions. Because this strategy is easily adaptable to current protocols for clinical-grade immune cell processing, its clinical testing is feasible and warranted.
Collapse
Affiliation(s)
- Takahiro Kamiya
- Department of Pediatrics and National University Cancer Institute Singapore, National University of Singapore, Singapore
| | - See Voon Seow
- Department of Pediatrics and National University Cancer Institute Singapore, National University of Singapore, Singapore
| | - Desmond Wong
- Department of Pediatrics and National University Cancer Institute Singapore, National University of Singapore, Singapore
| | | | - Dario Campana
- Department of Pediatrics and National University Cancer Institute Singapore, National University of Singapore, Singapore
| |
Collapse
|
321
|
Abstract
Genetically engineered T cells are powerful new medicines, offering hope for curative responses in patients with cancer. Chimeric antigen receptor (CAR) T cells were recently approved by the US Food and Drug Administration and are poised to enter the practice of medicine for leukemia and lymphoma, demonstrating that engineered immune cells can serve as a powerful new class of cancer therapeutics. The emergence of synthetic biology approaches for cellular engineering provides a broadly expanded set of tools for programming immune cells for enhanced function. Advances in T cell engineering, genetic editing, the selection of optimal lymphocytes, and cell manufacturing have the potential to broaden T cell-based therapies and foster new applications beyond oncology, in infectious diseases, organ transplantation, and autoimmunity.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
322
|
MacDonald KN, Piret JM, Levings MK. Methods to manufacture regulatory T cells for cell therapy. Clin Exp Immunol 2019; 197:52-63. [PMID: 30913302 DOI: 10.1111/cei.13297] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2019] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cell (Treg ) therapy has shown promise in early clinical trials for treating graft-versus-host disease, transplant rejection and autoimmune disorders. A challenge has been to isolate sufficiently pure Tregs and expand them to a clinical dose. However, there has been considerable progress in the development and optimization of these methods, resulting in a variety of manufacturing protocols being tested in clinical trials. In this review, we summarize methods that have been used to manufacture Tregs for clinical trials, including the choice of cell source and protocols for cell isolation and expansion. We also discuss alternative culture or genome editing methods for modulating Treg specificity, function or stability that could be applied to future clinical manufacturing protocols to increase the efficacy of Treg therapy.
Collapse
Affiliation(s)
- K N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - J M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, BC, Canada
| | - M K Levings
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
323
|
Foster JB, Barrett DM, Karikó K. The Emerging Role of In Vitro-Transcribed mRNA in Adoptive T Cell Immunotherapy. Mol Ther 2019; 27:747-756. [PMID: 30819612 PMCID: PMC6453504 DOI: 10.1016/j.ymthe.2019.01.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 01/29/2019] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Adoptive T cell therapy is a form of cellular therapy that utilizes human immune cells, often empowered by the expression of recombinant proteins, to attack selected targets present on tumor or infected cells. T cell-based immunotherapy has been progressing over the past several decades, and reached a milestone with the recent US Food and Drug Administration (FDA) approval of chimeric antigen receptor T cell therapy for relapsed and refractory leukemia and lymphoma. Although most studies have used viral vectors, a growing number of researchers have come to appreciate in vitro-transcribed (IVT) mRNA for the development, testing, and application of T cell-based immunotherapeutics. IVT mRNA offers inherent safety features, highly efficient recombinant protein translation, and the ability to control pharmacokinetic properties of the therapy. In this review, we discuss the history of IVT mRNA in adoptive T cell therapy, from tumor-infiltrating lymphocytes and T cell receptor-based therapies to chimeric antigen receptor therapy and gene-editing techniques, as well as prior and ongoing clinical trials.
Collapse
Affiliation(s)
- Jessica B Foster
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - David M Barrett
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Center for Childhood Cancer Research, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | |
Collapse
|
324
|
Sommer C, Boldajipour B, Kuo TC, Bentley T, Sutton J, Chen A, Geng T, Dong H, Galetto R, Valton J, Pertel T, Juillerat A, Gariboldi A, Pascua E, Brown C, Chin SM, Sai T, Ni Y, Duchateau P, Smith J, Rajpal A, Van Blarcom T, Chaparro-Riggers J, Sasu BJ. Preclinical Evaluation of Allogeneic CAR T Cells Targeting BCMA for the Treatment of Multiple Myeloma. Mol Ther 2019; 27:1126-1138. [PMID: 31005597 DOI: 10.1016/j.ymthe.2019.04.001] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Clinical success of autologous CD19-directed chimeric antigen receptor T cells (CAR Ts) in acute lymphoblastic leukemia and non-Hodgkin lymphoma suggests that CAR Ts may be a promising therapy for hematological malignancies, including multiple myeloma. However, autologous CAR T therapies have limitations that may impact clinical use, including lengthy vein-to-vein time and manufacturing constraints. Allogeneic CAR T (AlloCAR T) therapies may overcome these innate limitations of autologous CAR T therapies. Unlike autologous cell therapies, AlloCAR T therapies employ healthy donor T cells that are isolated in a manufacturing facility, engineered to express CARs with specificity for a tumor-associated antigen, and modified using gene-editing technology to limit T cell receptor (TCR)-mediated immune responses. Here, transcription activator-like effector nuclease (TALEN) gene editing of B cell maturation antigen (BCMA) CAR Ts was used to confer lymphodepletion resistance and reduced graft-versus-host disease (GvHD) potential. The safety profile of allogeneic BCMA CAR Ts was further enhanced by incorporating a CD20 mimotope-based intra-CAR off switch enabling effective CAR T elimination in the presence of rituximab. Allogeneic BCMA CAR Ts induced sustained antitumor responses in mice supplemented with human cytokines, and, most importantly, maintained their phenotype and potency after scale-up manufacturing. This novel off-the-shelf allogeneic BCMA CAR T product is a promising candidate for clinical evaluation.
Collapse
Affiliation(s)
- Cesar Sommer
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA.
| | - Bijan Boldajipour
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Tracy C Kuo
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Trevor Bentley
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Janette Sutton
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Amy Chen
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Tao Geng
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Holly Dong
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Roman Galetto
- Cellectis SA, 8 rue de la Croix Jarry, 75013 Paris, France
| | - Julien Valton
- Cellectis, Inc., 430 East 29th Street, New York, NY 10016, USA
| | - Thomas Pertel
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA
| | | | | | - Edward Pascua
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Colleen Brown
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Sherman M Chin
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Tao Sai
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Yajin Ni
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA
| | | | - Julianne Smith
- Cellectis, Inc., 430 East 29th Street, New York, NY 10016, USA
| | - Arvind Rajpal
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Thomas Van Blarcom
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Javier Chaparro-Riggers
- Pfizer Cancer Immunology Discovery, Pfizer Worldwide Research and Development, 230 E. Grand Avenue, South San Francisco, CA 94080, USA
| | - Barbra J Sasu
- Allogene Therapeutics, Inc., 210 E. Grand Avenue, South San Francisco, CA 94080, USA.
| |
Collapse
|
325
|
Emerging CRISPR/Cas9 applications for T-cell gene editing. Emerg Top Life Sci 2019; 3:261-275. [PMID: 33523139 DOI: 10.1042/etls20180144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 03/04/2019] [Accepted: 03/08/2019] [Indexed: 12/17/2022]
Abstract
Gene editing tools are being rapidly developed, accelerating many areas of cell and gene therapy research. Each successive gene editing technology promises increased efficacy, improved specificity, reduced manufacturing cost and design complexity; all of which are currently epitomised by the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein (Cas9) platform. Since its conceptualisation, CRISPR-based gene editing has been applied to existing methodologies and has further allowed the exploration of novel avenues of research. Implementation of CRISPR/Cas9 has been instrumental to recent progress in the treatment of cancer, primary immunodeficiency, and infectious diseases. To this end, T-cell therapies have attempted to harness and redirect antigen recognition function, and through gene editing, broaden T-cell targeting capabilities and enhance their potency. The purpose of this review is to provide insights into emerging applications of CRISPR/Cas9 in T-cell therapies, to briefly address concerns surrounding CRISPR-mediated indel formation, and to introduce CRISPR/Cas9 base editing technologies that hold vast potential for future research and clinical translation.
Collapse
|
326
|
Reisner Y, Or-Geva N. Veto cells for safer nonmyeloablative haploidentical HSCT and CAR T cell therapy. Semin Hematol 2019; 56:173-182. [PMID: 31202427 DOI: 10.1053/j.seminhematol.2019.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 03/18/2019] [Indexed: 12/15/2022]
Abstract
Haploidentical donors are a readily available source for mismatched hematopoietic bone marrow transplantation. The application of this regimen is constantly increasing with the advent of methods that overcome T-cell alloreactions that occur due to human-leukocyte-antigen disparity between host and donor. One successful method to overcome both graft rejection and graft-vs-host disease is transplantation of large numbers T-cell-depleted (TCD) haploidentical stem cell grafts (haploSCT), after myeloablative conditioning. The success of stem cell dose escalation is attributed to a unique immunoregulatory cell-property, termed "veto-activity." However, engraftment of mismatched hematopoietic stem cells following reduced-intensity conditioning still represents a major challenge. Here, we describe how the addition of post-transplant high-dose cyclophosphamide can promote immune tolerance induction after megadose TCD haploSCT, following nonmyeloablative conditioning. We also discuss ways of harnessing the immune regulatory properties of adoptively transferred "veto" cells to support mixed chimerism further and confer tolerance to cell-therapies, such as CAR-T cells. These approaches will soon be tested in phase 1-2 clinical studies and may prove to be a safe and efficacious treatment for many disorders such as hemoglobinopathies, autoimmune diseases, and as a prelude for organ tolerance. Moreover, this approach could pave the way for "off-the-shelf" cell-therapy agents, making them cheaper and easily obtainable.
Collapse
Affiliation(s)
- Yair Reisner
- Stem Cell Research, Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, TX.
| | - Noga Or-Geva
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Interdepartmental Program in Immunology, Stanford, TX
| |
Collapse
|
327
|
CAR-T Cells: Future Perspectives. Hemasphere 2019; 3:e188. [PMID: 31723827 PMCID: PMC6746028 DOI: 10.1097/hs9.0000000000000188] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 01/03/2023] Open
|
328
|
Liu J, Zhou G, Zhang L, Zhao Q. Building Potent Chimeric Antigen Receptor T Cells With CRISPR Genome Editing. Front Immunol 2019; 10:456. [PMID: 30941126 PMCID: PMC6433930 DOI: 10.3389/fimmu.2019.00456] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/20/2019] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have shown great promise in the treatment of hematological and solid malignancies. However, despite the success of this field, there remain some major challenges, including accelerated T cell exhaustion, potential toxicities, and insertional oncogenesis. To overcome these limitations, recent advances in CRISPR technology have enabled targetable interventions of endogenous genes in human CAR T cells. These CRISPR genome editing approaches have unleashed the therapeutic potential of CAR T cell therapy. Here, we summarize the potential benefits, safety concerns, and difficulties in the generation of gene-edited CAR T cells using CRISPR technology.
Collapse
Affiliation(s)
- Jie Liu
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Guangyu Zhou
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| | - Li Zhang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qi Zhao
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China.,Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
329
|
Affiliation(s)
- Matthew H Porteus
- From the Department of Pediatrics-Stem Cell Transplantation, Stanford University, Stanford, CA
| |
Collapse
|
330
|
Rahbarizadeh F, Ahmadvand D, Moghimi S. CAR T-cell bioengineering: Single variable domain of heavy chain antibody targeted CARs. Adv Drug Deliv Rev 2019; 141:41-46. [PMID: 31004624 DOI: 10.1016/j.addr.2019.04.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/05/2019] [Accepted: 04/15/2019] [Indexed: 10/27/2022]
Abstract
Redirecting the recognition specificity of T lymphocytes to designated tumour cell surface antigens by transferring chimeric antigen receptor (CAR) genes is becoming an effective strategy to combat cancer. Today, CAR T-cell therapy has proven successful in the treatment of haematological malignancies and the first CD19 CAR T-cell products has already entered the market. This success is expanding CAR design for broader malignancies including solid tumours. Nevertheless, CARs such as those built on antigen-specific single chain antibody variable fragment (scFv) may induce some adverse effects. Here, we briefly review CAR T-cell bioengineering and discuss selected important initiatives for improved T-cell reprogramming, function and safety. In this respect, we further elaborate on unconventional CARs structured on single variable domain of heavy chain (VHH) antibodies (single-domain antibodies) as an alternative to scFv, because of their interesting immunological and physicochemical characteristics and unique structure, which shows a high degree of homology with human VH3 gene family.
Collapse
|
331
|
Holzinger A, Abken H. CAR T Cells: A Snapshot on the Growing Options to Design a CAR. Hemasphere 2019; 3:e172. [PMID: 31723811 PMCID: PMC6745938 DOI: 10.1097/hs9.0000000000000172] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/14/2018] [Indexed: 12/27/2022] Open
Abstract
Adoptive cell therapy of malignant diseases with chimeric antigen receptor (CAR) modified T cells rapidly advanced from pre-clinical models to commercial approvals within 2 decades. CARs redirect patient's T cells towards cancer cells and activate the engineered cells for a cytolytic attack resulting in the destruction of the cognate target cell. CAR T cells have demonstrated their powerful capacities in inducing complete and lasting remissions of leukemia/lymphoma in an increasing number of trials worldwide. Since the early 90's, the design of CARs went through various steps of optimization until the very recent developments which include CARs with logic gating in the recognition of antigen patterns on target cells and TRUCKs with a target recognition induced delivery of immune modulating agents. Here we review the generations in CAR design, the impact of specific modifications, the strategies to improve the safety of CAR T cell therapy, and the challenges to adapt the CAR design for broader applications.
Collapse
Affiliation(s)
- Astrid Holzinger
- RCI, Regensburg Center for Interventional Immunology, Chair for Gene-Immune Therapy, University Hospital Regensburg, Regensburg, Germany
| | - Hinrich Abken
- RCI, Regensburg Center for Interventional Immunology, Chair for Gene-Immune Therapy, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
332
|
Sukari A, Abdallah N, Nagasaka M. Unleash the power of the mighty T cells-basis of adoptive cellular therapy. Crit Rev Oncol Hematol 2019; 136:1-12. [PMID: 30878123 DOI: 10.1016/j.critrevonc.2019.01.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/22/2019] [Accepted: 01/23/2019] [Indexed: 02/04/2023] Open
Abstract
Adoptive cellular therapy (ACT) is an immunotherapy which involves the passive transfer of lymphocytes into a lymphodepleted host after ex vivo stimulation and expansion. Tumor-infiltrating lymphocytes (TILs) have shown objective tumor responses mainly restricted to melanoma and rely on a laborious manufacturing process. These limitations led to emergence of engineered cells, where normal peripheral blood lymphocytes are modified to express T cell receptors (TCRs) or chimeric antigen receptors (CARs) specific for tumor-associated antigens (TAAs). To date, CD19-targeted chimeric antigen receptor T (CAR T) cells have been the most extensively studied, showing complete and durable responses in B-cell malignancies. Antitumor responses with engineered T cells have often been accompanied by undesired toxicities in clinical trials including cytokine release syndrome (CRS) and neurotoxicity. In this review, we provide an overview of adoptive cellular strategies, early and ongoing clinical trials, adverse events and strategies to mitigate side effects and overcome limitations.
Collapse
Affiliation(s)
- Ammar Sukari
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Nadine Abdallah
- Department of Internal Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Misako Nagasaka
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Advanced Medical Innovation, St. Marianna University Graduate School of Medicine, Kawasaki, Kanagawa, Japan
| |
Collapse
|
333
|
Lee JB, Kang H, Fang L, D'Souza C, Adeyi O, Zhang L. Developing Allogeneic Double-Negative T Cells as a Novel Off-the-Shelf Adoptive Cellular Therapy for Cancer. Clin Cancer Res 2019; 25:2241-2253. [DOI: 10.1158/1078-0432.ccr-18-2291] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/19/2018] [Accepted: 01/03/2019] [Indexed: 11/16/2022]
|
334
|
Guedan S, Calderon H, Posey AD, Maus MV. Engineering and Design of Chimeric Antigen Receptors. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 12:145-156. [PMID: 30666307 PMCID: PMC6330382 DOI: 10.1016/j.omtm.2018.12.009] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
T cells engineered with chimeric antigen receptors (CARs) have emerged as a potent new class of therapeutics for cancer, based on their remarkable potency in blood cancers. Since the first clinical reports of their efficacy emerged 7 years ago, investigators have focused on the mechanisms and properties that make CARs effective or toxic, and their effects on T cell biology. Novel CAR designs coupled with improvements in gene transfer technology, incorporating advances in gene editing, have the potential to increase access to engineered cell therapies, as well as improve their potency in solid tumors.
Collapse
Affiliation(s)
- Sonia Guedan
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Hugo Calderon
- Department of Hematology, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Avery D Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Parker Institute for Cellular Immunotherapy at the University of Pennsylvania, Philadelphia, PA, USA.,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
335
|
Lau CH. Applications of CRISPR-Cas in Bioengineering, Biotechnology, and Translational Research. CRISPR J 2018; 1:379-404. [PMID: 31021245 DOI: 10.1089/crispr.2018.0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
CRISPR technology is rapidly evolving, and the scope of CRISPR applications is constantly expanding. CRISPR was originally employed for genome editing. Its application was then extended to epigenome editing, karyotype engineering, chromatin imaging, transcriptome, and metabolic pathway engineering. Now, CRISPR technology is being harnessed for genetic circuits engineering, cell signaling sensing, cellular events recording, lineage information reconstruction, gene drive, DNA genotyping, miRNA quantification, in vivo cloning, site-directed mutagenesis, genomic diversification, and proteomic analysis in situ. It has also been implemented in the translational research of human diseases such as cancer immunotherapy, antiviral therapy, bacteriophage therapy, cancer diagnosis, pathogen screening, microbiota remodeling, stem-cell reprogramming, immunogenomic engineering, vaccine development, and antibody production. This review aims to summarize the key concepts of these CRISPR applications in order to capture the current state of play in this fast-moving field. The key mechanisms, strategies, and design principles for each technological advance are also highlighted.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Biomedical Engineering, City University of Hong Kong , Hong Kong, SAR, China
| |
Collapse
|
336
|
Zhao J, Lin Q, Song Y, Liu D. Universal CARs, universal T cells, and universal CAR T cells. J Hematol Oncol 2018; 11:132. [PMID: 30482221 PMCID: PMC6257951 DOI: 10.1186/s13045-018-0677-2] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 11/15/2018] [Indexed: 12/11/2022] Open
Abstract
Currently, the two approved T cell products with chimeric antigen receptors (CAR) are from autologous T cells. These CAR T cells approved for clinical use must be generated on a custom-made basis. This case-by-case autologous T cell production platform remains a significant limiting factor for large-scale clinical application due to the costly and lengthy production process. There is also an inherent risk of production failure. The individualized, custom-made autologous CAR T cell production process also posts constriction on the wide application on diverse tumor types. Therefore, universal allogeneic T cells are needed for the preparation of universal CAR T cells that can serve as the “off-the-shelf” ready-to-use therapeutic agents for large-scale clinical applications. Genome-editing technologies including ZFN (zinc finger nuclease), TALEN (transcription activator-like effector nuclease), and CRISPR-Cas9 are being used to generate the universal third-party T cells. In addition, split, universal, and programmable (SUPRA) CARs are being developed to enhance the flexibility and controllability of CAR T cells. The engineered universal T cells and universal CARs are paving the road for a totally new generation of CAR T cells capable of targeting multiple antigens and/ or being delivered to multiple recipients without re-editing of T cells. This may escalate to a new wave of revolution in cancer immunotherapy. This review summarized the latest advances on designs and development of universal CARs, universal T cells, and clinical application of universal CAR T cells.
Collapse
Affiliation(s)
- Juanjuan Zhao
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| | - Quande Lin
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| | - Yongping Song
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China
| | - Delong Liu
- The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008, China.
| |
Collapse
|
337
|
Chimeric antigen receptor-modified T cell therapy in chronic lymphocytic leukemia. J Hematol Oncol 2018; 11:130. [PMID: 30458878 PMCID: PMC6247712 DOI: 10.1186/s13045-018-0676-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 11/08/2018] [Indexed: 01/21/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL), a common type of B cell chronic lymphoproliferative disorder in adults, has witnessed enormous development in its treatment in recent years. New drugs such as ibrutinib, idelalisib, and venetoclax have achieved great success in treating relapsed and refractory (R/R) CLL. In addition, with the development of immunotherapy, chimeric antigen receptor-engineered T cells (CAR-T) therapy, a novel adoptive immune treatment, has also become more and more important in treating R/R CLL. It combines the advantages of T cells and B cells via ex vivo gene transfer technology and is able to bind targets recognized by specific antibodies without antigen presentation, thus breaking the restriction of major histocompatibility complex. So far, there have been lots of studies exploring the application of CAR-T therapy in CLL. In this review, we describe the structure of chimeric antigen receptor, the preclinical, and clinical results of CAR-T therapy against CLL, along with its adverse events and advances in efficacy.
Collapse
|
338
|
Thomas X, Paubelle E. Tisagenlecleucel-T for the treatment of acute lymphocytic leukemia. Expert Opin Biol Ther 2018; 18:1095-1106. [DOI: 10.1080/14712598.2018.1533951] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France
| | - Etienne Paubelle
- Hospices Civils de Lyon, Hematology Department, Lyon-Sud Hospital, Pierre Bénite, France
| |
Collapse
|
339
|
Graham C, Jozwik A, Pepper A, Benjamin R. Allogeneic CAR-T Cells: More than Ease of Access? Cells 2018; 7:cells7100155. [PMID: 30275435 PMCID: PMC6210057 DOI: 10.3390/cells7100155] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/27/2018] [Accepted: 09/27/2018] [Indexed: 02/07/2023] Open
Abstract
Patient derived anti-CD19 chimeric antigen receptor-T (CAR-T) cells are a powerful tool in achieving a complete remission in a range of B-cell malignancies, most notably B-acute lymphoblastic leukaemia (B-ALL) and diffuse large B-cell lymphoma (DLBCL). However, there are limitations, including inability to manufacture CAR-T cells from the patient's own T cells, disease progression and death prior to return of engineered cells. T cell dysfunction is known to occur in cancer patients, and several groups have recently described differences in CAR-T cells generated from chronic lymphocytic leukaemia (CLL) patients compared with those from a healthy donor. This is thought to contribute to the low response rate in this disease group. Healthy donor, gene-edited CAR-T cells which do not require human leucocyte antigen (HLA) matching have the potential to provide an 'off the shelf' product, overcoming the manufacturing difficulties of producing CAR-T cells for each individual patient. They may also provide a more functional, potent product for malignancies such as CLL, where T cell dysfunction is common and frequently cannot be fully reversed during the manufacturing process. Here we review the potential benefits and obstacles for healthy donor, allogeneic CAR-T cells.
Collapse
Affiliation(s)
- Charlotte Graham
- Department of Haematological Medicine, King's College London, London SE5 9NU, UK.
- Department of Haematology, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK.
| | - Agnieszka Jozwik
- Department of Haematological Medicine, King's College London, London SE5 9NU, UK.
| | - Andrea Pepper
- Department of Clinical and Experimental Medicine, Brighton and Sussex Medical School, University of Sussex, Falmer BN1 9PX, UK.
| | - Reuben Benjamin
- Department of Haematological Medicine, King's College London, London SE5 9NU, UK.
- Department of Haematology, King's College Hospital NHS Foundation Trust, London SE5 9RS, UK.
| |
Collapse
|
340
|
Han X, Wang Y, Han WD. Chimeric antigen receptor modified T-cells for cancer treatment. Chronic Dis Transl Med 2018; 4:225-243. [PMID: 30603741 PMCID: PMC6309024 DOI: 10.1016/j.cdtm.2018.08.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Indexed: 12/12/2022] Open
Abstract
T cells engineered with the chimeric antigen receptor (CAR) are rapidly emerging as an important immunotherapy for hematologic malignancies. The anti-cluster of differentiation (CD)19 CAR-T cell therapy has been remarkably successful against refractory/relapsed acute lymphoblastic leukemia (ALL), and a complete remission rate as high as 90% was observed, in both children and adults. Although the achievement of clinical efficacy using CAR-T cell therapy for solid tumors has encountered several obstacles that were associated with the multiple mechanisms contributing to an immunosuppressive microenvironment, investigators are exploring more optimized approaches to improve the efficiency of CAR-T in solid tumors. In addition, cytokine release syndrome (CRS) and neurotoxicity following CAR-T cell therapy can be severe or even fatal; therefore, the management of these toxicities is significant. Herein, we briefly review the structure of CAR-T and some novel CAR designs, the clinical application of CAR-T cell therapies, as well as the assessment and management of toxicities.
Collapse
Affiliation(s)
- Xiao Han
- Molecular & Immunological Department, Bio-therapeutic Department, The General Hospital of People's Liberation Army, Beijing 100853, China
| | - Yao Wang
- Molecular & Immunological Department, Bio-therapeutic Department, The General Hospital of People's Liberation Army, Beijing 100853, China
| | - Wei-Dong Han
- Molecular & Immunological Department, Bio-therapeutic Department, The General Hospital of People's Liberation Army, Beijing 100853, China
| |
Collapse
|
341
|
Jung IY, Lee J. Unleashing the Therapeutic Potential of CAR-T Cell Therapy Using Gene-Editing Technologies. Mol Cells 2018; 41:717-723. [PMID: 30110720 PMCID: PMC6125425 DOI: 10.14348/molcells.2018.0242] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/12/2018] [Accepted: 08/07/2018] [Indexed: 12/21/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy, an emerging immunotherapy, has demonstrated promising clinical results in hematological malignancies including B-cell malignancies. However, accessibility to this transformative medicine is highly limited due to the complex process of manufacturing, limited options for target antigens, and insufficient anti-tumor responses against solid tumors. Advances in gene-editing technologies, such as the development of Zinc Finger Nucleases (ZFNs), Transcription Activator-Like Effector Nucleases (TALENs), and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9), have provided novel engineering strategies to address these limitations. Development of next-generation CAR-T cells using gene-editing technologies would enhance the therapeutic potential of CAR-T cell treatment for both hematologic and solid tumors. Here we summarize the unmet medical needs of current CAR-T cell therapies and gene-editing strategies to resolve these challenges as well as safety concerns of gene-edited CAR-T therapies.
Collapse
|
342
|
Rein LA, Yang H, Chao NJ. Applications of Gene Editing Technologies to Cellular Therapies. Biol Blood Marrow Transplant 2018; 24:1537-1545. [DOI: 10.1016/j.bbmt.2018.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/23/2018] [Indexed: 12/26/2022]
|
343
|
Rasaiyaah J, Georgiadis C, Preece R, Mock U, Qasim W. TCRαβ/CD3 disruption enables CD3-specific antileukemic T cell immunotherapy. JCI Insight 2018; 3:99442. [PMID: 29997304 DOI: 10.1172/jci.insight.99442] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 06/06/2018] [Indexed: 12/27/2022] Open
Abstract
T cells engineered to express chimeric antigen receptors (CARs) against B cell antigens are being investigated as cellular immunotherapies. Similar approaches designed to target T cell malignancies have been hampered by the critical issue of T-on-T cytotoxicity, whereby fratricide or self-destruction of healthy T cells prohibits cell product manufacture. To date, there have been no reports of T cells engineered to target the definitive T cell marker, CD3 (3CAR). Recent improvements in gene editing now provide access to efficient disruption of such molecules on T cells, and this has provided a route to generation of 3CAR, CD3-specific CAR T cells. T cells were transduced with a lentiviral vector incorporating an anti-CD3ε CAR derived from OKT3, either before or after TALEN-mediated disruption of the endogenous TCRαβ/CD3 complex. Only transduction after disrupting assembly of TCRαβ/CD3 yielded viable 3CAR T cells, and these cultures were found to undergo self-enrichment for 3CAR+TCR-CD3- T cells without any further processing. Specific cytotoxicity against CD3ε was demonstrated against primary T cells and against childhood T cell acute lymphoblastic leukemia (T-ALL). 3CAR T cells mediated potent antileukemic effects in a human/murine chimeric model, supporting the application of cellular immunotherapy strategies against T cell malignancies. 3CAR provides a bridging strategy to achieve T cell eradication and leukemic remission ahead of conditioned allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Jane Rasaiyaah
- Molecular and Cellular Immunology Unit, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Christos Georgiadis
- Molecular and Cellular Immunology Unit, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Roland Preece
- Molecular and Cellular Immunology Unit, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Ulrike Mock
- Molecular and Cellular Immunology Unit, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Waseem Qasim
- Molecular and Cellular Immunology Unit, University College London (UCL) Great Ormond Street Institute of Child Health, London, United Kingdom.,Great Ormond Street Hospital BRC, London, United Kingdom
| |
Collapse
|
344
|
Singh N, Shi J, June CH, Ruella M. Genome-Editing Technologies in Adoptive T Cell Immunotherapy for Cancer. Curr Hematol Malig Rep 2018; 12:522-529. [PMID: 29039115 DOI: 10.1007/s11899-017-0417-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW In this review, we discuss the most recent developments in gene-editing technology and discuss their application to adoptive T cell immunotherapy. RECENT FINDINGS Engineered T cell therapies targeting cancer antigens have demonstrated significant efficacy in specific patient populations. Most impressively, CD19-directed chimeric antigen receptor T cells (CART19) have led to impressive responses in patients with B-cell leukemia and lymphoma. CTL019, or KYMRIAH™ (tisagenlecleucel), a CD19 CAR T cell product developed by Novartis and the University of Pennsylvania, was recently approved for clinical use by the Food and Drug Administration, representing a landmark in the application of adoptive T cell therapies. As CART19 enters routine clinical use, improving the efficacy of this exciting platform is the next step in broader application. Novel gene-editing technologies like CRISPR-Cas9 allow facile editing of specific genes within the genome, generating a powerful platform to further optimize the activity of engineered T cells.
Collapse
Affiliation(s)
- Nathan Singh
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Division of Hematology and Oncology, University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Junwei Shi
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA. .,Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. .,Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
345
|
Werner Sunderland M, Peggs KS. Successful translation and future prospects of TALEN editing for leukemia patients. Expert Opin Biol Ther 2018; 18:725-726. [PMID: 29860877 DOI: 10.1080/14712598.2018.1484105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Mariana Werner Sunderland
- a Cancer Immunology Unit , University College London Cancer Institute , London , UK.,b Research Department of Haematology , UCL Cancer Institute , London , UK
| | - Karl S Peggs
- a Cancer Immunology Unit , University College London Cancer Institute , London , UK.,b Research Department of Haematology , UCL Cancer Institute , London , UK
| |
Collapse
|
346
|
A Versatile Safeguard for Chimeric Antigen Receptor T-Cell Immunotherapies. Sci Rep 2018; 8:8972. [PMID: 29895885 PMCID: PMC5997667 DOI: 10.1038/s41598-018-27264-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/25/2018] [Indexed: 02/07/2023] Open
Abstract
CAR T-cell therapies hold great promise for treating a range of malignancies but are however challenged by the complexity of their production and by the adverse events related to their activity. Here we report the development of the CubiCAR, a tri-functional CAR architecture that enables CAR T-cell detection, purification and on-demand depletion by the FDA-approved antibody Rituximab. This novel architecture has the potential to streamline the manufacturing of CAR T-cells, allow their tracking and improve their overall safety.
Collapse
|
347
|
Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO Mol Med 2018; 9:1183-1197. [PMID: 28765140 PMCID: PMC5582407 DOI: 10.15252/emmm.201607485] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy, together with checkpoint inhibition, has been celebrated as a breakthrough technology due to the substantial benefit observed in clinical trials with patients suffering from relapsed or refractory B‐cell malignancies. In this review, we provide a comprehensive overview of the clinical trials performed so far worldwide and analyze parameters such as targeted antigen and indication, CAR molecular design, CAR T cell manufacturing, anti‐tumor activities, and related toxicities. More than 200 CAR T cell clinical trials have been initiated so far, most of which aim to treat lymphoma or leukemia patients using CD19‐specific CARs. An increasing number of studies address solid tumors as well. Notably, not all clinical trials conducted so far have shown promising results. Indeed, in a few patients CAR T cell therapy resulted in severe adverse events with fatal outcome. Of note, less than 10% of the ongoing CAR T cell clinical trials are performed in Europe. Taking lead from our analysis, we discuss the problems and general hurdles preventing efficient clinical development of CAR T cells as well as opportunities, with a special focus on the European stage.
Collapse
Affiliation(s)
- Jessica Hartmann
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany
| | - Martina Schüßler-Lenz
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, Langen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Attilio Bondanza
- Innovative immunotherapies, Ospedale San Raffaele, Milano, Italy
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Langen, Germany .,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
348
|
Urnov FD. Ctrl-Alt-inDel: genome editing to reprogram a cell in the clinic. Curr Opin Genet Dev 2018; 52:48-56. [PMID: 29879620 DOI: 10.1016/j.gde.2018.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/10/2018] [Accepted: 05/15/2018] [Indexed: 12/12/2022]
Abstract
Genome editing with engineered nucleases (zinc finger, TAL effector, or CRISPR/Cas9-based) enables `write' access to regulatory programs executed by primary human cells. A decade of its clinical development, along with a reduction of conventional gene therapy to medical and commercial practice, has made cell reprogramming via editing a viable clinical modality. Reviewed here are the first examples of this to enter the clinic: ex vivo edited T cells for infectious disease and cancer, and hematopoietic stem/progenitor cells for the hemoglobinopathies. Three ongoing developments will ensure that the range of edited and reprogrammed cells to enter the clinic, and the scope of target indications, will grow markedly in the next five years: our ability to identify disease-relevant targets in noncoding regulatory DNA, which is uniquely suited for editing-based cell program control; recent reduction to clinical practice of in vivo editing; and progress in engineering and manufacture of differentiated cells from pluripotent progenitors.
Collapse
Affiliation(s)
- Fyodor D Urnov
- Altius Institute for Biomedical Sciences, Seattle, WA, United States.
| |
Collapse
|
349
|
Gene-knocked out chimeric antigen receptor (CAR) T cells: Tuning up for the next generation cancer immunotherapy. Cancer Lett 2018; 423:95-104. [DOI: 10.1016/j.canlet.2018.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 12/15/2022]
|
350
|
Salmikangas P, Kinsella N, Chamberlain P. Chimeric Antigen Receptor T-Cells (CAR T-Cells) for Cancer Immunotherapy - Moving Target for Industry? Pharm Res 2018; 35:152. [PMID: 29855723 PMCID: PMC5982434 DOI: 10.1007/s11095-018-2436-z] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 05/23/2018] [Indexed: 12/19/2022]
Abstract
The first CD19 CAR T-cell products, Kymriah and Yescarta, are entering the US market and also being evaluated for marketing authorization in the EU. This breakthrough has expanded the interest and also investments towards novel chimeric antigen receptor (CAR) designs, both for hematological malignancies and solid tumors. At the same time, there is active development in moving from autologous products to allogeneic, off-the-shelf -products. New manufacturing technologies are also emerging for production of these complex genetically-modified cells and even decentralized manufacturing in hospitals is under consideration. However, the high potency of CAR T-cells is associated with toxicity and not all patients respond to the treatment. In addition, the number of patient and product variables impacting the clinical outcome is high. The race towards novel CAR T treatment options for cancer patients has begun, but without careful design of the constructs and overall understanding of the factors that impact the ultimate outcome in each case, the road towards commercial success may be long and winding. This review discusses the product- and patient-related variables that may pose challenges for the industry and developers both from the scientific and regulatory perspective.
Collapse
|