351
|
Cornel AM, van Til NP, Boelens JJ, Nierkens S. Strategies to Genetically Modulate Dendritic Cells to Potentiate Anti-Tumor Responses in Hematologic Malignancies. Front Immunol 2018; 9:982. [PMID: 29867960 PMCID: PMC5968097 DOI: 10.3389/fimmu.2018.00982] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic cell (DC) vaccination has been investigated as a potential strategy to target hematologic malignancies, while generating sustained immunological responses to control potential future relapse. Nonetheless, few clinical trials have shown robust long-term efficacy. It has been suggested that a combination of surmountable shortcomings, such as selection of utilized DC subsets, DC loading and maturation strategies, as well as tumor-induced immunosuppression may be targeted to maximize anti-tumor responses of DC vaccines. Generation of DC from CD34+ hematopoietic stem and progenitor cells (HSPCs) may provide potential in patients undergoing allogeneic HSPC transplantations for hematologic malignancies. CD34+ HSPC from the graft can be genetically modified to optimize antigen presentation and to provide sufficient T cell stimulatory signals. We here describe beneficial (gene)-modifications that can be implemented in various processes in T cell activation by DC, among which major histocompatibility complex (MHC) class I and MHC class II presentation, DC maturation and migration, cross-presentation, co-stimulation, and immunosuppression to improve anti-tumor responses.
Collapse
Affiliation(s)
- Annelisa M Cornel
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Niek P van Til
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Jaap Jan Boelens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands.,Pediatric Blood and Marrow Transplantation Program, University Medical Center Utrecht, Utrecht, Netherlands.,Blood and Marrow Transplantation Program, Princess Maxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Stefan Nierkens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
352
|
Tasian SK. Acute myeloid leukemia chimeric antigen receptor T-cell immunotherapy: how far up the road have we traveled? Ther Adv Hematol 2018; 9:135-148. [PMID: 29899889 DOI: 10.1177/2040620718774268] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy resistance and relapse remain significant sources of mortality for children and adults with acute myeloid leukemia (AML). Further intensification of conventional cytotoxic chemotherapy is likely not feasible due to the severity of acute and long-term side effects upon normal tissues commonly induced by these drugs. Successful development and implementation of new precision medicine treatment approaches for patients with AML, which may improve leukemia remission and diminish toxicity, is thus a major priority. Tumor antigen-redirected chimeric antigen receptor (CAR) T-cell immunotherapies have induced remarkable responses in patients with relapsed or chemorefractory B-lymphoblastic leukemia, and similar strategies are now under early clinical study in adults with relapsed/refractory AML. However, potential on target/off tumor toxicity of AML CAR T-cell immunotherapies, notably aplasia of normal myeloid cells, may limit broader implementation of such approaches. Careful selection of optimal target antigens, consideration of toxicity mitigation strategies, and development of methodologies to circumvent potential CAR T-cell resistance are essential for successful implementation of cellular immunotherapies for patients with high-risk AML.
Collapse
Affiliation(s)
- Sarah K Tasian
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania and Abramson Cancer Center, 3501 Civic Center Boulevard, CTRB, 3010, Philadelphia, PA, 19104, USA
| |
Collapse
|
353
|
Georgiadis C, Preece R, Nickolay L, Etuk A, Petrova A, Ladon D, Danyi A, Humphryes-Kirilov N, Ajetunmobi A, Kim D, Kim JS, Qasim W. Long Terminal Repeat CRISPR-CAR-Coupled "Universal" T Cells Mediate Potent Anti-leukemic Effects. Mol Ther 2018; 26:1215-1227. [PMID: 29605708 PMCID: PMC5993944 DOI: 10.1016/j.ymthe.2018.02.025] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/25/2018] [Accepted: 02/25/2018] [Indexed: 12/15/2022] Open
Abstract
Gene editing can be used to overcome allo-recognition, which otherwise limits allogeneic T cell therapies. Initial proof-of-concept applications have included generation of such "universal" T cells expressing chimeric antigen receptors (CARs) against CD19 target antigens combined with transient expression of DNA-targeting nucleases to disrupt the T cell receptor alpha constant chain (TRAC). Although relatively efficient, transgene expression and editing effects were unlinked, yields variable, and resulting T cell populations heterogeneous, complicating dosing strategies. We describe a self-inactivating lentiviral "terminal" vector platform coupling CAR expression with CRISPR/Cas9 effects through incorporation of an sgRNA element into the ΔU3 3' long terminal repeat (LTR). Following reverse transcription and duplication of the hybrid ΔU3-sgRNA, delivery of Cas9 mRNA resulted in targeted TRAC locus cleavage and allowed the enrichment of highly homogeneous (>96%) CAR+ (>99%) TCR- populations by automated magnetic separation. Molecular analyses, including NGS, WGS, and Digenome-seq, verified on-target specificity with no evidence of predicted off-target events. Robust anti-leukemic effects were demonstrated in humanized immunodeficient mice and were sustained longer than by conventional CAR+TCR+ T cells. Terminal-TRAC (TT) CAR T cells offer the possibility of a pre-manufactured, non-HLA-matched CAR cell therapy and will be evaluated in phase 1 trials against B cell malignancies shortly.
Collapse
Affiliation(s)
- Christos Georgiadis
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Roland Preece
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Lauren Nickolay
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Aniekan Etuk
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Anastasia Petrova
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK
| | - Dariusz Ladon
- NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, UK
| | | | | | | | - Daesik Kim
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Jin-Soo Kim
- Department of Chemistry, Seoul National University, Seoul, South Korea
| | - Waseem Qasim
- Molecular and Cellular Immunology Unit, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK; NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
354
|
Smith DM, Culme-Seymour EJ, Mason C. Evolving Industry Partnerships and Investments in Cell and Gene Therapies. Cell Stem Cell 2018; 22:623-626. [DOI: 10.1016/j.stem.2018.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
355
|
Köhl U, Arsenieva S, Holzinger A, Abken H. CAR T Cells in Trials: Recent Achievements and Challenges that Remain in the Production of Modified T Cells for Clinical Applications. Hum Gene Ther 2018; 29:559-568. [PMID: 29620951 DOI: 10.1089/hum.2017.254] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The adoptive transfer of chimeric antigen receptor (CAR)-modified T cells is attracting growing interest for the treatment of malignant diseases. Early trials with anti-CD19 CAR T cells have achieved spectacular remissions in B-cell leukemia and lymphoma, so far refractory, very recently resulting in the Food and Drug Administration approval of CD19 CAR T cells for therapy. With further applications and increasing numbers of patients, the reproducible manufacture of high-quality clinical-grade CAR T cells is becoming an ever greater challenge. New processing techniques, quality-control mechanisms, and logistic developments are required to meet both medical needs and regulatory restrictions. This paper summarizes the state-of-the-art in manufacturing CAR T cells and the current challenges that need to be overcome to implement this type of cell therapy in the treatment of a variety of malignant diseases and in a greater number of patients.
Collapse
Affiliation(s)
- Ulrike Köhl
- 1 Institute of Cellular Therapeutics , Hannover Medical School, Hannover, Germany.,2 Institute of Clinical Immunology, University Hospital Leipzig , Leipzig, Germany.,3 Fraunhofer Institute for Cell Therapy and Immunology , Leipzig, Germany
| | - Stanislava Arsenieva
- 1 Institute of Cellular Therapeutics , Hannover Medical School, Hannover, Germany.,2 Institute of Clinical Immunology, University Hospital Leipzig , Leipzig, Germany.,3 Fraunhofer Institute for Cell Therapy and Immunology , Leipzig, Germany
| | - Astrid Holzinger
- 4 Center for Molecular Medicine Cologne, University of Cologne , Cologne, Germany.,5 Department I for Internal Medicine, University Hospital Cologne , Cologne, Germany
| | - Hinrich Abken
- 4 Center for Molecular Medicine Cologne, University of Cologne , Cologne, Germany.,5 Department I for Internal Medicine, University Hospital Cologne , Cologne, Germany
| |
Collapse
|
356
|
McCreedy BJ, Senyukov VV, Nguyen KT. Off the shelf T cell therapies for hematologic malignancies. Best Pract Res Clin Haematol 2018; 31:166-175. [PMID: 29909917 DOI: 10.1016/j.beha.2018.03.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/02/2018] [Accepted: 03/08/2018] [Indexed: 10/17/2022]
Abstract
Adoptive transfer of autologous CAR-T cells can induce durable remissions in patients with relapsed/refractory hematologic malignancies. However, multiple challenges exist for manufacturing CAR-T cells from patients with advanced disease including inability to manufacture a product, disease progression or death while waiting for the CAR-T product to be available, and heterogeneity among autologous CAR-T products that contributes to unpredictable and variable clinical activity. Healthy donor T cells can provide a source for production of universal CAR-T cells when combined with gene editing to prevent expression of endogenous TCRs and avoid generation of GvHD in HLA mismatched recipients. Additional gene edits can be included to impart resistance to immunosuppression or improve trafficking to tumor sites. Recent advances in cell manufacturing and analytics technology can provide for consistent batch to batch manufacturing of gene edited allogeneic CAR-T cells in sufficient quantity to treat thousands of patients when needed as off the shelf products.
Collapse
Affiliation(s)
- Bruce J McCreedy
- Precision BioSciences, Inc., 302 East Pettigrew Street, Durham, NC 27701, USA.
| | - Vladimir V Senyukov
- Precision BioSciences, Inc., 302 East Pettigrew Street, Durham, NC 27701, USA.
| | - Kim T Nguyen
- Precision BioSciences, Inc., 302 East Pettigrew Street, Durham, NC 27701, USA.
| |
Collapse
|
357
|
Kamiya T, Wong D, Png YT, Campana D. A novel method to generate T-cell receptor-deficient chimeric antigen receptor T cells. Blood Adv 2018; 2:517-528. [PMID: 29507075 PMCID: PMC5851418 DOI: 10.1182/bloodadvances.2017012823] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 02/01/2018] [Indexed: 12/11/2022] Open
Abstract
Practical methods are needed to increase the applicability and efficacy of chimeric antigen receptor (CAR) T-cell therapies. Using donor-derived CAR-T cells is attractive, but expression of endogenous T-cell receptors (TCRs) carries the risk for graft-versus-host-disease (GVHD). To remove surface TCRαβ, we combined an antibody-derived single-chain variable fragment specific for CD3ε with 21 different amino acid sequences predicted to retain it intracellularly. After transduction in T cells, several of these protein expression blockers (PEBLs) colocalized intracellularly with CD3ε, blocking surface CD3 and TCRαβ expression. In 25 experiments, median TCRαβ expression in T lymphocytes was reduced from 95.7% to 25.0%; CD3/TCRαβ cell depletion yielded virtually pure TCRαβ-negative T cells. Anti-CD3ε PEBLs abrogated TCRαβ-mediated signaling, without affecting immunophenotype or proliferation. In anti-CD3ε PEBL-T cells, expression of an anti-CD19-41BB-CD3ζ CAR induced cytokine secretion, long-term proliferation, and CD19+ leukemia cell killing, at rates meeting or exceeding those of CAR-T cells with normal CD3/TCRαβ expression. In immunodeficient mice, anti-CD3ε PEBL-T cells had markedly reduced GVHD potential; when transduced with anti-CD19 CAR, these T cells killed engrafted leukemic cells. PEBL blockade of surface CD3/TCRαβ expression is an effective tool to prepare allogeneic CAR-T cells. Combined PEBL and CAR expression can be achieved in a single-step procedure, is easily adaptable to current cell manufacturing protocols, and can be used to target other T-cell molecules to further enhance CAR-T-cell therapies.
Collapse
Affiliation(s)
- Takahiro Kamiya
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Desmond Wong
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yi Tian Png
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Dario Campana
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
358
|
Vormittag P, Gunn R, Ghorashian S, Veraitch FS. A guide to manufacturing CAR T cell therapies. Curr Opin Biotechnol 2018; 53:164-181. [PMID: 29462761 DOI: 10.1016/j.copbio.2018.01.025] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/10/2018] [Accepted: 01/22/2018] [Indexed: 01/07/2023]
Abstract
In recent years, chimeric antigen receptor (CAR) modified T cells have been used as a treatment for haematological malignancies in several phase I and II trials and with Kymriah of Novartis and Yescarta of KITE Pharma, the first CAR T cell therapy products have been approved. Promising clinical outcomes have yet been tempered by the fact that many therapies may be prohibitively expensive to manufacture. The process is not yet defined, far from being standardised and often requires extensive manual handling steps. For academia, big pharma and contract manufacturers it is difficult to obtain an overview over the process strategies and their respective advantages and disadvantages. This review details current production processes being used for CAR T cells with a particular focus on efficacy, reproducibility, manufacturing costs and release testing. By undertaking a systematic analysis of the manufacture of CAR T cells from reported clinical trial data to date, we have been able to quantify recent trends and track the uptake of new process technology. Delivering new processing options will be key to the success of the CAR-T cells ensuring that excessive manufacturing costs do not disrupt the delivery of exciting new therapies to the wide possible patient cohort.
Collapse
Affiliation(s)
- Philipp Vormittag
- Karlsruhe Institute of Technology, Institute of Engineering in Life Sciences, Section IV: Biomolecular Separation Engineering, Fritz-Haber-Weg 2, 76131 Karlsruhe, Germany
| | - Rebecca Gunn
- University College London, Department of Biochemical Engineering, Gower Street, London WC1E 6BT, United Kingdom
| | - Sara Ghorashian
- Molecular and Cellular Immunology Section, UCL Institute of Child Health, 30 Guilford Street, London WC1N 1E, United Kingdom
| | - Farlan S Veraitch
- University College London, Department of Biochemical Engineering, Gower Street, London WC1E 6BT, United Kingdom.
| |
Collapse
|
359
|
Abstract
Chimeric antigen receptor (CAR) T-cells are redirected T-cells that can recognize cancer antigens in a major histocompatibility complex (MHC)-independent fashion. A typical CAR is comprised of two main functional domains: an extracellular antigen recognition domain, called a single-chain variable fragment (scFv), and an intracellular signaling domain. Based on the number of intracellular signaling molecules, CARs are categorized into four generations. CAR T-cell therapy has become a promising treatment for hematologic malignancies. However, results of its clinical trials on solid tumors have not been encouraging. Here, we described the structure of CARs and summarized the clinical trials of CD19-targeted CAR T-cells. The side effects, safety management, challenges, and future prospects of CAR T-cells for the treatment of cancer, particularly for solid tumors, were also discussed.
Collapse
Affiliation(s)
- Niaz Muhammad
- a Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences , Shaanxi Normal University , Xi'an , P.R. China
| | - Qinwen Mao
- b Department of Pathology , Northwestern University Feinberg School of Medicine , Chicago , IL , USA
| | - Haibin Xia
- a Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences , Shaanxi Normal University , Xi'an , P.R. China
| |
Collapse
|
360
|
Vivien R, Saïagh S, Lemarre P, Chabaud V, Jesson B, Godon C, Jarry U, Guillaume T, Chevallier P, Vié H, Clémenceau B. The doubling potential of T lymphocytes allows clinical-grade production of a bank of genetically modified monoclonal T-cell populations. Cytotherapy 2018; 20:436-452. [PMID: 29396255 DOI: 10.1016/j.jcyt.2017.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/29/2017] [Accepted: 12/02/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND AIMS To produce an anti-leukemic effect after hematopoietic stem cell transplantation we have long considered the theoretical possibility of using banks of HLA-DP specific T-cell clones transduced with a suicide gene. For that application as for any others, a clonal strategy is constrained by the population doubling (PD) potential of T cells, which has been rarely explored or exploited. METHODS We used clinical-grade conditions and two donors who were homozygous and identical for all HLA-alleles except HLA-DP. After mixed lymphocyte culture and transduction, we obtained 14 HLA-DP-specific T-cell clones transduced with the HSV-TK suicide gene. Clones were then selected on the basis of their specificity and functional characteristics and evaluated for their doubling potential. RESULTS After these steps of selection the clone NAT-DP4(TK), specific for HLA-DPB1*04:01/04:02, which produced high levels of interferon-γ (IFNγ), tumor necrosis factor (TNF), interleukin-2 (IL-2) and granulocyte-macrophage colony-stimulating factor (GM-CSF), was fully sequenced. It has two copies of the HSV-TK suicide transgene whose localizations were determined. Four billion NAT-DP4(TK) cells were frozen after 50 PDs. Thawed NAT-DP4(TK) cells retain the potential to undergo 50 additional PDs, a potential very far beyond that required to produce a biological effect. This PD potential was confirmed on 6/16 additional different T-cell clones. This type of well-defined clone can also support a second genetic modification with CAR constructs. CONCLUSION The possibility of choosing rare donors and exploiting the natural proliferative potential of T lymphocytes may dramatically reduce the clinical and immunologic complexity of adoptive transfer protocols that rely on the use of third-party T-cell populations.
Collapse
Affiliation(s)
- Régine Vivien
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM) 1232, Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France
| | - Soraya Saïagh
- Centre Hospitalier Universitaire (CHU) de Nantes, Hôtel Dieu, Unité de Thérapie Cellulaire et Génique (UTCG), Nantes, France
| | - Philippe Lemarre
- Centre Hospitalier Universitaire (CHU) de Nantes, Hôtel Dieu, Unité de Thérapie Cellulaire et Génique (UTCG), Nantes, France
| | | | - Béline Jesson
- Helixio Biopôle Clermont-Limagne, Saint-Beauzire, France
| | - Catherine Godon
- Centre Hospitalier Universitaire (CHU) de Nantes, Hôtel Dieu, Unité de Thérapie Cellulaire et Génique (UTCG), Nantes, France
| | - Ulrich Jarry
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM) 1232, Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France
| | - Thierry Guillaume
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM) 1232, Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France; Centre Hospitalier Universitaire (CHU) de Nantes, Hôtel Dieu, Unité de Thérapie Cellulaire et Génique (UTCG), Nantes, France
| | - Patrice Chevallier
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM) 1232, Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France; Centre Hospitalier Universitaire (CHU) de Nantes, Hôtel Dieu, Unité de Thérapie Cellulaire et Génique (UTCG), Nantes, France
| | - Henri Vié
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM) 1232, Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France; Etablissement Français du Sang (EFS), Pays de la Loire, Site de Nantes, Nantes, France.
| | - Béatrice Clémenceau
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM) 1232, Centre National de la Recherche Scientifique (CNRS), Université d'Angers, Université de Nantes, Nantes, France; Centre Hospitalier Universitaire (CHU) de Nantes, Hôtel Dieu, Unité de Thérapie Cellulaire et Génique (UTCG), Nantes, France.
| |
Collapse
|
361
|
Lo Presti V, Nierkens S, Boelens JJ, van Til NP. Use of cord blood derived T-cells in cancer immunotherapy: milestones achieved and future perspectives. Expert Rev Hematol 2018; 11:209-218. [PMID: 29359983 DOI: 10.1080/17474086.2018.1431119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Hematopoietic cell transplantation is a potentially lifesaving procedure for patients with hematological malignancies who are refractory to conventional chemotherapy and/or irradiation treatment. Umbilical cord blood (CB) transplantation, as a hematopoietic stem and progenitor cell (HSPC) source, has several advantages over bone marrow transplantation with respect to matching and prompt availability for transplantation. Additionally, CB has some inherent features, such as rapid expansion of T cells, lower prevalence of graft-versus-host disease and higher graft versus tumor efficacy that make this HSPC cell source more favorable over other HSPC sources. Areas covered: This review summarizes the current CB and CB derived T cell applications aiming to better disease control for hematological malignancies and discusses future directions to more effective therapies. Expert commentary: CB transplantation could be used as a platform to extract cord blood derived T cells for ex vivo expansion and/or gene modification to improve cellular immunotherapies. In addition, combining cord blood gene-engineered T cell products with vaccination strategies, such as cord blood derived dendritic cell based vaccines, may provide synergistic immunotherapies with enhanced anti-tumor effects.
Collapse
Affiliation(s)
- Vania Lo Presti
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Stefan Nierkens
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Jaap Jan Boelens
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands.,b Pediatric Blood and Marrow Transplantation Program , University Medical Center Utrecht , Utrecht , the Netherlands
| | - Niek P van Til
- a Laboratory of Translational Immunology , University Medical Center Utrecht , Utrecht , the Netherlands
| |
Collapse
|
362
|
Gomes-Silva D, Ramos CA. Cancer Immunotherapy Using CAR-T Cells: From the Research Bench to the Assembly Line. Biotechnol J 2018; 13:10.1002/biot.201700097. [PMID: 28960810 PMCID: PMC5966018 DOI: 10.1002/biot.201700097] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/20/2017] [Indexed: 11/08/2022]
Abstract
The focus of cancer treatment has recently shifted toward targeted therapies, including immunotherapy, which allow better individualization of care and are hoped to increase the probability of success for patients. Specifically, T cells genetically modified to express chimeric antigen receptors (CARs; CAR-T cells) have generated exciting results. Recent clinical successes with this cutting-edge therapy have helped to push CAR-T cells toward approval for wider use. However, several limitations need to be addressed before the widespread use of CAR-T cells as a standard treatment. Here, a succinct background on adoptive T-cell therapy (ATCT)is given. A brief overview of the structure of CARs, how they are introduced into T cells, and how CAR-T cell expansion and selection is achieved in vitro is then presented. Some of the challenges in CAR design are discussed, as well as the difficulties that arise in large-scale CAR-T cell manufacture that will need to be addressed to achieve successful commercialization of this type of cell therapy. Finally, developments already on the horizon are discussed.
Collapse
Affiliation(s)
- Diogo Gomes-Silva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, 77030, USA
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A Ramos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, 77030, USA
| |
Collapse
|
363
|
Qasim W, Zhan H, Samarasinghe S, Adams S, Amrolia P, Stafford S, Butler K, Rivat C, Wright G, Somana K, Ghorashian S, Pinner D, Ahsan G, Gilmour K, Lucchini G, Inglott S, Mifsud W, Chiesa R, Peggs KS, Chan L, Farzeneh F, Thrasher AJ, Vora A, Pule M, Veys P. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med 2018; 9:9/374/eaaj2013. [PMID: 28123068 DOI: 10.1126/scitranslmed.aaj2013] [Citation(s) in RCA: 655] [Impact Index Per Article: 93.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 09/13/2016] [Accepted: 01/03/2017] [Indexed: 12/17/2022]
Abstract
Autologous T cells engineered to express chimeric antigen receptor against the B cell antigen CD19 (CAR19) are achieving marked leukemic remissions in early-phase trials but can be difficult to manufacture, especially in infants or heavily treated patients. We generated universal CAR19 (UCART19) T cells by lentiviral transduction of non-human leukocyte antigen-matched donor cells and simultaneous transcription activator-like effector nuclease (TALEN)-mediated gene editing of T cell receptor α chain and CD52 gene loci. Two infants with relapsed refractory CD19+ B cell acute lymphoblastic leukemia received lymphodepleting chemotherapy and anti-CD52 serotherapy, followed by a single-dose infusion of UCART19 cells. Molecular remissions were achieved within 28 days in both infants, and UCART19 cells persisted until conditioning ahead of successful allogeneic stem cell transplantation. This bridge-to-transplantation strategy demonstrates the therapeutic potential of gene-editing technology.
Collapse
Affiliation(s)
- Waseem Qasim
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK. .,Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Hong Zhan
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sujith Samarasinghe
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Stuart Adams
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Persis Amrolia
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK.,Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Sian Stafford
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Katie Butler
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Christine Rivat
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Gary Wright
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Kathy Somana
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Sara Ghorashian
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Danielle Pinner
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Gul Ahsan
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Kimberly Gilmour
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Giovanna Lucchini
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Sarah Inglott
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - William Mifsud
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Robert Chiesa
- Great Ormond Street Hospital National Health Service Trust, London WC1N 1LE, UK
| | - Karl S Peggs
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Lucas Chan
- Division of Cancer Studies, Department of Haematological Medicine, King's College London, London SE5 9NU, UK
| | - Farzin Farzeneh
- Division of Cancer Studies, Department of Haematological Medicine, King's College London, London SE5 9NU, UK
| | - Adrian J Thrasher
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ajay Vora
- Sheffield Children's Hospital, Sheffield S10 2TH, UK
| | - Martin Pule
- Cancer Institute, University College London, London WC1E 6DD, UK
| | - Paul Veys
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
364
|
Posttransplant chimeric antigen receptor therapy. Blood 2018; 131:1045-1052. [PMID: 29358181 DOI: 10.1182/blood-2017-08-752121] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/18/2017] [Indexed: 12/27/2022] Open
Abstract
Therapeutic T-cell engineering is emerging as a powerful approach to treat refractory hematological malignancies. Its most successful embodiment to date is based on the use of second-generation chimeric antigen receptors (CARs) targeting CD19, a cell surface molecule found in most B-cell leukemias and lymphomas. Remarkable complete remissions have been obtained with autologous T cells expressing CD19 CARs in patients with relapsed, chemo-refractory B-cell acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. Allogeneic CAR T cells may also be harnessed to treat relapse after allogeneic hematopoietic stem cell transplantation. However, the use of donor T cells poses unique challenges owing to potential alloreactivity. We review different approaches to mitigate the risk of causing or aggravating graft-versus-host disease (GVHD), including CAR therapies based on donor leukocyte infusion, virus-specific T cells, T-cell receptor-deficient T cells, lymphoid progenitor cells, and regulatory T cells. Advances in CAR design, T-cell selection and gene editing are poised to enable the safe use of allogeneic CAR T cells without incurring GVHD.
Collapse
|
365
|
Legut M, Dolton G, Mian AA, Ottmann OG, Sewell AK. CRISPR-mediated TCR replacement generates superior anticancer transgenic T cells. Blood 2018; 131:311-322. [PMID: 29122757 PMCID: PMC5774207 DOI: 10.1182/blood-2017-05-787598] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/04/2017] [Indexed: 12/20/2022] Open
Abstract
Adoptive transfer of T cells genetically modified to express a cancer-specific T-cell receptor (TCR) has shown significant therapeutic potential for both hematological and solid tumors. However, a major issue of transducing T cells with a transgenic TCR is the preexisting expression of TCRs in the recipient cells. These endogenous TCRs compete with the transgenic TCR for surface expression and allow mixed dimer formation. Mixed dimers, formed by mispairing between the endogenous and transgenic TCRs, may harbor autoreactive specificities. To circumvent these problems, we designed a system where the endogenous TCR-β is knocked out from the recipient cells using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein-9 (Cas9) technology, simultaneously with transduction with a cancer-reactive receptor of choice. This TCR replacement strategy resulted in markedly increased surface expression of transgenic αβ and γδ TCRs, which in turn translated to a stronger, and more polyfunctional, response of engineered T cells to their target cancer cell lines. Additionally, the TCR-plus-CRISPR-modified T cells were up to a thousandfold more sensitive to antigen than standard TCR-transduced T cells or conventional model proxy systems used for studying TCR activity. Finally, transduction with a pan-cancer-reactive γδ TCR used in conjunction with CRISPR/Cas9 knockout of the endogenous αβ TCR resulted in more efficient redirection of CD4+ and CD8+ T cells against a panel of established blood cancers and primary, patient-derived B-cell acute lymphoblastic leukemia blasts compared with standard TCR transfer. Our results suggest that TCR transfer combined with genome editing could lead to new, improved generations of cancer immunotherapies.
Collapse
Affiliation(s)
- Mateusz Legut
- Division of Infection and Immunity, School of Medicine
- Systems Immunity Research Institute, and
| | - Garry Dolton
- Division of Infection and Immunity, School of Medicine
- Systems Immunity Research Institute, and
| | - Afsar Ali Mian
- Haematology, Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Oliver G Ottmann
- Haematology, Division of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Andrew K Sewell
- Division of Infection and Immunity, School of Medicine
- Systems Immunity Research Institute, and
| |
Collapse
|
366
|
Simon S, Charpentier M, Anegon I, Labarriere N. Immunotherapies in transplantation and cancer: 22nd NAT meeting/2nd NAT LabEx IGO joint meeting; 1-2 June 2017, Nantes, France. Immunotherapy 2018; 9:867-870. [PMID: 29338607 DOI: 10.2217/imt-2017-0094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This 22nd edition of the Nantes Actualités Transplantation annual meeting was co-organized for the second time with the LabEx Immuno-Graft Oncology network. This international meeting was held on 1 and 2 June 2017 in Nantes (western France). The topic of this 2-day meeting was 'Immunotherapies in transplantation and cancer'. This meeting brought together 17 international invited speakers, young researchers and 220 attendees mainly from Europe and North America. It was a unique opportunity to bring together the pioneers and leading immunologists in the fields of transplantation and cancer, focusing on shared mechanisms that control immune responses in organ or bone marrow transplantation and in cancer.
Collapse
Affiliation(s)
- Sylvain Simon
- CRCINA, Inserm UMR1232, 8 Quai Moncousu, 44007 Nantes Cedex 1, France.,LabEx IGO 'Immunotherapy, Graft, Oncology', Nantes, France
| | - Maud Charpentier
- CRCINA, Inserm UMR1232, 8 Quai Moncousu, 44007 Nantes Cedex 1, France.,LabEx IGO 'Immunotherapy, Graft, Oncology', Nantes, France
| | - Ignacio Anegon
- LabEx IGO 'Immunotherapy, Graft, Oncology', Nantes, France.,Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Nathalie Labarriere
- CRCINA, Inserm UMR1232, 8 Quai Moncousu, 44007 Nantes Cedex 1, France.,LabEx IGO 'Immunotherapy, Graft, Oncology', Nantes, France
| |
Collapse
|
367
|
Zych AO, Bajor M, Zagozdzon R. Application of Genome Editing Techniques in Immunology. Arch Immunol Ther Exp (Warsz) 2018; 66:289-298. [PMID: 29344676 PMCID: PMC6061149 DOI: 10.1007/s00005-018-0504-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/06/2018] [Indexed: 12/23/2022]
Abstract
The idea of using the effector immune cells to specifically fight cancer has recently evolved into an exciting concept of adoptive cell therapies. Indeed, genetically engineered T cells expressing on their surface recombinant, cancer-targeted receptors have been shown to induce promising response in oncological patients. However, in addition to exogenous expression of such receptors, there is also a need for disruption of certain genes in the immune cells to achieve more potent disease-targeted actions, to produce universal chimeric antigen receptor-based therapies or to study the signaling pathways in detail. In this review, we present novel genetic engineering methods, mainly TALEN and CRISPR/Cas9 systems, that can be used for such purposes. These unique techniques may contribute to creating more successful immune therapies against cancer or prospectively other diseases as well.
Collapse
Affiliation(s)
- Agata O Zych
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland.,Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Malgorzata Bajor
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Radoslaw Zagozdzon
- Department of Immunology, Transplantology and Internal Medicine, Medical University of Warsaw, Warsaw, Poland. .,Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland. .,Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland.
| |
Collapse
|
368
|
Xu D, Jin G, Chai D, Zhou X, Gu W, Chong Y, Song J, Zheng J. The development of CAR design for tumor CAR-T cell therapy. Oncotarget 2018; 9:13991-14004. [PMID: 29568411 PMCID: PMC5862632 DOI: 10.18632/oncotarget.24179] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 12/04/2017] [Indexed: 12/17/2022] Open
Abstract
In recent years, the chimeric antigen receptor modified T cells (Chimeric antigen receptor T cells, CAR-T) immunotherapy has developed rapidly, which has been considered the most promising therapy. Efforts to enhance the efficacy of CAR-based anti-tumor therapy have been made, such as the improvement of structures of CAR-T cells, including the development of extracellular antigen recognition receptors, intracellular co-stimulatory molecules and the combination application of CARs and synthetic small molecules. In addition, effects on the function of the CAR-T cells that the space distance between the antigen binding domains and tumor targets and the length of the spacer domains have are also being investigated. Given the fast-moving nature of this field, it is necessary to make a summary of the development of CAR-T cells. In this review, we mainly focus on the present design strategies of CAR-T cells with the hope that they can provide insights to increase the anti-tumor efficacy and safety.
Collapse
Affiliation(s)
- Dandan Xu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guoliang Jin
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaowan Zhou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Weiyu Gu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanyun Chong
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jingyuan Song
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
369
|
Toward Engineered Cells as Transformational and Broadly Available Medicines for the Treatment of Cancer. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
370
|
|
371
|
Abstract
Autologous, patient-specific chimeric antigen receptor T-cell (CART) therapy has emerged as a powerful and potentially curative therapy for cancer, especially for CD19-positive hematological malignancies. Indeed, on August 30, 2017, the University of Pennsylvania-designed CD19-directed CART (CART-19) cell therapy (CTL019, tisagenlecleucel-t, Kymriah - Novartis) became the first CART therapy approved by the Food and Drug Administration (FDA) for acute lymphoblastic leukemia. However, the development of CART technology and its wider application is partly limited by the patient-specific nature of such a platform and by the time required for manufacturing. The efficacious generation of universal allogeneic CART cells would overcome these limitations and represent a major advance in the field. However, several obstacles in the generation of universal CART cells need to be overcome, namely the risk of CART rejection and the risk of graft-versus-host disease mediated by the allogeneic CART. In this review, we discuss the different strategies being employed to generate universal CART and provide our perspective on the successful development of a truly off-the-shelf CART product.
Collapse
Affiliation(s)
- Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Smilow Center for Translational Research, 8-112, 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA.
| | - Saad S Kenderian
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
372
|
Yakoub-Agha I, Ferrand C, Chalandon Y, Ballot C, Castilla Llorente C, Deschamps M, Gauthier J, Labalette M, Larghero J, Maheux C, Moreau AS, Varlet P, Pétillon MO, Pinturaud M, Rubio MT, Chabannon C. Prérequis nécessaires pour la mise en place de protocoles de recherche clinique évaluant des thérapies cellulaires et géniques par lymphocytes T dotés de récepteur chimérique à l’antigène (CAR T-cells) : recommandations de la Société francophone de greffe de moelle et de thérapie cellulaire (SFGM-TC). Bull Cancer 2017; 104:S43-S58. [DOI: 10.1016/j.bulcan.2017.10.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 11/30/2022]
|
373
|
Or-Geva N, Gidron-Budovsky R, Radomir L, Edelstein Y, Singh AK, Sidlik-Muskatel R, Ophir E, Bachar-Lustig E, Reisner Y. Towards 'off-the-shelf' genetically modified T cells: prolonging functional engraftment in mice by CD8 veto T cells. Leukemia 2017; 32:1039-1041. [PMID: 29151584 DOI: 10.1038/leu.2017.332] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- N Or-Geva
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - R Gidron-Budovsky
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - L Radomir
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Y Edelstein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - A K Singh
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - R Sidlik-Muskatel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - E Ophir
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - E Bachar-Lustig
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Y Reisner
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
374
|
Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci Rep 2017; 7:14366. [PMID: 29085043 PMCID: PMC5662687 DOI: 10.1038/s41598-017-14749-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/17/2017] [Indexed: 01/04/2023] Open
Abstract
Adoptive transfer of high-affinity chimeric antigen receptor (CAR) T cells targeting hematological cancers has yielded impressive clinical results. However, safety concerns regarding target expression on healthy tissue and poor efficacy have hampered application to solid tumors. Here, a panel of affinity-variant CARs were constructed targeting overexpressed ICAM-1, a broad tumor biomarker, using its physiological ligand, LFA-1. Anti-tumor T cell potency in vitro was directly proportional to CAR affinity and ICAM-1 density. In a solid tumor mouse model allowing simultaneous monitoring of anti-tumor potency and systemic off-tumor toxicity, micromolar affinity CAR T cells demonstrated superior anti-tumor efficacy and safety compared to their nanomolar counterparts. Longitudinal T cell tracking by PET/CT and concurrent cytokine measurement revealed superior expansion and contraction kinetics of micromolar affinity CAR T cells. Therefore, we developed an ICAM-1 specific CAR with broad anti-tumor applicability that utilized a reduced affinity targeting strategy to significantly boost efficacy and safety.
Collapse
|
375
|
Gautron AS, Juillerat A, Guyot V, Filhol JM, Dessez E, Duclert A, Duchateau P, Poirot L. Fine and Predictable Tuning of TALEN Gene Editing Targeting for Improved T Cell Adoptive Immunotherapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2017; 9:312-321. [PMID: 29246309 PMCID: PMC5684446 DOI: 10.1016/j.omtn.2017.10.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/25/2022]
Abstract
Using a TALEN-mediated gene-editing approach, we have previously described a process for the large-scale manufacturing of “off-the-shelf” CAR T cells from third-party donor T cells by disrupting the gene encoding TCRα constant chain (TRAC). Taking advantage of a previously described strategy to control TALEN targeting based on the exclusion capacities of non-conventional RVDs, we have developed highly efficient and specific nucleases targeting a key T cell immune checkpoint, PD-1, to improve engineered CAR T cells’ functionalities. Here, we demonstrate that this approach allows combined TRAC and PDCD1 TALEN processing at the desired locus while eliminating low-frequency off-site processing. Thus, by replacing few RVDs, we provide here an easy and rapid redesign of optimal TALEN combinations. We anticipate that this method can greatly benefit multiplex editing, which is of key importance especially for therapeutic applications where high editing efficiencies need to be associated with maximal specificity and safety.
Collapse
Affiliation(s)
| | | | - Valérie Guyot
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| | | | - Emilie Dessez
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| | | | | | - Laurent Poirot
- Cellectis SA, 8 Rue de la Croix Jarry, 75013 Paris, France
| |
Collapse
|
376
|
Abramowski-Mock U, Delhove JM, Qasim W. Gene Modified T Cell Therapies for Hematological Malignancies. Hematol Oncol Clin North Am 2017; 31:913-926. [PMID: 28895856 DOI: 10.1016/j.hoc.2017.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This article focuses on clinical applications of T cells transduced to express recombinant T cell receptor and chimeric antigen receptor constructs directed toward hematological malignancies, and considers newer strategies incorporating gene-editing technologies to address GvHD and host-mediated rejection. Recent data from clinical trials are reviewed, and an overview is provided of current and emerging manufacturing processes; consideration is also given to new developments in the pipeline.
Collapse
Affiliation(s)
- Ulrike Abramowski-Mock
- Molecular and Cellular Immunology Unit, University college London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Juliette M Delhove
- Molecular and Cellular Immunology Unit, University college London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK
| | - Waseem Qasim
- Molecular and Cellular Immunology Unit, University college London, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, UK.
| |
Collapse
|
377
|
Abdeen AA, Saha K. Manufacturing Cell Therapies Using Engineered Biomaterials. Trends Biotechnol 2017; 35:971-982. [PMID: 28711155 PMCID: PMC5621598 DOI: 10.1016/j.tibtech.2017.06.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/09/2017] [Accepted: 06/13/2017] [Indexed: 02/06/2023]
Abstract
Emerging manufacturing processes to generate regenerative advanced therapies can involve extensive genomic and/or epigenomic manipulation of autologous or allogeneic cells. These cell engineering processes need to be carefully controlled and standardized to maximize safety and efficacy in clinical trials. Engineered biomaterials with smart and tunable properties offer an intriguing tool to provide or deliver cues to retain stemness, direct differentiation, promote reprogramming, manipulate the genome, or select functional phenotypes. This review discusses the use of engineered biomaterials to control human cell manufacturing. Future work exploiting engineered biomaterials has the potential to generate manufacturing processes that produce standardized cells with well-defined critical quality attributes appropriate for clinical testing.
Collapse
Affiliation(s)
- Amr A Abdeen
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - Krishanu Saha
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Medical History and Bioethics, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
378
|
Bak RO, Dever DP, Reinisch A, Cruz Hernandez D, Majeti R, Porteus MH. Multiplexed genetic engineering of human hematopoietic stem and progenitor cells using CRISPR/Cas9 and AAV6. eLife 2017; 6:e27873. [PMID: 28956530 PMCID: PMC5656432 DOI: 10.7554/elife.27873] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 09/26/2017] [Indexed: 12/21/2022] Open
Abstract
Precise and efficient manipulation of genes is crucial for understanding the molecular mechanisms that govern human hematopoiesis and for developing novel therapies for diseases of the blood and immune system. Current methods do not enable precise engineering of complex genotypes that can be easily tracked in a mixed population of cells. We describe a method to multiplex homologous recombination (HR) in human hematopoietic stem and progenitor cells and primary human T cells by combining rAAV6 donor delivery and the CRISPR/Cas9 system delivered as ribonucleoproteins (RNPs). In addition, the use of reporter genes allows FACS-purification and tracking of cells that have had multiple alleles or loci modified by HR. We believe this method will enable broad applications not only to the study of human hematopoietic gene function and networks, but also to perform sophisticated synthetic biology to develop innovative engineered stem cell-based therapeutics.
Collapse
Affiliation(s)
- Rasmus O Bak
- Department of PediatricsStanford UniversityStanfordUnited States
| | - Daniel P Dever
- Department of PediatricsStanford UniversityStanfordUnited States
| | - Andreas Reinisch
- Department of Medicine, Division of HematologyStanford UniversityStanfordUnited States
- Department of Medicine, Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordUnited States
- Department of Medicine, Cancer InstituteStanford UniversityStanfordUnited States
| | - David Cruz Hernandez
- Department of Medicine, Division of HematologyStanford UniversityStanfordUnited States
- Department of Medicine, Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordUnited States
- Department of Medicine, Cancer InstituteStanford UniversityStanfordUnited States
| | - Ravindra Majeti
- Department of Medicine, Division of HematologyStanford UniversityStanfordUnited States
- Department of Medicine, Institute for Stem Cell Biology and Regenerative MedicineStanford UniversityStanfordUnited States
- Department of Medicine, Cancer InstituteStanford UniversityStanfordUnited States
| | | |
Collapse
|
379
|
Chimeric Antigen Receptor T cells for B Cell Neoplasms: Choose the Right CAR for You. Curr Hematol Malig Rep 2017; 11:368-84. [PMID: 27475429 DOI: 10.1007/s11899-016-0336-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Genetic redirection of T lymphocytes allows us to unleash these potent cellular immune effectors against cancer. Chimeric antigen receptor (CAR) T cells are the best-in-class example that genetic engineering of T cells can lead to deep and durable responses, as has been shown in several clinical trials for CD19+ B cell malignancies. As a consequence, in the last few years, several academic institutions and commercial partners have started developing anti-CD19 CAR T cell products. Although most of these T cell products are highly effective in vivo, basic differences among them can generate different performance characteristics and thereby impact their long-term clinical outcome. Several strategies are being implemented in order to solve the current open issues of CART19 therapy: (i) increasing efficacy against indolent B cell leukemias and lymphomas, (ii) avoiding or preventing antigen-loss relapses, (iii) reducing and managing toxicity, and (iv) bringing this CART therapy to routine clinical practice. The field of CART therapies is thriving, and exciting new avenues are opening for both scientists and patients.
Collapse
|
380
|
Zhang Y, Mu W, Wang H. Gene editing in T cell therapy. J Genet Genomics 2017; 44:415-422. [DOI: 10.1016/j.jgg.2017.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/28/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
|
381
|
Chimeric antigen receptor T-cells for B-cell malignancies. Transl Res 2017; 187:59-82. [PMID: 28719798 DOI: 10.1016/j.trsl.2017.06.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/18/2017] [Accepted: 06/23/2017] [Indexed: 12/27/2022]
Abstract
The adoptive transfer of T-lymphocytes modified to express chimeric antigen receptors (CAR-Ts) has produced impressive clinical responses among patients with B-cell malignancies. This has led to a rapid expansion in the number of clinical trials over the past several years. Although CD19-specific CAR-Ts are the most extensively evaluated, CAR-Ts specific for other B-cell-associated targets have also shown promise. However, despite this success, toxicities associated with CAR-T administration remain a significant concern. There continues to be substantial heterogeneity among CAR-T products, and differences in both CAR designs and CAR-T production strategies can substantially affect clinical outcomes. Ongoing clinical studies will further elucidate these differences and many other innovative approaches are being evaluated at the preclinical level. In this review, we will discuss the background and rationale for the use of CAR-Ts, provide an overview of advances in the field, and examine the application of CAR-Ts to the treatment of B-cell malignancies, including a summary of clinical trials published to date.
Collapse
|
382
|
Baruch EN, Berg AL, Besser MJ, Schachter J, Markel G. Adoptive T cell therapy: An overview of obstacles and opportunities. Cancer 2017; 123:2154-2162. [PMID: 28543698 DOI: 10.1002/cncr.30491] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/12/2022]
Abstract
The therapeutic potential of adoptive cell therapy (ACT) in cancer patients was first acknowledged 3 decades ago, but it was an esoteric approach at the time. In recent years, technological advancements have transformed ACT into a viable therapeutic option that can be curative in some patients. In fact, current ACT response rates are 80% to 90% for hematological malignancies and 30% for metastatic melanoma refractory to multiple lines of therapy. Although these results are encouraging, there is still much to be done to fulfill ACT's potential, specifically with regard to improving clinical efficacy, expanding clinical indications, reducing toxicity, and increasing production and cost-effectiveness. This review addresses the current major obstacles to ACT and presents potential solutions. Cancer 2017;123:2154-62. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Erez Nissim Baruch
- The Ella Lemelbaum Institute of Immuno-oncology, Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Amy Lauren Berg
- The Ella Lemelbaum Institute of Immuno-oncology, Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel
| | - Michal Judith Besser
- The Ella Lemelbaum Institute of Immuno-oncology, Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Schachter
- The Ella Lemelbaum Institute of Immuno-oncology, Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gal Markel
- The Ella Lemelbaum Institute of Immuno-oncology, Institute of Oncology, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
383
|
Maartens JH, De-Juan-Pardo E, Wunner FM, Simula A, Voelcker NH, Barry SC, Hutmacher DW. Challenges and opportunities in the manufacture and expansion of cells for therapy. Expert Opin Biol Ther 2017; 17:1221-1233. [DOI: 10.1080/14712598.2017.1360273] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Joachim H. Maartens
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Elena De-Juan-Pardo
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Felix M. Wunner
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Antonio Simula
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
| | - Nicolas H. Voelcker
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Future Industries Institute, University of South Australia, Adelaide, Australia
| | - Simon C. Barry
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
- Molecular Immunology, Department of Gastroenterology, Women’s and Children’s Hospital, Adelaide, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Dietmar W. Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
- Cooperative Research Centre for Cell Therapy Manufacturing, Adelaide, Australia
- ARC Centre in Additive Biomanufacturing, Queensland University of Technology, Brisbane, Australia
| |
Collapse
|
384
|
Mussolino C, Alzubi J, Pennucci V, Turchiano G, Cathomen T. Genome and Epigenome Editing to Treat Disorders of the Hematopoietic System. Hum Gene Ther 2017; 28:1105-1115. [PMID: 28806883 DOI: 10.1089/hum.2017.149] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The possibility of editing complex genomes in a targeted fashion has revolutionized basic research as well as biomedical and biotechnological applications in the last 5 years. The targeted introduction of genetic changes has allowed researchers to create smart model systems for basic research, bio-engineers to modify crops and farm animals, and translational scientists to develop novel treatment approaches for inherited and acquired disorders for which curative treatment options are not yet available. With the rapid development of genome editing tools, in particular zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the CRISPR-Cas system, a wide range of therapeutic options have been-and will be-developed at an unprecedented speed, which will change the clinical routine of various disciplines in a revolutionary way. This review summarizes the fundamentals of genome editing and the current state of research. It particularly focuses on the advances made in employing engineered nucleases in hematopoietic stem cells for the treatment of primary immunodeficiencies and hemoglobinopathies, provides a perspective of combining gene editing with the chimeric antigen receptor T cell technology, and concludes by presenting targeted epigenome editing as a novel potential treatment option.
Collapse
Affiliation(s)
- Claudio Mussolino
- 1 Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg , Germany .,2 Center for Chronic Immunodeficiency, Medical Center - University of Freiburg , Germany
| | - Jamal Alzubi
- 1 Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg , Germany .,2 Center for Chronic Immunodeficiency, Medical Center - University of Freiburg , Germany
| | - Valentina Pennucci
- 1 Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg , Germany .,2 Center for Chronic Immunodeficiency, Medical Center - University of Freiburg , Germany
| | - Giandomenico Turchiano
- 1 Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg , Germany .,2 Center for Chronic Immunodeficiency, Medical Center - University of Freiburg , Germany
| | - Toni Cathomen
- 1 Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg , Germany .,2 Center for Chronic Immunodeficiency, Medical Center - University of Freiburg , Germany .,3 Faculty of Medicine, University of Freiburg , Freiburg, Germany
| |
Collapse
|
385
|
Refining strategies to translate genome editing to the clinic. Nat Med 2017; 23:415-423. [PMID: 28388605 DOI: 10.1038/nm.4313] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/27/2017] [Indexed: 12/17/2022]
Abstract
Recent progress in developing programmable nucleases, such as zinc-finger nucleases, transcription activator-like effector nucleases (TALENs) and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas nucleases, have paved the way for gene editing to enter clinical practice. This translation is a result of combining high nuclease activity with high specificity and successfully applying this technology in various preclinical disease models, including infectious disease, primary immunodeficiencies, hemoglobinopathies, hemophilia and muscular dystrophy. Several clinical gene-editing trials, both ex vivo and in vivo, have been initiated in the past 2 years, including studies that aim to knockout genes as well as to add therapeutic transgenes. Here we discuss the advances made in the gene-editing field in recent years, and specify priorities that need to be addressed to expand therapeutic genome editing to further disease entities.
Collapse
|
386
|
Engineering Natural Killer Cells for Cancer Immunotherapy. Mol Ther 2017; 25:1769-1781. [PMID: 28668320 PMCID: PMC5542803 DOI: 10.1016/j.ymthe.2017.06.012] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/09/2017] [Accepted: 06/10/2017] [Indexed: 01/21/2023] Open
Abstract
The past several years have seen tremendous advances in the engineering of immune effector cells as therapy for cancer. While chimeric antigen receptors (CARs) have been used extensively to redirect the specificity of autologous T cells against hematological malignancies with striking clinical results, studies of CAR-modified natural killer (NK) cells have been largely preclinical. In this review, we focus on recent advances in NK cell engineering, particularly on preclinical evidence suggesting that NK cells may be as effective as T cells in recognizing and killing targets after genetic modification. We will discuss strategies to introduce CARs into both primary NK cells and NK cell lines in an effort to provide antigen specificity, the challenges of manufacturing engineered NK cells, and evidence supporting the effectiveness of this approach from preclinical and early-phase clinical studies using CAR-engineered NK cells. CAR-NK cells hold great promise as a novel cellular immunotherapy against refractory malignancies. Notably, NK cells can provide an "off-the-shelf" product, eliminating the need for a personalized and patient-specific product that plagues current CAR-T cell therapies. The ability to more potently direct NK cell-mediated cytotoxicity against refractory tumors through the expression of CAR is likely to contribute to the recent paradigm shift in cancer treatment.
Collapse
|
387
|
Ren J, Zhang X, Liu X, Fang C, Jiang S, June CH, Zhao Y. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget 2017; 8:17002-17011. [PMID: 28199983 PMCID: PMC5370017 DOI: 10.18632/oncotarget.15218] [Citation(s) in RCA: 301] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 01/27/2017] [Indexed: 01/01/2023] Open
Abstract
The therapeutic potential of CRISPR system has already been demonstrated in many instances and begun to overlap with the rapidly expanding field of cancer immunotherapy, especially on the production of genetically modified T cell receptor or chimeric antigen receptor (CAR) T cells. Efficient genomic disruption of multiple gene loci to generate universal donor cells, as well as potent effector T cells resistant to multiple inhibitory pathways such as PD-1 and CTLA4 is an attractive strategy for cell therapy. In this study, we accomplished rapid and efficient multiplex genomic editing, and re-directing T cells with antigen specific CAR via a one-shot CRISPR protocol by incorporation of multiple gRNAs in a CAR lentiviral vector. High efficient double knockout of endogenous TCR and HLA class I could be easily achieved to generate allogeneic universal CAR T cells. We also generated Fas-resistant universal CAR T cells by triple gene disruption. Simultaneous gene editing of four gene loci using the one-shot CRISPR protocol to generate allogeneic universal T cells deficient of both PD1 and CTLA-4 was also attempted.
Collapse
Affiliation(s)
- Jiangtao Ren
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuhua Zhang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaojun Liu
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chongyun Fang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuguang Jiang
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yangbing Zhao
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
388
|
Fesnak AD, Hanley PJ, Levine BL. Considerations in T Cell Therapy Product Development for B Cell Leukemia and Lymphoma Immunotherapy. Curr Hematol Malig Rep 2017; 12:335-343. [PMID: 28762038 PMCID: PMC5693739 DOI: 10.1007/s11899-017-0395-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Based on laboratory and clinical research findings and investments in immunotherapy by many institutions in academia, government-funded laboratories, and industry, there is tremendous and deserved excitement in the field of cell and gene therapy. In particular, understanding of immune-mediated control of cancer has created opportunities to develop new forms of therapies based on engineered T cells. Unlike conventional drugs or biologics, the source material for these new therapies is collected from the patient or donor. The next step is commonly either enrichment to deplete unwanted cells, or methods to positively select T cells prior to polyclonal expansion or antigen-specific expansion. As the first generation of engineered T cell therapies have demonstrated proof of concept, the next stages of development will require the integration of automated technologies to enable more consistent manufacturing and the ability to produce therapies for more patients.
Collapse
Affiliation(s)
- Andrew D Fesnak
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-5156, USA.
| | - Patrick J Hanley
- Program for Cell Enhancement and Technologies for Immunotherapy, Center for Cancer and Immunology Research, Division of Blood and Marrow Transplantation, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System and The George Washington University, Washington, DC, 20010, USA
| | - Bruce L Levine
- Department of Pathology and Laboratory Medicine and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-5156, USA
| |
Collapse
|
389
|
Georgiadis C, Qasim W. Emerging applications of gene edited T cells for the treatment of leukemia. Expert Rev Hematol 2017; 10:753-755. [DOI: 10.1080/17474086.2017.1350575] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Christos Georgiadis
- Molecular and Cellular Immunology Unit, Institute of Child Health, London, UK
| | - Waseem Qasim
- Molecular and Cellular Immunology Unit, Institute of Child Health, London, UK
| |
Collapse
|
390
|
Dallos M, Tap WD, D'Angelo SP. Current status of engineered T-cell therapy for synovial sarcoma. Immunotherapy 2017; 8:1073-80. [PMID: 27485079 DOI: 10.2217/imt-2016-0026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Synovial sarcoma is a rare soft tissue sarcoma characterized by a t(X;18) translocation, which results in a SYT-SSX gene fusion. In the metastatic setting, chemotherapy has limited, durable efficacy prompting the necessity for new therapeutic modalities. One emerging new strategy involves T-cell-directed therapy such as tumor-infiltrating lymphocytes or the development of T cells that are genetically engineered to express a T-cell receptor against a cancer testis antigen. Of these approaches, engineered T cells that recognize NY-ESO-1 are the furthest along in development. Completed and on-going clinical trials have shown promise and there are efforts to continue to optimize the current approach.
Collapse
Affiliation(s)
- Matthew Dallos
- New York University Langone Medical Center, NY 10016, USA
| | - William D Tap
- Memorial Sloan Kettering Cancer Center, NY 10065, USA.,Weill Cornell Medical College, NY 10065, USA
| | - Sandra P D'Angelo
- Memorial Sloan Kettering Cancer Center, NY 10065, USA.,Weill Cornell Medical College, NY 10065, USA
| |
Collapse
|
391
|
Abstract
Lentivirus-mediated transduction of autologous T cells with a chimeric antigen receptor (CAR) to confer a desired epitope specificity as a targeted immunotherapy for cancer has been among the first human gene therapy techniques to demonstrate widespread therapeutic efficacy. Other approaches to using gene therapy to enhance antitumor immunity have been less specific and less effective. These have included amplification, marking, and cytokine transduction of tumor infiltrating lymphocytes, recombinant virus-based expression of tumor antigens as a tumor vaccine, and transduction of antigen-presenting cells with tumor antigens. Unlike any of those methods, the engineering of CAR T cells combine specific monoclonal antibody gene sequences to confer epitope specificity and other T-cell receptor and activation domains to create a self-contained single vector approach to produce a very specific antitumor response, as is seen with CD19-directed CAR T cells used to treat CD19-expressing B-cell malignancies. Recent success with these therapies is the culmination of a long step-wise iterative process of improvement in the design of CAR vectors. This review aims to summarize this long series of advances in the development of effective CAR vector since their initial development in the 1990s, and to describe emerging approaches to design that promise to enhance and widen the human gene therapy relevance of CAR T-cell therapy in the future.
Collapse
Affiliation(s)
- Olivia Wilkins
- 1 Department of Biology, Wheaton College , Norton, Massachusetts
| | - Allison M Keeler
- 1 Department of Biology, Wheaton College , Norton, Massachusetts.,2 Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| | - Terence R Flotte
- 2 Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School , Worcester, Massachusetts
| |
Collapse
|
392
|
Mo Z, Du P, Wang G, Wang Y. The Multi-Purpose Tool of Tumor Immunotherapy: Gene-Engineered T Cells. J Cancer 2017; 8:1690-1703. [PMID: 28775789 PMCID: PMC5535725 DOI: 10.7150/jca.18681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 02/27/2017] [Indexed: 02/05/2023] Open
Abstract
A detailed summary of the published clinical trials of chimeric antigen receptor T cells (CAR-T) and TCR-transduced T cells (TCR-T) was constructed to understand the development trend of adoptive T cell therapy (ACT). In contrast to TCR-T, the number of CAR-T clinical trials has increased dramatically in China in the last three years. The ACT seems to be very prosperous. But, the multidimensional interaction of tumor, tumor associated antigen (TAA) and normal tissue exacerbates the uncontrolled outcome of T cells gene therapy. It reminds us the importance that optimizing treatment security to prevent the fatal serious adverse events. How to balance the safety and effectiveness of the ACT? At least six measures can potentially optimize the safety of ACT. At the same time, with the application of gene editing techniques, more endogenous receptors are disrupted while more exogenous receptors are expressed on T cells. As a multi-purpose tool of tumor immunotherapy, gene-engineered T cells (GE-T) have been given different functional weapons. A network which is likely to link radiation therapy, tumor vaccines, CAR-T and TCR-T is being built. Moreover, more and more evidences indicated that the combination of the ACT and other therapies would further enhance the anti-tumor capacity of the GE-T.
Collapse
Affiliation(s)
- Zeming Mo
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| | - Peixin Du
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| | - Guoping Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| | - Yongsheng Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| |
Collapse
|
393
|
Keeler AM, ElMallah MK, Flotte TR. Gene Therapy 2017: Progress and Future Directions. Clin Transl Sci 2017; 10:242-248. [PMID: 28383804 PMCID: PMC5504480 DOI: 10.1111/cts.12466] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 03/29/2017] [Indexed: 12/13/2022] Open
Affiliation(s)
- A M Keeler
- Horae Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - M K ElMallah
- Horae Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - T R Flotte
- Horae Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
394
|
Chimeric Antigen Receptors: A Cell and Gene Therapy Perspective. Mol Ther 2017; 25:1117-1124. [PMID: 28456379 DOI: 10.1016/j.ymthe.2017.03.034] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 03/28/2017] [Accepted: 03/28/2017] [Indexed: 02/08/2023] Open
Abstract
Chimeric antigen receptors (CARs) are synthetic receptors that reprogram T lymphocytes to target chosen antigens. The targeting of CD19, a cell surface molecule expressed in the vast majority of leukemias and lymphomas, has been successfully translated in the clinic, earning CAR therapy a special distinction in the selection of "cancer immunotherapy" by Science as the breakthrough of the year in 2013. CD19 CAR therapy is predicated on advances in genetic engineering, T cell biology, tumor immunology, synthetic biology, target identification, cell manufacturing sciences, and regulatory compliance-the central tenets of CAR therapy. Here, we review two of these foundations: the genetic engineering approaches and cell types to engineer.
Collapse
|
395
|
Ren J, Zhao Y. Advancing chimeric antigen receptor T cell therapy with CRISPR/Cas9. Protein Cell 2017; 8:634-643. [PMID: 28434148 PMCID: PMC5563282 DOI: 10.1007/s13238-017-0410-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/30/2017] [Indexed: 02/06/2023] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (CRISPR/Cas9) system, an RNA-guided DNA targeting technology, is triggering a revolution in the field of biology. CRISPR/Cas9 has demonstrated great potential for genetic manipulation. In this review, we discuss the current development of CRISPR/Cas9 technologies for therapeutic applications, especially chimeric antigen receptor (CAR) T cell-based adoptive immunotherapy. Different methods used to facilitate efficient CRISPR delivery and gene editing in T cells are compared. The potential of genetic manipulation using CRISPR/Cas9 system to generate universal CAR T cells and potent T cells that are resistant to exhaustion and inhibition is explored. We also address the safety concerns associated with the use of CRISPR/Cas9 gene editing and provide potential solutions and future directions of CRISPR application in the field of CAR T cell immunotherapy. As an integration-free gene insertion method, CRISPR/Cas9 holds great promise as an efficient gene knock-in platform. Given the tremendous progress that has been made in the past few years, we believe that the CRISPR/Cas9 technology holds immense promise for advancing immunotherapy.
Collapse
Affiliation(s)
- Jiangtao Ren
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-5156, USA
| | - Yangbing Zhao
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104-5156, USA.
| |
Collapse
|
396
|
Abstract
PURPOSE OF REVIEW Alternative approaches to conventional drug-based cancer treatments have seen T cell therapies deployed more widely over the last decade. This is largely due to their ability to target and kill specific cell types based on receptor recognition. Introduction of recombinant T cell receptors (TCRs) using viral vectors and HLA-independent T cell therapies using chimeric antigen receptors (CARs) are discussed. This article reviews the tools used for genome editing, with particular emphasis on the applications of site-specific DNA nuclease mediated editing for T cell therapies. RECENT FINDINGS Genetic engineering of T cells using TCRs and CARs with redirected antigen-targeting specificity has resulted in clinical success of several immunotherapies. In conjunction, the application of genome editing technologies has resulted in the generation of HLA-independent universal T cells for allogeneic transplantation, improved T cell sustainability through knockout of the checkpoint inhibitor, programmed cell death protein-1 (PD-1), and has shown efficacy as an antiviral therapy through direct targeting of viral genomic sequences and entry receptors. SUMMARY The combined use of engineered antigen-targeting moieties and innovative genome editing technologies have recently shown success in a small number of clinical trials targeting HIV and hematological malignancies and are now being incorporated into existing strategies for other immunotherapies.
Collapse
Affiliation(s)
- Juliette M. K. M. Delhove
- Molecular Immunology Unit, UCL Great Ormond Street Institute of Child Health, University College London (UCL), 30 Guilford Street, London, WC1N 1EH UK
| | - Waseem Qasim
- Molecular Immunology Unit, UCL Great Ormond Street Institute of Child Health, University College London (UCL), 30 Guilford Street, London, WC1N 1EH UK
| |
Collapse
|
397
|
CARs: Synthetic Immunoreceptors for Cancer Therapy and Beyond. Trends Mol Med 2017; 23:430-450. [PMID: 28416139 DOI: 10.1016/j.molmed.2017.03.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/13/2017] [Accepted: 03/14/2017] [Indexed: 02/07/2023]
Abstract
Chimeric antigen receptors (CARs) are versatile synthetic receptors that provide T cells with engineered specificity. Clinical success in treating B-cell malignancies has demonstrated the therapeutic potential of CAR-T cells against cancer, and efforts are underway to expand the use of engineered T cells to the treatment of diverse medical conditions, including infections and autoimmune diseases. Here, we review current understanding of the molecular properties of CARs, how this knowledge informs the rational design and characterization of novel receptors, the successes and shortcomings of CAR-T cells in the clinic, and emerging solutions for the continued improvement of CAR-T cell therapy.
Collapse
|
398
|
Progenitor T-cell differentiation from hematopoietic stem cells using Delta-like-4 and VCAM-1. Nat Methods 2017; 14:531-538. [PMID: 28394335 DOI: 10.1038/nmeth.4258] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 03/07/2017] [Indexed: 12/28/2022]
Abstract
The molecular and cellular signals that guide T-cell development from hematopoietic stem and progenitor cells (HSPCs) remain poorly understood. The thymic microenvironment integrates multiple niche molecules to potentiate T-cell development in vivo. Recapitulating these signals in vitro in a stromal cell-free system has been challenging and limits T-cell generation technologies. Here, we describe a fully defined engineered in vitro niche capable of guiding T-lineage development from HSPCs. Synergistic interactions between Notch ligand Delta-like 4 and vascular cell adhesion molecule 1 (VCAM-1) were leveraged to enhance Notch signaling and progenitor T-cell differentiation rates. The engineered thymus-like niche enables in vitro production of mouse Sca-1+cKit+ and human CD34+ HSPC-derived CD7+ progenitor T-cells capable of in vivo thymus colonization and maturation into cytokine-producing CD3+ T-cells. This engineered thymic-like niche provides a platform for in vitro analysis of human T-cell development as well as clinical-scale cell production for future development of immunotherapeutic applications.
Collapse
|
399
|
CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep 2017; 7:737. [PMID: 28389661 PMCID: PMC5428439 DOI: 10.1038/s41598-017-00462-8] [Citation(s) in RCA: 527] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 02/28/2017] [Indexed: 02/07/2023] Open
Abstract
Immunotherapies with chimeric antigen receptor (CAR) T cells and checkpoint inhibitors (including antibodies that antagonize programmed cell death protein 1 [PD-1]) have both opened new avenues for cancer treatment, but the clinical potential of combined disruption of inhibitory checkpoints and CAR T cell therapy remains incompletely explored. Here we show that programmed death ligand 1 (PD-L1) expression on tumor cells can render human CAR T cells (anti-CD19 4-1BBζ) hypo-functional, resulting in impaired tumor clearance in a sub-cutaneous xenograft model. To overcome this suppressed anti-tumor response, we developed a protocol for combined Cas9 ribonucleoprotein (Cas9 RNP)-mediated gene editing and lentiviral transduction to generate PD-1 deficient anti-CD19 CAR T cells. Pdcd1 (PD-1) disruption augmented CAR T cell mediated killing of tumor cells in vitro and enhanced clearance of PD-L1+ tumor xenografts in vivo. This study demonstrates improved therapeutic efficacy of Cas9-edited CAR T cells and highlights the potential of precision genome engineering to enhance next-generation cell therapies.
Collapse
|
400
|
Seet CS, He C, Bethune MT, Li S, Chick B, Gschweng EH, Zhu Y, Kim K, Kohn DB, Baltimore D, Crooks GM, Montel-Hagen A. Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids. Nat Methods 2017; 14:521-530. [PMID: 28369043 PMCID: PMC5426913 DOI: 10.1038/nmeth.4237] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/03/2017] [Indexed: 01/08/2023]
Abstract
Studies of human T cell development require robust model systems that recapitulate the full span of thymopoiesis, from hematopoietic stem and progenitor cells (HSPCs) through to mature T cells. Existing in vitro models induce T cell commitment from human HSPCs; however, differentiation into mature CD3+TCRab+ single positive (SP) CD8+ or CD4+ cells is limited. We describe here a serum-free, artificial thymic organoid (ATO) system that supports highly efficient and reproducible in vitro differentiation and positive selection of conventional human T cells from all sources of HSPCs. ATO-derived T cells exhibited mature naïve phenotypes, a diverse TCR repertoire, and TCR-dependent function. ATOs initiated with TCR-engineered HSPCs produced T cells with antigen specific cytotoxicity and near complete lack of endogenous TCR Vβ expression, consistent with allelic exclusion of Vβ loci. ATOs provide a robust tool for studying human T cell development and stem cell based approaches to engineered T cell therapies.
Collapse
Affiliation(s)
- Christopher S Seet
- Division of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine (DGSOM), University of California Los Angeles (UCLA), Los Angeles, California, USA
| | - Chongbin He
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA
| | - Michael T Bethune
- Division of Biology and Biological Engineering, California Institute of Technology (Caltech), Pasadena, California, USA
| | - Suwen Li
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA
| | - Brent Chick
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA
| | - Eric H Gschweng
- Department of Microbiology, Immunology and Molecular Genetics, DGSOM, UCLA, Los Angeles, California, USA
| | - Yuhua Zhu
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA
| | - Kenneth Kim
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA
| | - Donald B Kohn
- Department of Microbiology, Immunology and Molecular Genetics, DGSOM, UCLA, Los Angeles, California, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, DGSOM, UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology (Caltech), Pasadena, California, USA
| | - Gay M Crooks
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA.,Division of Pediatric Hematology-Oncology, Department of Pediatrics, DGSOM, UCLA, Los Angeles, California, USA.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California, USA.,Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| | - Amélie Montel-Hagen
- Department of Pathology and Laboratory Medicine, DGSOM, UCLA, Los Angeles, California, USA
| |
Collapse
|