1
|
Targeting PTPN22 does not enhance the efficacy of CAR T cells in solid tumours. Mol Cell Biol 2022; 42:e0044921. [PMID: 35041491 DOI: 10.1128/mcb.00449-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adoptive cell therapy with chimeric antigen receptor (CAR) T cells has revolutionised the treatment of certain B cell malignancies, but has been in ineffective against solid tumours. Recent studies have highlighted the potential of targeting negative regulators of T cell signalling to enhance the efficacy and extend the utility of CAR T cells to solid tumours. Autoimmunity-linked protein tyrosine phosphatase N22 (PTPN22) has been proposed as a target for cancer immunotherapy. Here we have used CRISPR/Cas9 gene-editing to generate PTPN22-deficient (Ptpn22Δ/Δ) mice (C57BL/6) and assessed the impact of PTPN22 deficiency on the cytotoxicity and efficacy of CAR T cells in vitro and in vivo. As reported previously, PTPN22 deficiency was accompanied by the promotion of effector T cell responses ex vivo and the repression of syngeneic tumour growth in vivo. However, PTPN22-deficiency did not enhance the cytotoxic activity of murine CAR T cells targeting the extracellular domain of the human oncoprotein HER2 in vitro. Moreover, PTPN22-deficient α-HER2 CAR T cells or ovalbumin-specific OT-I CD8+ T cells adoptively transferred into mice bearing HER2+ mammary tumours or ovalbumin-expressing mammary or colorectal tumours respectively were no more effective than their wild type counterparts in suppressing tumour growth. The deletion of PTPN22 using CRISPR/Cas9 gene-editing also did not affect the cytotoxic activity of human CAR T cells targeting the Lewis Y antigen that is expressed by many human solid tumours. Therefore, PTPN22-deficiency does not enhance the anti-tumour activity of CAR T cells in solid organ malignancies.
Collapse
|
2
|
Wiede F, Lu K, Du X, Liang S, Hochheiser K, Dodd GT, Goh PK, Kearney C, Meyran D, Beavis PA, Henderson MA, Park SL, Waithman J, Zhang S, Zhang Z, Oliaro J, Gebhardt T, Darcy PK, Tiganis T. PTPN2 phosphatase deletion in T cells promotes anti-tumour immunity and CAR T-cell efficacy in solid tumours. EMBO J 2020; 39:e103637. [PMID: 31803974 PMCID: PMC6960448 DOI: 10.15252/embj.2019103637] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/04/2019] [Accepted: 11/08/2019] [Indexed: 12/18/2022] Open
Abstract
Although adoptive T-cell therapy has shown remarkable clinical efficacy in haematological malignancies, its success in combating solid tumours has been limited. Here, we report that PTPN2 deletion in T cells enhances cancer immunosurveillance and the efficacy of adoptively transferred tumour-specific T cells. T-cell-specific PTPN2 deficiency prevented tumours forming in aged mice heterozygous for the tumour suppressor p53. Adoptive transfer of PTPN2-deficient CD8+ T cells markedly repressed tumour formation in mice bearing mammary tumours. Moreover, PTPN2 deletion in T cells expressing a chimeric antigen receptor (CAR) specific for the oncoprotein HER-2 increased the activation of the Src family kinase LCK and cytokine-induced STAT-5 signalling, thereby enhancing both CAR T-cell activation and homing to CXCL9/10-expressing tumours to eradicate HER-2+ mammary tumours in vivo. Our findings define PTPN2 as a target for bolstering T-cell-mediated anti-tumour immunity and CAR T-cell therapy against solid tumours.
Collapse
Affiliation(s)
- Florian Wiede
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Kun‐Hui Lu
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Xin Du
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Shuwei Liang
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | - Katharina Hochheiser
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Garron T Dodd
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
| | - Pei K Goh
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| | | | | | - Paul A Beavis
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | | | - Simone L Park
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Jason Waithman
- Telethon Kids InstituteUniversity of Western AustraliaPerthWAAustralia
| | - Sheng Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyInstitute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Zhong‐Yin Zhang
- Department of Medicinal Chemistry and Molecular PharmacologyInstitute for Drug DiscoveryPurdue UniversityWest LafayetteINUSA
| | - Jane Oliaro
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Thomas Gebhardt
- Department of Microbiology and ImmunologyThe University of MelbourneMelbourneVic.Australia
- Peter Doherty Institute for Infection and ImmunityMelbourneVic.Australia
| | - Phillip K Darcy
- Peter MacCallum Cancer CentreMelbourneVic.Australia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneMelbourneVic.Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery InstituteMonash UniversityClaytonVic.Australia
- Department of Biochemistry and Molecular BiologyMonash UniversityClaytonVic.Australia
- Peter MacCallum Cancer CentreMelbourneVic.Australia
| |
Collapse
|
3
|
Schubert ML, Hückelhoven A, Hoffmann JM, Schmitt A, Wuchter P, Sellner L, Hofmann S, Ho AD, Dreger P, Schmitt M. Chimeric Antigen Receptor T Cell Therapy Targeting CD19-Positive Leukemia and Lymphoma in the Context of Stem Cell Transplantation. Hum Gene Ther 2016; 27:758-771. [PMID: 27479233 DOI: 10.1089/hum.2016.097] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Novel therapies with chimeric antigen receptor (CAR)-transduced T cells (TCs) sparked new hope for patients with relapsed or refractory CD19-positive leukemia or lymphoma even after stem cell therapies. This review focuses on CARs recognizing the B cell antigen CD19. Both retroviral and lentiviral vectors are used, encoding various anti-CD19 CAR constructs comprising costimulatory molecules such as CD28, CD137/4-1BB, and OX40 either alone (second-generation CARs) or in combination (third-generation CARs). Current, up-to-date published studies on anti-CD19 CAR therapy for acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), and non-Hodgkin lymphoma (NHL) with observed side effects are discussed and an outlook on 58 ongoing trials is given. Clinical responses were achieved in up to 81% of ALL, 50% of CLL, and 40% of NHL patients. Factors with potential influence on the clinical outcome might be the design of the vector, the preconditioning regimen, and the number and quality of transfused CAR TCs. The applicability of clinical CAR TC therapy might include relapse after allogeneic stem cell transplantation (alloSCT), and ineligibility for or "bridging" until alloSCT. In summary, CAR therapy represents a highly promising treatment option even in heavily pretreated patients.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Angela Hückelhoven
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Jean-Marc Hoffmann
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Patrick Wuchter
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Leopold Sellner
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Susanne Hofmann
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital , Heidelberg, Germany
| |
Collapse
|
4
|
Gilham DE, Anderson J, Bridgeman JS, Hawkins RE, Exley MA, Stauss H, Maher J, Pule M, Sewell AK, Bendle G, Lee S, Qasim W, Thrasher A, Morris E. Adoptive T-cell therapy for cancer in the United kingdom: a review of activity for the British Society of Gene and Cell Therapy annual meeting 2015. Hum Gene Ther 2015; 26:276-85. [PMID: 25860661 PMCID: PMC4442586 DOI: 10.1089/hum.2015.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/31/2015] [Indexed: 12/25/2022] Open
Abstract
Adoptive T-cell therapy is delivering objective clinical responses across a number of cancer indications in the early phase clinical setting. Much of this clinical activity is taking place at major clinical academic centers across the United States. This review focuses upon cancer-focused cell therapy activity within the United Kingdom as a contribution to the 2015 British Society of Gene and Cell Therapy annual general meeting. This overview reflects the diversity and expansion of clinical and preclinical studies within the United Kingdom while considering the background context of this work against new infrastructural developments and the requirements of nationalized healthcare delivery within the UK National Health Service.
Collapse
Affiliation(s)
- David Edward Gilham
- Clinical and Experimental Immunotherapy Group, Institute of Cancer Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
| | - John Anderson
- UCL Institute of Child Health, London WC1N 1EH, United Kingdom
| | | | - Robert Edward Hawkins
- Clinical and Experimental Immunotherapy Group, Institute of Cancer Sciences, The University of Manchester, Manchester M20 4BX, United Kingdom
- Cellular Therapeutics Ltd., UMIC Bio-incubator, Manchester M13 9XX, United Kingdom
| | - Mark Adrian Exley
- MCCIR, Faculty of Medicine, The University of Manchester, Manchester M13 9NT, United Kingdom
| | - Hans Stauss
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| | - John Maher
- Department of Research Oncology, Bermondsey Wing, Guy's Hospital, London SE1 9RT, United Kingdom
| | - Martin Pule
- Research Department of Haematology, UCL Cancer Institute, London WC1E 6DD, United Kingdom
| | - Andrew Kelvin Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital, Cardiff CF14 4XN, United Kingdom
| | - Gavin Bendle
- School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Steven Lee
- School of Cancer Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Waseem Qasim
- Molecular and Cellular Immunology Institute, Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Adrian Thrasher
- Molecular and Cellular Immunology Institute, Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Emma Morris
- Department of Immunology, University College London, Royal Free Hospital, London NW3 2PF, United Kingdom
| |
Collapse
|
5
|
Inclusion of an IgG1-Fc spacer abrogates efficacy of CD19 CAR T cells in a xenograft mouse model. Gene Ther 2015; 22:391-403. [DOI: 10.1038/gt.2015.4] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 11/03/2014] [Accepted: 01/06/2015] [Indexed: 12/15/2022]
|
6
|
Kershaw MH, Westwood JA, Slaney CY, Darcy PK. Clinical application of genetically modified T cells in cancer therapy. Clin Transl Immunology 2014; 3:e16. [PMID: 25505964 PMCID: PMC4232070 DOI: 10.1038/cti.2014.7] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 04/01/2014] [Accepted: 04/01/2014] [Indexed: 02/08/2023] Open
Abstract
Immunotherapies are emerging as highly promising approaches for the treatment of cancer. In these approaches, a variety of materials are used to boost immunity against malignant cells. A key component of many of these approaches is functional tumor-specific T cells, but the existence and activity of sufficient T cells in the immune repertoire is not always the case. Recent methods of generating tumor-specific T cells include the genetic modification of patient lymphocytes with receptors to endow them with tumor specificity. These T cells are then expanded in vitro followed by infusion of the patient in adoptive cell transfer protocols. Genes used to modify T cells include those encoding T-cell receptors and chimeric antigen receptors. In this review, we provide an introduction to the field of genetic engineering of T cells followed by details of their use against cancer in the clinic.
Collapse
Affiliation(s)
- Michael H Kershaw
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia ; Department of Immunology, Monash University , Prahran, Victoria, Australia
| | - Jennifer A Westwood
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia
| | - Clare Y Slaney
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia
| | - Phillip K Darcy
- Sir Peter MacCallum Cancer Centre, Department of Oncology, University of Melbourne , Melbourne, Victoria, Australia ; Department of Immunology, Monash University , Prahran, Victoria, Australia
| |
Collapse
|
7
|
Stärck L, Popp K, Pircher H, Uckert W. Immunotherapy with TCR-Redirected T Cells: Comparison of TCR-Transduced and TCR-Engineered Hematopoietic Stem Cell–Derived T Cells. THE JOURNAL OF IMMUNOLOGY 2013; 192:206-13. [DOI: 10.4049/jimmunol.1202591] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
8
|
Advances in siRNA delivery to T-cells: potential clinical applications for inflammatory disease, cancer and infection. Biochem J 2013; 455:133-47. [DOI: 10.1042/bj20130950] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The specificity of RNAi and its ability to silence ‘undruggable’ targets has made inhibition of gene expression in T-cells with siRNAs an attractive potential therapeutic strategy for the treatment of inflammatory disease, cancer and infection. However, delivery of siRNAs into primary T-cells represents a major hurdle to their use as potential therapeutic agents. Recent advances in siRNA delivery through the use of electroporation/nucleofection, viral vectors, peptides/proteins, nanoparticles, aptamers and other agents have now enabled efficient gene silencing in primary T-cells both in vitro and in vivo. Overcoming such barriers in siRNA delivery offers exciting new prospects for directly targeting T-cells systemically with siRNAs, or adoptively transferring T-cells back into patients following ex vivo manipulation with siRNAs. In the present review, we outline the challenges in delivering siRNAs into primary T-cells and discuss the mechanism and therapeutic opportunities of each delivery method. We emphasize studies that have exploited RNAi-mediated gene silencing in T-cells for the treatment of inflammatory disease, cancer and infection using mouse models. We also discuss the potential therapeutic benefits of manipulating T-cells using siRNAs for the treatment of human diseases.
Collapse
|
9
|
Chicaybam L, Sodre AL, Curzio BA, Bonamino MH. An efficient low cost method for gene transfer to T lymphocytes. PLoS One 2013; 8:e60298. [PMID: 23555950 PMCID: PMC3608570 DOI: 10.1371/journal.pone.0060298] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 02/25/2013] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Gene transfer to T lymphocytes has historically relied on retro and lentivirus, but recently transposon-based gene transfer is rising as a simpler and straight forward approach to achieve stable transgene expression. Transfer of expression cassettes to T lymphocytes remains challenging, being based mainly on commercial kits. AIMS We herein report a convenient and affordable method based on in house made buffers, generic cuvettes and utilization of the widely available Lonza nucleofector II device to promote efficient gene transfer to T lymphocytes. RESULTS This approach renders high transgene expression levels in primary human T lymphocytes (mean 45%, 41-59%), the hard to transfect murine T cells (mean 38%, 36-42% for C57/BL6 strain) and human Jurkat T cell line. Cell viability levels after electroporation allowed further manipulations such as in vitro expansion and Chimeric Antigen Receptor (CAR) mediated gain of function for target cell lysis. CONCLUSIONS We describe here an efficient general protocol for electroporation based modification of T lymphocytes. By opening access to this protocol, we expect that efficient gene transfer to T lymphocytes, for transient or stable expression, may be achieved by an increased number of laboratories at lower and affordable costs.
Collapse
Affiliation(s)
- Leonardo Chicaybam
- Programa de Carcinogênese Molecular, Coordenação de Pesquisa (CPQ), Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
- Instituto de Pesquisa Clínica Evandro Chagas (IPEC), Fundação Instituto Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Andressa Laino Sodre
- Programa de Carcinogênese Molecular, Coordenação de Pesquisa (CPQ), Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Bianca Azevedo Curzio
- Programa de Carcinogênese Molecular, Coordenação de Pesquisa (CPQ), Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Martin Hernan Bonamino
- Programa de Carcinogênese Molecular, Coordenação de Pesquisa (CPQ), Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| |
Collapse
|
10
|
|
11
|
Abstract
INTRODUCTION Despite extensive research, cancer continues to be a leading cause of death worldwide and is expected to continue to rise as a result of an aging population. Therefore, new therapies are constantly being developed. Par-4 is a naturally occurring tumor suppressor protein that is capable of inducing apoptosis in cancer, but not normal cells. For this reason, Par-4 offers an attractive target for development of cancer therapy, particularly of difficult to treat cancers. AREAS COVERED The mechanisms by which Par-4 induces cell death are summarized. The ways that Par-4 is controlled in cancer cells are discussed. We discuss how different research groups have developed ways to overexpress and/or activate Par-4 in vitro and in vivo. The studies described demonstrate that when Par-4 levels and/or activity are increased, susceptibility to apoptosis is enhanced and tumor growth is inhibited. EXPERT OPINION Par-4 is a promising therapeutic protein that can be overexpressed and/or activated to induce apoptosis in a cancer-selective manner. This cancer selectivity is important given that the side-effects of chemotherapeutics can be as debilitating as cancer itself. However, there are key issues that need to be addressed to optimize the effects of Par-4 in patients.
Collapse
Affiliation(s)
- Rosalyn B Irby
- Penn State Hershey Cancer Institute, 500 University Drive, Hershey, PA 17033, USA.
| | | |
Collapse
|
12
|
Colovos C, Villena-Vargas J, Adusumilli PS. Safety and stability of retrovirally transduced chimeric antigen receptor T cells. Immunotherapy 2012; 4:899-902. [PMID: 23046233 DOI: 10.2217/imt.12.91] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Evaluation of: Scholler J, Brady TL, Binder-Scholl G et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci. Transl Med. 4(132), 132-153 (2012). Adoptive cellular therapy with genetically engineered T cells is predicated on generating effective and persisting T-cell-mediated immunity. Gammaretroviral vector-mediated gene transfer in T cells is the technological basis for promising therapy in both HIV and cancer. Because of concerns over delayed adverse events caused by persisting retroviral vector-engineered cells, the US FDA mandates long-term follow-up of clinical trials. Scholler et al. report FDA-mandated safety data and demonstrate that retrovirally transduced T cells persist in the blood of patients for more than a decade after treatment. These persisting T cells proliferated ex vivo in the presence of antigens and showed no evidence of either insertional oncogenesis or clonal expansion in 11 subjects followed for up to 11 years, supporting the long-term safety and persistence of retrovirally transduced T cells.
Collapse
Affiliation(s)
- Christos Colovos
- Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | | |
Collapse
|
13
|
Ramos CA, Dotti G. Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy. Expert Opin Biol Ther 2011; 11:855-73. [PMID: 21463133 DOI: 10.1517/14712598.2011.573476] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Chimeric antigen receptors (CARs) usually combine the antigen binding site of a monoclonal antibody with the signal activating machinery of a T cell, freeing antigen recognition from MHC restriction and thus breaking one of the barriers to more widespread application of cellular therapy. Similar to treatment strategies employing monoclonal antibodies, T cells expressing CARs are highly targeted, but additionally offer the potential benefits of active trafficking to tumor sites, in vivo expansion and long-term persistence. Furthermore, gene transfer allows the introduction of countermeasures to tumor immune evasion and of safety mechanisms. AREAS COVERED The basic structure of so-called first and later generation CARs and their potential advantages over other immune therapy systems. How these molecules can be grafted into immune cells (including retroviral and non-retroviral transduction methods) and strategies to improve the in vivo persistence and function of immune cells expressing CARs. Examples of tumor-associated antigens that have been targeted in preclinical models and clinical experience with these modified cells. Safety issues surrounding CAR gene transfer into T cells and potential solutions to them. EXPERT OPINION Because of recent advances in immunology, genetics and cell processing, CAR-modified T cells will likely play an increasing role in the cellular therapy of cancer, chronic infections and autoimmune disorders.
Collapse
Affiliation(s)
- Carlos A Ramos
- Center for Cell and Gene Therapy, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|
14
|
Almåsbak H, Rian E, Hoel HJ, Pulè M, Wälchli S, Kvalheim G, Gaudernack G, Rasmussen AM. Transiently redirected T cells for adoptive transfer. Cytotherapy 2010; 13:629-40. [PMID: 21174490 DOI: 10.3109/14653249.2010.542461] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS T cells can be redirected to reject cancer by retroviral transduction with a chimeric antigen receptor (CAR) or by administration of a bispecific T cell engager (BiTE). We demonstrate that transfection of T cells with messenger (m) RNA coding for CAR is an alternative strategy. METHODS We describe the pre-clinical evaluation of a method based on transient modification of expanded T cells with a CD19 CAR directed against B-cell malignancies. CAR mRNA was generated under cell-free conditions in a scalable process using recombinant RNA polymerase. Efficient and non-toxic square-wave electroporation was used to load the mRNA into the cytoplasm of T cells with no risk of insertional mutagenesis. RESULTS After transfection >80% of T cells were viable, with 94% CAR expression. Transfected T cells were cytolytic to CD19(+) targets and produced interferon (IFN)-γ in response. Killing of CD19(+) target cells was demonstrated even at day 8 with undetectable CAR expression. Increasing the concentration of mRNA resulted in higher surface CAR expression, better killing and more IFN-γ release but at the expense of increased activation-induced cell death. Finally, we demonstrated that a second transgene could be introduced by co-electroporation of CXCR4 or CCR7 with CAR to also modify chemotactic responses. CONCLUSIONS We advocate the transient redirection approach as well suited to meet safety aspects for early phase studies, prior to trials using stably transduced cells once CAR has been proven safe. The simplicity of this methodology also facilitates rapid screening of candidate targets and novel receptors in pre-clinical studies.
Collapse
Affiliation(s)
- Hilde Almåsbak
- Section for Immunology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
IMPORTANCE OF THE FIELD Adoptive therapy with T cell receptor- (TCR-) redirected T cells has shown efficacy in mouse tumor models and first responses in cancer patients. One prerequisite to elicit effective anti-tumor reactivity is the transfer of high-avidity T cells. Their generation, however, faces several technical difficulties. Target antigens are often expressed at low levels and their recognition requires the use of high-affine receptors. Yet, mainly low-affinity TCRs have been isolated from tumor-infiltrating lymphocytes. Furthermore, upon transfer into a T cell the introduced receptor has to compete with the endogenous TCR. AREAS COVERED IN THIS REVIEW This review discusses how the functional avidity of TCR-modified T cells can be enhanced by i) increasing the amount of introduced TCR heterodimers on the cell surface; and ii) generating receptors with high affinity. Risks of TCR gene therapy and possible safety mechanisms are discussed. WHAT THE READER WILL GAIN The reader will gain an overview of the technical developments in TCR and T cell engineering. TAKE HOME MESSAGE Despite technical obstacles, many advances have been made in the generation of high-avidity T cells expressing enhanced TCRs. Mouse studies and clinical trials will evaluate the effect of these improvements.
Collapse
Affiliation(s)
- Elisa Kieback
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Strasse 10, D-13092 Berlin, Germany
| | | |
Collapse
|
16
|
Neeson P, Shin A, Tainton KM, Guru P, Prince HM, Harrison SJ, Peinert S, Smyth MJ, Trapani JA, Kershaw MH, Darcy PK, Ritchie DS. Ex vivo culture of chimeric antigen receptor T cells generates functional CD8+ T cells with effector and central memory-like phenotype. Gene Ther 2010; 17:1105-16. [PMID: 20428216 DOI: 10.1038/gt.2010.59] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The anti-tumor efficacy of adoptively transferred T cells requires their in vivo persistence and memory polarization. It is unknown if human chimeric antigen receptor (CAR)-expressing T cells can also undergo memory polarization. We examined the functional status of CAR CD8(+) T cells, re-directed to Lewis Y antigen (LeY-T), throughout a period of ex vivo expansion. Immediately before culture CD8(+) T cells comprised a mixture of phenotypes including naive (CD45RA(+)/CCR7(+)/CD27(+)/CD28(+)/perforin-), central memory (CM, CD45RA(-)/CCR7(lo)/CD27(+)/CD28(+)/perforin(lo)), effector memory (EM, CD45RA(-)/CCR7(-)/CD27(+)/CD28(+)/perforin(mod)) and effector (Eff, CD45RA(+)/CCR7(-)/CD27(-)/CD28(-)/perforin(hi)) cells. After transduction and expansion culture of peripheral blood mononuclear cells from normal donors or multiple myeloma patients, CD8(+) LeY-T cells polarized to EM- and CM-like phenotype. CD8(+) LeY-T cells differed from starting CD8(+) CM and EM T cells in that CD27, but not CD28, was downregulated. In addition, CD8(+) LeY-T cells expressed high levels of perforin, similar to starting CD8(+) Eff. CD8(+) LeY-T cells also showed hallmarks of both memory and Eff function, underwent homeostatic proliferation in response to interleukin (IL)-15, and showed interferon (IFN)-γ production and cytotoxicity in response to Le-Y antigen on OVCAR-3 (human ovarian adenocarcinoma) cells. This study confirms CD8(+) LeY-T cells have a CM- and EM-like phenotype and heterogeneous function consistent with potential to persist in vivo after adoptive transfer.
Collapse
Affiliation(s)
- P Neeson
- Hematology and Immunology Translational Research Lab, Peter MacCallum Cancer Center, Melbourne, Australia.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther 2010; 17:678-86. [PMID: 20200563 DOI: 10.1038/gt.2010.21] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We have evaluated the carbohydrate antigen Lewis(Y) (Le(Y)) as a potential target for T-cell immunotherapy of hematological neoplasias. Analysis of 81 primary bone marrow samples revealed moderate Le(Y) expression on plasma cells of myeloma patients and myeloblasts of patients with acute myeloid leukemia (AML) (52 and 46% of cases, respectively). We developed a retroviral vector construct encoding a chimeric T-cell receptor that recognizes the Le(Y) antigen in a major histocompatibility complex-independent manner and delivers co-stimulatory signals to achieve T-cell activation. We have shown efficient transduction of peripheral blood-derived T cells with this construct, resulting in antigen-restricted interferon-gamma secretion and cell lysis of Le(Y)-expressing tumor cells. In vivo activity of gene-modified T cells was demonstrated in the delayed growth of myeloma xenografts in NOD/SCID mice, which prolonged survival. Therefore, targeting Le(Y)-positive malignant cells with T cells expressing a chimeric receptor recognizing Le(Y) was effective both in vitro and in a myeloma mouse model. Consequently, we plan to use T cells manufactured under Good Manufacturing Practice conditions in a phase I immunotherapy study for patients with Le(Y)-positive myeloma or AML.
Collapse
|
18
|
Abstract
Adoptive transfer of T cells with restricted tumor specificity provides a promising approach to immunotherapy of cancers. However, the isolation of autologous cytotoxic T cells that recognize tumor-associated antigens is time consuming and fails in many instances. Alternatively, gene modification with tumor antigen-specific T-cell receptors (TCR) or chimeric antigen receptors (CARs) can be used to redirect the specificity of large numbers of immune cells toward the malignant cells. Chimeric antigen receptors are composed of the single-chain variable fragment (scFv) of a tumor-recognizing antibody cloned in frame with human T-cell signaling domains (e.g., CD3zeta, CD28, OX40, 4-1BB), thus combining the specificity of antibodies with the effector functions of cytotoxic T cells. Upon antigen binding, the intracellular signaling domains of the CAR initiate cellular activation mechanisms including cytokine secretion and cytolysis of the antigen-positive target cell.In this chapter, we provide detailed protocols for large-scale ex vivo expansion of T cells and manufacturing of medium-scale batches of CAR-expressing T cells for translational research by mRNA electroporation. An anti-CD19 chimeric receptor for the targeting of leukemias and lymphomas was used as a model system. We are currently scaling up the protocols to adapt them to cGMP production of a large number of redirected T cells for clinical applications.
Collapse
Affiliation(s)
- Hilde Almåsbak
- Department of Immunology, Radiumhospitalet-Rikshospitalet, University Hospital, Oslo, Norway
| | | | | |
Collapse
|
19
|
Peinert S, Kershaw MH, Prince HM. Chimeric T cells for adoptive immunotherapy of cancer: using what have we learned to plan for the future. Immunotherapy 2009; 1:905-12. [DOI: 10.2217/imt.09.69] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
| | - Michael H Kershaw
- Peter MacCallum Cancer Centre, Australia and University of Melbourne, Australia
| | - H Miles Prince
- Department of Haematology, Peter MacCallum Cancer Centre, St Andrews Place, East Melbourne, Victoria 3002, Australia
| |
Collapse
|
20
|
Weisser NE, Hall JC. Applications of single-chain variable fragment antibodies in therapeutics and diagnostics. Biotechnol Adv 2009; 27:502-20. [PMID: 19374944 DOI: 10.1016/j.biotechadv.2009.04.004] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Revised: 03/03/2009] [Accepted: 04/04/2009] [Indexed: 11/18/2022]
Abstract
Antibodies (Abs) are some of the most powerful tools in therapy and diagnostics and are currently one of the fastest growing classes of therapeutic molecules. Recombinant antibody (rAb) fragments are becoming popular therapeutic alternatives to full length monoclonal Abs since they are smaller, possess different properties that are advantageous in certain medical applications, can be produced more economically and are easily amendable to genetic manipulation. Single-chain variable fragment (scFv) Abs are one of the most popular rAb format as they have been engineered into larger, multivalent, bi-specific and conjugated forms for many clinical applications. This review will show the tremendous versatility and importance of scFv fragments as they provide the basic antigen binding unit for a multitude of engineered Abs for use as human therapeutics and diagnostics.
Collapse
Affiliation(s)
- Nina E Weisser
- Department of Environmental Biology, University of Guelph, Guelph, ON N1G2W1, Canada
| | | |
Collapse
|
21
|
Symes JC, Siatskas C, Fowler DH, Medin JA. Retrovirally transduced murine T lymphocytes expressing FasL mediate effective killing of prostate cancer cells. Cancer Gene Ther 2008; 16:439-52. [PMID: 19096446 DOI: 10.1038/cgt.2008.96] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Adoptively transferred T cells possess anticancer activities partially mediated by T-cell FasL engagement of Fas tumor targets. However, antigen-induced T-cell activation and clonal expansion, which stimulates FasL activity, is often inefficient in tumors. As a gene therapy approach to overcome this obstacle, we have created oncoretroviral vectors to overexpress FasL or non-cleavable FasL (ncFasL) on murine T cells of a diverse T-cell receptor repertoire. Expression of c-FLIP was also engineered to prevent apoptosis of transduced cells. Retroviral transduction of murine T lymphocytes has historically been problematic, and we describe optimized T-cell transduction protocols involving CD3/CD28 co-stimulation of T cells, transduction on ice using concentrated oncoretrovirus, and culture with IL-15. Genetically modified T cells home to established prostate cancer tumors in vivo. Co-stimulated T cells expressing FasL, ncFasL and ncFasL/c-FLIP each mediated cytotoxicity in vitro against RM-1 and LNCaP prostate cancer cells. To evaluate the compatibility of this approach with current prostate cancer therapies, we exposed RM-1, LNCaP, and TRAMP-C1 cells to radiation, mitoxantrone, or docetaxel. Fas and H-2(b) expression were upregulated by these methods. We have developed a novel FasL-based immuno-gene therapy for prostate cancer that warrants further investigation given the apparent constitutive and inducible Fas pathway expression in this malignancy.
Collapse
Affiliation(s)
- J C Symes
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | | | | |
Collapse
|