1
|
Rothemejer FH, Lauritsen NP, Søgaard OS, Tolstrup M. Strategies for enhancing CAR T cell expansion and persistence in HIV infection. Front Immunol 2023; 14:1253395. [PMID: 37671164 PMCID: PMC10475529 DOI: 10.3389/fimmu.2023.1253395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/04/2023] [Indexed: 09/07/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapies are tremendously successful in hematological malignancies and show great promise as treatment and curative strategy for HIV. A major determinant for effective CAR T cell therapy is the persistence of CAR T cells. Particularly, antigen density and target cell abundance are crucial for the engagement, engraftment, and persistence of CAR T cells. The success of HIV-specific CAR T cells is challenged by limited antigen due to low cell surface expression of viral proteins and the scarcity of chronically infected cells during antiretroviral therapy. Several strategies have been explored to increase the efficacy of CAR T cells by enhancing expansion and persistence of the engineered cells. This review highlights the challenges of designing CAR T cells against HIV and other chronic viral infections. We also discuss potential strategies to enhance CAR T cell expansion and persistence in the setting of low antigen exposure.
Collapse
Affiliation(s)
- Frederik Holm Rothemejer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Nanna Pi Lauritsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Ole Schmeltz Søgaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Martin Tolstrup
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
2
|
Omer B, Castillo PA, Tashiro H, Shum T, Huynh MTA, Cardenas M, Tanaka M, Lewis A, Sauer T, Parihar R, Lapteva N, Schmueck-Henneresse M, Mukherjee M, Gottschalk S, Rooney CM. Chimeric Antigen Receptor Signaling Domains Differentially Regulate Proliferation and Native T Cell Receptor Function in Virus-Specific T Cells. Front Med (Lausanne) 2018; 5:343. [PMID: 30619856 PMCID: PMC6297364 DOI: 10.3389/fmed.2018.00343] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 11/22/2018] [Indexed: 11/16/2022] Open
Abstract
The efficacy of T cells expressing chimeric antigen receptors (CARs) for solid tumors has been limited by insufficient CAR T cell expansion and persistence. The use of virus-specific T cells (VSTs) as carriers for CARs may overcome this limitation since CAR-VSTs can be boosted by viral vaccines or oncolytic viruses. However, there is limited understanding of the optimal combination of endodomains and their influence on the native T cell receptor (TCR) in VSTs. We therefore compared the function of GD2.CARs expressing the TCR zeta chain (ζ) alone or combined with endodomains from CD28 and 4-1BB in varicella zoster virus-specific (VZV) T cells. VZVSTs expressing GD2-CARs recognized VZV-derived peptides and killed GD2-expressing tumor cells. However, after repeated stimulation through their native TCR, the expansion of GD2-CAR.CD28ζ-VZVSTs was 3.3-fold greater (p < 0.001) than non-transduced VZVSTs, whereas GD2-CARζ- and GD2-CAR.41BBζ inhibited VZVST expansion (p < 0.01). Compared to control VZVSTs, GD2-CAR.ζ VZVSTs showed a greater frequency of apoptotic (p < 0.01) T cells, whereas prolonged downregulation of the native αβ TCR was observed in GD2-CAR.41BBζ VZVSTs (p < 0.001). We confirmed that CD28ζ can best maintain TCR function by expressing GD2.CARs in Epstein-Barr virus-specific T cells and CD19-CARs in VZVSTs. In response to CAR stimulation VSTs with CD28ζ endodomains also showed the greatest expansion (6 fold > GD2-CAR.41BBζ VZVSTs (p < 0.001), however anti-tumor efficacy was superior in GD2-CAR.41BBζ-VZVSTs. These findings demonstrate that CAR signaling domains can enhance or diminish the function of the native TCR and indicate that only CD28ζ may preserve the function of the native TCR in tonically signaling CAR-VSTs.
Collapse
Affiliation(s)
- Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Paul A Castillo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Haruko Tashiro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Thomas Shum
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Mai T A Huynh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Mara Cardenas
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Miyuki Tanaka
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Andrew Lewis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Tim Sauer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Robin Parihar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| | - Natalia Lapteva
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Michael Schmueck-Henneresse
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Malini Mukherjee
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital, Texas Children's Hospital, Houston, TX, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States.,Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
3
|
Jacqueline C, Bonnefoy N, Charrière GM, Thomas F, Roche B. Personal history of infections and immunotherapy: Unexpected links and possible therapeutic opportunities. Oncoimmunology 2018; 7:e1466019. [PMID: 30221066 PMCID: PMC6136881 DOI: 10.1080/2162402x.2018.1466019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 01/17/2023] Open
Abstract
The recent breakthroughs in the understanding of tumor immune biology have given rise to a new generation of immunotherapies, harnessing the immune system to eliminate tumors. As the typology and frequency of encountered infections are susceptible to shape the immune system, it could also impact the efficiency of immunotherapy. In this review, we report evidences for an indirect link between personal history of infection and different strategies of immunotherapy. In the current context of interest rise for personalized medicine, we discuss the potential medical applications of considering personal history of infection to design immunotherapeutic strategies.
Collapse
Affiliation(s)
- Camille Jacqueline
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- MIVEGEC, IRD, CNRS, Université Montpellier, Montpellier, France
| | - Nathalie Bonnefoy
- IRCM, INSERM, Université de Montpellier, ICM, F-34298, Montpellier, France
| | - Guillaume M. Charrière
- IHPE, UMR 5244, CNRS, Ifremer, Université de Perpignan Via Domitia, Université de Montpellier, Montpellier, 34095, France
| | - Frédéric Thomas
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- MIVEGEC, IRD, CNRS, Université Montpellier, Montpellier, France
| | - Benjamin Roche
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- UMMISCO, IRD/ Sorbonne Université, Bondy, France
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| |
Collapse
|
4
|
Achard C, Surendran A, Wedge ME, Ungerechts G, Bell J, Ilkow CS. Lighting a Fire in the Tumor Microenvironment Using Oncolytic Immunotherapy. EBioMedicine 2018; 31:17-24. [PMID: 29724655 PMCID: PMC6013846 DOI: 10.1016/j.ebiom.2018.04.020] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 04/20/2018] [Indexed: 02/09/2023] Open
Abstract
Oncolytic virus (OV) therapy is potentially a game-changing cancer treatment that has garnered significant interest due to its versatility and multi-modal approaches towards tumor eradication. In the field of cancer immunotherapy, the immunological phenotype of the tumor microenvironment (TME) is an important determinant of disease prognosis and therapeutic success. There is accumulating data that OVs are capable of dramatically altering the TME immune landscape, leading to improved antitumor activity alone or in combination with assorted immune modulators. Herein, we review how OVs disrupt the immunosuppressive TME and can be used strategically to create a "pro-immune" microenvironment that enables and promotes potent, long-lasting host antitumor immune responses.
Collapse
Affiliation(s)
- Carole Achard
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada
| | - Abera Surendran
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Marie-Eve Wedge
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Guy Ungerechts
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Medical Oncology and Translational Oncology, National Center for Tumor Diseases (NCT), and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - John Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Carolina S Ilkow
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.
| |
Collapse
|
5
|
Xin G, Schauder DM, Jing W, Jiang A, Joshi NS, Johnson B, Cui W. Pathogen boosted adoptive cell transfer immunotherapy to treat solid tumors. Proc Natl Acad Sci U S A 2017; 114:740-745. [PMID: 28069963 PMCID: PMC5278465 DOI: 10.1073/pnas.1614315114] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Because of insufficient migration and antitumor function of transferred T cells, especially inside the immunosuppressive tumor microenvironment (TME), the efficacy of adoptive cell transfer (ACT) is much curtailed in treating solid tumors. To overcome these challenges, we sought to reenergize ACT (ReACT) with a pathogen-based cancer vaccine. To bridge ACT with a pathogen, we genetically engineered tumor-specific CD8 T cells in vitro with a second T-cell receptor (TCR) that recognizes a bacterial antigen. We then transferred these dual-specific T cells in combination with intratumoral bacteria injection to treat solid tumors in mice. The dual-specific CD8 T cells expanded vigorously, migrated to tumor sites, and robustly eradicated primary tumors. The mice cured from ReACT also developed immunological memory against tumor rechallenge. Mechanistically, we have found that this combined approach reverts the immunosuppressive TME and recruits CD8 T cells with an increased number and killing ability to the tumors.
Collapse
Affiliation(s)
- Gang Xin
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53213
| | - David M Schauder
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53213
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Weiqing Jing
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Aimin Jiang
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Nikhil S Joshi
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
| | - Bryon Johnson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Weiguo Cui
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53213;
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226
| |
Collapse
|
6
|
Slaney CY, von Scheidt B, Davenport AJ, Beavis PA, Westwood JA, Mardiana S, Tscharke DC, Ellis S, Prince HM, Trapani JA, Johnstone RW, Smyth MJ, Teng MW, Ali A, Yu Z, Rosenberg SA, Restifo NP, Neeson P, Darcy PK, Kershaw MH. Dual-specific Chimeric Antigen Receptor T Cells and an Indirect Vaccine Eradicate a Variety of Large Solid Tumors in an Immunocompetent, Self-antigen Setting. Clin Cancer Res 2016; 23:2478-2490. [PMID: 27965307 DOI: 10.1158/1078-0432.ccr-16-1860] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 11/16/2022]
Abstract
Purpose: While adoptive transfer of T cells bearing a chimeric antigen receptor (CAR) can eliminate substantial burdens of some leukemias, the ultimate challenge remains the eradication of large solid tumors for most cancers. We aimed to develop an immunotherapy approach effective against large tumors in an immunocompetent, self-antigen preclinical mouse model.Experimental Design: In this study, we generated dual-specific T cells expressing both a CAR specific for Her2 and a TCR specific for the melanocyte protein (gp100). We used a regimen of adoptive cell transfer incorporating vaccination (ACTIV), with recombinant vaccinia virus expressing gp100, to treat a range of tumors including orthotopic breast tumors and large liver tumors.Results: ACTIV therapy induced durable complete remission of a variety of Her2+ tumors, some in excess of 150 mm2, in immunocompetent mice expressing Her2 in normal tissues, including the breast and brain. Vaccinia virus induced extensive proliferation of T cells, leading to massive infiltration of T cells into tumors. Durable tumor responses required the chemokine receptor CXCR3 and exogenous IL2, but were independent of IFNγ. Mice were resistant to tumor rechallenge, indicating immune memory involving epitope spreading. Evidence of limited neurologic toxicity was observed, associated with infiltration of cerebellum by T cells, but was only transient.Conclusions: This study supports a view that it is possible to design a highly effective combination immunotherapy for solid cancers, with acceptable transient toxicity, even when the target antigen is also expressed in vital tissues. Clin Cancer Res; 23(10); 2478-90. ©2016 AACR.
Collapse
Affiliation(s)
- Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Bianca von Scheidt
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Alexander J Davenport
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Paul A Beavis
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Jennifer A Westwood
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Sherly Mardiana
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - David C Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Sarah Ellis
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - H Miles Prince
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Ricky W Johnstone
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Michele W Teng
- Cancer Immunoregulation and Immunotherapy Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Aesha Ali
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Zhiya Yu
- Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland
| | - Steven A Rosenberg
- Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland
| | - Paul Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Clayton, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia.,Department of Immunology, Monash University, Clayton, Australia
| |
Collapse
|
7
|
Haji-Fatahaliha M, Hosseini M, Akbarian A, Sadreddini S, Jadidi-Niaragh F, Yousefi M. CAR-modified T-cell therapy for cancer: an updated review. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2016; 44:1339-1349. [PMID: 26068778 DOI: 10.3109/21691401.2015.1052465] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/24/2015] [Indexed: 01/21/2023]
Abstract
The use of chimeric antigen receptor (CAR)-modified T cells is a promising approach for cancer immunotherapy. These genetically modified receptors contain an antigen-binding moiety, a hinge region, a transmembrane domain, and an intracellular costimulatory domain resulting in T-cell activation subsequent to antigen binding. Optimal tumor removal through CAR-modified T cells requires suitable target antigen selection, co-stimulatory signaling domain, and the ability of CAR T cells to traffic, persist, and retain antitumor function after adoptive transfer. There are several elements which can improve antitumor function of CAR T cells, including signaling, conditioning chemotherapy and irradiation, tumor burden of the disease, T-cell phenotype, and supplementary cytokine usage. This review outlines four generations of CAR. The pre-clinical and clinical studies showed that this technique has a great potential for treatment of solid and hematological malignancies. The main purpose of the current review is to focus on the pre-clinical and clinical developments of CAR-based immunotherapy.
Collapse
Affiliation(s)
- Mostafa Haji-Fatahaliha
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Maryam Hosseini
- b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Asiye Akbarian
- d Department of Microbiology , Faculty of Medicine, Tehran University of Medical Sciences , Tehran , Iran
| | - Sanam Sadreddini
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| | - Farhad Jadidi-Niaragh
- e Department of Immunology , School of Public Health, Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Yousefi
- a Drug Applied Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- b Immunology Research Center, Tabriz University of Medical Sciences , Tabriz , Iran
- c Department of Immunology , Faculty of Medicine, Tabriz University of Medical Sciences , Tabriz , Iran
| |
Collapse
|
8
|
Capitalizing on Cancer Specific Replication: Oncolytic Viruses as a Versatile Platform for the Enhancement of Cancer Immunotherapy Strategies. Biomedicines 2016; 4:biomedicines4030021. [PMID: 28536388 PMCID: PMC5344262 DOI: 10.3390/biomedicines4030021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 02/07/2023] Open
Abstract
The past decade has seen considerable excitement in the use of biological therapies in treating neoplastic disease. In particular, cancer immunotherapy and oncolytic virotherapy have emerged as two frontrunners in this regard with the first FDA approvals for agents in both categories being obtained in the last 5 years. It is becoming increasingly apparent that these two approaches are not mutually exclusive and that much of the therapeutic benefit obtained from the use of oncolytic viruses (OVs) is in fact the result of their immunotherapeutic function. Indeed, OVs have been shown to recruit and activate an antitumor immune response and much of the current work in this field centers around increasing this activity through strategies such as engineering genes for immunomodulators into OV backbones. Because of their broad immunostimulatory functions, OVs can also be rationally combined with a variety of other immunotherapeutic approaches including cancer vaccination strategies, adoptive cell transfer and checkpoint blockade. Therefore, while they are important therapeutics in their own right, the true power of OVs may lie in their ability to enhance the effectiveness of a wide range of immunotherapies.
Collapse
|
9
|
Uslu U, Schuler G, Dörrie J, Schaft N. Combining a chimeric antigen receptor and a conventional T-cell receptor to generate T cells expressing two additional receptors (TETARs) for a multi-hit immunotherapy of melanoma. Exp Dermatol 2016; 25:872-879. [PMID: 27246630 DOI: 10.1111/exd.13095] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
The adoptive transfer of engineered T cells represents an important approach in immunotherapy of melanoma. However, relapse of the tumor can occur due to immune-escape mechanisms developed by the tumor cells, for example antigen loss, downregulation of the major histocompatibility complex presentation machinery and defects in antigen processing. To counteract these mechanisms, we combined a T-cell receptor and a chimeric antigen receptor, specific for different common melanoma antigens, gp100 (PMEL) and MCSP (HMW-MAA), to generate functional CD8+ T cells expressing two additional receptors (TETARs) by electroporation of receptor-encoding mRNA. These TETARs produced cytokines and were lytic upon recognition of each of their cognate antigens, while no reciprocal inhibition of the receptors occurred. When stimulated with target cells, which express both antigens, an enhanced effect was suggested. The confirmation that chimeric antigen receptors and T-cell receptors can be functionally combined opens up new avenues in cancer immunotherapy, and the generation of TETARs helps by-passing major mechanisms by which tumor cells escape immune recognition.
Collapse
Affiliation(s)
- Ugur Uslu
- Department of Dermatology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Erlangen, Germany.
| |
Collapse
|
10
|
Abstract
T cells have the capacity to eradicate diseased cells, but tumours present considerable challenges that render T cells ineffectual. Cancer cells often make themselves almost 'invisible' to the immune system, and they sculpt a microenvironment that suppresses T cell activity, survival and migration. Genetic engineering of T cells can be used therapeutically to overcome these challenges. T cells can be taken from the blood of cancer patients and then modified with genes encoding receptors that recognize cancer-specific antigens. Additional genes can be used to enable resistance to immunosuppression, to extend survival and to facilitate the penetration of engineered T cells into tumours. Using genetic modification, highly active, self-propagating 'slayers' of cancer cells can be generated.
Collapse
Affiliation(s)
- Michael H Kershaw
- Cancer Immunology Research Program, Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria 3010, Australia. michael.kershaw@ petermac.org
| | | | | |
Collapse
|
11
|
Maher J. Immunotherapy of malignant disease using chimeric antigen receptor engrafted T cells. ISRN ONCOLOGY 2012; 2012:278093. [PMID: 23304553 PMCID: PMC3523553 DOI: 10.5402/2012/278093] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 11/14/2012] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor- (CAR-) based immunotherapy has been under development for almost 25 years, over which period it has progressed from a new but cumbersome technology to an emerging therapeutic modality for malignant disease. The approach involves the genetic engineering of fusion receptors (CARs) that couple the HLA-independent binding of cell surface target molecules to the delivery of a tailored activating signal to host immune cells. Engineered CARs are delivered most commonly to peripheral blood T cells using a range of vector systems, most commonly integrating viral vectors. Preclinical refinement of this approach has proceeded over several years to the point that clinical testing is now being undertaken at several centres, using increasingly sophisticated and therapeutically successful genetic payloads. This paper considers several aspects of the pre-clinical and clinical development of CAR-based immunotherapy and how this technology is acquiring an increasing niche in the treatment of both solid and haematological malignancies.
Collapse
Affiliation(s)
- John Maher
- CAR Mechanics Group, Department of Research Oncology, King's Health Partners Integrated Cancer Centre, King's College London, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK
- Department of Immunology, Barnet and Chase Farm Hospitals NHS Trust, Barnet, Hertfordshire EN5 3DJ, UK
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
12
|
Daniel-Meshulam I, Ya'akobi S, Ankri C, Cohen CJ. How (specific) would like your T-cells today? Generating T-cell therapeutic function through TCR-gene transfer. Front Immunol 2012; 3:186. [PMID: 22783259 PMCID: PMC3390604 DOI: 10.3389/fimmu.2012.00186] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/15/2012] [Indexed: 01/02/2023] Open
Abstract
T-cells are central players in the immune response against both pathogens and cancer. Their specificity is solely dictated by the T-cell receptor (TCR) they clonally express. As such, the genetic modification of T lymphocytes using pathogen- or cancer-specific TCRs represents an appealing strategy to generate a desired immune response from peripheral blood lymphocytes. Moreover, notable objective clinical responses were observed in terminally ill cancer patients treated with TCR-gene modified cells in several clinical trials conducted recently. Nevertheless, several key aspects of this approach are the object of intensive research aimed at improving the reliability and efficacy of this strategy. Herein, we will survey recent studies in the field of TCR-gene transfer dealing with the improvement of this approach and its application for the treatment of malignant, autoimmune, and infectious diseases.
Collapse
Affiliation(s)
- Inbal Daniel-Meshulam
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences , Bar-Ilan University, Ramat Gan, Israel
| | | | | | | |
Collapse
|
13
|
Marr LA, Gilham DE, Campbell JDM, Fraser AR. Immunology in the clinic review series; focus on cancer: double trouble for tumours: bi-functional and redirected T cells as effective cancer immunotherapies. Clin Exp Immunol 2012; 167:216-25. [PMID: 22235997 PMCID: PMC3278687 DOI: 10.1111/j.1365-2249.2011.04517.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2011] [Indexed: 01/04/2023] Open
Abstract
Cancer is one of the most important pathological conditions facing mankind in the 21st century, and is likely to become the most important cause of death as improvements continue in health, diet and life expectancy. The immune response is responsible for controlling nascent cancer through immunosurveillance. If tumours escape this control, they can develop into clinical cancer. Although surgery and chemo- or radiotherapy have improved survival rates significantly, there is a drive to reharness immune responses to treat disease. As T cells are one of the key immune cells in controlling cancer, research is under way to enhance their function and improve tumour targeting. This can be achieved by transduction with tumour-specific T cell receptor (TCR) or chimaeric antigen receptors (CAR) to generate redirected T cells. Virus-specific cells can also be transduced with TCR or CAR to create bi-functional T cells with specificity for both virus and tumour. In this review we outline the development and optimization of redirected and bi-functional T cells, and outline the results from current clinical trials using these cells. From this we discuss the challenges involved in generating effective anti-tumour responses while avoiding concomitant damage to normal tissues and organs.
Collapse
Affiliation(s)
- L A Marr
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | | | | |
Collapse
|
14
|
The construction of chimeric T-Cell receptor with spacer base of modeling study of VHH and MUC1 interaction. J Biomed Biotechnol 2011; 2011:578128. [PMID: 21869862 PMCID: PMC3160013 DOI: 10.1155/2011/578128] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 05/24/2011] [Accepted: 06/02/2011] [Indexed: 01/07/2023] Open
Abstract
Adaptive cell immunotherapy with the use of chimeric receptors leads to the best and most specific response against tumors. Chimeric receptors consist of a signaling fragment, extracellular spacer, costimulating domain, and an antibody. Antibodies cause immunogenicity; therefore, VHH is a good replacement for ScFv in chimeric receptors. Since peptide sequences have an influence on chimeric receptors, the effect of peptide domains on each other's conformation were investigated. CD3Zeta, CD28, VHH and CD8α, and FcgIIα are used as signaling moieties, costimulating domain, antibody, and spacers, respectively. To investigate the influence of the ligation of spacers on the conformational structure of VHH, models of VHH were constructed. Molecular dynamics simulation was run to study the influence of the presence of spacers on the conformational changes in the binding sites of VHH. Root mean square deviation and root mean square fluctuation of critical segments in the binding site showed no noticeable differences with those in the native VHH. Results from molecular docking revealed that the presence of spacer FcgIIα causes an increasing effect on VHH with MUC1 interaction. Each of the constructs was transformed into the Jurkat E6.1. Expression analysis and evaluation of their functions were examined. The results showed good expression and function.
Collapse
|
15
|
Westwood JA, Berry LJ, Wang LX, Duong CP, Pegram HJ, Darcy PK, Kershaw MH. Enhancing adoptive immunotherapy of cancer. Expert Opin Biol Ther 2010; 10:531-45. [PMID: 20132063 DOI: 10.1517/14712591003610622] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Conventional therapies, including surgery, chemotherapy and radiotherapy have contributed much to cancer treatment. However, these treatment modalities fail in a large proportion of patients, and there is a great need for effective alternate therapies. Adoptive immunotherapy can be effective against some cancers that have failed all other treatment options, even when disease burdens are massive. AREAS COVERED IN THIS REVIEW This review gives a brief introduction of the historical origins of adoptive immunotherapy and then provides details of strategies for increasing the potency of cell transfer. Approaches for enhancing adoptive immunotherapy include: selecting the right type of cell; providing cytokine support; preconditioning patients and tuning the tumor microenvironment. The review also provides insights into the safety, feasibility and costs of this form of therapy. WHAT THE READER WILL GAIN This article will give the reader an appreciation of the potential of adoptive immunotherapy, as well as an understanding of some limitations and current approaches for optimizing the effectiveness of this approach. TAKE HOME MESSAGE With recent developments in knowledge of the interactions between the immune system and tumors, the field of adoptive immunotherapy is now poised to make dramatic contributions to cancer therapy.
Collapse
Affiliation(s)
- Jennifer A Westwood
- Peter MacCallum Cancer Centre, Cancer Immunology Research Program, St. Andrews Place, Melbourne, Victoria 3002, Australia
| | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Davies DM, Maher J. Adoptive T-cell immunotherapy of cancer using chimeric antigen receptor-grafted T cells. Arch Immunol Ther Exp (Warsz) 2010; 58:165-78. [PMID: 20373147 DOI: 10.1007/s00005-010-0074-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Accepted: 10/27/2009] [Indexed: 12/25/2022]
Abstract
Harnessing the power of the immune system to target cancer has long been a goal of tumor immunologists. One avenue under investigation is the modification of T cells to express a chimeric antigen receptor (CAR). Expression of such a receptor enables T-cell specificity to be redirected against a chosen tumor antigen. Substantial research in this field has been carried out, incorporating a wide variety of malignancies and tumor-associated antigens. Ongoing investigations will ensure this area continues to expand at a rapid pace. This review will explain the evolution of CAR technology over the last two decades in addition to detailing the associated benefits and disadvantages. The outcome of recent phase I clinical trials and the impact that these have had upon the direction of future research in this field will also be addressed.
Collapse
Affiliation(s)
- David Marc Davies
- King's College London School of Medicine, Research Oncology Section, Division of Cancer Studies, Third Floor Bermondsey Wing, Guy's Hospital Campus, St Thomas Street, London SE1 9RT, UK
| | | |
Collapse
|
18
|
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
|
19
|
Westwood JA, Kershaw MH. Genetic redirection of T cells for cancer therapy. J Leukoc Biol 2010; 87:791-803. [PMID: 20179152 DOI: 10.1189/jlb.1209824] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Adoptive immunotherapy can induce dramatic tumor regressions in patients with melanoma or viral-induced malignancies, but extending this approach to many common cancers has been hampered by a lack of naturally occurring tumor-specific T cells. In this review, we describe recent advances in the genetic modification of T cells using genes encoding cell-surface receptors specific for tumor-associated antigen. Using genetic modification, the many functional properties of T cells, including cytokine secretion and cytolytic capacity, are redirected from their endogenous specificity toward the elimination of tumor cells. Advances in gene design, vectors, and cell production are discussed, and details of the progress in clinical application of this approach are provided.
Collapse
Affiliation(s)
- Jennifer A Westwood
- Cancer Immunology Research Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | |
Collapse
|
20
|
Berry LJ, Moeller M, Darcy PK. Adoptive immunotherapy for cancer: the next generation of gene-engineered immune cells. ACTA ACUST UNITED AC 2009; 74:277-89. [PMID: 19775368 DOI: 10.1111/j.1399-0039.2009.01336.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Adoptive cellular immunotherapy involving transfer of tumor-reactive T cells has shown some notable antitumor responses in a minority of cancer patients. In particular, transfer of tumor-infiltrating lymphocytes has resulted in long-term objective responses in patients with advanced melanoma. However, the inability to isolate sufficient numbers of tumor-specific T cells from most malignancies has restricted the broad utility of this approach. An emerging approach to circumvent this limitation involves the genetic modification of effector cells with T cell receptor (TCR) transgenes or chimeric single-chain variable fragment (scFv) receptors that can specifically redirect T cells to tumor. There has been much progress in the design of TCR and scFv receptors to enhance the antigen-specific activation of effector cells and their trafficking and persistence in vivo. Considerable effort has been directed toward improving the safety of this approach and reducing the immunogenicity of the receptor. This review discusses the latest developments in the field of adoptive immunotherapy using genetically modified immune cells that have been transduced with either TCR or scFv receptor transgenes and used in preclinical and clinical settings as anticancer agents.
Collapse
Affiliation(s)
- L J Berry
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Victoria, Australia
| | | | | |
Collapse
|
21
|
The Lewis-Y carbohydrate antigen is expressed by many human tumors and can serve as a target for genetically redirected T cells despite the presence of soluble antigen in serum. J Immunother 2009; 32:292-301. [PMID: 19242371 DOI: 10.1097/cji.0b013e31819b7c8e] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
In this study we aimed to determine the suitability of the Lewis-Y carbohydrate antigen as a target for immunotherapy using genetically redirected T cells. Using the 3S193 monoclonal antibody and immunohistochemistry, Lewis-Y was found to be expressed on a range of tumors including 42% squamous cell lung carcinoma, 80% lung adenocarcinoma, 25% ovarian carcinoma, and 25% colorectal adenocarcinoma. Expression levels varied from low to intense on between 1% and 90% of tumor cells. Lewis- was also found in soluble form in sera from both normal donors and cancer patients using a newly developed enzyme-linked immunosorbent assay. Serum levels in patients was often less than 1 ng/mL, similar to normal donors, but approximately 30% of patients had soluble Lewis-Y levels exceeding 1 ng/mL and up to 9 ng/mL. Lewis-Y-specific human T cells were generated by genetic modification with a chimeric receptor encoding a single-chain humanized antibody linked to the T-cell signaling molecules, T-cell receptor-zeta, and CD28. T cells responded against the Lewis-Y antigen by cytokine secretion and cytolysis in response to tumor cells. Importantly, the T-cell response was not inhibited by patient serum containing soluble Lewis-Y. This study demonstrates that Lewis-Y is expressed on a large number of tumors and Lewis-Y-specific T cells can retain antitumor function in the presence of patient serum, indicating that this antigen is a suitable target for this form of therapy.
Collapse
|
22
|
Sadelain M, Brentjens R, Rivière I. The promise and potential pitfalls of chimeric antigen receptors. Curr Opin Immunol 2009; 21:215-23. [PMID: 19327974 DOI: 10.1016/j.coi.2009.02.009] [Citation(s) in RCA: 373] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 02/25/2009] [Indexed: 12/26/2022]
Abstract
One important purpose of T cell engineering is to generate tumor-targeted T cells through the genetic transfer of antigen-specific receptors, which consist of either physiological, MHC-restricted T cell receptors (TCRs) or non MHC-restricted chimeric antigen receptors (CARs). CARs combine antigen-specificity and T cell activating properties in a single fusion molecule. First generation CARs, which included as their signaling domain the cytoplasmic region of the CD3zeta or Fc receptor gamma chain, effectively redirected T cell cytotoxicity but failed to enable T cell proliferation and survival upon repeated antigen exposure. Receptors encompassing both CD28 and CD3zeta are the prototypes for second generation CARs, which are now rapidly expanding to a diverse array of receptors with different functional properties. First generation CARs have been tested in phase I clinical studies in patients with ovarian cancer, renal cancer, lymphoma, and neuroblastoma, where they have induced modest responses. Second generation CARs, which are just now entering the clinical arena in the B cell malignancies and other cancers, will provide a more significant test for this approach. If the immunogenicity of CARs can be averted, the versatility of their design and HLA-independent antigen recognition will make CARs tools of choice for T cell engineering for the development of targeted cancer immunotherapies.
Collapse
Affiliation(s)
- Michel Sadelain
- Center for Cell Engineering, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | | | |
Collapse
|