1001
|
|
1002
|
Abstract
Building on the principals that the adoptive transfer of T cells can lead to the regression of established tumors in humans, investigators are now further manipulating these cells using genetic engineering. Two decades of human gene transfer experiments have resulted in the translation of laboratory technology into robust clinical applications. The purpose of this review is to give the reader an introduction to the 2 major approaches being developed to redirect effector T-cell specificity. Primary human T cells can be engineered to express exogenous T-cell receptors or chimeric antigen receptors directed against multiple human tumor antigens. Initial clinical trial results have demonstrated that both T-cell receptor- and chimeric antigen receptor-engineered T cells can be administered to cancer patients and mediate tumor regression.
Collapse
Affiliation(s)
- Richard A Morgan
- Surgery Branch, National Cancer Institute, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
1003
|
Abstract
Living cells have evolved a broad array of complex signalling responses, which enables them to survive diverse environmental challenges and execute specific physiological functions. Our increasingly sophisticated understanding of the molecular mechanisms of cell signalling networks in eukaryotes has revealed a remarkably modular organization and synthetic biologists are exploring how this can be exploited to engineer cells with novel signalling behaviours. This approach is beginning to reveal the logic of how cells might evolve innovative new functions and moves us towards the exciting possibility of engineering custom cells with precise sensing-response functions that could be useful in medicine and biotechnology.
Collapse
Affiliation(s)
- Wendell A Lim
- Howard Hughes Medical Institute and Department of Cellular and Molecular Pharmacology, University of California San Francisco, California 94158, USA.
| |
Collapse
|
1004
|
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
|
1005
|
Hawkins RE, Gilham DE, Debets R, Eshhar Z, Taylor N, Abken H, Schumacher TN. Development of Adoptive Cell Therapy for Cancer: A Clinical Perspective. Hum Gene Ther 2010; 21:665-72. [DOI: 10.1089/hum.2010.086] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Robert E. Hawkins
- Cellular Therapy Group, School of Cancer and Enabling Sciences, The Paterson Institute of Cancer Research, The University of Manchester, Manchester M20 4BX, United Kingdom
| | - David E. Gilham
- Cellular Therapy Group, School of Cancer and Enabling Sciences, The Paterson Institute of Cancer Research, The University of Manchester, Manchester M20 4BX, United Kingdom
| | - Reno Debets
- Laboratory of Experimental Tumor Immunology, Department of Medical Oncology, Erasmus MC-Daniel den Hoed Cancer Center, 3075EA Rotterdam, The Netherlands
| | - Zelig Eshhar
- The Weizmann Institute of Science, Department of Immunology, 76100 Rehovot, Israel
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, 34293 Montpellier, France
| | - Hinrich Abken
- Klinik I für Innere Medizin and Zentrum für Molekulare Medizin Köln, Universitat zu Köln, 50931 Köln, Germany
| | - Ton N. Schumacher
- The Division of Immunology, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
1006
|
Chimeric antigen receptor-engineered T cells for immunotherapy of cancer. J Biomed Biotechnol 2010; 2010:956304. [PMID: 20467460 PMCID: PMC2864912 DOI: 10.1155/2010/956304] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Accepted: 02/15/2010] [Indexed: 11/18/2022] Open
Abstract
CD4+ and CD8+ T lymphocytes are powerful components of adaptive immunity, which essentially contribute to the elimination of tumors. Due to their cytotoxic capacity, T cells emerged as attractive candidates for specific immunotherapy of cancer. A promising approach is the genetic modification of T cells with chimeric antigen receptors (CARs). First generation CARs consist of a binding moiety specifically recognizing a tumor cell surface antigen and a lymphocyte activating signaling chain. The CAR-mediated recognition induces cytokine production and tumor-directed cytotoxicity of T cells. Second and third generation CARs include signal sequences from various costimulatory molecules resulting in enhanced T-cell persistence and sustained antitumor reaction. Clinical trials revealed that the adoptive transfer of T cells engineered with first generation CARs represents a feasible concept for the induction of clinical responses in some tumor patients. However, further improvement is required, which may be achieved by second or third generation CAR-engrafted T cells.
Collapse
|
1007
|
Redirecting T-cell specificity by introducing a tumor-specific chimeric antigen receptor. Blood 2010; 116:1035-44. [PMID: 20439624 DOI: 10.1182/blood-2010-01-043737] [Citation(s) in RCA: 230] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Infusions of antigen-specific T cells have yielded therapeutic responses in patients with pathogens and tumors. To broaden the clinical application of adoptive immunotherapy against malignancies, investigators have developed robust systems for the genetic modification and characterization of T cells expressing introduced chimeric antigen receptors (CARs) to redirect specificity. Human trials are under way in patients with aggressive malignancies to test the hypothesis that manipulating the recipient and reprogramming T cells before adoptive transfer may improve their therapeutic effect. These examples of personalized medicine infuse T cells designed to meet patients' needs by redirecting their specificity to target molecular determinants on the underlying malignancy. The generation of clinical grade CAR(+) T cells is an example of bench-to-bedside translational science that has been accomplished using investigator-initiated trials operating largely without industry support. The next-generation trials will deliver designer T cells with improved homing, CAR-mediated signaling, and replicative potential, as investigators move from the bedside to the bench and back again.
Collapse
|
1008
|
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
|
1009
|
Marcu-Malina V, van Dorp S, Kuball J. Re-targeting T-cells against cancer by gene-transfer of tumor-reactive receptors. Expert Opin Biol Ther 2010; 9:579-91. [PMID: 19368527 DOI: 10.1517/14712590902887018] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Adoptive transfer of T-lymphocytes is a promising treatment for a variety of malignancies, but is often not feasible due to difficulties in generating T-cells reactive with the targeted antigen from patients. To facilitate rapid generation of cells for therapy, T-cells can be programmed with genes encoding for an antigen-specific T-cell receptor (TCR) or chimeric receptors. OBJECTIVE To discuss the molecular design and selected pitfalls of TCR gene modified T-cells and T-cells expressing chimeric receptors, so called T-bodies. METHODS A selected review of the recent literature. CONCLUSION Clinical trials report so far only limited efficacy of adoptively transferred genetically modified T-cells. However, the recent progress in engineering tumor-reactive T cells is providing a promising basis to further explore this treatment modality.
Collapse
Affiliation(s)
- Victoria Marcu-Malina
- Department of Hematology and VanCreveld Clinic, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | |
Collapse
|
1010
|
Abstract
Cell-based therapies with various lymphocytes and antigen-presenting cells are promising approaches for cancer immunotherapy. The transfusion of T lymphocytes, also called adoptive cell therapy (ACT), is an effective treatment for viral infections, has induced regression of cancer in early stage clinical trials, and may be a particularly important and efficacious modality in the period following hematopoietic stem cell transplantation (HSCT). Immune reconstitution post-SCT is often slow and incomplete, which in turn leads to an increased risk of infection and may impact relapse risk in patients with malignant disease. Immunization post-HSCT is frequently unsuccessful, due to the prolonged lymphopenia, especially of CD4 T cells, seen following transplant. ACT has the potential to enhance antitumor and overall immunity, and augment vaccine efficacy in the post-transplant setting. The ability to genetically engineer lymphocyte subsets has the further potential to improve the natural immune response, correct impaired immunity, and redirect T cells to an antitumor effector response. This chapter focuses on various applications of ACT for cancer immunotherapy, and we discuss some of the latest progress and hurdles in translating these technologies to the clinic.
Collapse
Affiliation(s)
- Stephan A Grupp
- Division of Oncology and Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | |
Collapse
|
1011
|
Abstract
Standard therapies for many common cancers remain toxic and are often ineffective. Cellular immunotherapy has the potential to be a highly targeted alternative, with low toxicity to normal tissues but a high capacity to eradicate tumor. In this chapter we describe approaches that generate cellular therapies using active immunization with cells, proteins, peptides, or nucleic acids, as well as efforts that use adoptive transfer of effector cells that directly target antigens on malignant cells. Many of these approaches are proving successful in hematologic malignancy and in melanoma. In this chapter we discuss the advantages and limitations of each and how over the next decade investigators will attempt to broaden their reach, increase their efficacy, and simplify their application.
Collapse
Affiliation(s)
- Fatma V Okur
- Baylor College of Medicine, Methodist Hospital and Texas Children's Hospital, Houston, TX, USA
| | | |
Collapse
|
1012
|
June CH, Blazar BR, Riley JL. Engineering lymphocyte subsets: tools, trials and tribulations. Nat Rev Immunol 2009; 9:704-16. [PMID: 19859065 DOI: 10.1038/nri2635] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cell-based therapies with various lymphocyte subsets hold promise for the treatment of several diseases, including cancer and disease resulting from inflammation and infection. The ability to genetically engineer lymphocyte subsets has the potential to improve the natural immune response and correct impaired immunity. In this Review we focus on the lymphocyte subsets that have been modified genetically or by other means for therapeutic benefit, on the technologies used to engineer lymphocytes and on the latest progress and hurdles in translating these technologies to the clinic.
Collapse
Affiliation(s)
- Carl H June
- Department of Pathology and Laboratory Medicine and Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
1013
|
Abstract
BACKGROUND Chemotherapy-resistant lymphomas can be cured with allogeneic hematopoietic cell transplantation, demonstrating the susceptibility of these tumors to T cell mediated immune responses. However, high rates of transplant-related morbidity and mortality limit this approach. Efforts have, therefore, been made to develop alternative T cell based therapies, and there is growing evidence that adoptive therapy with T cells targeted to lymphoma-associated antigens may be a safe and effective new method for treating this group of diseases. OBJECTIVE/METHODS We review publications on adoptive therapy with ex vivo expanded T cells targeting viral antigens, as well as genetically modified autologous T cells, as strategies for the treatment of lymphoma, with the goal of providing an overview of these approaches. RESULTS/CONCLUSIONS Epstein-Barr virus specific T cell therapy is an effective and safe method of treating Epstein-Barr virus associated lymphomas; however, most lymphoma subtypes do not express EBV antigens. For these diseases, adoptive immunotherapy with genetically modified T cells expressing chimeric T cell receptors targeting lymphoma-associated antigens such as CD19 and CD20 appears to be a promising alternative. Recent innovations including enhanced co-stimulation, exogenous cytokine administration and use of memory T cells promise to overcome many of the limitations and pitfalls initially encountered with this approach.
Collapse
Affiliation(s)
- Brian G Till
- Research Associate, Acting Instructor, University of Washington, Fred Hutchinson Cancer Research Center, Department of Medicine, Seattle, WA 98109, USA.
| | | |
Collapse
|
1014
|
Alonso-Camino V, Sánchez-Martín D, Compte M, Álvarez-Vallina LSL. Lymphocyte display: a novel antibody selection platform based on T cell activation. PLoS One 2009; 4:e7174. [PMID: 19777065 PMCID: PMC2747005 DOI: 10.1371/journal.pone.0007174] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 08/30/2009] [Indexed: 12/02/2022] Open
Abstract
Since their onset, display technologies have proven useful for the selection of antibodies against a variety of targets; however, most of the antibodies selected with the currently available platforms need to be further modified for their use in humans, and are restricted to accessible antigens. Furthermore, these platforms are not well suited for in vivo selections. We present here a novel cell based antibody display platform, which takes advantage of the functional capabilities of T lymphocytes. The display of antibodies on the surface of T lymphocytes, as a part of a chimeric-immune receptor (CIR) mediating signaling, may ideally link the antigen-antibody interaction to a demonstrable change in T cell phenotype, due to subsequent expression of the early T cell activation marker CD69. In this proof-of-concept, an in vitro selection was carried out using a human T cell line lentiviral-transduced to express a tumor-specific CIR on the surface, against a human tumor cell line expressing the carcinoembryonic antigen. Based on an effective interaction between the CIR and the tumor antigen, we demonstrated that combining CIR-mediated activation with FACS sorting of CD69+ T cells, it is possible to isolate binders to tumor specific cell surface antigen, with an enrichment factor of at least 103-fold after two rounds, resulting in a homogeneous population of T cells expressing tumor-specific CIRs.
Collapse
Affiliation(s)
- Vanesa Alonso-Camino
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - David Sánchez-Martín
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Marta Compte
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | |
Collapse
|
1015
|
Kochenderfer JN, Feldman SA, Zhao Y, Xu H, Black MA, Morgan RA, Wilson WH, Rosenberg SA. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother 2009; 32:689-702. [PMID: 19561539 PMCID: PMC2747302 DOI: 10.1097/cji.0b013e3181ac6138] [Citation(s) in RCA: 329] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
T cells can be engineered to express the genes of chimeric antigen receptors (CARs) that recognize tumor-associated antigens. We constructed and compared 2 CARs that contained a single chain variable region moiety that recognized CD19. One CAR contained the signaling moiety of the 4-1BB molecule and the other did not. We selected the CAR that did not contain the 4-1BB moiety for further preclinical development. We demonstrated that gammaretroviruses encoding this receptor could transduce human T cells. Anti-CD19-CAR-transduced CD8+ and CD4+ T cells produced interferon-gamma and interleukin-2 specifically in response to CD19+ target cells. The transduced T cells specifically killed primary chronic lymphocytic leukemia (CLL) cells. We transduced T cells from CLL patients that had been previously treated with chemotherapy. We induced these T cells to proliferate sufficiently to provide enough cells for clinical adoptive T cell transfer with a protocol consisting of an initial stimulation with an anti-CD3 monoclonal antibody (OKT3) before transduction followed by a second OKT3 stimulation 7 days after transduction. This protocol was successfully adapted for use in CLL patients with high peripheral blood leukemia cell counts by depleting CD19+ cells before the initial OKT3 stimulation. In preparation for a clinical trial that will enroll patients with advanced B cell malignancies, we generated a producer cell clone that produces retroviruses encoding the anti-CD19 CAR, and we produced sufficient retroviral supernatant for the proposed clinical trial under good manufacturing practice conditions.
Collapse
MESH Headings
- Antigens, CD19/genetics
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic/immunology
- Drug Evaluation, Preclinical
- Enzyme-Linked Immunosorbent Assay
- Gammaretrovirus/genetics
- Genetic Vectors/genetics
- Humans
- Immunotherapy/methods
- Interferon-gamma/metabolism
- Interleukin-2/metabolism
- K562 Cells
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Muromonab-CD3/immunology
- Receptors, Antigen/genetics
- Receptors, Antigen/immunology
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- James N Kochenderfer
- Surgery Branch of the National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
1016
|
Bullain SS, Sahin A, Szentirmai O, Sanchez C, Lin N, Baratta E, Waterman P, Weissleder R, Mulligan RC, Carter BS. Genetically engineered T cells to target EGFRvIII expressing glioblastoma. J Neurooncol 2009; 94:373-82. [PMID: 19387557 PMCID: PMC2730985 DOI: 10.1007/s11060-009-9889-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2008] [Accepted: 04/02/2009] [Indexed: 10/20/2022]
Abstract
Glioblastoma remains a significant therapeutic challenge, warranting further investigation of novel therapies. We describe an immunotherapeutic strategy to treat glioblastoma based on adoptive transfer of genetically modified T-lymphocytes (T cells) redirected to kill EGFRvIII expressing gliomas. We constructed a chimeric immune receptor (CIR) specific to EGFRvIII, (MR1-zeta). After in vitro selection and expansion, MR1-zeta genetically modified primary human T-cells specifically recognized EGFRvIII-positive tumor cells as demonstrated by IFN-gamma secretion and efficient tumor lysis compared to control CIRs defective in EGFRvIII binding (MRB-zeta) or signaling (MR1-delzeta). MR1-zeta expressing T cells also inhibited EGFRvIII-positive tumor growth in vivo in a xenografted mouse model. Successful targeting of EGFRvIII-positive tumors via adoptive transfer of genetically modified T cells may represent a new immunotherapy strategy with great potential for clinical applications.
Collapse
Affiliation(s)
- Szofia S. Bullain
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| | - Ayguen Sahin
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| | - Oszkar Szentirmai
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| | - Carlos Sanchez
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| | - Ning Lin
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| | - Elizabeth Baratta
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| | - Peter Waterman
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Richard C. Mulligan
- Harvard Medical School Department of Genetics, 77 Louis Pasteur Avenue, Boston, MA 02115, USA
| | - Bob S. Carter
- Neurosurgical Service, Massachusetts General Hospital, 55 Fruit Street, Yawkey 9026, Boston, MA 02114, USA
| |
Collapse
|
1017
|
Brenner MK, Okur FV. Overview of gene therapy clinical progress including cancer treatment with gene-modified T cells. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2009; 2009:675-681. [PMID: 20008253 PMCID: PMC2929387 DOI: 10.1182/asheducation-2009.1.675] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
It is now twenty years since the first legal gene transfer studies were approved, and there has been considerable disappointment in the slow rate of progress that followed the initial studies. Gradually, however, as the limitations of available vectors are acknowledged and overcome, and with advances in our understanding of the molecular and cell biology of genetic diseases and of cancer, unequivocal successes are now being reported. In this paper we describe the remaining major roadblocks to successful gene therapy and outline approaches to overcome them. We also illustrate how genetically modified immune system cells are already being used for the effective treatment of hematological and other malignancies, and how these approaches are being modified so that they can be effective in treating a broader range of malignancies.
Collapse
Affiliation(s)
- Malcolm K Brenner
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital, Houston, TX 77030-2303, USA.
| | | |
Collapse
|
1018
|
Abstract
The development of effective immunotherapy strategies for glioma requires adequate understanding of the unique immunological microenvironment in the central nervous system (CNS) and CNS tumors. Although the CNS is often considered to be an immunologically privileged site and poses unique challenges for the delivery of effector cells and molecules, recent advances in technology and discoveries in CNS immunology suggest novel mechanisms that may significantly improve the efficacy of immunotherapy against gliomas. In this review, we first summarize recent advances in the CNS and CNS tumor immunology. We address factors that may promote immune escape of gliomas. We also review advances in passive and active immunotherapy strategies for glioma, with an emphasis on lessons learned from recent early-phase clinical trials. We also discuss novel immunotherapy strategies that have been recently tested in non-CNS tumors and show great potential for application to gliomas. Finally, we discuss how each of these promising strategies can be combined to achieve clinical benefit for patients with gliomas.
Collapse
Affiliation(s)
- Hideho Okada
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | |
Collapse
|
1019
|
Willemsen R, Chames P, Schooten E, Gratama JW, Debets R. Selection of human antibody fragments directed against tumor T-cell epitopes for adoptive T-cell therapy. Cytometry A 2008; 73:1093-9. [DOI: 10.1002/cyto.a.20644] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
1020
|
Elkord E, Hawkins RE, Stern PL. Immunotherapy for gastrointestinal cancer: current status and strategies for improving efficacy. Expert Opin Biol Ther 2008; 8:385-95. [PMID: 18352844 DOI: 10.1517/14712598.8.4.385] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Despite improvement in conventional strategies for treating gastrointestinal (GI) carcinoma, large numbers of patients still suffer from incurable or progressive disease. OBJECTIVE Here we consider the prospects for circumventing limitations and maximising the efficacy of different immunotherapies. METHODS We summarise different cancer vaccines and targeted drugs and highlight the scientific rationale of using immunotherapy for targeting GI cancers, in addition to the potential strategies for improving immunotherapeutic efficacy. RESULTS/CONCLUSION Many cancer vaccines and antibody-directed therapies have been tested in early phase clinical trials and demonstrated proof of concept and safety. As yet few have been properly evaluated for clinical efficacy; although adoptive transfer of tumour-associated-antigen-specific T cells has shown dramatic clinical responses in some patients. The recognition of a role for T regulatory cells in limiting anti-tumour immunity has provided momentum for developing strategies to over-ride such immunoinhibitory effects. There is some evidence that conventional therapies may work by influencing these negative factors and allowing expression of immune control mechanisms. An important developing area for clinical evaluation is the testing of combined conventional and immunotherapeutic modalities which may provide for synergy; thereby circumventing the limitations of individualised treatments and generating additional clinical benefits.
Collapse
Affiliation(s)
- Eyad Elkord
- University of Manchester, Paterson Institute for Cancer Research, Department of Medical Oncology, Wilmslow Road, Manchester M20 4BX, UK.
| | | | | |
Collapse
|
1021
|
Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer 2008; 8:299-308. [PMID: 18354418 PMCID: PMC2553205 DOI: 10.1038/nrc2355] [Citation(s) in RCA: 1207] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adoptive cell therapy (ACT) using autologous tumour-infiltrating lymphocytes has emerged as the most effective treatment for patients with metastatic melanoma and can mediate objective cancer regression in approximately 50% of patients. The use of donor lymphocytes for ACT is an effective treatment for immunosuppressed patients who develop post-transplant lymphomas. The ability to genetically engineer human lymphocytes and use them to mediate cancer regression in patients, which has recently been demonstrated, has opened possibilities for the extension of ACT immunotherapy to patients with a wide variety of cancer types and is a promising new approach to cancer treatment.
Collapse
Affiliation(s)
- Steven A Rosenberg
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, Maryland 20892, USA.
| | | | | | | | | |
Collapse
|
1022
|
|
1023
|
Abstract
"T-bodies" are genetically engineered T cells armed with chimeric receptors whose extracellular recognition unit is comprised of an antibody-derived recognition domain and whose intracellular region is derived from lymphocyte stimulating moiety(ies). The structure of the prototypic chimeric receptor, also known as a chimeric immune receptor, is modular, designed to accomodate various functional domains and thereby to enable choice of specificity and controlled activation of T cells. The preferred antibody-derived recognition unit is a single chain variable fragment (scFv) that combines the specificity and binding residues of both the heavy and light chain variable regions of a monoclonal antibody. The most common lymphocyte activation moieties include a T-cell costimulatory (e.g. CD28) domain in tandem with a T-cell triggering (e.g. CD3zeta) moiety. By arming effector lymphocytes (such as T cells and natural killer cells) with such chimeric receptors, the engineered cell is redirected with a predefined specificity to any desired target antigen, in a non-HLA restricted manner. Chimeric receptor (CR) constructs are introduced ex vivo into T cells from peripheral lymphocytes of a given patient using retroviral vectors. Following infusion of the resulting T-bodies back into the patient, they traffic, reach their target site, and upon interaction with their target cell or tissue, they undergo activation and perform their predefined effector function. Therapeutic targets for the T-body approach include cancer and HIV-infected cells, or autoimmune effector cells. To date, the most investigated area is cancer therapy. Here, the T-bodies are advantageous because their tumor recognition is not HLA-specific and, therefore, the same constructs can be used for a wide spectrum of patients and cancers.
Collapse
Affiliation(s)
- Z Eshhar
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
1024
|
Turatti F, Figini M, Balladore E, Alberti P, Casalini P, Marks JD, Canevari S, Mezzanzanica D. Redirected activity of human antitumor chimeric immune receptors is governed by antigen and receptor expression levels and affinity of interaction. J Immunother 2007; 30:684-93. [PMID: 17893561 DOI: 10.1097/cji.0b013e3180de5d90] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Novel Ab-based immunotherapeutic strategies have exploited T-cell receptor-like chimeric immune receptors (CIR) expressed on the surface of transduced human peripheral blood mononuclear cell (PBMC) to redirect potent non-major histocompatibility complex-dependent cytotoxicity to tumor cells expressing a tumor-associated antigens. We transduced human PBMC with 2 fully human CIRs that trigger through the zeta-chain of CD3 and contain either one of two human scFv specific for the same epitope on the extracellular domain of HER2 but with distinctly different affinities (KD 1616 and 1 nM) for this antigen. Potent direct CIR-mediated killing and in vitro tumor growth inhibition mediated by transduced PBMC were observed against targets expressing different levels of HER2. High-affinity CIR showed stronger ability to bind Ag and retain binding than low-affinity CIR. When lytic potential of the 2 CIRs was evaluated, their efficiency was comparable under conditions of high CIR and Ag expression, whereas low-affinity CIR was more efficient than high-affinity CIR in conditions of limiting Ag and CIR expression levels. When tumor growth inhibition was evaluated, Ag and CIR levels, rather than CIR affinity appeared relevant. Ag-driven CIR activation resulted in the production of soluble factors mediating efficient bystander effect. By carefully defining CIR surface expression and increasing affinity for a specific target antigen, it may be possible to selectively exclude CIR-mediated activity against targets expressing low levels of antigen, as normal cells. On the contrary, low antigen-expressing tumor variants could be eliminated by decreasing CIR affinity. Tuning CIR expression and affinity might help in discriminating different biologic contexts.
Collapse
MESH Headings
- Antibody Affinity
- Antigens, Neoplasm/immunology
- Cell Line
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Humans
- Immunoglobulin Variable Region/immunology
- Leukocytes, Mononuclear/immunology
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transduction, Genetic
Collapse
Affiliation(s)
- Fabio Turatti
- Molecular Therapies Unit, Department of Experimental Oncology, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
1025
|
Abstract
Adoptive transfer of antigen-specific T cells is a promising approach for preventing progressive viral infections in immunosuppressed hosts. By contrast, effective T-cell therapy of malignant disease has proven to be much more difficult to achieve. This, in part, reflects the difficulty of isolating high avidity T cells specific for tumor-associated antigens, many of which are self-antigens that have induced some level of tolerance in the host. Even when tumor-reactive T cells can be isolated, the ability of these cells to survive in vivo and traffic to tumor sites is often impaired. Additionally, most tumors employ multiple mechanisms to escape T-cell recognition, including interference in antigen presentation, secretion of inhibitory factors and recruitment of regulatory or immunosuppressive cells. The genetic modification of T cells prior to transfer provides a potential means to overcome many of these obstacles and enhance the efficacy of T-cell therapy. This review article discusses the rationale for genetic modification of T cells, the critical steps involved in gene transfer, and potential advantages and disadvantages of strategies that are now being examined to engineer improved effector T cells for the treatment of human infectious and malignant disease.
Collapse
Affiliation(s)
- Carolina Berger
- Fred Hutchinson Cancer Research Center, Program in Immunology, Seattle, WA 98109-1024, USA.
| | | | | | | |
Collapse
|
1026
|
Flynn JP, O'Hara MH, Gandhi SJ. Preclinical rationale for combining radiation therapy and immunotherapy beyond checkpoint inhibitors (i.e., CART). Transl Lung Cancer Res 2007; 6:159-168. [PMID: 28529898 DOI: 10.21037/tlcr.2017.03.07] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
An increasing appreciation for the role of the immune system in targeting cancer cells over the last decade has led to the development of several immunomodulatory agents aimed at enhancing the systemic antitumor immune response. One such method is the use of T cells that are genetically engineered to express chimeric antigen receptors (CARs). The remarkable success of this approach in advanced hematologic malignancies has garnered much enthusiasm for using this novel tool in treating other cancers. However, multiple challenges have hampered the application of this therapy to a broader set of solid tumors, most notably lung cancer. Immunotherapy has already shown great success in lung cancer, and is now the first-line treatment in PD-L1 expressing metastatic disease. Given the mounting evidence that radiation therapy plays a crucial role in amplifying the immune response elicited by immunomodulatory agents, there is potential for radiation to help in overcoming some of these challenges. In this review, we describe the basic principles of CAR T cell therapy and examine its successes and challenges to date. We then discuss the preclinical and clinical data supporting the use of radiation with immunomodulatory agents with a focus on preclinical rationale for combining CAR T cells and radiation therapy in future experiments with a focus on lung cancer.
Collapse
Affiliation(s)
- James P Flynn
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark H O'Hara
- Division of Hematology Oncology, Department of Medicine, Perelman School of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Saumil J Gandhi
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
1027
|
Abstract
Adoptive transfer of antigen-specific T lymphocytes is a powerful therapy for the treatment of opportunistic disease and some virus-associated malignancies such as Epstein-Barr virus-positive post-transplant lymphoproliferative disease. However, this strategy has been less successful in patients with nonviral cancers owing to their many and varied immune evasion mechanisms. These mechanisms include downregulation of target antigens and antigen-presenting machinery, secretion of inhibitory cytokines, and recruitment of regulatory immune cells to the tumor site. With increased understanding of the tumor microenvironment and the behavior and persistence of ex vivo-manipulated, adoptively transferred T cells, two novel approaches for increasing the efficacy of T cell therapy have been proposed. The first involves genetic modification of tumor-specific T cells to improve their biological function, for example by augmenting their ability to recognize tumor cells or their resistance to tumor-mediated immunosuppression. The second requires modifications to the host environment to improve the homeostatic expansion of infused T cells or to eliminate inhibitory T cell subsets. In this review, we discuss current, promising strategies to improve adoptive T cell therapy for the treatment of cancer.
Collapse
Affiliation(s)
- Ann M Leen
- Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas 77030, USA
| | | | | |
Collapse
|
1028
|
Vera J, Savoldo B, Vigouroux S, Biagi E, Pule M, Rossig C, Wu J, Heslop HE, Rooney CM, Brenner MK, Dotti G. T lymphocytes redirected against the kappa light chain of human immunoglobulin efficiently kill mature B lymphocyte-derived malignant cells. Blood 2006; 108:3890-7. [PMID: 16926291 PMCID: PMC1895462 DOI: 10.1182/blood-2006-04-017061] [Citation(s) in RCA: 234] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2006] [Accepted: 07/21/2006] [Indexed: 01/04/2023] Open
Abstract
There has been interest in generating T cells expressing chimeric artificial receptors (CARs) targeting CD19/CD20 antigens to treat B-cell lymphomas. If successful, however, this approach would likely impair humoral immunity because T cells may persist long-term. Most low-grade lymphoma and chronic lymphocytic leukemia (B-CLL) cells express monoclonal immunoglobulins carrying either kappa or lambda light chains. We, therefore, explored whether T lymphocytes could be genetically modified to target the tumor-associated light chain, sparing B lymphocytes expressing the reciprocal light chain, and consequently reduce impairment of humoral immunity. We found that T lymphocytes expressing the anti-kappa light chain CAR showed cytotoxic activity against Igkappa(+) tumor cell lines and B-CLL cells both in vitro and in vivo. We also found that the incorporation of the CD28 endodomain within the CAR enhanced the in vitro and in vivo expansion of transgenic T cells after tumor-associated antigen stimulation. Free Igkappa(+) did not compromise the ability of redirected T lymphocytes to eliminate Igkappa(+) tumors because these free immunoglobulins served to sustain proliferation of CAR-CD28 transgenic T cells. Thus, adoptive transfer of T lymphocytes targeting the appropriate light chain could be a useful immunotherapy approach to treat B-lymphocyte malignancies that clonally express immunoglobulin without entirely compromising humoral immunity.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Animals
- Antibody Formation/immunology
- Antigens, CD19/genetics
- Antigens, CD19/immunology
- Antigens, CD20/genetics
- Antigens, CD20/immunology
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Cell Proliferation
- Gene Expression Regulation, Leukemic/immunology
- Humans
- Immunoglobulin kappa-Chains/immunology
- Immunoglobulin lambda-Chains/immunology
- K562 Cells
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Mice
- Mice, SCID
- Neoplasm Proteins/immunology
- Protein Structure, Tertiary/genetics
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Juan Vera
- Center for Cell and Gene Therapy, Baylor College of Medicine, Fannin St, MC 3-3320, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1029
|
Milone MC, June CH. Adoptive immunotherapy: new ways to skin the cat? Clin Immunol 2006; 117:101-3. [PMID: 16214079 DOI: 10.1016/j.clim.2005.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2005] [Accepted: 08/24/2005] [Indexed: 11/22/2022]
Affiliation(s)
- Michael C Milone
- Abramson Family Cancer Research Institute and the Department of Pathology and Laboratory Medicine, University of Pennsylvania, PA 19104, USA.
| | | |
Collapse
|
1030
|
Tonn T, Seifried E. Natural Killer Cells for the Treatment of Malignancies*. Transfus Med Hemother 2006. [DOI: 10.1159/000091112] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
1031
|
Friedmann-Morvinski D, Eshhar Z. Adoptive immunotherapy of cancer using effector lymphocytes redirected with antibody specificity. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.uct.2006.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
1032
|
Abstract
A central goal immunologists has been to develop targeted therapies that will induce or maintain immunologic tolerance in the absence of potentially harmful immunosuppression. The ability to isolate and expand regulatory T-cell populations with immune suppressive activity will enable new forms of adoptive immunotherapy that may achieve this long held dream. Assuming that certain technical challenges regarding the manufacturing of regulatory T cells can be overcome, a wide variety of clinical applications can be envisioned using adoptively transferred CD4(+)CD25(+) regulatory T cells. It is likely that suppressor T cells will first be tested for their ability to prevent or treat graft-versus-host disease (GVHD) following allogeneic bone marrow or stem cell transplantation. A related approach will be clinical studies to induce allogeneic or xenogeneic tolerance using regulatory T cells in solid organ transplantation. A more technically challenging approach will be the use of regulatory T-cell therapy for autoimmune disorders. Finally on the horizon are approaches that will use genetically engineered lymphocytes to replace regulatory T cells in the immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, and potentially to create more potent regulatory T (Treg) cells with enhanced suppressive activity.
Collapse
Affiliation(s)
- Carl H June
- The Abramson Family Cancer Research Institute, The Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | |
Collapse
|
1033
|
Kershaw MH, Teng MWL, Smyth MJ, Darcy PK. Supernatural T cells: genetic modification of T cells for cancer therapy. Nat Rev Immunol 2006; 5:928-40. [PMID: 16322746 DOI: 10.1038/nri1729] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapy is receiving much attention as a means of treating cancer, but complete, durable responses remain rare for most malignancies. The natural immune system seems to have limitations and deficiencies that might affect its ability to control malignant disease. An alternative to relying on endogenous components in the immune repertoire is to generate lymphocytes with abilities that are greater than those of natural T cells, through genetic modification to produce 'supernatural' T cells. This Review describes how such T cells can circumvent many of the barriers that are inherent in the tumour microenvironment while optimizing T-cell specificity, activation, homing and antitumour function.
Collapse
Affiliation(s)
- Michael H Kershaw
- Peter MacCallum Cancer Centre, Saint Andrews Place, East Melbourne, Victoria 3002, Australia.
| | | | | | | |
Collapse
|
1034
|
Biglari A, Southgate TD, Fairbairn LJ, Gilham DE. Human monocytes expressing a CEA-specific chimeric CD64 receptor specifically target CEA-expressing tumour cells in vitro and in vivo. Gene Ther 2006; 13:602-10. [PMID: 16397508 DOI: 10.1038/sj.gt.3302706] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is one means by which macrophages (as well as natural killer cells and granulocytes) elicit a cytotoxic response. This is achieved via interaction of the Fc-gamma-receptor (CD64) with the Fc portion of antibody bound to target cells. We have created a chimeric CD64 molecule that incorporates a single chain Fv molecule, targeted against human carcinoembryonic antigen (CEA), fused to the membrane spanning and cytosolic domains of human CD64. Following adenoviral transfer to primary human monocytes, this chimeric CD64 receptor induced antigen-specific cytokine secretion during culture on immobilised CEA protein or on CEA-expressing tumour cells. Moreover, CEA targeted, but not control, monocytes effectively retarded CEA-positive tumour cell growth in vitro. Importantly, targeted monocyte cultures significantly reduced in vivo tumour growth rates in xenograft studies resulting in improved survival rates over that of control monocyte cultures. These data suggest that genetically directing monocytes against tumour antigens may be a useful means of achieving an immunotherapeutic response.
Collapse
Affiliation(s)
- A Biglari
- Cancer Research UK Gene Therapy Group and Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester, UK
| | | | | | | |
Collapse
|
1035
|
Abstract
It is generally accepted that the immune system plays an important role in controlling tumour development. However, the interplay between tumour and immune system is complex, as demonstrated by the fact that tumours can successfully establish and develop despite the presence of T cells in tumour. An improved understanding of how tumours evade T-cell surveillance, coupled with technical developments allowing the culture and manipulation of T cells, has driven the exploration of therapeutic strategies based on the adoptive transfer of tumour-specific T cells. The isolation, expansion and re-infusion of large numbers of tumour-specific T cells generated from tumour biopsies has been shown to be feasible. Indeed, impressive clinical responses have been documented in melanoma patients treated with these T cells. These studies and others demonstrate the potential of T cells for the adoptive therapy of cancer. However, the significant technical issues relating to the production of natural tumour-specific T cells suggest that the application of this approach is likely to be limited at the moment. With the advent of retroviral gene transfer technology, it has become possible to efficiently endow T cells with antigen-specific receptors. Using this strategy, it is potentially possible to generate large numbers of tumour reactive T cells rapidly. This review summarises the current gene therapy approaches in relation to the development of adoptive T-cell-based cancer treatments, as these methods now head towards testing in the clinical trial setting.
Collapse
Affiliation(s)
- W Mansoor
- Cancer Research UK, Department of Medical Oncology, University of Manchester, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - D E Gilham
- Cancer Research UK, Department of Medical Oncology, University of Manchester, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - F C Thistlethwaite
- Cancer Research UK, Department of Medical Oncology, University of Manchester, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Withington, Manchester M20 4BX, UK
| | - R E Hawkins
- Cancer Research UK, Department of Medical Oncology, University of Manchester, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Withington, Manchester M20 4BX, UK
- Cancer Research UK, Department of Medical Oncology, University of Manchester, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Wilmslow Road, Withington, Manchester M20 4BX, UK. E-mail:
| |
Collapse
|
1036
|
Guest RD, Hawkins RE, Kirillova N, Cheadle EJ, Arnold J, O'Neill A, Irlam J, Chester KA, Kemshead JT, Shaw DM, Embleton MJ, Stern PL, Gilham DE. The role of extracellular spacer regions in the optimal design of chimeric immune receptors: evaluation of four different scFvs and antigens. J Immunother 2005; 28:203-11. [PMID: 15838376 DOI: 10.1097/01.cji.0000161397.96582.59] [Citation(s) in RCA: 249] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Human peripheral blood lymphocytes can be transduced to express antigen-dependent CD3zeta chimeric immune receptors (CIRs), which function independently of the T-cell receptor (TCR). Although the exact function of these domains is unclear, previous studies imply that an extracellular spacer region is required for optimal CIR activity. In this study, four scFvs (in the context of CIRs with or without extracellular spacer regions) were used to target the human tumor-associated antigens carcinoembryonic antigen (CEA), neural cell adhesion molecule (NCAM), the oncofetal antigen 5T4, and the B-cell antigen CD19. In all cases human T-cell populations expressing the CIRs were functionally active against their respective targets, but the anti-5T4 and anti-NCAM CIRs showed enhanced specific cytokine release and cytotoxicity only when possessing an extracellular spacer region. In contrast, the anti-CEA and anti-CD19 CIRs displayed optimal cytokine release activity only in the absence of an extracellular spacer. Interestingly, mapping of the scFv epitopes has revealed that the anti-CEA scFv binds close to the amino-terminal of CEA, which is easily accessible to the CIR. In contrast, CIRs enhanced by a spacer domain appear to bind to epitopes residing closer to the cell membrane, suggesting that a more flexible extracellular domain may be required to permit the efficient binding of such epitopes. These results show that a spacer is not necessary for optimal activity of CIRs but that the optimal design varies.
Collapse
Affiliation(s)
- Ryan D Guest
- Cancer Research UK, Department of Medical Oncology, University of Manchester and Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1037
|
Teillaud JL. Engineering of monoclonal antibodies and antibody-based fusion proteins: successes and challenges. Expert Opin Biol Ther 2005; 5 Suppl 1:S15-27. [PMID: 16187937 DOI: 10.1517/14712598.5.1.s15] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Monoclonal antibodies (mAbs) and antibody-based fusion molecules have now come of age as therapeutics. Eighteen mAbs and two fusion molecules are on the market. mAbs directed against new targets are progressing at a rapid rate with the help of proteomics and genomics approaches. Many technical efforts have been made to generate a second-generation mAb with decreased immunogenicity and with optimised effector functions. The development of molecular engineering techniques applied to antibody molecules has also made it possible to design fusion molecules exhibiting different modules with bifunctional activities. Different approaches developed over the last two decades to generate and optimise therapeutic antibodies and antibody-based fusion molecules are described, with a particular focus on antibodies and fusion proteins used in oncology and inflammatory diseases. Some current technical challenges and trends are also discussed.
Collapse
Affiliation(s)
- Jean-Luc Teillaud
- University Paris 6-Pierre & Marie Curie, Unité INSERM 255, Centre de Recherches Biomédicales des Cordeliers, 15 rue de l'Ecole de Médecine, 75270 Paris Cedex 06, France.
| |
Collapse
|
1038
|
McKee MD, Roszkowski JJ, Nishimura MI. T cell avidity and tumor recognition: implications and therapeutic strategies. J Transl Med 2005; 3:35. [PMID: 16174302 PMCID: PMC1262785 DOI: 10.1186/1479-5876-3-35] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2005] [Accepted: 09/20/2005] [Indexed: 12/22/2022] Open
Abstract
In the last two decades, great advances have been made studying the immune response to human tumors. The identification of protein antigens from cancer cells and better techniques for eliciting antigen specific T cell responses in vitro and in vivo have led to improved understanding of tumor recognition by T cells. Yet, much remains to be learned about the intricate details of T cell – tumor cell interactions. Though the strength of interaction between T cell and target is thought to be a key factor influencing the T cell response, investigations of T cell avidity, T cell receptor (TCR) affinity for peptide-MHC complex, and the recognition of peptide on antigen presenting targets or tumor cells reveal complex relationships. Coincident with these investigations, therapeutic strategies have been developed to enhance tumor recognition using antigens with altered peptide structures and T cells modified by the introduction of new antigen binding receptor molecules. The profound effects of these strategies on T cell – tumor interactions and the clinical implications of these effects are of interest to both scientists and clinicians. In recent years, the focus of much of our work has been the avidity and effector characteristics of tumor reactive T cells. Here we review concepts and current results in the field, and the implications of therapeutic strategies using altered antigens and altered effector T cells.
Collapse
Affiliation(s)
- Mark D McKee
- Department of Surgery, The University of Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
1039
|
Pulè MA, Straathof KC, Dotti G, Heslop HE, Rooney CM, Brenner MK. A chimeric T cell antigen receptor that augments cytokine release and supports clonal expansion of primary human T cells. Mol Ther 2005; 12:933-41. [PMID: 15979412 DOI: 10.1016/j.ymthe.2005.04.016] [Citation(s) in RCA: 378] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2004] [Revised: 04/15/2005] [Accepted: 04/19/2005] [Indexed: 01/15/2023] Open
Abstract
The transduction of primary T cells to express chimeric T cell receptors (cTCR) for redirected targeting of tumor cells is an attractive strategy for generating tumor-specific T cells for adoptive therapy. However, tumor cells rarely provide costimulatory signals and hence cTCRs that transmit just a CD3zeta signal can only initiate target cell killing and interferon-gamma release and fail to induce full activation. Although incorporation of a CD28 component results in IL-2 release and limited proliferation, T cell activation remains incomplete. OX40 transmits a potent and prolonged T cell activation signal and is crucial for maintaining an immunological response. We hypothesize that the CD28-OX40-CD3zeta tripartite cytoplasmic domain will provide a full complement of activation, proliferation, and survival signals for enhanced anti-tumor activity.
Collapse
Affiliation(s)
- Martin A Pulè
- Center for Cell and Gene Therapy, Baylor College of Medicine, 6621 Fannin MC3-3320, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
1040
|
Cheadle EJ, Gilham DE, Thistlethwaite FC, Radford JA, Hawkins RE. Killing of non-Hodgkin lymphoma cells by autologous CD19 engineered T cells. Br J Haematol 2005; 129:322-32. [PMID: 15842655 DOI: 10.1111/j.1365-2141.2005.05456.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Adoptive immunotherapy with tumour-specific T cells is an emerging technology that may be applicable to a broad range of cancers. However, tumours can avoid T cell-mediated attack through multiple mechanisms including downregulation of major histocompatability complex (MHC). Consequently, engineering T cells to target intact protein antigen directly, thus bypassing the need for MHC presentation, can facilitate T cell targeting of tumour cells. Peripheral blood lymphocytes from nine of nine patients with non-Hodgkin lymphoma (NHL) were successfully gene-modified to express a receptor consisting of a CD19 single chain variable fragment (scFv) fused to the T cell CD3zeta signalling molecule. These T cells were functionally active against the CD19(+) Raji Burkitt's lymphoma cell line. Importantly, engineered T cells from seven of nine NHL patients efficiently lysed autologous lymph node tumour biopsy cells. There was a clear correlation between levels of CD19 expression on the tumour and effective killing by the engineered T cells. For two patients with a low or absent CD19(+) cells within the biopsy, no significant killing was observed. These results demonstrate that patients with CD19(+) NHL would be suitable candidates for this form of therapy in the setting of a phase I clinical trial.
Collapse
Affiliation(s)
- Eleanor J Cheadle
- Cancer Research UK Department of Medical Oncology, University of Manchester and Paterson Institute for Cancer Research, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | | | | | | | |
Collapse
|
1041
|
Friedmann-Morvinski D, Bendavid A, Waks T, Schindler D, Eshhar Z. Redirected primary T cells harboring a chimeric receptor require costimulation for their antigen-specific activation. Blood 2005; 105:3087-93. [PMID: 15626734 DOI: 10.1182/blood-2004-09-3737] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractChimeric receptor (CR)–redirected lymphocytes (T bodies) have great potential in the eradication of tumor cells. To extend this approach to target cells that do not express surface ligands to costimulatory receptors (eg, cancer cells), we have generated an antibody-based tripartite chimeric receptor (TPCR) that contains scFv linked to the costimulatory molecule, CD28 without its ligand-binding domain, and to the cytoplasmic moiety of the FcRγ subunit. In this study, we tested the ability of 2,4,6-trinitrophenyl (TNP)–specific TPCR to drive primary, naïve T cells derived from CR-transgenic (Tg) mice to undergo full activation. As a control, we used Tg mice expressing a similar transgene but lacking the signaling region of CD28 (Tg-TPCRΔCD28). Only T cells from the TPCR-Tg and not the CD28-truncated TPCR-Tg mice could undergo activation following stimulation on hapten-modified target cells not expressing B7. Moreover, when stimulated with TNP protein displayed on plastic, the TPCR-Tg T cells expressing the entire TPCR gene became fully activated for proliferation, interleukin 2 production, protection from apoptosis, and killing of TNP-modified target cells. Finally, TPCR-Tg mice manifested a delayed-type hypersensitivity response following skin challenge in the absence of priming. Taken together, our results suggest that the TPCR is the receptor configuration of choice for clinical applications using primary T or stem cells.
Collapse
|
1042
|
Masiero S, Del Vecchio C, Gavioli R, Mattiuzzo G, Cusi MG, Micheli L, Gennari F, Siccardi A, Marasco WA, Palù G, Parolin C. T-cell engineering by a chimeric T-cell receptor with antibody-type specificity for the HIV-1 gp120. Gene Ther 2004; 12:299-310. [PMID: 15496956 DOI: 10.1038/sj.gt.3302413] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immune-based approaches of cell therapy against viral pathogens such as the human immunodeficiency virus type 1 (HIV-1) could be of primary importance for the control of this viral infection. Here, we designed a chimeric cell surface receptor (105TCR) to provide primary human T-lymphocytes with antibody-type specificity for the HIV-1 envelope glycoprotein. This receptor includes the single chain Fv domain of the neutralizing anti-gp120 human monoclonal antibody F105, CD8alpha hinge and the transmembrane and the cytoplasmic domains of TCRzeta. Our results show that 105TCR is expressed at the cellular surface and is capable of recognizing the HIV-1 envelope glycoprotein inducing highly efficient effector T-cell responses, including extracellular signal-regulated kinase phosphorylation and cytokine secretion. Moreover, human primary CD8+ T-lymphocytes transduced by oncoretroviral and lentiviral vectors containing the 105TCR gene are able to mediate in vitro-specific cytolysis of envelope-expressing cells and HIV-1-infected CD4+ T-lymphocytes. These findings suggest that 105TCR is particularly suited for in vivo efficacy studies.
Collapse
Affiliation(s)
- S Masiero
- Department of Histology, Microbiology and Medical Biotechnologies, Section of Microbiology and Virology, University of Padova, Via A Gabelli 63, Padova 35121, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1043
|
Ma Q, Safar M, Holmes E, Wang Y, Boynton AL, Junghans RP. Anti-prostate specific membrane antigen designer T cells for prostate cancer therapy. Prostate 2004; 61:12-25. [PMID: 15287090 DOI: 10.1002/pros.20073] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND Designer T cells are T lymphocytes engineered toward specific antibody-type membrane antigens through chimeric immunoglobulin-T-cell receptor (IgTCR) genes that have been used for adoptive cellular immunotherapy. We have extended this approach to prostate specific membrane antigen (PSMA) as a means to attack prostate cancer. METHODS A chimeric anti-PSMA IgTCR gene was constructed based on an anti-PSMA monoclonal antibody, 3D8. Both T-cell lines and primary cultured human T lymphocytes were transduced with the chimeric anti-PSMA IgTCR construct and were analyzed for IgTCR expression, specific activation by PSMA, cytotoxicity against PSMA-expressing tumor cells in vitro, and retardation of tumor growth in an animal model. RESULTS The IgTCR was incorporated into the TCR-CD3 complex and formed a functional chimeric complex. The IgTCR-modified T cells were specifically activated through the chimeric receptor with PSMA as measured by IL-2 production and increased CD25 expression and specifically lysed the PSMA-expressing prostate cancer cells in vitro as well as retarded tumor growth in an animal model. CONCLUSIONS The anti-PSMA designer T cells exhibit an antibody-type specificity that can recognize PSMA expressing tumor cells in a MHC-independent fashion, resulting in T-cell activation, target cell lysis in vitro and inhibition of tumor growth in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antigens, Surface/immunology
- Cell Line, Tumor
- Cloning, Molecular
- Cytotoxicity Tests, Immunologic
- Flow Cytometry
- Glutamate Carboxypeptidase II/immunology
- Humans
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Heavy Chains/immunology
- Immunoglobulin Light Chains/genetics
- Immunoglobulin Light Chains/immunology
- Immunotherapy, Adoptive/methods
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Precipitin Tests
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Qiangzhong Ma
- Biotherapeutics Development Lab, Division of Hematology-Oncology, Beth Israel Deaconess Medical Center and Harvard Institute of Human Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
1044
|
Dermime S, Gilham DE, Shaw DM, Davidson EJ, Meziane EK, Armstrong A, Hawkins RE, Stern PL. Vaccine and antibody-directed T cell tumour immunotherapy. Biochim Biophys Acta Rev Cancer 2004; 1704:11-35. [PMID: 15238242 DOI: 10.1016/j.bbcan.2004.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2003] [Revised: 01/22/2004] [Accepted: 03/31/2004] [Indexed: 10/26/2022]
Abstract
Clearer evidence for immune surveillance in malignancy and the identification of many new tumour-associated antigens (TAAs) have driven novel vaccine and antibody-targeted responses for therapy in cancer. The exploitation of active immunisation may be particularly favourable for TAA where tolerance is incomplete but passive immunisation may offer an additional strategy where the immune repertoire is affected by either tolerance or immune suppression. This review will consider how to utilise both active and passive types of therapy delivered by T cells in the context of the failure of tumour-specific immunity by presenting cancer patients. This article will outline the progress, problems and prospects of several different vaccine and antibody-targeted approaches for immunotherapy of cancer where proof of principle pre-clinical studies have been or will soon be translated into the clinic. Two examples of vaccination-based therapies where both T cell- and antibody-mediated anti-tumour responses are likely to be relevant and two examples of oncofoetal antigen-specific antibody-directed T cell therapies are described in the following sections: (1) therapeutic vaccination against human papillomavirus (HPV) antigens in cervical neoplasia; (2) B cell lymphoma vaccines including against immunoglobulin idiotype; (3) oncofoetal antigens as tumour targets for redirecting T cells with antibody strategies.
Collapse
Affiliation(s)
- Said Dermime
- Immunology, Cancer Research UK Groups, Paterson Institute for Cancer Research and University of Manchester, Christie Hospital NHS Trust, Manchester M20 4BX, UK
| | | | | | | | | | | | | | | |
Collapse
|
1045
|
Gyobu H, Tsuji T, Suzuki Y, Ohkuri T, Chamoto K, Kuroki M, Miyoshi H, Kawarada Y, Katoh H, Takeshima T, Nishimura T. Generation and targeting of human tumor-specific Tc1 and Th1 cells transduced with a lentivirus containing a chimeric immunoglobulin T-cell receptor. Cancer Res 2004; 64:1490-5. [PMID: 14973062 DOI: 10.1158/0008-5472.can-03-2780] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD4+ Th cells, in particular IFN-gamma-producing Th1 cells, play a critical role in the activation and maintenance of Tc1 cells that are essential for tumor eradication. Here, we report the generation of artificial tumor-specific Th1 and Tc1 cells from nonspecifically activated T cells using a lentiviral transduction system. Anti-CD3-activated T cells from healthy human donors were transduced with a lentivirus containing a chimeric immunoglobulin T-cell receptor gene composed of single-chain variable fragments derived from an anticarcinoembryonic antigen (CEA)-specific monoclonal antibody fused to an intracellular signaling domain derived from the cytoplasmic portions of membrane-bound CD28 and CD3zeta. These artificial tumor-specific Tc1 and Th1 cells, termed Tc1- and Th1-T bodies, respectively, could be targeted to CEA+ tumor cells independently of MHC restriction. Specifically, Tc1-T bodies demonstrated high cytotoxicity and produced IFN-gamma in response to CEA+ tumor cell lines but not CEA- tumors. Although Th1-T bodies exhibited low cytotoxicity, they secreted high levels of IFN-gamma and interleukin-2 in response to CEA+ tumor cells. Such CEA+ tumor-specific activation was not observed in mock gene-transduced nonspecific Tc1 and Th1 cells. Moreover, Tc1- and Th1-T bodies exhibited strong antitumor activities against CEA+ human lung cancer cells implanted into RAG2(-/-) mice. Furthermore, combined therapy with Tc1- and Th1-T bodies resulted in enhanced antitumor activities in vivo. Taken together, our findings demonstrate that Tc1- and Th1-T bodies represent a promising alternative to current methods for the development of effective adoptive immunotherapies.
Collapse
Affiliation(s)
- Hiroshi Gyobu
- Division of Immunoregulation, Institute for Genetic Medicine, Hokkaido University, N-15 W-7, Kita-ku, Sapporo 060-0815, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
1046
|
Sheen AJ, Sherlock DJ, Irlam J, Hawkins RE, Gilham DE. T lymphocytes isolated from patients with advanced colorectal cancer are suitable for gene immunotherapy approaches. Br J Cancer 2003; 88:1119-27. [PMID: 12671714 PMCID: PMC2376387 DOI: 10.1038/sj.bjc.6600857] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Despite improvements in treatment, the 5-year survival for metastatic colorectal cancer remains poor. Novel approaches such as gene immunotherapy are being investigated to improve treatment. Retroviral gene transfer methods have been shown to transduce primary human T lymphocytes effectively resulting in the expression of therapeutic genes. However, a number of defects have been identified in T lymphocytes isolated from patients bearing tumour, which may have critical implications for the development of gene-targeted T cells as an anticancer therapy. To address this issue, primary T lymphocytes were isolated from patients with advanced colorectal cancer and tested for their ability to be transduced and to express subsequently a chimeric immune receptor consisting of a single-chain antibody fragment antigen-binding moiety specific for carcinoembryonic antigen (CEA) fused to the T cell receptor (TCR) CD3zeta chain. In 10 out of 10 patients, T lymphocytes were transduced, expanded in the absence of selection and tested for functional activity against CEA-expressing tumour cells. In each case, functional-specific cytotoxic activity was observed. Negligible activity was found in control cultures. This study highlights the feasibility of patient-derived T lymphocytes as a source of immune cells for autologous gene immunotherapy approaches.
Collapse
Affiliation(s)
- A J Sheen
- Cancer Research UK Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
- Department of Surgery, North Manchester Healthcare NHS Trust, Manchester M8 5RB, UK
| | - D J Sherlock
- Department of Surgery, North Manchester Healthcare NHS Trust, Manchester M8 5RB, UK
| | - J Irlam
- Cancer Research UK Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - R E Hawkins
- Cancer Research UK Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
- Cancer Research UK Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK. E-mail:
| | - D E Gilham
- Cancer Research UK Department of Medical Oncology, Paterson Institute for Cancer Research, Christie Hospital NHS Trust, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
1047
|
Willemsen RA, Debets R, Chames P, Bolhuis RLH. Genetic engineering of T cell specificity for immunotherapy of cancer. Hum Immunol 2003; 64:56-68. [PMID: 12507815 DOI: 10.1016/s0198-8859(02)00730-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The ultimate goal of immunotherapy of cancer is to make use of the immune system of patients to eliminate malignant cells. Research has mainly focused on the generation of effective antigen specific T-cell responses because of the general belief that T-cell immunity is essential in controlling tumor growth and protection against viral infections. However, the isolation of antigen specific T cells for therapeutic application is a laborious task and it is often impossible to derive autologous tumor specific T cells to be used for adoptive immunotherapy. Therefore, strategies were developed to genetically transfer tumor specific immune receptors into patients T cells. To this end, chimeric receptors were constructed that comprise antibody fragments specific for tumor associated antigens, linked to genes encoding signaling domains of the T-cell receptor (TCR) or Fc receptor. T cells expressing such chimeric antibody receptors recapitulate the immune specific responses mediated by the introduced receptor. Recently, we introduced chimeric TCR genes into primary human T lymphocytes and demonstrated that these T cell transductants acquired the exquisite major histocompatibility complex (MHC) restricted tumor specificity dictated by the introduced TCR. Importantly, the introduction of chimeric TCR bypasses problems associated with the introduction of nonmodified TCR genes, such as pairing of introduced TCR chains with endogenous TCR chains and unstable TCRalpha expression. A novel strategy which is completely independent of available tumor specific T-cell clones for cloning of the TCR genes was recently used to transfer MHC restricted tumor specificity to T cells. Human "TCR-like" Fab fragments obtained by in vitro selection of Fab phages on soluble peptide/MHC complexes were functionally expressed on human T lymphocytes, resulting in MHC restricted, tumor specific lysis and cytokine production. In addition, affinity maturation of the antibody fragment on Fab phages allows improvement of the tumor cell killing capacity of chimeric Fab receptor engrafted T cells. Developments in retroviral transfer technology now enables the generation of large numbers of antigen specific T cells that can be used for adoptive transfer to cancer patients. In this article we summarize the developments in adoptive T cell immunogenetic therapy and discuss the limitations and perspectives to improve this technology toward clinical application.
Collapse
Affiliation(s)
- Ralph A Willemsen
- Department of Clinical and Tumor Immunology, Erasmus Medical Center-Daniel den Hoed, Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
1048
|
|
1049
|
Retargeting of natural killer–cell cytolytic activity to ErbB2-expressing cancer cells results in efficient and selective tumor cell destruction. Blood 2002. [DOI: 10.1182/blood.v100.4.1265.h81602001265_1265_1273] [Citation(s) in RCA: 185] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The continuously growing natural killer (NK) cell line NK-92 is highly cytotoxic against malignant cells of various origins without affecting normal human cells. Based on this selectivity, the potential of NK-92 cells for adoptive therapy is currently being investigated in phase I clinical studies. To further enhance the antitumoral activity of NK-92 cells and expand the range of tumor entities suitable for NK-92–based therapies, here by transduction with a retroviral vector we have generated genetically modified NK-92 cells expressing a chimeric antigen receptor specific for the tumor-associated ErbB2 (HER2/neu) antigen, which is overexpressed by many tumors of epithelial origin. The chimeric antigen receptor consists of the ErbB2-specific scFv(FRP5) antibody fragment, a flexible hinge region derived from CD8, and transmembrane and intracellular regions of the CD3 ζ chain. Transduced NK-92-scFv(FRP5)-ζ cells express high levels of the fusion protein on the cell surface as determined by fluorescence-activated cell-scanning (FACS) analysis. In europium release assays, no difference in cytotoxic activity of NK-92 and NK-92-scFv(FRP5)-ζ cells toward ErbB2-negative targets was found. However, even at low effector-to-target ratios, NK-92-scFv(FRP5)-ζ cells specifically and efficiently lysed established and primary ErbB2-expressing tumor cells that were completely resistant to cytolytic activity of parental NK-92 cells. These results demonstrate that efficient retargeting of NK-92 cytotoxicity can be achieved and might allow the generation of potent cell-based therapeutics for the treatment of ErbB2-expressing malignancies.
Collapse
|
1050
|
Eaton D, Gilham DE, O'Neill A, Hawkins RE. Retroviral transduction of human peripheral blood lymphocytes with Bcl-X(L) promotes in vitro lymphocyte survival in pro-apoptotic conditions. Gene Ther 2002; 9:527-35. [PMID: 11948378 DOI: 10.1038/sj.gt.3301685] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2001] [Accepted: 01/28/2002] [Indexed: 11/08/2022]
Abstract
The prolonged in vivo survival of genetically modified effector cells is crucial to the success of any (gene-modified) adoptive cellular immunotherapy approach. In cancer clinical trials to date, however, the detection of surviving circulating gene-modified T cells has required highly sensitive techniques. In vitro studies of T cell co-stimulation have shown that up-regulation of the anti-apoptosis gene Bcl-X(L) by ligation of CD28 promotes T cell survival, but not proliferation. Here we have investigated the ability to modulate resistance to apoptosis and improve cell survival by transducing human peripheral blood lymphocytes using a retroviral vector that expresses Bcl-X(L). We show that Jurkat cells transduced with Bcl-X(L) retrovirus were partially resistant to Fas (CD95) antibody-induced apoptosis. Subsequent in vitro assays with transduced primary human lymphocytes demonstrates that over-expression of Bcl-X(L) promotes the survival of lymphocytes cultured in the absence of interleukin-2. Activation-induced apoptosis with anti-CD3(epsilon) antibody, OKT3 is also modulated. Furthermore, Bcl-X(L) over-expression in human lymphocytes delays the onset of apoptosis induced by long-term co-culture with tumour cell lines. Despite this improved in vitro survival, in a preliminary experiment to assess safety, no signs of malignancy or autoimmunity were observed in NOD/SCID mice injected with Bcl-X(L) transduced lymphocytes. These results indicate that expression of Bcl-X(L) in lymphocyte therapy either alone or in conjunction with an additional therapeutic gene could enhance persistence of cells in vivo thereby potentially improving the clinical outcome of adoptive cellular therapy.
Collapse
Affiliation(s)
- D Eaton
- CRC Department of Medical Oncology, Paterson Institute for Cancer Research, University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|