1801
|
San Miguel JF, Paiva B, Lasarte JJ. Engineering Anti-myeloma Responses Using Affinity-Enhanced TCR-Engineered T Cells. Cancer Cell 2015; 28:281-3. [PMID: 26373276 DOI: 10.1016/j.ccell.2015.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
NY-ESO-1 TCR-engineered T cells have shown activity in solid tumors. Recent work supports their use in multiple myeloma by showing that ex vivo antigen-specific expanded T cells traffic to and persist in bone marrow, are well tolerated, and produce promising response rates when infused after stem cell transplantation.
Collapse
Affiliation(s)
- Jesus F San Miguel
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain.
| | - Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| | - Juan-Jose Lasarte
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada (CIMA), IDISNA, 31008 Pamplona, Spain
| |
Collapse
|
1802
|
|
1803
|
Stauss HJ, Morris EC, Abken H. Cancer gene therapy with T cell receptors and chimeric antigen receptors. Curr Opin Pharmacol 2015; 24:113-8. [PMID: 26342910 DOI: 10.1016/j.coph.2015.08.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/10/2015] [Accepted: 08/18/2015] [Indexed: 10/23/2022]
Abstract
Viral and non-viral gene transfer technologies have been used to efficiently generate therapeutic T cells with desired cancer-specificity. Chimeric antigen receptors (CARs) redirect T cell specificity toward antibody-recognized antigens expressed on the surface of cancer cells, while T cell receptors (TCRs) extend the range of targets to include intracellular tumor antigens. CAR redirected T cells specific for the B cell differentiation antigen CD19 have shown dramatic efficacy in the treatment of B cell malignancies, while TCR-redirected T cells have shown benefits in patients suffering from solid cancer. In this review we will present strategies to optimize CAR and TCR function, and discuss the importance of target antigen selection to enhance tumor specificity, while reducing on-target and off-target toxicity.
Collapse
Affiliation(s)
- Hans J Stauss
- Institute of Immunity and Transplantation, Royal Free Campus, University College London, Rowland Hill Street, London NW3 2PF, UK.
| | - Emma C Morris
- Institute of Immunity and Transplantation, Royal Free Campus, University College London, Rowland Hill Street, London NW3 2PF, UK
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Clinic I for Internal Medicine, University Hospital Cologne, Cologne, Germany.
| |
Collapse
|
1804
|
de Guillebon E, Tartour E. Immunité antitumorale (mécanismes, immunoediting, immunosurveillance). ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2542-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
1805
|
Xu TS, Li XY, Xie ZH, Li XG, Zhang HY. Poly(o-phenylenediamine) nanosphere-conjugated capture antibody immobilized on a glassy carbon electrode for electrochemical immunoassay of carcinoembryonic antigen. Mikrochim Acta 2015. [DOI: 10.1007/s00604-015-1625-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
1806
|
Bole-Richard E, Deschamps M, Ferrand C, Robinet E. Editorial: Improving the safety of cell therapy products by suicide gene transfer. Front Pharmacol 2015; 6:174. [PMID: 26379549 PMCID: PMC4548201 DOI: 10.3389/fphar.2015.00174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/03/2022] Open
Affiliation(s)
- Elodie Bole-Richard
- Institut National de la Santé et de la Recherche Médicale UMR 1098, Etablissement Français du Sang Bourgogne/Franche-Comté Besançon, France ; SFR FRD 4234 Ingénierie et Biologie Cellulaire et Tissulaire, Université de Franche-Comté Besançon, France
| | - Marina Deschamps
- Institut National de la Santé et de la Recherche Médicale UMR 1098, Etablissement Français du Sang Bourgogne/Franche-Comté Besançon, France ; SFR FRD 4234 Ingénierie et Biologie Cellulaire et Tissulaire, Université de Franche-Comté Besançon, France
| | - Christophe Ferrand
- Institut National de la Santé et de la Recherche Médicale UMR 1098, Etablissement Français du Sang Bourgogne/Franche-Comté Besançon, France ; SFR FRD 4234 Ingénierie et Biologie Cellulaire et Tissulaire, Université de Franche-Comté Besançon, France
| | - Eric Robinet
- Institut National de la Santé et de la Recherche Médicale UMR 1110, Institut de Recherche sur les Maladies Virales et Hépatiques Strasbourg, France ; Institut Hospitalo-Universitaire de Strasbourg Strasbourg, France
| |
Collapse
|
1807
|
Sun W, Wei X, Niu A, Ma X, Li JJ, Gao D. Enhanced anti-colon cancer immune responses with modified eEF2-derived peptides. Cancer Lett 2015; 369:112-23. [PMID: 26304717 DOI: 10.1016/j.canlet.2015.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/22/2022]
Abstract
Eukaryotic elongation factor-2 (eEF2) is overexpressed in many human cancers and is an attractive target for cancer immunotherapy. The eEF2 derived polypeptides have been shown to be able to induce cytotoxic T lymphocytes from healthy donor. Here, we demonstrate the evidence indicating that modification of a segment of peptides from wild type eEF2-derived immunogenic peptides is able to further enhance its capacity of inducing antigen-specific cytotoxic T lymphocytes (CTLs) against colon cancer cells. Using peptide-MHC binding algorithms, potential HLA-A2.1-restricted epitopes capable of inducing specific CD8(+) CTLs were identified. By analyzing HLA-A2.1 affinity and immunogenicity, we further identified one novel immunogenic peptide, P739-747 (RLMEPIYLV), that elicited specific CTL responses in HLA-A2.1/K(b) transgenic mice and culture with peripheral blood lymphocytes from colon cancer patients. Furthermore, replacing certain amino acids (at positions 1, 3, 7) within the P739-747 sequence improved the immunogenicity against eEF2. Several analogs containing the auxiliary HLA-A*0201 anchor residues were able to stably bind to HLA-A*0201 and enhance CTL responses compared with the native sequence; two of them showed increased anti-tumor effects during the adoptive immunotherapy in vivo. Thus, these results support that modified immunogenic analogs are promising candidates for peptide-based cancer vaccination and immunotherapy.
Collapse
Affiliation(s)
- Weihong Sun
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China.
| | - Xiaofang Wei
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Airong Niu
- Department of Clinical Laboratory, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Xuezhen Ma
- Department of Oncology, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Jian Jian Li
- Department of Radiation Oncology, NCI-designated Compressive Cancer Center, University of California Davis, Sacramento, CA 95817, USA.
| | - Daiqing Gao
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| |
Collapse
|
1808
|
Abstract
Tumors originate from a number of genetic events that deregulate homeostatic mechanisms controlling normal cell behavior. The immune system, devoted to patrol the organism against pathogenic events, can identify transformed cells, and in several cases cause their elimination. It is however clear that several mechanisms encompassing both central and peripheral tolerance limit antitumor immunity, often resulting into progressive diseases. Adoptive T-cell therapy with either allogeneic or autologous T cells can transfer therapeutic immunity. To date, genetic engineering of T cells appears to be a powerful tool for shaping tumor immunity. In this review, we discuss the most recent achievements in the areas of suicide gene therapy, and TCR-modified T cells and chimeric antigen receptor gene-modified T cells. We provide an overview of current strategies aimed at improving the safety and efficacy of these approaches, with an outlook on prospective developments.
Collapse
Affiliation(s)
- Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Anna Mondino
- Lymphocyte Activation Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
1809
|
Melero I, Berman DM, Aznar MA, Korman AJ, Pérez Gracia JL, Haanen J. Evolving synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer 2015. [PMID: 26205340 DOI: 10.1038/nrc3973] [Citation(s) in RCA: 509] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Immunotherapy has now been clinically validated as an effective treatment for many cancers. There is tremendous potential for synergistic combinations of immunotherapy agents and for combining immunotherapy agents with conventional cancer treatments. Clinical trials combining blockade of cytotoxic T lymphocyte-associated antigen 4 (CTLA4) and programmed cell death protein 1 (PD1) may serve as a paradigm to guide future approaches to immuno-oncology combination therapy. In this Review, we discuss progress in the synergistic design of immune-targeting combination therapies and highlight the challenges involved in tailoring such strategies to provide maximal benefit to patients.
Collapse
Affiliation(s)
- Ignacio Melero
- Centro de Investigación Médica Aplicada (CIMA) and Clínica Universitaria, Avenida Pío XII, 55 E-31008, Universidad de Navarra, Pamplona, Spain
| | - David M Berman
- Bristol-Myers Squibb, 3551 Lawrenceville Princeton, New Jersey 08648, USA
| | - M Angela Aznar
- Centro de Investigación Médica Aplicada (CIMA) and Clínica Universitaria, Avenida Pío XII, 55 E-31008, Universidad de Navarra, Pamplona, Spain
| | - Alan J Korman
- Bristol-Myers Squibb Biologics Discovery California, 700 Bay Road, Redwood City, California 94063, USA
| | - José Luis Pérez Gracia
- Centro de Investigación Médica Aplicada (CIMA) and Clínica Universitaria, Avenida Pío XII, 55 E-31008, Universidad de Navarra, Pamplona, Spain
| | - John Haanen
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| |
Collapse
|
1810
|
Palumbo A, Da Costa NDOM, Bonamino MH, Pinto LFR, Nasciutti LE. Genetic instability in the tumor microenvironment: a new look at an old neighbor. Mol Cancer 2015; 14:145. [PMID: 26227631 PMCID: PMC4521350 DOI: 10.1186/s12943-015-0409-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 07/08/2015] [Indexed: 12/19/2022] Open
Abstract
The recent exponential increase in our knowledge of cellular and molecular mechanisms involved in carcinogenesis has largely failed to translate into new therapies and clinical practices. This lack of success may result in part from the fact that most studies focus on tumor cells as potential therapeutic targets and neglect the complex microenvironment that undergoes profound changes during tumor development. Furthermore, an unfortunate association of factors such as tumor genetic complexity, overestimation of biomarker and drug potentials, as well as a poor understanding of tumor microenvironment in diagnosis and prognosis leads to the current levels of treatment failure regarding a vast majority of cancer types. A growing body of evidence points to the importance of the functional diversity of immune and structural cells during tumor development. In this sense, the lack of technologies that would allow for molecular screening of individual stromal cell types poses a major challenge for the development of therapies targeting the tumor microenvironment. Progress in microenvironment genetic studies represents a formidable opportunity for the development of new selective drugs because stromal cells have lower mutation rates than malignant cells, and should prove to be good targets for therapy.
Collapse
Affiliation(s)
- Antonio Palumbo
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Prédio de Ciências da Saúde - Cidade Universitária, Ilha do Fundão, A. Carlos Chagas, 373 - bloco F, sala 26, 21941-902, Rio de Janeiro, RJ, Brasil. .,Programa de Carcinogênese Molecular, Instituto Nacional de Câncer José de Alencar Gomes da Silva, Rua André Cavalcanti, 37 - 6° andar - Centro, 20231-050, Rio de Janeiro, RJ, Brasil.
| | - Nathalia de Oliveira Meireles Da Costa
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer José de Alencar Gomes da Silva, Rua André Cavalcanti, 37 - 6° andar - Centro, 20231-050, Rio de Janeiro, RJ, Brasil.
| | - Martin Hernan Bonamino
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer José de Alencar Gomes da Silva, Rua André Cavalcanti, 37 - 6° andar - Centro, 20231-050, Rio de Janeiro, RJ, Brasil. .,Fundação Oswaldo Cruz, Vice-presidência de Pesquisa e Laboratórios de Referência, Rio de Janeiro, Brasil, Av. Brasil, 4365 - Pavilhão Mourisco - Manguinhos, 21040-900, Rio de Janeiro, RJ, Brasil.
| | - Luis Felipe Ribeiro Pinto
- Programa de Carcinogênese Molecular, Instituto Nacional de Câncer José de Alencar Gomes da Silva, Rua André Cavalcanti, 37 - 6° andar - Centro, 20231-050, Rio de Janeiro, RJ, Brasil.
| | - Luiz Eurico Nasciutti
- Laboratório de Interações Celulares, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Prédio de Ciências da Saúde - Cidade Universitária, Ilha do Fundão, A. Carlos Chagas, 373 - bloco F, sala 26, 21941-902, Rio de Janeiro, RJ, Brasil.
| |
Collapse
|
1811
|
Lawler SE, Chiocca EA. Oncolytic Virus-Mediated Immunotherapy: A Combinatorial Approach for Cancer Treatment. J Clin Oncol 2015. [PMID: 26215964 DOI: 10.1200/jco.2015.62.5244] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Sean E Lawler
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | |
Collapse
|
1812
|
Pahl J, Cerwenka A. Tricking the balance: NK cells in anti-cancer immunity. Immunobiology 2015; 222:11-20. [PMID: 26264743 DOI: 10.1016/j.imbio.2015.07.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 07/15/2015] [Accepted: 07/22/2015] [Indexed: 01/21/2023]
Abstract
Natural Killer (NK) cells are classically considered innate immune effector cells involved in the first line of defense against infected and malignant cells. More recently, NK cells have emerged to acquire properties of adaptive immunity in response to certain viral infections such as expansion of specific NK cell subsets and long-lasting virus-specific responses to secondary challenges. NK cells distinguish healthy cells from abnormal cells by measuring the net input of activating and inhibitory signals perceived from target cells through NK cell surface receptors. Acquisition of activating ligands in combination with reduced expression of MHC class I molecules on virus-infected and cancer cells activates NK cell cytotoxicity and release of immunostimulatory cytokines like IFN-γ. In the cancer microenvironment however, NK cells become functionally impaired by inhibitory factors produced by immunosuppressive immune cells and cancer cells. Here we review recent progress on the role of NK cells in cancer immunity. We describe regulatory factors of the tumor microenvironment on NK cell function which determine cancer cell destruction or escape from immune recognition. Finally, recent strategies that focus on exploiting NK cell anti-cancer responses for immunotherapeutic approaches are outlined.
Collapse
Affiliation(s)
- Jens Pahl
- Innate Immunity Group, D080, German Cancer Research Center, DKFZ Im Neuenheimer Feld 280, 69221 Heidelberg, Germany.
| | - Adelheid Cerwenka
- Innate Immunity Group, D080, German Cancer Research Center, DKFZ Im Neuenheimer Feld 280, 69221 Heidelberg, Germany.
| |
Collapse
|
1813
|
Abstract
There is rapidly growing interest in learning how to engineer immune cells, such as T lymphocytes, because of the potential of these engineered cells to be used for therapeutic applications such as the recognition and killing of cancer cells. At the same time, our knowhow and capability to logically engineer cellular behavior is growing rapidly with the development of synthetic biology. Here we describe how synthetic biology approaches are being used to rationally alter the behavior of T cells to optimize them for therapeutic functions. We also describe future developments that will be important in order to construct safe and precise T cell therapeutics.
Collapse
Affiliation(s)
- Chia-Yung Wu
- Dept. of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, United States
| | - Levi J Rupp
- Dept. of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, United States
| | - Kole T Roybal
- Dept. of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, United States
| | - Wendell A Lim
- Dept. of Cellular & Molecular Pharmacology, University of California San Francisco, San Francisco, CA 94158, United States; Howard Hughes Medical Institute, University of California San Francisco, San Francisco, CA 94158, United States.
| |
Collapse
|
1814
|
Thomas S, Klobuch S, Podlech J, Plachter B, Hoffmann P, Renzaho A, Theobald M, Reddehase MJ, Herr W, Lemmermann NAW. Evaluating Human T-Cell Therapy of Cytomegalovirus Organ Disease in HLA-Transgenic Mice. PLoS Pathog 2015; 11:e1005049. [PMID: 26181057 PMCID: PMC4504510 DOI: 10.1371/journal.ppat.1005049] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/25/2015] [Indexed: 01/05/2023] Open
Abstract
Reactivation of human cytomegalovirus (HCMV) can cause severe disease in recipients of hematopoietic stem cell transplantation. Although preclinical research in murine models as well as clinical trials have provided 'proof of concept' for infection control by pre-emptive CD8 T-cell immunotherapy, there exists no predictive model to experimentally evaluate parameters that determine antiviral efficacy of human T cells in terms of virus control in functional organs, prevention of organ disease, and host survival benefit. We here introduce a novel mouse model for testing HCMV epitope-specific human T cells. The HCMV UL83/pp65-derived NLV-peptide was presented by transgenic HLA-A2.1 in the context of a lethal infection of NOD/SCID/IL-2rg-/- mice with a chimeric murine CMV, mCMV-NLV. Scenarios of HCMV-seropositive and -seronegative human T-cell donors were modeled by testing peptide-restimulated and T-cell receptor-transduced human T cells, respectively. Upon transfer, the T cells infiltrated host tissues in an epitope-specific manner, confining the infection to nodular inflammatory foci. This resulted in a significant reduction of viral load, diminished organ pathology, and prolonged survival. The model has thus proven its potential for a preclinical testing of the protective antiviral efficacy of HCMV epitope-specific human T cells in the evaluation of new approaches to an immunotherapy of CMV disease. Pre-emptive CD8 T-cell therapy of human cytomegalovirus (HCMV) disease in immunocompromised recipients of hematopoietic stem cell transplantation gave promising results in clinical trials, but limited efficacy and the need of HCMV-seropositive memory cell donors has so far prevented adoptive cell transfer from becoming clinical routine. Further development is currently hampered by the lack of experimental animal models that allow preclinical testing of the protective efficacy of human T cells in functional organs. While humanized mouse models with human tissue implants are technically and statistically demanding, and are limited to studying human T-cell activation and local virus control in the implants, a more feasible model for control of systemic infection and prevention of multiple-organ CMV disease is regrettably missing. Here we introduce such a model based on infection of genetically immunocompromised, HLA-A2.1-transgenic NOD/SCID/IL-2rg-/- mice with a chimeric murine CMV engineered to express the HCMV NLV-peptide epitope. Mimicking the scenario of HCMV-unexperienced donors, human T cells transduced with a human T-cell receptor specific for HLA-A.2.1-presented NLV peptide controlled systemic infection and moderated organ disease resulting in a survival benefit. The model promises to become instrumental in defining T-cell properties that determine their protective efficacy for a further development of adoptive immunotherapy of post-transplantation CMV infection.
Collapse
Affiliation(s)
- Simone Thomas
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
- Regensburg Center of Interventional Immunology, University of Regensburg, Regensburg, Germany
- Department of Internal Medicine III, Hematology, Oncology and Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- * E-mail:
| | - Sebastian Klobuch
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
- Department of Internal Medicine III, Hematology, Oncology and Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jürgen Podlech
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Bodo Plachter
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Petra Hoffmann
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
- Regensburg Center of Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - Angelique Renzaho
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Theobald
- Department of Internal Medicine III, Hematology, Oncology and Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Matthias J. Reddehase
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital of Regensburg, Regensburg, Germany
- Regensburg Center of Interventional Immunology, University of Regensburg, Regensburg, Germany
- Department of Internal Medicine III, Hematology, Oncology and Pneumology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Niels A. W. Lemmermann
- Institute for Virology and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
1815
|
Polyakova A, Kuznetsova K, Moshkovskii S. Proteogenomics meets cancer immunology: mass spectrometric discovery and analysis of neoantigens. Expert Rev Proteomics 2015; 12:533-41. [DOI: 10.1586/14789450.2015.1070100] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
1816
|
Manzo T, Heslop HE, Rooney CM. Antigen-specific T cell therapies for cancer. Hum Mol Genet 2015; 24:R67-73. [PMID: 26160910 DOI: 10.1093/hmg/ddv270] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/07/2015] [Indexed: 12/13/2022] Open
Abstract
Adoptively transferred antigen-specific T cells that recognize tumor antigens through their native receptors have many potential benefits as treatment for virus-associated diseases and malignancies, due to their ability to selectively recognize tumor antigens, expand and persist to provide long-term protection. Infusions of T cells targeting Epstein-Barr virus (EBV) antigens have shown encouraging response rates in patients with post-transplant lymphoproliferative disease as well as EBV-positive lymphomas and nasopharyngeal cancer, although a recent study also showed that human papilloma virus-reactive T cells can induce complete regression of metastatic cervical cancer. This strategy is also being evaluated to target non-viral tumor-associated antigens. Targeting these less immunogenic antigens is more challenging, as tumor antigens are generally weak, and high avidity T cells specific for self-antigens are deleted in the thymus, but tumor responses have been reported. Current research focusses on defining factors that promote in vivo persistence of transferred cells and ameliorate the immunosuppressive microenvironment. To this end, investigators are evaluating the effects of combining adoptive transfer of antigen-specific T cells with other immunotherapy moieties such as checkpoint inhibitors. Genetic modification of infused T cells may also be used to overcome tumor evasion mechanisms, and vaccines may be used to promote in vivo proliferation.
Collapse
Affiliation(s)
- Teresa Manzo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX 77030, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX 77030, USA
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX 77030, USA
| |
Collapse
|
1817
|
Boyiadzis M, Whiteside TL, Pavletic SZ. Immunotherapy for acute leukemia. Aging (Albany NY) 2015; 7:354-5. [PMID: 26143183 PMCID: PMC4505159 DOI: 10.18632/aging.100768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Michael Boyiadzis
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Theresa L Whiteside
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Steven Z Pavletic
- National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
1818
|
Adachi K, Tamada K. Immune checkpoint blockade opens an avenue of cancer immunotherapy with a potent clinical efficacy. Cancer Sci 2015; 106:945-50. [PMID: 25981182 PMCID: PMC4556381 DOI: 10.1111/cas.12695] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/08/2015] [Accepted: 05/12/2015] [Indexed: 12/18/2022] Open
Abstract
Recent progress in tumor immunology has revealed that tumors generate immunologically restrained milieu during the process of their growth, which facilitates the escape of tumors from host immune systems. Immune checkpoint molecules, which transduce co-inhibitory signals to immuno-competent cells, are one of the most important components conferring the immunosuppressive capacity in the tumor microenvironment. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death-1 (PD-1) are typical immune checkpoint molecules intimately involved in the suppression of anti-tumor immunity. Antibodies against those molecules have been developed, such as ipilimumab (anti-CTLA-4 antibody), nivolumab and pembrolizumab (anti-PD-1 antibody), and have been approved by regulatory agencies and used in some countries. Treatment with these antibodies demonstrates previously unobserved clinical efficacies superior to the conventional therapies. In this review, we first discuss the escape mechanisms of cancer from host immune systems, and then focus on the recent advances in immune checkpoint blockade therapy and on the new findings of related immune reactions, aiming to provide a better understanding of the novel cancer immunotherapies.
Collapse
Affiliation(s)
- Keishi Adachi
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| |
Collapse
|
1819
|
McCracken MN, Cha AC, Weissman IL. Molecular Pathways: Activating T Cells after Cancer Cell Phagocytosis from Blockade of CD47 "Don't Eat Me" Signals. Clin Cancer Res 2015; 21:3597-601. [PMID: 26116271 DOI: 10.1158/1078-0432.ccr-14-2520] [Citation(s) in RCA: 152] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 06/04/2015] [Indexed: 11/16/2022]
Abstract
Recent advances with immunotherapy agents for the treatment of cancer have provided remarkable, and in some cases, curative results. Our laboratory has identified CD47 as an important "don't eat me" signal expressed on malignant cells. Blockade of the CD47:SIRP-α axis between tumor cells and innate immune cells (monocytes, macrophages, and dendritic cells) increases tumor cell phagocytosis in both solid tumors (including, but not limited to, bladder, breast, colon, lung, and pancreatic) and hematologic malignancies. These phagocytic innate cells are also professional antigen-presenting cells (APC), providing a link from innate to adaptive antitumor immunity. Preliminary studies have demonstrated that APCs present antigens from phagocytosed tumor cells, causing T-cell activation. Therefore, agents that block the CD47:SIRP-α engagement are attractive therapeutic targets as a monotherapy or in combination with additional immune-modulating agents for activating antitumor T cells in vivo.
Collapse
Affiliation(s)
- Melissa N McCracken
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California. Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Adriel C Cha
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California. Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Institute of Biomedical Studies, Baylor University, Waco, Texas
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California. Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, California. Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California. Department of Pathology, Stanford University Medical Center, Stanford, California.
| |
Collapse
|
1820
|
Meir R, Shamalov K, Betzer O, Motiei M, Horovitz-Fried M, Yehuda R, Popovtzer A, Popovtzer R, Cohen CJ. Nanomedicine for Cancer Immunotherapy: Tracking Cancer-Specific T-Cells in Vivo with Gold Nanoparticles and CT Imaging. ACS NANO 2015; 9:6363-72. [PMID: 26039633 DOI: 10.1021/acsnano.5b01939] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Application of immune cell-based therapy in routine clinical practice is challenging due to the poorly understood mechanisms underlying success or failure of treatment. Development of accurate and quantitative imaging techniques for noninvasive cell tracking can provide essential knowledge for elucidating these mechanisms. We designed a novel method for longitudinal and quantitative in vivo cell tracking, based on the superior visualization abilities of classical X-ray computed tomography (CT), combined with state-of-the-art nanotechnology. Herein, T-cells were transduced to express a melanoma-specific T-cell receptor and then labeled with gold nanoparticles (GNPs) as a CT contrast agent. The GNP-labeled T-cells were injected intravenously to mice bearing human melanoma xenografts, and whole-body CT imaging allowed examination of the distribution, migration, and kinetics of T-cells. Using CT, we found that transduced T-cells accumulated at the tumor site, as opposed to nontransduced cells. Labeling with gold nanoparticles did not affect T-cell function, as demonstrated both in vitro, by cytokine release and proliferation assays, and in vivo, as tumor regression was observed. Moreover, to validate the accuracy and reliability of the proposed cell tracking technique, T-cells were labeled both with green fluorescent protein for fluorescence imaging, and with GNPs for CT imaging. A remarkable correlation in signal intensity at the tumor site was observed between the two imaging modalities, at all time points examined, providing evidence for the accuracy of our CT cell tracking abilities. This new method for cell tracking with CT offers a valuable tool for research, and more importantly for clinical applications, to study the fate of immune cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Rinat Meir
- †Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Katerina Shamalov
- ‡Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Oshra Betzer
- †Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Menachem Motiei
- †Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Miryam Horovitz-Fried
- ‡Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Ronen Yehuda
- §The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Aron Popovtzer
- ∥Davidoff Cancer Center, Rabin Medical Center, Beilinson Campus, Petah Tiqwa 49100, Israel
| | - Rachela Popovtzer
- †Faculty of Engineering and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Cyrille J Cohen
- ‡Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 52900, Israel
| |
Collapse
|
1821
|
Abstract
Scientific advances in the last decade have demonstrated the critical role of host immune system in the elimination and suppression of cancer cells. Better knowledge of signaling pathways has enabled the development of new cancer immunotherapy. The discovery of negative feedback mechanisms following the lymphocyte activation has promoted the development of new antibodies targeting molecule inhibitors such as PD1, PDL1 or CTLA-4. Dramatic results were obtained with melanoma. Checkpoint inhibitors (pembrolizumab and ipilimumab) have many advantages in terms of rate of objective response and overall survival. Recent studies in translational research aimed to understand and analyze mechanisms of action of anti-PD1/anti-PDL1. Expression of PDL1 in the tumor is associated with a significantly greater objective response rate (immunohistochemistry). Nevertheless, limits with tumor immunohistochemical analysis encourage new biomarkers research. Other immunotherapy approaches, such as cell and gene therapies using engineered T cells call for further advancements to broaden their applicability. However, these therapies are very expensive and their manufacturing process very restrictive, which could lately limit their use in case of inefficiency of checkpoint inhibitors or when lymphocytic infiltration in tumor is absent. In this case, the objective would be to engineer ex vivo the patient's immune system by restoring the ability of T cells to identify and suppress tumor cells. Currently, two gene-reprogramming tools are under development: chimeric antigen receptor and TCR modified T cells.
Collapse
|
1822
|
Harnessing the Microbiome to Enhance Cancer Immunotherapy. J Immunol Res 2015; 2015:368736. [PMID: 26101781 PMCID: PMC4458560 DOI: 10.1155/2015/368736] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/10/2015] [Indexed: 12/20/2022] Open
Abstract
The microbiota plays a key role in regulating the innate and adaptive immune system. Herein, we review the immunological aspects of the microbiota in tumor immunity in mice and man, with a focus on toll-like receptor (TLR) agonists, vaccines, checkpoint modulators, chemotherapy, and adoptive T cell transfer (ACT) therapies. We propose innovative treatments that may safely harness the microbiota to enhance T cell-based therapies in cancer patients. Finally, we highlight recent developments in tumor immunotherapy, particularly novel ways to modulate the microbiome and memory T cell responses to human malignancies.
Collapse
|
1823
|
Aranda F, Buqué A, Bloy N, Castoldi F, Eggermont A, Cremer I, Fridman WH, Fucikova J, Galon J, Spisek R, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Adoptive cell transfer for oncological indications. Oncoimmunology 2015; 4:e1046673. [PMID: 26451319 DOI: 10.1080/2162402x.2015.1046673] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 04/25/2015] [Indexed: 12/15/2022] Open
Abstract
One particular paradigm of anticancer immunotherapy relies on the administration of (potentially) tumor-reactive immune effector cells. Generally, these cells are obtained from autologous peripheral blood lymphocytes (PBLs) ex vivo (in the context of appropriate expansion, activation and targeting protocols), and re-infused into lymphodepleted patients along with immunostimulatory agents. In spite of the consistent progress achieved throughout the past two decades in this field, no adoptive cell transfer (ACT)-based immunotherapeutic regimen is currently approved by regulatory agencies for use in cancer patients. Nonetheless, the interest of oncologists in ACT-based immunotherapy continues to increase. Accumulating clinical evidence indicates indeed that specific paradigms of ACT, such as the infusion of chimeric antigen receptor (CAR)-expressing autologous T cells, are associated with elevated rates of durable responses in patients affected by various neoplasms. In line with this notion, clinical trials investigating the safety and therapeutic activity of ACT in cancer patients are being initiated at an ever increasing pace. Here, we review recent preclinical and clinical advances in the development of ACT-based immunotherapy for oncological indications.
Collapse
Affiliation(s)
- Fernando Aranda
- Group of Immune Receptors of the Innate and Adaptive System; Institut d'Investigacions Biomédiques August Pi i Sunyer (IDIBAPS) ; Barcelona, Spain
| | - Aitziber Buqué
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Francesca Castoldi
- INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Faculté de Medicine; Université Paris Sud/Paris XI ; Le Kremlin-Bicêtre, France ; Sotio a.c. ; Prague, Czech Republic
| | | | - Isabelle Cremer
- INSERM; U1138 ; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Wolf Hervé Fridman
- INSERM; U1138 ; Paris, France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France
| | - Jitka Fucikova
- Sotio a.c. ; Prague, Czech Republic ; Dept. of Immunology; 2nd Faculty of Medicine and University Hospital Motol; Charles University ; Prague, Czech Republic
| | - Jérôme Galon
- INSERM; U1138 ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris, France
| | - Radek Spisek
- Sotio a.c. ; Prague, Czech Republic ; Dept. of Immunology; 2nd Faculty of Medicine and University Hospital Motol; Charles University ; Prague, Czech Republic
| | - Eric Tartour
- Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris, France ; INSERM; U970 ; Paris, France ; Paris-Cardiovascular Research Center (PARCC) ; Paris, France ; Service d'Immunologie Biologique; Hôpital Européen Georges Pompidou (HEGP); AP-HP ; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1015; CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris, France ; Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM; U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Université Paris Descartes/Paris V , Sorbonne Paris Cité , Paris, France
| |
Collapse
|
1824
|
Brown MC, Gromeier M. Oncolytic immunotherapy through tumor-specific translation and cytotoxicity of poliovirus. DISCOVERY MEDICINE 2015; 19:359-365. [PMID: 26105699 PMCID: PMC4780852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Achieving tumor-specific, robust, and durable effector cytotoxic immune responses is key to successful immunotherapy. This has been accomplished with adoptive cell transfer of ex vivo-expanded autologous tumor-infiltrating or engineered T cells, or with immune checkpoint inhibitors, enhancing inherent T cell reactivity. A natural ability to recruit effector responses makes tumor-targeting ('oncolytic') viruses attractive as immunotherapy vehicles. However, most viruses actively block inflammatory and immunogenic events; or, host innate immune responses may prevent immune initiating events in the first place. Moreover, the mechanisms of how virus infection can produce effector responses against host (tumor) neo-antigens are unclear. We are pioneering oncolytic immunotherapy based on poliovirus, which has no specific mechanism to interfere with host immune activation, exhibits lytic cytotoxicity in the presence of an antiviral interferon response and pre-existing immunity, and engages a powerful innate immune sensor implicated in recruiting cytotoxic T cell responses. Central to this approach is a unique confluence of factors that drive tumor-specific viral translation and cytotoxicity.
Collapse
Affiliation(s)
- Michael C Brown
- Department of Surgery Division of Neurosurgery and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Matthias Gromeier
- Department of Surgery Division of Neurosurgery and Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
1825
|
Abstract
Effective immunotherapy promotes the killing of cancer cells by cytotoxic T cells. This requires not only that cancer-specific T cells be generated, but also that these T cells physically contact cancer cells. The coexistence in some patients of cancer cells and T cells that recognize them indicates that tumors may exhibit the phenomenon of immune privilege, in which immunogenic tissue is protected from immune attack. Here, we review the evidence that stromal cells of the tumor microenvironment mediate this restriction by excluding T cells from the vicinity of cancer cells. Overcoming this T cell checkpoint may thus enable optimal immunotherapy.
Collapse
Affiliation(s)
- Johanna A Joyce
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Douglas T Fearon
- Cold Spring Harbor Laboratory, New York, NY 11724, USA. Department of Microbiology and Immunology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medical School, New York, NY 10065, USA.
| |
Collapse
|
1826
|
Salem M, El-Shanshory M, El-Naggar RED, Abdou S, Attia M, Zidan AA, Zidan M. Reduction in the numbers of CD33+myeloid population in Egyptian children with B-linage acute lymphoblastic leukemia and its recovery after induction of chemotherapy. CLINICAL CANCER INVESTIGATION JOURNAL 2015. [DOI: 10.4103/2278-0513.164716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|