1
|
Nguyen-Hoai T, Kobelt D, Hohn O, Vu MD, Schlag PM, Dörken B, Norley S, Lipp M, Walther W, Pezzutto A, Westermann J. HER2/neu DNA vaccination by intradermal gene delivery in a mouse tumor model: Gene gun is superior to jet injector in inducing CTL responses and protective immunity. Oncoimmunology 2021; 1:1537-1545. [PMID: 23264900 PMCID: PMC3525609 DOI: 10.4161/onci.22563] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
DNA vaccines are potential tools for the induction of immune responses against both infectious disease and cancer. The dermal application of DNA vaccines is of particular interest since the epidermal and dermal layers of the skin are characterized by an abundance of antigen-presenting cells (APCs). The aim of our study was to compare tumor protection as obtained by two different methods of intradermal DNA delivery (gene gun and jet injector) in a well-established HER2/neu mouse tumor model. BALB/c mice were immunized twice with a HER2/neu-coding plasmid by gene gun or jet injector. Mice were then subcutaneously challenged with HER2/neu+ syngeneic D2F2/E2 tumor cells. Protection against subsequent challenges with tumor cells as well as humoral and T-cell immune responses induced by the vaccine were monitored. Gene gun immunization was far superior to jet injector both in terms of tumor protection and induction of HER2/neu-specific immune responses. After gene gun immunization, 60% of the mice remained tumor-free until day 140 as compared with 25% after jet injector immunization. Furthermore, gene gun vaccination was able to induce both a strong TH1-polarized T-cell response with detectable cytotoxic T-lymphocyte (CTL) activity and a humoral immune response against HER2/neu, whereas the jet injector was not. Although the disadvantages that were associated with the use of the jet injector in our model may be overcome with methodological modifications and/or in larger animals, which exhibit a thicker skin and/or subcutaneous muscle tissue, we conclude that gene gun delivery constitutes the method of choice for intradermal DNA delivery in preclinical mouse models and possibly also for the clinical development of DNA-based vaccines.
Collapse
Affiliation(s)
- Tam Nguyen-Hoai
- Deptartment of Hematology, Oncology, and Tumor Immunology Charité; University Medicine Berlin; Campus Berlin-Buch, Campus Benjamin Franklin and Campus Virchow-Klinikum; Berlin, Germany ; Max Delbrück Center for Molecular Medicine; Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Carreira B, Acúrcio RC, Matos AI, Peres C, Pozzi S, Vaskovich‐Koubi D, Kleiner R, Bento M, Satchi‐Fainaro R, Florindo HF. Nanomedicines as Multifunctional Modulators of Melanoma Immune Microenvironment. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Barbara Carreira
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ana I. Matos
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Daniella Vaskovich‐Koubi
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Ron Kleiner
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Mariana Bento
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| | - Ronit Satchi‐Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine Tel Aviv University Tel Aviv 6997801 Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa) Faculty of Pharmacy, University of Lisbon Av. Prof. Gama Pinto Lisboa 1649‐003 Portugal
| |
Collapse
|
3
|
Onodi F, Maherzi-Mechalikh C, Mougel A, Ben Hamouda N, Taboas C, Gueugnon F, Tran T, Nozach H, Marcon E, Gey A, Terme M, Bouzidi A, Maillere B, Kerzerho J, Tartour E, Tanchot C. High Therapeutic Efficacy of a New Survivin LSP-Cancer Vaccine Containing CD4 + and CD8 + T-Cell Epitopes. Front Oncol 2018; 8:517. [PMID: 30483475 PMCID: PMC6243131 DOI: 10.3389/fonc.2018.00517] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 10/22/2018] [Indexed: 12/22/2022] Open
Abstract
The efficacy of an antitumoral vaccine relies both on the choice of the antigen targeted and on its design. The tumor antigen survivin is an attractive target to develop therapeutic cancer vaccines because of its restricted over-expression and vital functions in most human tumors. Accordingly, several clinical trials targeting survivin in various cancer indications have been conducted. Most of them relied on short peptide-based vaccines and showed promising, but limited clinical results. In this study, we investigated the immunogenicity and therapeutic efficacy of a new long synthetic peptide (LSP)-based cancer vaccine targeting the tumor antigen survivin (SVX). This SVX vaccine is composed of three long synthetic peptides containing several CD4+ and CD8+ T-cell epitopes, which bind to various HLA class II and class I molecules. Studies in healthy individuals showed CD4+ and CD8+ T-cell immunogenicity of SVX peptides in human, irrespective of the individual's HLA types. Importantly, high frequencies of spontaneous T-cell precursors specific to SVX peptides were also detected in the blood of various cancer patients, demonstrating the absence of tolerance against these peptides. We then demonstrated SVX vaccine's high therapeutic efficacy against four different established murine tumor models, associated with its capacity to generate both specific cytotoxic CD8+ and multifunctional Th1 CD4+ T-cell responses. When tumors were eradicated, generated memory T-cell responses protected against rechallenge allowing long-term protection against relapses. Treatment with SVX vaccine was also found to reshape the tumor microenvironment by increasing the tumor infiltration of both CD4+ and CD8+ T cells but not Treg cells therefore tipping the balance toward a highly efficient immune response. These results highlight that this LSP-based SVX vaccine appears as a promising cancer vaccine and warrants its further clinical development.
Collapse
Affiliation(s)
- Fanny Onodi
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| | - Chahrazed Maherzi-Mechalikh
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Alice Mougel
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Nadine Ben Hamouda
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Service d'immunologie Biologique, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Charlotte Taboas
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| | - Fabien Gueugnon
- VAXEAL Research, Evry, France.,CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | - Thi Tran
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| | - Herve Nozach
- CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | - Elodie Marcon
- CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | - Alain Gey
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Service d'immunologie Biologique, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Magali Terme
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Bernard Maillere
- CEA-Saclay, Institut des Sciences du Vivant Frederic Joliot, Service d'Ingénierie Moléculaire des Protéines, Gif Sur Yvette, France
| | | | - Eric Tartour
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service d'immunologie Biologique, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Corinne Tanchot
- INSERM U970, PARCC (Paris-Cardiovascular Research Center), Paris, France
| |
Collapse
|
4
|
Romero P, Banchereau J, Bhardwaj N, Cockett M, Disis ML, Dranoff G, Gilboa E, Hammond SA, Hershberg R, Korman AJ, Kvistborg P, Melief C, Mellman I, Palucka AK, Redchenko I, Robins H, Sallusto F, Schenkelberg T, Schoenberger S, Sosman J, Türeci Ö, Van den Eynde B, Koff W, Coukos G. The Human Vaccines Project: A roadmap for cancer vaccine development. Sci Transl Med 2016; 8:334ps9. [PMID: 27075624 DOI: 10.1126/scitranslmed.aaf0685] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer vaccine development has been vigorously pursued for 40 years. Immunity to tumor antigens can be elicited by most vaccines tested, but their clinical efficacy remains modest. We argue that a concerted international effort is necessary to understand the human antitumor immune response and achieve clinically effective cancer vaccines.
Collapse
Affiliation(s)
- Pedro Romero
- Ludwig Cancer Research at University of Lausanne, 1066 Epalinges, Switzerland
| | | | - Nina Bhardwaj
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Mary L Disis
- University of Washington School of Medicine, Seattle, WA 98109-4714, USA
| | - Glenn Dranoff
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Eli Gilboa
- Sylvester Comprehensive Cancer Center, Miami, FL 33136, USA
| | | | - Robert Hershberg
- Celgene Immuno-Oncology Center of Excellence, 1616 Eastlake Avenue, Suite 500, Seattle, WA 98102, USA
| | - Alan J Korman
- Bristol-Myers Squibb, Biologics Discovery California, Redwood City, CA 94063, USA
| | - Pia Kvistborg
- Netherlands Cancer Institute, 1066CX Amsterdam, Netherlands
| | - Cornelis Melief
- ISA Pharmaceuticals & Leiden University Medical Center, 2333 ZA Leiden, Netherlands
| | | | - A Karolina Palucka
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA. Baylor Institute for Immunology Research, Dallas, TX 75204, USA
| | | | - Harlan Robins
- Adaptive Biotechnologies, Fred Hutchinson Cancer Research Center, Seattle, WA 98102, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | | | - Stephen Schoenberger
- Center for Personalized Cancer Immunotherapy, La Jolla Institute for Allergy and Immunology & UCSD Moores Cancer Center, La Jolla, San Diego, CA 92037, USA
| | - Jeffrey Sosman
- Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN 37232, USA
| | - Özlem Türeci
- CI3 Cluster for Individualized Immunotherapy, Kupferbergterasse 17-19, 55131 Mainz, Germany
| | - Benoît Van den Eynde
- Ludwig Institute for Cancer Research, Brussels branch, Brussels, BRU 1200, Belgium. Université Catholique de Louvain, Avenue Hippocrate 10, 1200 Woluwe-Saint-Lambert, Belgium. University of Oxford, Nuffield Department of Medicine, Ludwig Institute for Cancer Research, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Wayne Koff
- International AIDS Vaccines Initiative, 125 Broad Street, 9th Floor, New York, NY 10004, USA
| | - George Coukos
- Ludwig Cancer Research at University of Lausanne, 1066 Epalinges, Switzerland.
| |
Collapse
|
5
|
Rapid and Continued T-Cell Differentiation into Long-term Effector and Memory Stem Cells in Vaccinated Melanoma Patients. Clin Cancer Res 2016; 23:3285-3296. [DOI: 10.1158/1078-0432.ccr-16-1708] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/30/2016] [Accepted: 11/05/2016] [Indexed: 11/16/2022]
|
6
|
Murahashi M, Hijikata Y, Yamada K, Tanaka Y, Kishimoto J, Inoue H, Marumoto T, Takahashi A, Okazaki T, Takeda K, Hirakawa M, Fujii H, Okano S, Morita M, Baba E, Mizumoto K, Maehara Y, Tanaka M, Akashi K, Nakanishi Y, Yoshida K, Tsunoda T, Tamura K, Nakamura Y, Tani K. Phase I clinical trial of a five-peptide cancer vaccine combined with cyclophosphamide in advanced solid tumors. Clin Immunol 2016; 166-167:48-58. [PMID: 27072896 DOI: 10.1016/j.clim.2016.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 12/18/2022]
Abstract
We designed a phase I trial to investigate the safety, immune responses and clinical benefits of a five-peptide cancer vaccine in combination with chemotherapy. Study subjects were patients positive for HLA-A2402 with locally advanced, metastatic, and/or recurrent gastrointestinal, lung or cervical cancer. Eighteen patients including nine cases of colorectal cancer were treated with escalating doses of cyclophosphamide 4days before vaccination. Five HLA-A2402-restricted, tumor-associated antigen (TAA) epitope peptides from KOC1, TTK, URLC10, DEPDC1 and MPHOSPH1 were injected weekly for 4weeks. Treatment was well tolerated without any adverse events above grade 3. Analysis of peripheral blood lymphocytes showed that the number of regulatory T cells dropped from baseline after administration of cyclophosphamide and confirmed that TAA-specific T cell responses were associated significantly with longer overall survival. This phase I clinical trial demonstrated safety and promising immune responses that correlated with vaccine-induced T-cell responses. Therefore, this approach warrants further clinical studies.
Collapse
Affiliation(s)
- Mutsunori Murahashi
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Yasuki Hijikata
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Kazunari Yamada
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshihiro Tanaka
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Junji Kishimoto
- Digital Medicine Initiative, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Inoue
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tomotoshi Marumoto
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Atsushi Takahashi
- Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Toshihiko Okazaki
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan
| | - Kazuyoshi Takeda
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Hiroshi Fujii
- Department of Pathology, Kyushu University, Fukuoka, Japan
| | - Shinji Okano
- Department of Pathology, Kyushu University, Fukuoka, Japan
| | - Masaru Morita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eishi Baba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kazuhiro Mizumoto
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masao Tanaka
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoichi Nakanishi
- Institute of Diseases of Chest, Kyushu University, Fukuoka, Japan
| | - Koji Yoshida
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Takuya Tsunoda
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kazuo Tamura
- Division of Oncology, Hematology and Infectious Diseases, Department of Internal Medicine, Fukuoka University, Fukuoka, Japan
| | - Yusuke Nakamura
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kenzaburo Tani
- Department of Advanced Cell and Molecular Therapy, Kyushu University Hospital, Fukuoka, Japan; Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
7
|
Legat A, Maby-El Hajjami H, Baumgaertner P, Cagnon L, Abed Maillard S, Geldhof C, Iancu EM, Lebon L, Guillaume P, Dojcinovic D, Michielin O, Romano E, Berthod G, Rimoldi D, Triebel F, Luescher I, Rufer N, Speiser DE. Vaccination with LAG-3Ig (IMP321) and Peptides Induces Specific CD4 and CD8 T-Cell Responses in Metastatic Melanoma Patients—Report of a Phase I/IIa Clinical Trial. Clin Cancer Res 2015; 22:1330-40. [DOI: 10.1158/1078-0432.ccr-15-1212] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/01/2015] [Indexed: 11/16/2022]
|
8
|
Lepore M, de Lalla C, Gundimeda SR, Gsellinger H, Consonni M, Garavaglia C, Sansano S, Piccolo F, Scelfo A, Häussinger D, Montagna D, Locatelli F, Bonini C, Bondanza A, Forcina A, Li Z, Ni G, Ciceri F, Jenö P, Xia C, Mori L, Dellabona P, Casorati G, De Libero G. A novel self-lipid antigen targets human T cells against CD1c(+) leukemias. ACTA ACUST UNITED AC 2014; 211:1363-77. [PMID: 24935257 PMCID: PMC4076585 DOI: 10.1084/jem.20140410] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
T cells that recognize self-lipids presented by CD1c are frequent in the peripheral blood of healthy individuals and kill transformed hematopoietic cells, but little is known about their antigen specificity and potential antileukemia effects. We report that CD1c self-reactive T cells recognize a novel class of self-lipids, identified as methyl-lysophosphatidic acids (mLPAs), which are accumulated in leukemia cells. Primary acute myeloid and B cell acute leukemia blasts express CD1 molecules. mLPA-specific T cells efficiently kill CD1c(+) acute leukemia cells, poorly recognize nontransformed CD1c-expressing cells, and protect immunodeficient mice against CD1c(+) human leukemia cells. The identification of immunogenic self-lipid antigens accumulated in leukemia cells and the observed leukemia control by lipid-specific T cells in vivo provide a new conceptual framework for leukemia immune surveillance and possible immunotherapy.
Collapse
Affiliation(s)
- Marco Lepore
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudia de Lalla
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - S Ramanjaneyulu Gundimeda
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Heiko Gsellinger
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Michela Consonni
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Claudio Garavaglia
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sebastiano Sansano
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Francesco Piccolo
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Andrea Scelfo
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Daniel Häussinger
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Daniela Montagna
- Laboratorio di Immunologia, Dipartimento di Pediatria, Università di Pavia and Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology-Oncology, IRCCS Bambino Gesù Hospital, 00165 Rome, Italy
| | - Chiara Bonini
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Attilio Bondanza
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Alessandra Forcina
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Zhiyuan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Guanghui Ni
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Fabio Ciceri
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Paul Jenö
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland
| | - Chengfeng Xia
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore 138648
| | - Paolo Dellabona
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giulia Casorati
- Experimental Immunology Unit, Division of Immunology, Transplantation, and Infectious Diseases, Experimental Hematology Unit, and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel; Nuclear Magnetic Resonance Laboratory, Department of Chemistry; and Department of Biochemistry, Biozentrum; University of Basel, 4056 Basel, Switzerland Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research, Singapore 138648
| |
Collapse
|
9
|
Kawano M, Matsui M, Handa H. SV40 virus-like particles as an effective delivery system and its application to a vaccine carrier. Expert Rev Vaccines 2014; 12:199-210. [DOI: 10.1586/erv.12.149] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
10
|
Tan S, Sasada T, Bershteyn A, Yang K, Ioji T, Zhang Z. Combinational delivery of lipid-enveloped polymeric nanoparticles carrying different peptides for anti-tumor immunotherapy. Nanomedicine (Lond) 2013; 9:635-47. [PMID: 23905577 DOI: 10.2217/nnm.13.67] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
AIM The authors aimed to investigate whether nanotechnology-based delivery of antigenic peptides is feasible for efficiently inducing anti-tumor cytotoxic T lymphocyte responses through vaccination. MATERIALS & METHODS Three different murine melanoma antigens were entrapped in lipid-coated poly(D,L-lactide-co-glycolide) nanoparticles (NPs) by the double emulsion method. RESULTS The loading efficiency of hydrophilic peptides was greatly improved when lipids were introduced to formulate lipid-coated NPs. The lipid-coated NPs carrying a single peptide and/or combinations of multiple lipid-coated NPs carrying antigenic peptides were characterized in vitro and in vivo in a C57/BL6 (B6) mouse model. Both the single melanoma antigen peptide-loaded NPs and combinational delivery of lipid-coated NPs carrying different peptides could induce antigen-specific T-cell responses. However, single peptide-loaded NPs failed to significantly delay the growth of subcutaneously inoculated B16 melanoma cells in a prophylactic setting. By contrast, the combinational delivery of lipid-coated NPs carrying different peptides significantly suppressed growth of inoculated B16 melanoma cells.
Collapse
Affiliation(s)
- Songwei Tan
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | | | | | | | | | | |
Collapse
|
11
|
Abstract
The spontaneous T-cell responses to the KIF20A cancer-associated antigen found by Tomita and colleagues among peripheral blood mononuclear cells of patients with cancer, but not healthy people, involve both CD4 and CD8 T cells. Synthetic long peptides of KIF20A stimulate synergy between these two T-cell types to promote cancer cell killing.
Collapse
Affiliation(s)
- Cornelis J M Melief
- ISA Pharmaceuticals and Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
12
|
Buhrman JD, Slansky JE. Improving T cell responses to modified peptides in tumor vaccines. Immunol Res 2013; 55:34-47. [PMID: 22936035 DOI: 10.1007/s12026-012-8348-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immune recognition and elimination of cancerous cells is the primary goal of cancer immunotherapy. However, obstacles including immune tolerance and tumor-induced immunosuppression often limit beneficial immune responses. Vaccination is one proposed intervention that may help to overcome these issues and is an active area of study in cancer immunotherapy. Immunizing with tumor antigenic peptides is a promising, straight-forward vaccine strategy hypothesized to boost preexisting antitumor immunity. However, tumor antigens are often weak T cell agonists, attributable to several mechanisms, including immune self-tolerance and poor immunogenicity of self-derived tumor peptides. One strategy for overcoming these mechanisms is vaccination with mimotopes, or peptide mimics of tumor antigens, which alter the antigen presentation and/or T cell activation to increase the expansion of tumor-specific T cells. Evaluation of mimotope vaccine strategies has revealed that even subtle alterations in peptide sequence can dramatically alter antigen presentation and T cell receptor recognition. Most of this research has been performed using T cell clones, which may not be accurate representations of the naturally occurring antitumor response. The relationship between clones generated after mimotope vaccination and the polyclonal T cell repertoire is unclear. Our work with mimotopes in a mouse model of colon carcinoma has revealed important insights into these issues. We propose that the identification of mimotopes based on stimulation of the naturally responding T cell repertoire will dramatically improve the efficacy of mimotope vaccination.
Collapse
Affiliation(s)
- Jonathan D Buhrman
- Integrated Department of Immunology, University of Colorado School of Medicine, National Jewish Health, Denver, CO 80206, USA
| | | |
Collapse
|
13
|
Silva JM, Videira M, Gaspar R, Préat V, Florindo HF. Immune system targeting by biodegradable nanoparticles for cancer vaccines. J Control Release 2013; 168:179-99. [PMID: 23524187 DOI: 10.1016/j.jconrel.2013.03.010] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 03/11/2013] [Accepted: 03/14/2013] [Indexed: 01/08/2023]
Abstract
The concept of therapeutic cancer vaccines is based on the activation of the immune system against tumor cells after the presentation of tumor antigens. Nanoparticles (NPs) have shown great potential as delivery systems for cancer vaccines as they potentiate the co-delivery of tumor-associated antigens and adjuvants to dendritic cells (DCs), insuring effective activation of the immune system against tumor cells. In this review, the immunological mechanisms behind cancer vaccines, including the role of DCs in the stimulation of T lymphocytes and the use of Toll-like receptor (TLR) ligands as adjuvants will be discussed. An overview of each of the three essential components of a therapeutic cancer vaccine - antigen, adjuvant and delivery system - will be provided with special emphasis on the potential of particulate delivery systems for cancer vaccines, in particular those made of biodegradable aliphatic polyesters, such as poly(lactic-co-glycolic acid) (PLGA) and poly-ε-caprolactone (PCL). Some of the factors that can influence NP uptake by DCs, including size, surface charge, surface functionalization and route of administration, will also be considered.
Collapse
Affiliation(s)
- Joana M Silva
- iMed.UL, Research Institute for Medicines and Pharmaceutical Sciences, Faculty of Pharmacy, University of Lisbon, 1649-003 Lisbon, Portugal
| | | | | | | | | |
Collapse
|
14
|
Aarntzen EHJG, Srinivas M, Radu CG, Punt CJA, Boerman OC, Figdor CG, Oyen WJG, de Vries IJM. In vivo imaging of therapy-induced anti-cancer immune responses in humans. Cell Mol Life Sci 2012; 70:2237-57. [PMID: 23052208 PMCID: PMC3676735 DOI: 10.1007/s00018-012-1159-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 08/27/2012] [Accepted: 09/03/2012] [Indexed: 12/16/2022]
Abstract
Immunotherapy aims to re-engage and revitalize the immune system in the fight against cancer. Research over the past decades has shown that the relationship between the immune system and human cancer is complex, highly dynamic, and variable between individuals. Considering the complexity, enormous effort and costs involved in optimizing immunotherapeutic approaches, clinically applicable tools to monitor therapy-induced immune responses in vivo are most warranted. However, the development of such tools is complicated by the fact that a developing immune response encompasses several body compartments, e.g., peripheral tissues, lymph nodes, lymphatic and vascular systems, as well as the tumor site itself. Moreover, the cells that comprise the immune system are not static but constantly circulate through the vascular and lymphatic system. Molecular imaging is considered the favorite candidate to fulfill this task. The progress in imaging technologies and modalities has provided a versatile toolbox to address these issues. This review focuses on the detection of therapy-induced anticancer immune responses in vivo and provides a comprehensive overview of clinically available imaging techniques as well as perspectives on future developments. In the discussion, we will focus on issues that specifically relate to imaging of the immune system and we will discuss the strengths and limitations of the current clinical imaging techniques. The last section provides future directions that we envision to be crucial for further development.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Recombinant lipidated HPV E7 induces a Th-1-biased immune response and protective immunity against cervical cancer in a mouse model. PLoS One 2012; 7:e40970. [PMID: 22815882 PMCID: PMC3399806 DOI: 10.1371/journal.pone.0040970] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Accepted: 06/18/2012] [Indexed: 02/02/2023] Open
Abstract
The E7 oncoprotein of human papillomavirus (HPV) is an ideal target for developing immunotherapeutic strategies against HPV-associated tumors. However, because protein-based immunogens alone are poor elicitors of the cytotoxic T-lymphocyte (CTL) responses, they have been difficult to exploit for therapeutic purposes. In this study, we report that a recombinant lipoprotein consisting of inactive E7 (E7m) biologically linked to a bacterial lipid moiety (rlipo-E7m) induces the maturation of mouse bone marrow-derived dendritic cells through toll-like receptor 2 (TLR2), skews the immune responses toward the Th1 responses and induces E7-specific CTL responses. We further studied the ability of rlipo-E7m to provide protection against a TC-1 tumor cell challenge in an animal model. Mice prophylactically immunized with two 10-µg doses of rlipo-E7m were found to be free of TC-1 tumor growth. Experiments in a therapeutic immunization model showed that the tumor volume in mice receiving a single dose of rlipo-E7m was less than 0.01 cm3 on day 40, whereas the tumor volume in mice treated with rE7m was 2.28±1.21 cm3. The tumor volume of the entire control group was over 3 cm3. In addition, we demonstrated that the CD8+ T cells play a major role in anti-tumor immunity when administration of rlipo-E7m. These results demonstrate that rlipo-E7m could be a promising candidate for treating HPV-associated tumors.
Collapse
|
16
|
Bae J, Smith R, Daley J, Mimura N, Tai YT, Anderson KC, Munshi NC. Myeloma-specific multiple peptides able to generate cytotoxic T lymphocytes: a potential therapeutic application in multiple myeloma and other plasma cell disorders. Clin Cancer Res 2012; 18:4850-60. [PMID: 22753586 DOI: 10.1158/1078-0432.ccr-11-2776] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE The efficacy of peptide vaccines may be enhanced by stimulating immune cells with multiple peptides derived from distinct tumor-associated antigens. We have evaluated the heteroclitic XBP1-US(184-192) (YISPWILAV), heteroclitic XBP1-SP(367-375) (YLFPQLISV), native CD138(260-268) (GLVGLIFAV), and native CS1(239-247) (SLFVLGLFL) peptides, which have strong HLA-A2 affinity and immunogenicity in combination, for their ability to elicit multiple myeloma antigen-specific responses. EXPERIMENTAL DESIGN Multipeptide-specific cytotoxic T lymphocytes (MP-CTL) were generated by the stimulation of CD3(+) T lymphocytes from HLA-A2(+) individuals with either autologous mature dendritic cells or T2 cells pulsed with a cocktail of these four peptides. RESULTS The peptide cocktail did not compromise tumor antigen-specific activity of CTLs. MP-CTLs displayed increased total, effector memory (CCR7(-)CD45RO(+)), and activated (CD69(+)) CD3(+)CD8(+) T lymphocytes. In addition, MP-CTL showed IFN-γ production, cell proliferation, and cytotoxicity against HLA-A2(+) multiple myeloma cells, including cells of HLA-A2(+) patients with multiple myeloma. Importantly, MP-CTLs showed specific responses in functional assays to each relevant peptide but not to an irrelevant HLA-A2-specific CMV pp65 (NLVPMVATV) peptide. CONCLUSIONS These results highlight the potential therapeutic application of vaccination with a cocktail of HLA-A2-specific peptides to induce CTLs with a broad spectrum of immune responses against multiple myeloma antigens.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Lövgren T, Baumgaertner P, Wieckowski S, Devêvre E, Guillaume P, Luescher I, Rufer N, Speiser DE. Enhanced cytotoxicity and decreased CD8 dependence of human cancer-specific cytotoxic T lymphocytes after vaccination with low peptide dose. Cancer Immunol Immunother 2012; 61:817-26. [PMID: 22080404 PMCID: PMC11029156 DOI: 10.1007/s00262-011-1140-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 10/24/2011] [Indexed: 02/04/2023]
Abstract
In mice, vaccination with high peptide doses generates higher frequencies of specific CD8+ T cells, but with lower avidity compared to vaccination with lower peptide doses. To investigate the impact of peptide dose on CD8+ T cell responses in humans, melanoma patients were vaccinated with 0.1 or 0.5 mg Melan-A/MART-1 peptide, mixed with CpG 7909 and Incomplete Freund's adjuvant. Neither the kinetics nor the amplitude of the Melan-A-specific CD8+ T cell responses differed between the two vaccination groups. Also, CD8+ T cell differentiation and cytokine production ex vivo were similar in the two groups. Interestingly, after low peptide dose vaccination, Melan-A-specific CD8+ T cells showed enhanced degranulation upon peptide stimulation, as assessed by CD107a upregulation and perforin release ex vivo. In accordance, CD8+ T cell clones derived from low peptide dose-vaccinated patients showed significantly increased degranulation and stronger cytotoxicity. In parallel, Melan-A-specific CD8+ T cells and clones from low peptide dose-vaccinated patients expressed lower CD8 levels, despite similar or even stronger binding to tetramers. Furthermore, CD8+ T cell clones from low peptide dose-vaccinated patients bound CD8 binding-deficient tetramers more efficiently, suggesting that they may express higher affinity TCRs. We conclude that low peptide dose vaccination generated CD8+ T cell responses with stronger cytotoxicity and lower CD8 dependence.
Collapse
Affiliation(s)
- Tanja Lövgren
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Petra Baumgaertner
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Sébastien Wieckowski
- University Hospital Center and University of Lausanne (CHUV), Lausanne, Switzerland
| | - Estelle Devêvre
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Philippe Guillaume
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Immanuel Luescher
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Nathalie Rufer
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
- University Hospital Center and University of Lausanne (CHUV), Lausanne, Switzerland
| | - Daniel E. Speiser
- Clinical Tumor Biology and Immunotherapy Unit, Ludwig Center of the University of Lausanne, Hôpital Orthopédique 05/1552, Av. Pierre-Decker 4, 1011 Lausanne, Switzerland
| |
Collapse
|
18
|
van der Burg SH, Kalos M, Gouttefangeas C, Janetzki S, Ottensmeier C, Welters MJP, Romero P, Britten CM, Hoos A. Harmonization of immune biomarker assays for clinical studies. Sci Transl Med 2012; 3:108ps44. [PMID: 22072636 DOI: 10.1126/scitranslmed.3002785] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Assays that measure a patient's immune response play an increasingly important role in the development of immunotherapies. The inherent complexity of these assays and independent protocol development between laboratories result in high data variability and poor reproducibility. Quality control through harmonization--based on integration of laboratory-specific protocols with standard operating procedures and assay performance benchmarks--is one way to overcome these limitations. Harmonization guidelines can be widely implemented to address assay performance variables. This process enables objective interpretation and comparison of data across clinical trial sites and also facilitates the identification of relevant immune biomarkers, guiding the development of new therapies.
Collapse
Affiliation(s)
- Sjoerd H van der Burg
- Department of Clinical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Bae J, Song W, Smith R, Daley J, Tai YT, Anderson KC, Munshi NC. A novel immunogenic CS1-specific peptide inducing antigen-specific cytotoxic T lymphocytes targeting multiple myeloma. Br J Haematol 2012; 157:687-701. [PMID: 22533610 DOI: 10.1111/j.1365-2141.2012.09111.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/15/2012] [Indexed: 01/19/2023]
Abstract
The CS1 antigen provides a unique target for the development of an immunotherapeutic strategy to treat patients with multiple myeloma (MM). This study aimed to identify HLA-A2(+) immunogenic peptides from the CS1 antigen, which induce peptide-specific cytotoxic T lymphocytes (CTL) against HLA-A2(+) MM cells. We identified a novel immunogenic HLA-A2-specific CS1(239-247) (SLFVLGLFL) peptide, which induced CS1-specific CTL (CS1-CTL) to MM cells. The CS1-CTL showed a distinct phenotype, with an increased percentage of effector memory and activated CTL and a decreased percentage of naïve CTL. CS1(239-247) peptide-specific CD8(+) T cells were detected by DimerX analyses and demonstrated functional activities specific to the peptide. The CTL displayed HLA-A2-restricted and antigen-specific cytotoxicity, proliferation, degranulation and γ-interferon (IFN-γ) production against both primary MM cells and MM cell lines. In addition, the effector memory cells subset (CD45RO(+) CCR7(-) /CD3(+) CD8(+) ) within CS1-CTL showed a higher level of CD107a degranulation and IFN-γ production as compared to effector cells (CD45RO(-) CCR7(-) /CD3(+) CD8(+) ) against HLA-A2(+) primary MM cells or MM cell lines. In conclusion, this study introduced a novel immunogenic HLA-A2-specific CS1(239-247) peptide capable of inducing antigen-specific CTL against MM cells that will provide a framework for its application as a novel MM immunotherapy.
Collapse
Affiliation(s)
- Jooeun Bae
- Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Dendritic cells (DCs) control the initiation and differentiation of T cells. In the steady state, DCs mediate tolerance. To achieve immunization, the tolerogenic function of DCs must be switched off by inducing their maturation with appropriate "adjuvants." Dendritic cells form a system composed of distinct subsets that differ in their expression of endocytic and signaling receptors. These subsets have different capacities to differentiate and polarize T cells and to cross-present antigen to expand CD8+ T cells. Optimization of vaccines is possible by exploiting the unique biological properties of DCs.
Collapse
|
21
|
Sharma P, Wagner K, Wolchok JD, Allison JP. Novel cancer immunotherapy agents with survival benefit: recent successes and next steps. Nat Rev Cancer 2011; 11:805-12. [PMID: 22020206 PMCID: PMC3426440 DOI: 10.1038/nrc3153] [Citation(s) in RCA: 478] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The US Food and Drug Administration (FDA) recently approved two novel immunotherapy agents, sipuleucel-T and ipilimumab, which showed a survival benefit for patients with metastatic prostate cancer and melanoma, respectively. The mechanisms by which these agents provideclinical benefit are not completely understood. However, knowledge of these mechanisms will be crucial for probing human immune responses and tumour biology in order to understand what distinguishes responders from non-responders. The following next steps are necessary: first, the development of immune-monitoring strategies for the identification of relevant biomarkers; second, the establishment of guidelines for the assessment of clinical end points; and third, the evaluation of combination therapy strategies to improve clinical benefit.
Collapse
Affiliation(s)
- Padmanee Sharma
- Department of Genitourinary Medical Oncology, University of Texas M D Anderson Cancer Center, Box 0018-7, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
22
|
Suchorska WM, Dams-Kozlowska H, Kazimierczak U, Wysocki PJ, Mackiewicz A. Hyper-interleukin-11 novel designer molecular adjuvant targeting gp130 for whole cell cancer vaccines. Expert Opin Biol Ther 2011; 11:1555-67. [PMID: 21995459 DOI: 10.1517/14712598.2011.627852] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Hyper-IL-11 (H11) is a fusion protein comprising IL-11 and soluble IL-11 receptor directly targeting gp130. We evaluated efficacy of H11 as a molecular adjuvant in therapeutic whole tumor cell vaccine formulation. METHODS H11 was tested in ectopic and orthotopic murine renal cell carcinoma (RENCA) models. H11 cDNA was transduced into RENCA cells (RENCA-H11). Mice were immunized with RENCA-H11 or control vaccine (RENCA-IRR) in prophylactic, adjuvant and therapeutic settings. Tumor formation, survival and immune mechanisms activated by H11 were studied. RESULTS Biologically active H11 was secreted by RENCA-H11 cells. Immunization with RENCA-H11 resulted in mounting specific anti-RENCA response. Treatment of tumor bearing mice in adjuvant setting prevented disease recurrence in therapeutic setting eradicated tumors. In induction phase H11 inhibited T-regulatory cell formation and activated recruitment and maturation of dendritic cells. Downstream of immunization tumors were densely infiltrated by CD8(+), CD4(+), NK cells, cells expressing CD8(+)CD69(+) and CD4(+)CD62L(low). CONCLUSIONS H11 is a good candidate for adjuvant of whole tumor cell vaccines. Direct targeting of gp130 leads to induction of specific and long lasting anticancer immune response. Enhancement of tumor antigen presentation, abrogation of immune tolerance, and activation of NK cells and generation of memory cells lead to eradication of existing tumors.
Collapse
|
23
|
Hong M, Puaux AL, Huang C, Loumagne L, Tow C, Mackay C, Kato M, Prévost-Blondel A, Avril MF, Nardin A, Abastado JP. Chemotherapy induces intratumoral expression of chemokines in cutaneous melanoma, favoring T-cell infiltration and tumor control. Cancer Res 2011; 71:6997-7009. [PMID: 21948969 DOI: 10.1158/0008-5472.can-11-1466] [Citation(s) in RCA: 184] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
T-cell infiltration is known to impact tumor growth and is associated with cancer patient survival. However, the molecular cues that favor T-cell infiltration remain largely undefined. Here, using a genetically engineered mouse model of melanoma, we show that CXCR3 ligands and CCL5 synergize to attract effector T cells into cutaneous metastases, and their expression inhibits tumor growth. Treatment of tumor-bearing mice with chemotherapy induced intratumoral expression of these chemokines and favored T-cell infiltration into cutaneous tumors. In patients with melanoma, these chemokines were also upregulated in chemotherapy-sensitive lesions following chemotherapy, and correlated with T-cell infiltration, tumor control, and patient survival. We found that dacarbazine, temozolomide, and cisplatin induced expression of T-cell-attracting chemokines in several human melanoma cell lines in vitro. These data identify the induction of intratumoral expression of chemokines as a novel cell-extrinsic mechanism of action of chemotherapy that results in the recruitment of immune cells with antitumor activity. Therefore, identifying chemotherapeutic drugs able to induce the expression of T-cell-attracting chemokines in cancer cells may represent a novel strategy to improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Michelle Hong
- Singapore Immunology Network, BMSI, A-STAR, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Aurisicchio L, Ciliberto G. Emerging cancer vaccines: the promise of genetic vectors. Cancers (Basel) 2011; 3:3687-713. [PMID: 24212974 PMCID: PMC3759217 DOI: 10.3390/cancers3033687] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 09/09/2011] [Accepted: 09/14/2011] [Indexed: 01/18/2023] Open
Abstract
Therapeutic vaccination against cancer is an important approach which, when combined with other therapies, can improve long-term control of cancer. In fact, the induction of adaptive immune responses against Tumor Associated Antigens (TAAs) as well as innate immunity are important factors for tumor stabilization/eradication. A variety of immunization technologies have been explored in last decades and are currently under active evaluation, such as cell-based, protein, peptide and heat-shock protein-based cancer vaccines. Genetic vaccines are emerging as promising methodologies to elicit immune responses against a wide variety of antigens, including TAAs. Amongst these, Adenovirus (Ad)-based vectors show excellent immunogenicity profile and have achieved immunological proof of concept in humans. In vivo electroporation of plasmid DNA (DNA-EP) is also a desirable vaccine technology for cancer vaccines, as it is repeatable several times, a parameter required for the long-term maintenance of anti-tumor immunity. Recent findings show that combinations of different modalities of immunization (heterologous prime/boost) are able to induce superior immune reactions as compared to single-modality vaccines. In this review, we will discuss the challenges and requirements of emerging cancer vaccines, particularly focusing on the genetic cancer vaccines currently under active development and the promise shown by Ad and DNA-EP heterologous prime-boost.
Collapse
Affiliation(s)
- Luigi Aurisicchio
- Takis, via di Castel Romano 100, 00128 Rome, Italy; E-Mail:
- BIOGEM scarl, via Camporeale, 83031 Ariano Irpino (AV), Italy
| | - Gennaro Ciliberto
- Takis, via di Castel Romano 100, 00128 Rome, Italy; E-Mail:
- Dipartimento di Medicina Sperimentale e Clinica, Università degli studi di Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
| |
Collapse
|
25
|
Ma Y, Conforti R, Aymeric L, Locher C, Kepp O, Kroemer G, Zitvogel L. How to improve the immunogenicity of chemotherapy and radiotherapy. Cancer Metastasis Rev 2011; 30:71-82. [PMID: 21298323 DOI: 10.1007/s10555-011-9283-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Chemotherapy or radiotherapy could induce various tumor cell death modalities, releasing tumor-derived antigen as well as danger signals that could either be captured for triggering antitumor immune response or ignored. Exploring the interplay among therapeutic drugs, tumor cell death and the immune cells should improve diagnostic, prognostic, predictive, and therapeutic management of tumor. We summarized some of the cell death-derived danger signals and the mechanism for host to sense and response to cell death in the tumor microenvironment. Based on the recent clinical or experimental findings, several strategies have been suggested to improve the immunogenicity of cell death and augment antitumor immunity.
Collapse
Affiliation(s)
- Yuting Ma
- INSERM, U1015, 94805 Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
26
|
Baumgaertner P, Jandus C, Rivals JP, Derré L, Lövgren T, Baitsch L, Guillaume P, Luescher IF, Berthod G, Matter M, Rufer N, Michielin O, Speiser DE. Vaccination-induced functional competence of circulating human tumor-specific CD8 T-cells. Int J Cancer 2011; 130:2607-17. [PMID: 21796616 DOI: 10.1002/ijc.26297] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 06/10/2011] [Indexed: 02/04/2023]
Abstract
T-cells specific for foreign (e.g., viral) antigens can give rise to strong protective immune responses, whereas self/tumor antigen-specific T-cells are thought to be less powerful. However, synthetic T-cell vaccines composed of Melan-A/MART-1 peptide, CpG and IFA can induce high frequencies of tumor-specific CD8 T-cells in PBMC of melanoma patients. Here we analyzed the functionality of these T-cells directly ex vivo, by multiparameter flow cytometry. The production of multiple cytokines (IFNγ, TNFα, IL-2) and upregulation of LAMP-1 (CD107a) by tumor (Melan-A/MART-1) specific T-cells was comparable to virus (EBV-BMLF1) specific CD8 T-cells. Furthermore, phosphorylation of STAT1, STAT5 and ERK1/2, and expression of CD3 zeta chain were similar in tumor- and virus-specific T-cells, demonstrating functional signaling pathways. Interestingly, high frequencies of functionally competent T-cells were induced irrespective of patient's age or gender. Finally, CD8 T-cell function correlated with disease-free survival. However, this result is preliminary since the study was a Phase I clinical trial. We conclude that human tumor-specific CD8 T-cells can reach functional competence in vivo, encouraging further development and Phase III trials assessing the clinical efficacy of robust vaccination strategies.
Collapse
Affiliation(s)
- Petra Baumgaertner
- Clinical Tumor Immune-Biology Unit, Ludwig Center for Cancer Research of the University of Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Antigen sensitivity and T-cell receptor avidity as critical determinants of HIV control. Curr Opin HIV AIDS 2011; 6:157-62. [PMID: 21399498 DOI: 10.1097/coh.0b013e3283453dfd] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Induction of highly effective T cells capable of performing elite control of HIV replication represents a major goal of vaccinology. Here, we review the recent evidence supporting the central role of antigen sensitivity and T-cell receptor (TCR) avidity in determining anti-HIV T-cell efficacy. We discuss why the modulation of these factors represents an interesting approach for the rational design of HIV vaccines. RECENT FINDINGS The qualitative attributes of T-cell efficacy against HIV are closely related to the sensitivity of the cells for their cognate antigen, which appears essential to control viral replication in HIV-infected patients and is in turn strongly influenced by TCR avidity. High antigen sensitivity and TCR avidity present also potential caveats, notably T-cell clonal exhaustion and rapid emergence of escape variants. SUMMARY The central role of antigen sensitivity and TCR avidity in determining the quality of T-cell responses against HIV represents a new development in our understanding of the immune control of HIV, and the quest for an effective vaccine. Strategies to improve T-cell efficacy in vaccination approaches may rely on selecting T cells with high antigen sensitivity during priming.
Collapse
|
28
|
Wu HY, Chen YM, Lin L, Lin YG, Qiu QA, Liu N. Lentinan enhances the efficacy of the DCF regimen in patients with advanced gastric cancer: an analysis of 40 cases. Shijie Huaren Xiaohua Zazhi 2011; 19:2176-2180. [DOI: 10.11569/wcjd.v19.i20.2176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate whether lentinan enhances the curative effects of the DCF (docetaxel, cisplatin and fluorouracil) regimen in patients with advanced gastric cancer.
METHODS: Eighty patients with stages III and IV gastric cancer who were treated at our hospital between January 2007 and December 2008 were randomly divided into two groups to receive DCF chemotherapy alone or DCF chemotherapy in combination with lentinan. The curative effects, routine blood parameters, lymphocyte subsets and quality of life were compared between the two groups.
RESULTS: The disease control rate was significantly higher in the lentinan plus DCF group than in the DCF group (87.5% vs 75.0%, P < 0.05), while the incidence of side effects (including neutropenia, leukopenia, anemia, thrombocytopenia and infection) was significantly lower in the lentinan plus DCF group than in the DCF group (P < 0.05). No significant differences were noted in routine blood parameters, lymphocyte subsets and quality of life before chemotherapy between the two groups (all P > 0.05). After chemotherapy, the numbers of WBC, lymphocytes, CD3+, CD4+, CD8+, NK cells and quality of life were significantly improved in the lentinan plus DCF group compared to the DCF group (all P < 0.05).
CONCLUSION: Lentinan significantly enhanced the efficacy and safety of the DCF regimen in patients with advanced gastric cancer.
Collapse
|
29
|
Schlapbach C, Yerly D, Daubner B, Yawalkar N, Hunger RE. Telomerase-specific GV1001 peptide vaccination fails to induce objective tumor response in patients with cutaneous T cell lymphoma. J Dermatol Sci 2011; 62:75-83. [PMID: 21377838 DOI: 10.1016/j.jdermsci.2011.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 12/31/2022]
Abstract
BACKGROUND There is currently no curative therapy for cutaneous T cell lymphoma (CTCL). New therapies are therefore needed. Telomerase, the enzyme that allows for unrestricted cell divisions of cancer cells, is a promising target for cancer therapy. The telomerase-specific peptide vaccination GV1001 has shown promising results in previous studies. Since telomerase is expressed in malignant cells of CTCL, GV1001 vaccination in CTCL is a promising new therapeutic approach. OBJECTIVE We sought to investigate the efficacy of GV1001 vaccination in CTCL patients and characterize the induced immune response. METHODS Six CTCL patients were vaccinated with the GV-peptide using granulocyte/macrophage colony-stimulating factor as adjuvant. Objective clinical response and the T cell response were assessed. RESULTS None of the patients demonstrated objective clinical response to the vaccination whereas one patient showed disease progression. 1/6 patients acquired a GV1001-specifc T cell response with a Th1 cytokine profile and expression of skin-homing receptors. This hTERT-specific T cell response was not associated with beneficial modulation of the tumor-infiltrating leukocytes. Furthermore, removal of regulatory T cells did not enhance responsiveness to GV1001 in vitro in any of the patients analyzed. CONCLUSIONS Our results suggest that the GV1001 vaccination is not effective in CTCL patients and disease progression in 1/6 patients raises concerns about its safety. By analyzing skin-homing properties of GV1001-specific T cells and the involvement of regulatory T cells we nevertheless provide insight into vaccine-induced immune responses which may help to improve vaccine strategies in CTCL.
Collapse
Affiliation(s)
- Christoph Schlapbach
- Department of Dermatology, Inselspital, University of Bern, 3010 Bern, Switzerland.
| | | | | | | | | |
Collapse
|
30
|
Tacken PJ, Figdor CG. Targeted antigen delivery and activation of dendritic cells in vivo: steps towards cost effective vaccines. Semin Immunol 2011; 23:12-20. [PMID: 21269839 DOI: 10.1016/j.smim.2011.01.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 01/05/2011] [Indexed: 12/13/2022]
Abstract
During the past decade, the immunotherapeutic potential of ex vivo generated professional antigen presenting dendritic cells (DCs) has been explored in the clinic. Albeit safe, clinical results have thus far been limited. A major disadvantage of current cell-based dendritic cell (DC) therapies, preventing universal implementation of this form of immunotherapy, is the requirement that vaccines need to be tailor made for each individual. Targeted delivery of antigens to DC surface receptors in vivo would circumvent this laborious and expensive ex vivo culturing steps involved with these cell-based therapies. In addition, the opportunity to target natural and often rare DC subsets in vivo might have advantages over loading more artificial ex vivo cultured DCs. Preclinical studies show targeting antigens to DCs effectively induces humoral responses, while cellular responses are induced provided a DC maturation or activation stimulus is co-administered. Here, we discuss strategies to target antigens to distinct DC subsets and to simultaneously employ adjuvants to activate these cells to induce immunity.
Collapse
Affiliation(s)
- Paul J Tacken
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Medical Centre, Postbox 9101, 6500 HB Nijmegen, The Netherlands.
| | | |
Collapse
|
31
|
Archaeosome adjuvant overcomes tolerance to tumor-associated melanoma antigens inducing protective CD8 T cell responses. Clin Dev Immunol 2011; 2010:578432. [PMID: 21318177 PMCID: PMC3034908 DOI: 10.1155/2010/578432] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 12/15/2010] [Accepted: 12/23/2010] [Indexed: 01/16/2023]
Abstract
Vesicles comprised of the ether glycerolipids of the archaeon Methanobrevibacter smithii (archaeosomes) are potent adjuvants for evoking CD8+ T cell responses. We therefore explored the ability of archaeosomes to overcome immunologic tolerance to self-antigens. Priming and boosting of mice with archaeosome-antigen evoked comparable CD8+ T cell response and tumor protection to an alternate boosting strategy utilizing live bacterial vectors for antigen delivery. Vaccination with melanoma antigenic peptides TRP181-189 and Gp10025-33 delivered in archaeosomes resulted in IFN-γ producing antigen-specific CD8+ T cells with strong cytolytic capability and protection against subcutaneous B16 melanoma. Targeting responses against multiple antigens afforded prolonged median survival against melanoma challenge. Entrapment of multiple peptides within the same vesicle or admixed formulations were both effective at evoking CD8+ T cells against each antigen. Melanoma-antigen archaeosome formulations also afforded therapeutic protection against established B16 tumors when combined with depletion of T-regulatory cells. Overall, we demonstrate that archaeosome adjuvants constitute an effective choice for formulating cancer vaccines.
Collapse
|
32
|
Cesson V, Rivals JP, Escher A, Piotet E, Thielemans K, Posevitz V, Dojcinovic D, Monnier P, Speiser D, Bron L, Romero P. MAGE-A3 and MAGE-A4 specific CD4(+) T cells in head and neck cancer patients: detection of naturally acquired responses and identification of new epitopes. Cancer Immunol Immunother 2011; 60:23-35. [PMID: 20857101 PMCID: PMC11028544 DOI: 10.1007/s00262-010-0916-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Accepted: 08/31/2010] [Indexed: 12/13/2022]
Abstract
Frequent expression of cancer testis antigens (CTA) has been consistently observed in head and neck squamous cell carcinomas (HNSCC). For instance, in 52 HNSCC patients, MAGE-A3 and -A4 CTA were expressed in over 75% of tumors, regardless of the sites of primary tumors such as oral cavity or hypopharynx. Yet, T-cell responses against these CTA in tumor-bearing patients have not been investigated in detail. In this study, we assessed the naturally acquired T-cell response against MAGE-A3 and -A4 in nonvaccinated HNSCC patients. Autologous antigen-presenting cells pulsed with overlapping peptide pools were used to detect and isolate MAGE-A3 and MAGE-A4 specific CD4(+) T cells from healthy donors and seven head and neck cancer patients. CD4(+) T-cell clones were characterized by cytokine secretion. We could detect and isolate MAGE-A3 and MAGE-A4 specific CD4(+) T cells from 7/7 cancer patients analyzed. Moreover, we identified six previously described and three new epitopes for MAGE-A3. Among them, the MAGE-A3(111-125) and MAGE-A3(161-175) epitopes were shown to be naturally processed and presented by DC in association with HLA-DP and DR, respectively. All of the detected MAGE-A4 responses were specific for new helper epitopes. These data suggest that naturally acquired CD4(+) T-cell responses against CT antigens often occur in vivo in HNSCC cancer patients and provide a rationale for the development of active immunotherapeutic approaches in this type of tumor.
Collapse
Affiliation(s)
- Valérie Cesson
- Ludwig Institute for Cancer Research Ltd., Lausanne Branch, University Hospital (CHUV), Hôpital Orthopédique Niv. 5, aile est, Av. Pierre Decker 4, 1011 Lausanne, Switzerland
| | - Jean-Paul Rivals
- Service of Head and Neck Surgery, University Hospital, Lausanne, Switzerland
| | - Anette Escher
- Service of Head and Neck Surgery, University Hospital, Lausanne, Switzerland
| | - Elsa Piotet
- Service of Head and Neck Surgery, University Hospital, Lausanne, Switzerland
| | - Kris Thielemans
- Department of Physiology-Immunology, Medical School of the Vrje Universiteit Brussel, Brussels, Belgium
| | - Vilmos Posevitz
- Ludwig Institute for Cancer Research Ltd., Lausanne Branch, University Hospital (CHUV), Hôpital Orthopédique Niv. 5, aile est, Av. Pierre Decker 4, 1011 Lausanne, Switzerland
| | - Danijel Dojcinovic
- Ludwig Institute for Cancer Research Ltd., Lausanne Branch, 1066 Epalinges, Switzerland
| | - Philippe Monnier
- Service of Head and Neck Surgery, University Hospital, Lausanne, Switzerland
| | - Daniel Speiser
- Ludwig Institute for Cancer Research Ltd., Lausanne Branch, University Hospital (CHUV), Hôpital Orthopédique Niv. 5, aile est, Av. Pierre Decker 4, 1011 Lausanne, Switzerland
| | - Luc Bron
- Service of Head and Neck Surgery, University Hospital, Lausanne, Switzerland
| | - Pedro Romero
- Ludwig Institute for Cancer Research Ltd., Lausanne Branch, University Hospital (CHUV), Hôpital Orthopédique Niv. 5, aile est, Av. Pierre Decker 4, 1011 Lausanne, Switzerland
| |
Collapse
|