151
|
Bear AS, Kennedy LC, Young JK, Perna SK, Mattos Almeida JP, Lin AY, Eckels PC, Drezek RA, Foster AE. Elimination of metastatic melanoma using gold nanoshell-enabled photothermal therapy and adoptive T cell transfer. PLoS One 2013; 8:e69073. [PMID: 23935927 PMCID: PMC3720863 DOI: 10.1371/journal.pone.0069073] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 06/05/2013] [Indexed: 11/19/2022] Open
Abstract
Ablative treatments such as photothermal therapy (PTT) are attractive anticancer strategies because they debulk accessible tumor sites while simultaneously priming antitumor immune responses. However, the immune response following thermal ablation is often insufficient to treat metastatic disease. Here we demonstrate that PTT induces the expression of proinflammatory cytokines and chemokines and promotes the maturation of dendritic cells within tumor-draining lymph nodes, thereby priming antitumor T cell responses. Unexpectedly, however, these immunomodulatory effects were not beneficial to overall antitumor immunity. We found that PTT promoted the infiltration of secondary tumor sites by CD11b+Ly-6G/C+ myeloid-derived suppressor cells, consequently failing to slow the growth of poorly immunogenic B16-F10 tumors and enhancing the growth of distant lung metastases. To exploit the beneficial effects of PTT activity against local tumors and on antitumor immunity whilst avoiding the adverse consequences, we adoptively transferred gp100-specific pmel T cells following PTT. The combination of local control by PTT and systemic antitumor immune reactivity provided by adoptively transferred T cells prevented primary tumor recurrence post-ablation, inhibited tumor growth at distant sites, and abrogated the outgrowth of lung metastases. Hence, the combination of PTT and systemic immunotherapy prevented the adverse effects of PTT on metastatic tumor growth and optimized overall tumor control.
Collapse
Affiliation(s)
- Adham S. Bear
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
| | - Laura C. Kennedy
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Joseph K. Young
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, United States of America
| | - Serena K. Perna
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas, United States of America
| | | | - Adam Y. Lin
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Phillip C. Eckels
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas, United States of America
| | - Rebekah A. Drezek
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
- Department of Electrical and Computer Engineering, Rice University, Houston, Texas, United States of America
- * E-mail: (RAD); (AEF)
| | - Aaron E. Foster
- Center for Cell and Gene Therapy, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas, United States of America
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas, United States of America
- Bellicum Pharmaceuticals, Houston, Texas, United States of America
- * E-mail: (RAD); (AEF)
| |
Collapse
|
152
|
Ascierto PA, Grimaldi AM, Acquavella N, Borgognoni L, Calabrò L, Cascinelli N, Cesano A, Del Vecchio M, Eggermont AM, Faries M, Ferrone S, Fox BA, Gajewski TF, Galon J, Gnjatic S, Gogas H, Kashani-Sabet M, Kaufman HL, Larkin J, Lo RS, Mantovani A, Margolin K, Melief C, McArthur G, Palmieri G, Puzanov I, Ribas A, Seliger B, Sosman J, Suenaert P, Tarhini AA, Trinchieri G, Vidal-Vanaclocha F, Wang E, Ciliberto G, Mozzillo N, Marincola FM, Thurin M. Future perspectives in melanoma research. Meeting report from the "Melanoma Bridge. Napoli, December 2nd-4th 2012". J Transl Med 2013; 11:137. [PMID: 23731854 PMCID: PMC3681569 DOI: 10.1186/1479-5876-11-137] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 05/19/2013] [Indexed: 02/08/2023] Open
Abstract
Recent insights into the genetic and somatic aberrations have initiated a new era of rapidly evolving targeted and immune-based treatments for melanoma. After decades of unsuccessful attempts to finding a more effective cure in the treatment of melanoma now we have several drugs active in melanoma. The possibility to use these drugs in combination to improve responses to overcome the resistance, to potentiate the action of immune system with the new immunomodulating antibodies, and identification of biomarkers that can predict the response to a particular therapy represent new concepts and approaches in the clinical management of melanoma. The third "Melanoma Research: "A bridge from Naples to the World" meeting, shortened as "Bridge Melanoma Meeting" took place in Naples, December 2 to 4th, 2012. The four topics of discussion at this meeting were: advances in molecular profiling and novel biomarkers, combination therapies, novel concepts toward integrating biomarkers and therapies into contemporary clinical management of patients with melanoma across the entire spectrum of disease stage, and the knowledge gained from the biology of tumor microenvironment across different tumors as a bridge to impact on prognosis and response to therapy in melanoma. This international congress gathered more than 30 international faculty members who in an interactive atmosphere which stimulated discussion and exchange of their experience regarding the most recent advances in research and clinical management of melanoma patients.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Istituto Nazionale Tumori, Fondazione “G. Pascale”, Naples, Italy
| | | | - Nicolas Acquavella
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MA, USA
| | - Lorenzo Borgognoni
- Plastic and Reconstructive Surgery, Regional Melanoma Refferral Center – S.M. Annunziata Hospital, Florence, Italy
| | - Luana Calabrò
- Medical Oncology and Immunotherapy, University Hospital of Siena, Istituto Toscano Tumori, Siena, Italy
| | | | | | - Michele Del Vecchio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Mark Faries
- John Wayne Cancer Institute, Santa Monica, CA, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bernard A Fox
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | | | - Jérôme Galon
- INSERM, U872, Laboratory of Integrative Cancer Immunology, Paris F-75006, France
- Université Paris Descartes, Paris, France
- Centre de Recherche des Cordeliers, Université Pierre et Marie Curie Paris 6, Paris, France
| | - Sacha Gnjatic
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Helen Gogas
- 1st Department of Medicine, Medical School, University of Athens, Athens, Greece
| | - Mohammed Kashani-Sabet
- Center for Melanoma Research and Treatment, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | | | | | - Roger S Lo
- Dermatology/Medicine, UCLA Geffen School of Medicine and Jonsson Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Kim Margolin
- Fred Hutchinson Cancer Research Center, Seattle Cancer Care Alliance, University of Washington, Seattle, WA, USA
| | - Cornelis Melief
- Leiden University Medical Center and ISA Pharmaceuticals, Leiden, The Netherlands
| | - Grant McArthur
- Peter MacCallum Cancer Centre, East Melbourne, Australia
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Biomolecular Chemistry, National Research Council, Sassari, Italy
| | - Igor Puzanov
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Antoni Ribas
- Tumor Immunology Program, Jonsson Comprehensive Cancer Center (JCCC), David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jeff Sosman
- Vanderbilt-Ingram Comprehensive Cancer Center, Nashville, TN, USA
| | - Peter Suenaert
- Global Early Clinical Development, Clinical Immunotherapeutics, Immunotherapeutics, GlaxoSmithKline Vaccines, Rixensart, Belgium
| | - Ahmad A Tarhini
- University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Giorgio Trinchieri
- Cancer and Inflammation Program, Center for Cancer Research, NCI, NIH, Frederick, MD, USA
| | - Fernando Vidal-Vanaclocha
- Institute of Applied Molecular Medicine (IMMA), CEU-San Pablo University and HM-Hospitals School of Medicine, Boadilla del Monte, 28668, Madrid, Spain
| | - Ena Wang
- Infectious Disease and Immunogenetics Section (IDIS), Department of Transfusion Medicine, Clinical Center and Center for Human Immunology (CHI), NIH, Bethesda, MD, USA
| | | | - Nicola Mozzillo
- Istituto Nazionale Tumori, Fondazione “G. Pascale”, Naples, Italy
| | | | | |
Collapse
|
153
|
Tüting T. T cell immunotherapy for melanoma from bedside to bench to barn and back: how conceptual advances in experimental mouse models can be translated into clinical benefit for patients. Pigment Cell Melanoma Res 2013; 26:441-56. [PMID: 23617831 DOI: 10.1111/pcmr.12111] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/18/2013] [Indexed: 12/27/2022]
Abstract
A solid scientific basis now supports the concept that cytotoxic T lymphocytes can specifically recognize and destroy melanoma cells. Over the last decades, clinicians and basic scientists have joined forces to advance our concepts of melanoma immunobiology. This has catalyzed the rational development of therapeutic approaches to enforce melanoma-specific T cell responses. Preclinical studies in experimental mouse models paved the way for their successful translation into clinical benefit for patients with metastatic melanoma. A more thorough understanding of how melanomas develop resistance to T cell immunotherapy is necessary to extend this success. This requires a continued interdisciplinary effort of melanoma biologists and immunologists that closely connects clinical observations with in vitro investigations and appropriate in vivo mouse models: From bedside to bench to barn and back.
Collapse
Affiliation(s)
- Thomas Tüting
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
154
|
Cappello P, Rolla S, Chiarle R, Principe M, Cavallo F, Perconti G, Feo S, Giovarelli M, Novelli F. Vaccination with ENO1 DNA prolongs survival of genetically engineered mice with pancreatic cancer. Gastroenterology 2013; 144:1098-106. [PMID: 23333712 DOI: 10.1053/j.gastro.2013.01.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 11/30/2012] [Accepted: 01/10/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDA) is an aggressive tumor, and patients typically present with late-stage disease; rates of 5-year survival after pancreaticoduodenectomy are low. Antibodies against α-enolase (ENO1), a glycolytic enzyme, are detected in more than 60% of patients with PDA, and ENO1-specific T cells inhibit the growth of human pancreatic xenograft tumors in mice. We investigated whether an ENO1 DNA vaccine elicits antitumor immune responses and prolongs survival of mice that spontaneously develop autochthonous, lethal pancreatic carcinomas. METHODS We injected and electroporated a plasmid encoding ENO1 (or a control plasmid) into Kras(G12D)/Cre (KC) mice and Kras(G12D)/Trp53(R172H)/Cre (KPC) mice at 4 weeks of age (when pancreatic intraepithelial lesions are histologically evident). Antitumor humoral and cellular responses were analyzed by histology, immunohistochemistry, enzyme-linked immunosorbent assays, flow cytometry, and enzyme-linked immunosorbent spot and cytotoxicity assays. Survival was analyzed by Kaplan-Meier analysis. RESULTS The ENO1 vaccine induced antibody and a cellular response and increased survival times by a median of 138 days in KC mice and 42 days in KPC mice compared with mice given the control vector. On histologic analysis, the vaccine appeared to slow tumor progression. The vaccinated mice had increased serum levels of anti-ENO1 immunoglobulin G, which bound the surface of carcinoma cells and induced complement-dependent cytotoxicity. ENO1 vaccination reduced numbers of myeloid-derived suppressor cells and T-regulatory cells and increased T-helper 1 and 17 responses. CONCLUSIONS In a genetic model of pancreatic carcinoma, vaccination with ENO1 DNA elicits humoral and cellular immune responses against tumors, delays tumor progression, and significantly extends survival. This vaccination strategy might be developed as a neoadjuvant therapy for patients with PDA.
Collapse
Affiliation(s)
- Paola Cappello
- Center for Experimental Research and Medical Studies, Città della Salute e della Scienza di Torino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
155
|
Engels B, Engelhard VH, Sidney J, Sette A, Binder DC, Liu RB, Kranz DM, Meredith SC, Rowley DA, Schreiber H. Relapse or eradication of cancer is predicted by peptide-major histocompatibility complex affinity. Cancer Cell 2013; 23:516-26. [PMID: 23597565 PMCID: PMC3658176 DOI: 10.1016/j.ccr.2013.03.018] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 02/21/2013] [Accepted: 03/19/2013] [Indexed: 01/24/2023]
Abstract
Cancers often relapse after adoptive therapy, even though specific T cells kill cells from the same cancer efficiently in vitro. We found that tumor eradication by T cells required high affinities of the targeted peptides for major histocompatibility complex (MHC) class I. Affinities of at least 10 nM were required for relapse-free regression. Only high-affinity peptide-MHC interactions led to efficient cross-presentation of antigen, thereby stimulating cognate T cells to secrete cytokines. These findings highlight the importance of targeting peptides with high affinity for MHC class I when designing T cell-based immunotherapy.
Collapse
Affiliation(s)
- Boris Engels
- Department of Pathology, Committee on Immunology and Committee on Cancer Biology, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
156
|
Hailemichael Y, Dai Z, Jaffarzad N, Ye Y, Medina MA, Huang XF, Dorta-Estremera SM, Greeley NR, Nitti G, Peng W, Liu C, Lou Y, Wang Z, Ma W, Rabinovich B, Sowell RT, Schluns KS, Davis RE, Hwu P, Overwijk WW. Persistent antigen at vaccination sites induces tumor-specific CD8⁺ T cell sequestration, dysfunction and deletion. Nat Med 2013; 19:465-72. [PMID: 23455713 PMCID: PMC3618499 DOI: 10.1038/nm.3105] [Citation(s) in RCA: 364] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/25/2013] [Indexed: 12/22/2022]
Abstract
To understand why cancer vaccine-induced T cells often fail to eradicate tumors, we studied immune responses in mice vaccinated with gp100 melanoma peptide in incomplete Freund’s adjuvant (IFA), commonly used in clinical cancer vaccine trials. Peptide/IFA vaccination primed tumor-specific CD8+ T cells, which accumulated not in tumors but at the persisting, antigen-rich vaccination site. Once there, primed T cells became dysfunctional and underwent antigen-driven, Interferon-γ (IFN-γ) and Fas ligand (FasL)-mediated apoptosis, resulting in hyporesponsiveness to subsequent vaccination. Provision of anti-CD40 antibody, Toll-like receptor 7 (TLR7) agonist and interleukin-2 (IL-2) reduced T cell apoptosis but did not prevent vaccination site sequestration. A non-persisting vaccine formulation shifted T cell localization towards tumors, inducing superior anti-tumor activity while reducing systemic T cell dysfunction and promoting memory formation. Persisting peptide/IFA vaccine depots can induce specific T cell sequestration, dysfunction and deletion at vaccination sites; short-lived formulations may overcome these limitations and result in greater therapeutic efficacy of peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Yared Hailemichael
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Rodriguez GM, D'Urbano D, Bobbala D, Chen XL, Yeganeh M, Ramanathan S, Ilangumaran S. SOCS1 prevents potentially skin-reactive cytotoxic T lymphocytes from gaining the ability to cause inflammatory lesions. J Invest Dermatol 2013; 133:2013-22. [PMID: 23443260 DOI: 10.1038/jid.2013.86] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2012] [Revised: 01/28/2013] [Accepted: 02/01/2013] [Indexed: 12/21/2022]
Abstract
Suppressor of cytokine signaling 1 (SOCS1) is a critical regulator of T lymphocyte homeostasis. SOCS1-deficient mice accumulate CD8(+) T cells, which display a memory-like phenotype and proliferate strongly to IL-15. Socs1(-/-) mice develop inflammatory skin lesions, however, the underlying mechanisms are not well understood. In order to investigate the role of SOCS1 in regulating CD8(+) T cells potentially reactive to tissue antigens (Ags) of the skin, we generated Socs1(-/-) mice expressing MHC-I-restricted Pmel-1 transgenic TCR specific to the melanoma-derived gp100 Ag, which is also expressed by normal melanocytes. Socs1(-/-) Pmel-1 cells express increased levels of memory markers CD44, Ly6C, CD122, and CD62L, and show downregulation of TCR and upregulation of CD5, suggesting in vivo TCR stimulation. However, stimulation of Socs1(-/-)Pmel-1 cells with gp100-derived peptide induced only marginal proliferation in vitro despite eliciting strong effector functions, which was associated with elevated Blimp-1 induction. Following adoptive transfer to Rag1(-/-) mice, Socs1(-/-)Pmel-1 cells underwent lymphopenia-induced proliferation and caused severe skin pathology characterized by inflammatory lesions in ears, muzzle, extremities, and eyes. These findings underscore the importance of SOCS1 in regulating potentially skin-reactive cytotoxic T lymphocytes, which could get activated under conditions that promote Ag-nonspecific, cytokine-driven proliferation.
Collapse
Affiliation(s)
- Galaxia Maria Rodriguez
- Division of Immunology, Department of Pediatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
158
|
Kwong B, Gai SA, Elkhader J, Wittrup KD, Irvine DJ. Localized immunotherapy via liposome-anchored Anti-CD137 + IL-2 prevents lethal toxicity and elicits local and systemic antitumor immunity. Cancer Res 2013; 73:1547-58. [PMID: 23436794 PMCID: PMC3594475 DOI: 10.1158/0008-5472.can-12-3343] [Citation(s) in RCA: 154] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunostimulatory agonists such as anti-CD137 and interleukin (IL)-2 have elicited potent antitumor immune responses in preclinical studies, but their clinical use is limited by inflammatory toxicities that result upon systemic administration. We hypothesized that by rigorously restricting the biodistribution of immunotherapeutic agents to a locally accessible lesion and draining lymph node(s), effective local and systemic antitumor immunity could be achieved in the absence of systemic toxicity. We anchored anti-CD137 and an engineered IL-2Fc fusion protein to the surfaces of PEGylated liposomes, whose physical size permitted dissemination in the tumor parenchyma and tumor-draining lymph nodes but blocked entry into the systemic circulation following intratumoral injection. In the B16F10 melanoma model, intratumoral liposome-coupled anti-CD137 + IL-2Fc therapy cured a majority of established primary tumors while avoiding the lethal inflammatory toxicities caused by equivalent intratumoral doses of soluble immunotherapy. Immunoliposome therapy induced protective antitumor memory and elicited systemic antitumor immunity that significantly inhibited the growth of simultaneously established distal tumors. Tumor inhibition was CD8(+) T-cell-dependent and was associated with increased CD8(+) T-cell infiltration in both treated and distal tumors, enhanced activation of tumor antigen-specific T cells in draining lymph nodes, and a reduction in regulatory T cells in treated tumors. These data suggest that local nanoparticle-anchored delivery of immuno-agonists represents a promising strategy to improve the therapeutic window and clinical applicability of highly potent but otherwise intolerable regimens of cancer immunotherapy. Cancer Res; 73(5); 1547-58. ©2012 AACR.
Collapse
Affiliation(s)
- Brandon Kwong
- Koch Institute for Integrative Cancer Research, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | | |
Collapse
|
159
|
Lin AY, Lunsford J, Bear AS, Young JK, Eckels P, Luo L, Foster AE, Drezek RA. High-density sub-100-nm peptide-gold nanoparticle complexes improve vaccine presentation by dendritic cells in vitro. NANOSCALE RESEARCH LETTERS 2013; 8:72. [PMID: 23402570 PMCID: PMC3579702 DOI: 10.1186/1556-276x-8-72] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 02/02/2013] [Indexed: 05/21/2023]
Abstract
Nanocarriers have been explored to improve the delivery of tumor antigens to dendritic cells (DCs). Gold nanoparticles are attractive nanocarriers because they are inert, non-toxic, and can be readily endocytosed by DCs. Here, we designed novel gold-based nanovaccines (AuNVs) using a simple self-assembling bottom-up conjugation method to generate high-peptide density delivery and effective immune responses with limited toxicity. AuNVs were synthesized using a self-assembling conjugation method and optimized using DC-to-splenocyte interferon-γ enzyme-linked immunosorbent spot assays. The AuNV design has shown successful peptide conjugation with approximately 90% yield while remaining smaller than 80 nm in diameter. DCs uptake AuNVs with minimal toxicity and are able to process the vaccine peptides on the particles to stimulate cytotoxic T lymphocytes (CTLs). These high-peptide density AuNVs can stimulate CTLs better than free peptides and have great potential as carriers for various vaccine types.
Collapse
Affiliation(s)
- Adam Yuh Lin
- Department of Bioengineering, Rice University, 77005, Houston, TX USA
| | - Jessica Lunsford
- Center for Cell and Gene Therapy, Baylor College of Medicine, 77030, Houston, TX, USA
| | - Adham Sean Bear
- Center for Cell and Gene Therapy, Baylor College of Medicine, 77030, Houston, TX, USA
| | - Joseph Keith Young
- Department of Electrical and Computer Engineering, Rice University, 77005, Houston, TX USA
| | - Phillip Eckels
- Center for Cell and Gene Therapy, Baylor College of Medicine, 77030, Houston, TX, USA
| | - Laureen Luo
- Department of Bioengineering, Rice University, 77005, Houston, TX USA
| | - Aaron Edward Foster
- Center for Cell and Gene Therapy, Baylor College of Medicine, 77030, Houston, TX, USA
| | - Rebekah Anna Drezek
- Department of Bioengineering, Rice University, 77005, Houston, TX USA
- Department of Electrical and Computer Engineering, Rice University, 77005, Houston, TX USA
| |
Collapse
|
160
|
Cafri G, Sharbi-Yunger A, Tzehoval E, Eisenbach L. Production of LacZ inducible T cell hybridoma specific for human and mouse gp100₂₅₋₃₃ peptides. PLoS One 2013; 8:e55583. [PMID: 23383339 PMCID: PMC3562179 DOI: 10.1371/journal.pone.0055583] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 12/28/2012] [Indexed: 11/18/2022] Open
Abstract
Identification and quantification of immunogenic peptides and tumor-derived epitopes presented on MHC-I molecules are essential for basic studies and vaccines generation. Although lymphocytes derived from transgenic mice can serve as sensitive detectors of processes of antigen presentation and recognition, they are not always available. The use of cell lines might be extremely useful. In this study, we generated a lacZ inducible CD8⁺ hybridoma (BUSA14) capable of recognizing both human and mouse gp100₂₅₋₃₃ melanoma antigens presented on dendritic and tumor cell lines. This hybridoma expresses a variety of membranal T cell markers and secretes IL-2 and TNFα. Thus, BUSA14 offers a quantifiable, cheap and straightforward tool for studying peptide presentation by MHC-I molecules on the cell surface.
Collapse
Affiliation(s)
- Gal Cafri
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Adi Sharbi-Yunger
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Esther Tzehoval
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | - Lea Eisenbach
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
161
|
Yu X, Guo C, Yi H, Qian J, Fisher PB, Subjeck JR, Wang XY. A multifunctional chimeric chaperone serves as a novel immune modulator inducing therapeutic antitumor immunity. Cancer Res 2013; 73:2093-103. [PMID: 23333935 DOI: 10.1158/0008-5472.can-12-1740] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Converting the immunosuppressive tumor environment into one that is favorable to the induction of antitumor immunity is indispensable for effective cancer immunotherapy. Here, we strategically incorporate a pathogen (i.e., flagellin)-derived, NF-κB-stimulating "danger" signal into the large stress protein or chaperone Grp170 (HYOU1/ORP150) that was previously shown to facilitate antigen crosspresentation. This engineered chimeric molecule (i.e., Flagrp170) is capable of transporting tumor antigens and concurrently inducing functional activation of dendritic cells (DC). Intratumoral administration of adenoviruses expressing Flagrp170 induces a superior antitumor response against B16 melanoma and its distant lung metastasis compared with unmodified Grp170 and flagellin. The enhanced tumor destruction is accompanied with significantly increased tumor infiltration by CD8(+) cells as well as elevation of IFN-γ and interleukin (IL)-12 levels in the tumor sites. In situ Ad.Flagrp170 therapy provokes systemic activation of CTLs that recognize several antigens naturally expressing in melanoma (e.g., gp100/PMEL and TRP2/DCT). The mechanistic studies using CD11c-DTR transgenic mice and Batf3-deficient mice reveal that CD8α(+) DCs are required for the improved T-cell crosspriming. Antibody neutralization assays show that IL-12 and IFN-γ are essential for the Flagrp170-elicited antitumor response, which also involves CD8(+) T cells and natural killer cells. The therapeutic efficacy of Flagrp170 and its immunostimulating activity are also confirmed in mouse prostate cancer and colon carcinoma. Together, targeting the tumor microenvironment with this chimeric chaperone is highly effective in mobilizing or restoring antitumor immunity, supporting the potential therapeutic use of this novel immunomodulator in the treatment of metastatic diseases.
Collapse
Affiliation(s)
- Xiaofei Yu
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | | | | | | | | | | | | |
Collapse
|
162
|
Ly LV, Sluijter M, van der Burg SH, Jager MJ, van Hall T. Effective cooperation of monoclonal antibody and peptide vaccine for the treatment of mouse melanoma. THE JOURNAL OF IMMUNOLOGY 2012. [PMID: 23203930 DOI: 10.4049/jimmunol.1200135] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
mAbs binding to tumor-associated surface Ags are therapeutically applied in a range of malignancies. Therapeutic vaccination only recently met with clinical success, and the first cancer vaccine received U.S. Food and Drug Administration approval last year. To improve current protocols, we combined peptide vaccines with mAb to the tyrosinase-related protein (TRP)-1 surface Ag for the treatment of B16F10 skin melanoma. Vaccine formulations with synthetic long peptides failed to elicit strong CD8 T cell responses to self-differentiation Ags gp100 and TRP-2, whereas altered peptide sequences recruited gp100-specific CD8 T cells from the endogenous repertoire with frequencies of 40%. However, these high frequencies were reached too late; large, progressively growing melanomas had already emerged. Addition of the TRP-1-directed mAb TA99 to the treatment protocol mediated eradication of s.c. lesions. The mode of action of the Ab did not depend on complement factor C3 and did not lead to improved Ag presentation and CD8 T cell immunity; rather, it recruited FcγR-bearing innate immune cells during early tumor control, thereby creating a window of time for the generation of protective cellular immunity. These data support the concept of combination therapy, in which passive transfer of mAbs is supplemented with cancer peptide vaccines. Moreover, we advocate that tumor Ag-specific T cell immunity directed against self-proteins can be exploited from the endogenous repertoire.
Collapse
Affiliation(s)
- Long V Ly
- Department of Ophthalmology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
163
|
Watson AM, Mylin LM, Thompson MM, Schell TD. Modification of a tumor antigen determinant to improve peptide/MHC stability is associated with increased immunogenicity and cross-priming a larger fraction of CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:5549-60. [PMID: 23175697 DOI: 10.4049/jimmunol.1102221] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Altered peptide ligands (APLs) with enhanced binding to MHC class I can increase the CD8(+) T cell response to native Ags, including tumor Ags. In this study, we investigate the influence of peptide-MHC (pMHC) stability on recruitment of tumor Ag-specific CD8(+) T cells through cross-priming. Among the four known H-2(b)-restricted CD8(+) T cell determinants within SV40 large tumor Ag (TAg), the site V determinant ((489)QGINNLDNL(497)) forms relatively low-stability pMHC and is characteristically immunorecessive. Absence of detectable site V-specific CD8(+) T cells following immunization with wild-type TAg is due in part to inefficient cross-priming. We mutated nonanchor residues within the TAg site V determinant that increased pMHC stability but preserved recognition by both TCR-transgenic and polyclonal endogenous T cells. Using a novel approach to quantify the fraction of naive T cells triggered through cross-priming in vivo, we show that immunization with TAg variants expressing higher-stability determinants increased the fraction of site V-specific T cells cross-primed and effectively overcame the immunorecessive phenotype. In addition, using MHC class I tetramer-based enrichment, we demonstrate for the first time, to our knowledge, that endogenous site V-specific T cells are primed following wild-type TAg immunization despite their low initial frequency, but that the magnitude of T cell accumulation is enhanced following immunization with a site V variant TAg. Our results demonstrate that site V APLs cross-prime a higher fraction of available T cells, providing a potential mechanism for high-stability APLs to enhance immunogenicity and accumulation of T cells specific for the native determinant.
Collapse
Affiliation(s)
- Alan M Watson
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
164
|
Roy BM, Zhukov DV, Maynard JA. Flanking residues are central to DO11.10 T cell hybridoma stimulation by ovalbumin 323-339. PLoS One 2012; 7:e47585. [PMID: 23110081 PMCID: PMC3479146 DOI: 10.1371/journal.pone.0047585] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 09/18/2012] [Indexed: 11/30/2022] Open
Abstract
T cell activation requires formation of a tri-molecular interaction between a major histocompatibility complex (MHC), peptide, and T cell receptor. In a common model system, the ovalbumin epitope 323–339 binds the murine class II MHC, I-Ad, in at least three distinct registers. The DO11.10 T cell recognizes the least stable of these, as determined by peptide-MHC dissociation rates. Using exogenous peptides and peptide insertions into a carrier protein in combination with IL-2 secretion assays, we show that the alternate registers do not competitively inhibit display of the active register four. In contrast, this weakly binding register is stabilized by the presence of n-terminal flanking residues active in MHC binding. The DO11.10 hybridoma is sensitive to the presence of specific wild-type residues extending to at least the P-3 peptide position. Transfer of the P-4 to P-2 flanking residues to a hen egg lysozyme epitope also presented by I-Ad increases the activity of that epitope substantially. These results illustrate the inherent complexity in delineating the interaction of multiple registers based on traditional thermodynamic measurements and demonstrate the potential of flanking residue modification for increasing the activity of weakly bound epitopes. The latter technique represents an alternative to substitution of anchor residues within a weakly bound register, which we show can significantly decrease the activity of the epitope to a responding T cell.
Collapse
Affiliation(s)
- Benjamin M. Roy
- Department of Chemical Engineering, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Chemical Engineering, University of Texas at Austin, Austin Texas, United States of America
| | - Dmitriy V. Zhukov
- Department of Chemical Engineering, University of Texas at Austin, Austin Texas, United States of America
| | - Jennifer A. Maynard
- Department of Chemical Engineering, University of Texas at Austin, Austin Texas, United States of America
- * E-mail:
| |
Collapse
|
165
|
Rommelfanger DM, Wongthida P, Diaz RM, Kaluza KM, Thompson JM, Kottke TJ, Vile RG. Systemic combination virotherapy for melanoma with tumor antigen-expressing vesicular stomatitis virus and adoptive T-cell transfer. Cancer Res 2012; 72:4753-64. [PMID: 22836753 PMCID: PMC3893932 DOI: 10.1158/0008-5472.can-12-0600] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oncolytic virotherapy offers the potential to treat tumors both as a single agent and in combination with traditional modalities such as chemotherapy and radiotherapy. Here we describe an effective, fully systemic treatment regimen, which combines virotherapy, acting essentially as an adjuvant immunotherapy, with adoptive cell transfer (ACT). The combination of ACT with systemic administration of a vesicular stomatitis virus (VSV) engineered to express the endogenous melanocyte antigen glycoprotein 100 (gp100) resulted in regression of established melanomas and generation of antitumor immunity. Tumor response was associated with in vivo T-cell persistence and activation as well as treatment-related vitiligo. However, in a proportion of treated mice, initial tumor regressions were followed by recurrences. Therapy was further enhanced by targeting an additional tumor antigen with the VSV-antigen + ACT combination strategy, leading to sustained response in 100% of mice. Together, our findings suggest that systemic virotherapy combined with antigen-expressing VSV could be used to support and enhance clinical immunotherapy protocols with adoptive T-cell transfer, which are already used in the clinic.
Collapse
|
166
|
Burkhardt UE, Sloots A, Jakobi V, Wei WZ, Cavallo F, Kloke BP, Wels WS. IL-15 augments antitumoral activity of an ErbB2/HER2 cancer vaccine targeted to professional antigen-presenting cells. Cancer Immunol Immunother 2012; 61:1473-84. [PMID: 22331080 PMCID: PMC11029045 DOI: 10.1007/s00262-012-1215-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/27/2012] [Indexed: 01/01/2023]
Abstract
Targeted delivery of tumor-associated antigens to professional antigen-presenting cells (APC) is being explored as a strategy to enhance the antitumoral activity of cancer vaccines. Here, we generated a cell-based system for continuous in vivo production of a CTLA-4-ErbB2 fusion protein as a therapeutic vaccine. The chimeric CTLA-4-ErbB2 molecule contains the extracellular domain of CTLA-4 for specific targeting to costimulatory B7 molecules on the surface of APC, genetically fused to residues 1-222 of human ErbB2 (HER2) as an antigenic determinant. In wild-type BALB/c mice, inoculation of syngeneic epithelial cells continuously secreting the CTLA-4-ErbB2 fusion vaccine in the vicinity of subcutaneously growing ErbB2-expressing renal cell carcinomas resulted in the rejection of established tumors, accompanied by the induction of ErbB2-specific antibodies and cytotoxic T cells. In contrast, treatment with CTLA-4-ErbB2 vaccine-secreting producer cells alone was insufficient to induce tumor rejection in ErbB2-transgenic WAP-Her-2 F1 mice, which are characterized by pronounced immunological tolerance to the human self-antigen. When CTLA-4-ErbB2 producer cells were modified to additionally secrete interleukin (IL)-15, antigen-specific antitumoral activity of the vaccine in WAP-Her-2 F1 mice was restored, documented by an increase in survival, and marked inhibition of the growth of established ErbB2-expressing, but not antigen-negative tumors. Our results demonstrate that continuous in vivo expression of an APC-targeted ErbB2 fusion protein results in antigen-specific immune responses and antitumoral activity in tumor-bearing hosts, which is augmented by the pleiotropic cytokine IL-15. This provides a rationale for further development of this approach for specific cancer immunotherapy.
Collapse
Affiliation(s)
- Ute E. Burkhardt
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
| | - Arjen Sloots
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
| | - Vera Jakobi
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
| | - Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201 USA
| | - Federica Cavallo
- Department of Clinical and Biological Sciences, Molecular Biotechnology Center, University of Turin, 10126 Turin, Italy
| | - Björn-Philipp Kloke
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
| | - Winfried S. Wels
- Chemotherapeutisches Forschungsinstitut Georg-Speyer-Haus, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany
| |
Collapse
|
167
|
Multi-Compartmental Vaccine Delivery System for Enhanced Immune Response to gp100 Peptide Antigen in Melanoma Immunotherapy. Pharm Res 2012; 29:3393-403. [DOI: 10.1007/s11095-012-0834-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 07/09/2012] [Indexed: 12/28/2022]
|
168
|
Agonistic antibody to CD40 boosts the antitumor activity of adoptively transferred T cells in vivo. J Immunother 2012; 35:276-82. [PMID: 22421945 DOI: 10.1097/cji.0b013e31824e7f43] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CD40, a member of the tumor necrosis factor receptor superfamily, is broadly expressed on antigen-presenting cells and other cells, including fibroblasts and endothelial cells. Binding of CD40 and its natural ligand CD40L (CD154) triggers cytokine secretion, and increased expression of costimulatory molecules is required for T-cell activation and proliferation. However, to our knowledge, the use of agonistic antibodies to CD40 to boost adoptively transferred T cells in vivo has not been investigated. The purpose of this study was to determine whether anti-CD40 monoclonal antibody (mAb) in combination with interleukin (IL)-2 could improve the efficacy of in vitro-activated T cells to enhance antitumor activity. Mice bearing B16 melanoma tumors expressing the gp100 tumor antigen were treated with cultured, activated T cells transgenic for a T-cell receptor specifically recognizing gp100, with or without anti-CD40 mAb. In this model, the combination of anti-CD40 mAb with IL-2 led to expansion of adoptively transferred T cells and induced a more robust antitumor response. Furthermore, the expression of CD40 on bone marrow-derived cells and the presence of CD80/CD86 in the host were required for the expansion of adoptively transferred T cells. The use of neutralizing mAb to IL-12 provided direct evidence that enhanced IL-12 secretion induced by anti-CD40 mAb was crucial for the expansion of adoptively transferred T cells. Collectively, these findings provide a rationale to evaluate the potential application of anti-CD40 mAb in adoptive T-cell therapy for cancer.
Collapse
|
169
|
Eissler N, Ruf P, Mysliwietz J, Lindhofer H, Mocikat R. Trifunctional bispecific antibodies induce tumor-specific T cells and elicit a vaccination effect. Cancer Res 2012; 72:3958-66. [PMID: 22745368 DOI: 10.1158/0008-5472.can-12-0146] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A major goal of tumor immunotherapy is the induction of long-lasting systemic T-cell immunity. Bispecific antibodies (bsAbs) that lack the immunoglobulin Fc region confer T-cell-mediated killing of tumor cells but do not induce long-term memory. In contrast, trifunctional bsAbs comprise an appropriate Fc region and, therefore, not only recruit T cells but also accessory cells that bear activating Fcγ receptors (FcγR), providing additional T-cell-activating signals and securing presentation of tumor-derived antigens to T cells. In this study, we show that trifunctional bsAbs induce a polyvalent T-cell response and, therefore, a vaccination effect. Mice were treated with melanoma cells and with a trifunctional bsAb directed against the melanoma target antigen ganglioside GD2 in addition to murine CD3. The trifunctional bsAb activated dendritic cells and induced a systemic immune response that was not replicated by treatment with the F(ab')2-counterpart lacking the Fc region. Restimulation of spleen and lymph node cells in vitro yielded T-cell lines that specifically produced interferon-γ in response to tumor. In addition, trifunctional bsAb-induced T cells recognized various specific peptides derived from melanoma-associated antigens. Moreover, these polyvalent responses proved to be tumor-suppressive and could not be induced by the corresponding bsF(ab')2-fragment. Taken together, our findings provide preclinical proof of concept that trifunctional bsAbs can induce tumor-specific T cells with defined antigen specificity.
Collapse
Affiliation(s)
- Nina Eissler
- Helmholtz-Zentrum München, Institut für Molekulare Immunologie; Trion Research GmbH, München, Germany
| | | | | | | | | |
Collapse
|
170
|
Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol 2012; 12:269-81. [PMID: 22437939 PMCID: PMC6292222 DOI: 10.1038/nri3191] [Citation(s) in RCA: 1260] [Impact Index Per Article: 96.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunotherapy based on the adoptive transfer of naturally occurring or gene-engineered T cells can mediate tumour regression in patients with metastatic cancer. Here, we discuss progress in the use of adoptively transferred T cells, focusing on how they can mediate tumour cell eradication. Recent advances include more accurate targeting of antigens expressed by tumours and the associated vasculature, and the successful use of gene engineering to re-target T cells before their transfer into the patient. We also describe how new research has helped to identify the particular T cell subsets that can most effectively promote tumour eradication.
Collapse
Affiliation(s)
- Nicholas P Restifo
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | |
Collapse
|
171
|
Cho HI, Reyes-Vargas E, Delgado JC, Celis E. A potent vaccination strategy that circumvents lymphodepletion for effective antitumor adoptive T-cell therapy. Cancer Res 2012; 72:1986-95. [PMID: 22367213 DOI: 10.1158/0008-5472.can-11-3246] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adoptive cell therapy using tumor-reactive T lymphocytes is a promising approach for treating advanced cancer. Successful tumor eradication depends primarily on the expansion and survival of the adoptively transferred T cells. Lymphodepletion using total body irradiation (TBI) and administering high-dose interleukin (IL)-2 have been used with adoptive cell therapy to promote T-cell expansion and survival to achieve maximal therapeutic effects. However, TBI and high-dose IL-2 increase the risk for major complications that impact overall survival. Here we describe an alternative approach to TBI and high-dose IL-2 for optimizing adoptive cell therapy, resulting in dramatic therapeutic effects against established melanomas in mice. Administration of a potent, noninfectious peptide vaccine after adoptive cell therapy dramatically increased antigen-specific T-cell numbers leading to enhancement in the survival of melanoma-bearing mice. Furthermore, combinations of peptide vaccination with PD1 blockade or IL-2/anti-IL-2 antibody complexes led to complete disease eradication and long-term survival in mice with large tumors receiving adoptive cell therapy. Our results indicate that PD1 blockade and IL-2/anti-IL-2 complexes enhance both the quantitative and qualitative aspects of the T-cell responses induced by peptide vaccination after adoptive cell therapy. These findings could be useful for the optimization of adoptive cell therapy in cancer patients without the need of toxic adjunct procedures.
Collapse
Affiliation(s)
- Hyun-Il Cho
- Immunology Program, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | | | | | | |
Collapse
|
172
|
Harris JE, Harris TH, Weninger W, Wherry EJ, Hunter CA, Turka LA. A mouse model of vitiligo with focused epidermal depigmentation requires IFN-γ for autoreactive CD8⁺ T-cell accumulation in the skin. J Invest Dermatol 2012; 132:1869-76. [PMID: 22297636 PMCID: PMC3343174 DOI: 10.1038/jid.2011.463] [Citation(s) in RCA: 273] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Vitiligo is an autoimmune disease of the skin causing disfiguring patchy depigmentation of the epidermis and, less commonly, hair. Therapeutic options for vitiligo are limited, reflecting in part limited knowledge of disease pathogenesis. Existing mouse models of vitiligo consist of hair depigmentation but lack prominent epidermal involvement, which is the hallmark of human disease. They are thus unable to provide a platform to fully investigate disease mechanisms and treatment. CD8+ T cells have been implicated in the pathogenesis of vitiligo and expression of interferon-gamma (IFN-γ) is increased in the lesional skin of patients, however it is currently unknown what role IFN-γ plays in disease. Here, we have developed an adoptive transfer mouse model of vitiligo using melanocyte-specific CD8+ T cells, which recapitulates the human condition by inducing epidermal depigmentation while sparing the hair. Like active lesions in human vitiligo, histology of depigmenting skin reveals a patchy mononuclear infiltrate and single-cell infiltration of the epidermis. Depigmentation is accompanied by accumulation of autoreactive CD8+ T cells in the skin, quantifiable loss of tyrosinase transcript, and local IFN-γ production. Neutralization of IFN-γ with antibody prevents CD8+ T cell accumulation and depigmentation, suggesting a therapeutic potential for this approach.
Collapse
Affiliation(s)
- John E Harris
- Division of Dermatology, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, USA.
| | | | | | | | | | | |
Collapse
|
173
|
Amato RJ, Stepankiw M. Clinical Efficacy of TroVax in the Treatment of Progressive Castration-resistant Prostate Cancer. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2012; 6:67-73. [PMID: 22253556 PMCID: PMC3256982 DOI: 10.4137/cmo.s7654] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
With approximately 240,890 new cases expected in 2011, prostate cancer remains the leading cause of non-melanoma cancer deaths in men. Immunotherapies using viral vector-based delivery systems targeting tumor-specific antigens are being studied. Viral vector-based delivery systems present tumor-targeted antigens (TAAs) to the immune system while breaking self-tolerance. Modified vaccinia ankara has been combined with the oncofetal antigen 5T4 to create TroVax for the treatment of castration-resistant prostate cancer (CRPC). The 5T4 antigen is highly expressed in a large number of carcinomas, including prostate cancer, but is rarely expressed in healthy tissue. TroVax has been demonstrated to be safe and highly immunogenic, both as monotherapy and in combination with other standard of care therapies in colorectal, renal cell, and prostate cancer. With minimal side effects and the ability to produce a strong immunogenic response, TroVax (MVA-5T4) is a viable addition to the treatment of prostate cancer.
Collapse
Affiliation(s)
- Robert J Amato
- Division of Oncology, Department of Internal Medicine, University of Texas Health Science Center at Houston (Medical School), Memorial Hermann Cancer Center at Houston
| | | |
Collapse
|
174
|
Meadors JL, Cui Y, Chen QR, Song YK, Khan J, Merlino G, Tsokos M, Orentas RJ, Mackall CL. Murine rhabdomyosarcoma is immunogenic and responsive to T-cell-based immunotherapy. Pediatr Blood Cancer 2011; 57:921-9. [PMID: 21462302 PMCID: PMC7401311 DOI: 10.1002/pbc.23048] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 01/03/2011] [Indexed: 02/01/2023]
Abstract
BACKGROUND Immunotherapies targeting cellular immunity are currently approved for treatment of melanoma, renal cell carcinoma, and prostate cancer. Studies on the immunogenicity and immune responsiveness of pediatric tumors are limited, therefore, it remains unclear to what extent T-cell-based immunotherapy holds promise for pediatric solid tumors. PROCEDURE A new rhabdomyosarcoma cell line (M3-9-M) was derived from an embryonal rhabdomyosarcoma (ERMS) occurring in a C57BL/6 mouse transgenic for hepatocyte growth factor and heterozygous for mutated p53. Primary tumors and metastases derived from M3-9-M were studied for similarities to human ERMS, and for immunogenicity and immune responsiveness. RESULTS Primary and metastatic tumors develop after orthotopic injection of M3-9-M into immunocompetent C57BL/6 mice, which mirror human ERMS with regard to histology, gene expression, and metastatic behavior. Whole cell vaccination using irradiated M3-9-M cells or M3-9-M-pulsed dendritic cells (DC)-induced tumor-specific T-cell responses that prevent tumor growth following low-dose tumor injection, and slow tumor growth following higher doses. Administration of anti-CD25 moAbs to deplete CD4(+)CD25(+)FOXP3(+) regulatory T cells prior to tumor vaccination enhanced the potency of the ERMS tumor vaccine. Adoptive immunotherapy with M3-9-M primed T cells plus DC-based vaccination resulted in complete eradication of day 10 M3-9-M derived tumors. CONCLUSIONS M3-9-M derived murine ERMS is immunogenic and immunoresponsive; regulatory T cells contribute to immune evasion by murine rhabdomyosarcoma. Adoptive immunotherapy with DC vaccination can eradicate low tumor burdens. Future work will seek to identify the tumor-associated antigens that mediate protective and therapeutic immunity in this model.
Collapse
Affiliation(s)
- Joanna L. Meadors
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Yonghzi Cui
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Qing-Rong Chen
- Oncogenomics Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Young K. Song
- Oncogenomics Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Javed Khan
- Oncogenomics Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Glenn Merlino
- Cancer Modeling Section, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Maria Tsokos
- Laboratory of Pathology, Pediatric Tumor Biology and Ultrastructural Pathology Section, National Cancer Institute, Bethesda, Maryland
| | - Rimas J. Orentas
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Crystal L. Mackall
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
175
|
Liu Y, Komohara Y, Domenick N, Ohno M, Ikeura M, Hamilton RL, Horbinski C, Wang X, Ferrone S, Okada H. Expression of antigen processing and presenting molecules in brain metastasis of breast cancer. Cancer Immunol Immunother 2011; 61:789-801. [PMID: 22065046 DOI: 10.1007/s00262-011-1137-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Accepted: 10/20/2011] [Indexed: 01/10/2023]
Abstract
Defects in human leukocyte antigen class I antigen processing machinery (APM) component expression can have a negative impact on the clinical course of tumors and the response to T cell-based immunotherapy. Since brain metastases of breast cancer are of increasing clinical significance, the APM component expression levels and CD8(+) T cell infiltration patterns were analyzed in primary breast and metastatic brain lesions of breast cancer by immunohistochemistry. Comparison of unpaired 50 primary and 33 brain metastases showed lower expression of β2-microglobulin, transporter associated with antigen processing (TAP) 1, TAP2 and calnexin in the brain lesions. Although no significant differences were found in APM component scores between primary breast and brain lesions in 15 paired cases, primary breast lesions of which patients eventually developed brain metastases showed lower levels of β2-microglobulin, TAP1 and calnexin compared with breast lesions without known brain metastases. The extent of CD8(+) T cell infiltration was significantly higher in the lesions without metastasis compared with the ones with brain metastases, and was positively associated with the expression of TAP1 and calnexin. Furthermore, mouse tumor cells stably transfected with silencing hairpin (sh)RNA for TAP1 demonstrated a decreased susceptibility to cytotoxic T lymphocytes in vitro and enhanced spontaneous brain metastasis in vivo. These data support the functional significance of TAP1 expression in tumor cells. Taken together, our data suggest that patients with low or defective TAP1 or calnexin in primary breast cancers may be at higher risks for developing brain metastasis due to the defects in T cell-based immunosurveillance.
Collapse
Affiliation(s)
- Yan Liu
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Yi H, Guo C, Yu X, Gao P, Qian J, Zuo D, Manjili MH, Fisher PB, Subjeck JR, Wang XY. Targeting the immunoregulator SRA/CD204 potentiates specific dendritic cell vaccine-induced T-cell response and antitumor immunity. Cancer Res 2011; 71:6611-20. [PMID: 21914786 PMCID: PMC3213980 DOI: 10.1158/0008-5472.can-11-1801] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although dendritic cell (DC) vaccines offer promise as cancer immunotherapy, further improvements are needed to amplify their clinical therapeutic efficacy. The pattern recognition scavenger receptor SRA/CD204 attenuates the ability of DCs to activate CD8(+) T-cell responses. Therefore, we examined the impact of SRA/CD204 on antitumor responses generated by DC vaccines and we also evaluated the feasibility of enhancing DC vaccine potency by SRA/CD204 blockade. DCs from SRA/CD204-deficient mice were more immunogenic in generating antitumor responses to B16 melanoma, compared with DCs from wild-type mice. Similarly, siRNA-mediated knockdown of SRA/CD204 by lentiviral vectors improved the ability of wild-type DCs to stimulate the expansion and activation of CD8(+) T cells specific for idealized or established melanoma antigens in mice. Using SRA/CD204-silenced DCs to generate antigen-targeted vaccines, we documented a marked increase in the level of antitumor immunity achieved against established B16 tumors and metastases. This increase was associated with enhanced activation of antigen specific CTLs, greater tumor infiltration by CD8(+) T cells and NK cells, and increased intratumoral ratios of both CD4(+) and CD8(+) T-effector cells to CD4(+)CD25(+) T-regulatory cells. Our studies establish that downregulating SRA/CD204 strongly enhances DC-mediated antitumor immunity. In addition, they provide a rationale to enhance DC vaccine potency through SRA/CD204-targeting approaches that can improve clinical outcomes in cancer treatment.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Combined Modality Therapy
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Drug Screening Assays, Antitumor
- Genetic Therapy
- Genetic Vectors/pharmacology
- Genetic Vectors/therapeutic use
- Immunotherapy, Active
- Immunotherapy, Adoptive
- Lentivirus/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/secondary
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/therapeutic use
- Scavenger Receptors, Class A/antagonists & inhibitors
- Scavenger Receptors, Class A/deficiency
- Scavenger Receptors, Class A/genetics
- Scavenger Receptors, Class A/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- Vaccination
Collapse
Affiliation(s)
- Huanfa Yi
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - Xiaofei Yu
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - Ping Gao
- College of Life Science, Beijing Normal University, Beijing 100875, China
| | - Jie Qian
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - Daming Zuo
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - Masoud H. Manjili
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - Paul B. Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| | - John R. Subjeck
- Department of Cellular Stress Biology, Roswell Park Cancer Institute, Buffalo, NY14263, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
- VCU Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA23298, USA
| |
Collapse
|
177
|
Derouazi M, Wang Y, Marlu R, Epaulard O, Mayol JF, Pasqual N, Le Gouellec A, Polack B, Toussaint B. Optimal epitope composition after antigen screening using a live bacterial delivery vector: application to TRP-2. Bioeng Bugs 2011; 1:51-60. [PMID: 21327126 DOI: 10.4161/bbug.1.1.9482] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 07/09/2009] [Accepted: 07/09/2009] [Indexed: 01/03/2023] Open
Abstract
Immunotherapeutic approaches, based on the generation of tumor-specific cytotoxic T-lymphocytes (CTL), are currently emerging as promising strategies of anti-tumor therapy. The potential use of attenuated bacteria as engineered vectors for vaccine development offers several advantages, including the stimulation of innate immunity. We developed an attenuated live bacterial vector using the type III secretion system (TTSS) of Pseudomonas aeruginosa to deliver in vivo tumor antigens. Using an inducible and rapid expression plasmid, vaccination with several antigens of different length and epitope composition, including TRp-2, gp100 and MUC18, was evaluated against glioma tumor cells. We observed similar CTL immunity and T-cell receptor (TCR) repertoire diversity with the vaccines, TRP2(125-243), TRP2L(125-376) and TRP2S(291-376). However, only immunization with TRP2L(125-376) induced significant anti-tumor immunity. Taken together, our data indicate the importance of the epitopes composition and/or peptide length of these peptides for inducing cytotoxic T-lymphocyte (CTL) mediated immunity. Characteristics that consistently improved anti-tumor immunity include: long peptides with immunodominant and cryptic CD8(+) epitopes, and strong CD4(+) Th epitopes. Our bacterial vector is versatile, easy-to-use and quick to produce. This vector is suitable for rapid screening and evaluation of antigens of varying length and epitope composition.
Collapse
Affiliation(s)
- Madiha Derouazi
- Therex, TIMC-IMAG, CNRS Université Joseph Fourier; La Tronche, France
| | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Kemmler CB, Clambey ET, Kedl RM, Slansky JE. Elevated tumor-associated antigen expression suppresses variant peptide vaccine responses. THE JOURNAL OF IMMUNOLOGY 2011; 187:4431-9. [PMID: 21940675 DOI: 10.4049/jimmunol.1101555] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Variant peptide vaccines are used clinically to expand T cells that cross-react with tumor-associated Ags (TAA). To investigate the effects of elevated endogenous TAA expression on variant peptide-induced responses, we used the GP70 TAA model. Although young BALB/c mice display T cell tolerance to the TAA GP70(423-431) (AH1), expression of GP70 and suppression of AH1-specific responses increases with age. We hypothesized that as TAA expression increases, the AH1 cross-reactivity of variant peptide-elicited T cell responses diminishes. Controlling for immunosenescence, we showed that elevated GP70 expression suppressed AH1 cross-reactive responses elicited by two AH1 peptide variants. A variant that elicited almost exclusively AH1 cross-reactive T cells in young mice elicited few or no T cells in aging mice with Ab-detectable GP70 expression. In contrast, a variant that elicited a less AH1 cross-reactive T cell response in young mice successfully expanded AH1 cross-reactive T cells in all aging mice tested. However, these T cells bound the AH1/MHC complex with a relatively short half-life and responded poorly to ex vivo stimulation with the AH1 peptide. Variant peptide vaccine responses were also suppressed when AH1 peptide is administered tolerogenically to young mice before vaccination. Analyses of variant-specific precursor T cells from naive mice with Ab-detectable GP70 expression determined that these T cells expressed PD-1 and had downregulated IL-7Rα expression, suggesting they were anergic or undergoing deletion. Although variant peptide vaccines were less effective as TAA expression increases, data presented in this article also suggest that complementary immunotherapies may induce the expansion of T cells with functional TAA recognition.
Collapse
Affiliation(s)
- Charles B Kemmler
- Integrated Department of Immunology, School of Medicine, University of Colorado Denver and National Jewish Health, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
179
|
Byrne KT, Turk MJ. New perspectives on the role of vitiligo in immune responses to melanoma. Oncotarget 2011; 2:684-94. [PMID: 21911918 PMCID: PMC3248219 DOI: 10.18632/oncotarget.323] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 09/09/2011] [Indexed: 01/09/2023] Open
Abstract
Melanoma-associated vitiligo is the best-studied example of the linkage between tumor immunity and autoimmunity. Although vitiligo is an independent positive prognostic factor for melanoma patients, the autoimmune destruction of melanocytes was long thought to be merely a side effect of robust anti-tumor immunity. However, new data reveal a key role for vitiligo in supporting T cell responses to melanoma. This research perspective reviews the history of melanoma-associated vitiligo in patients, the experimental studies that form the basis for understanding this relationship, and the unique characteristics of melanoma-specific CD8 T cells found in hosts with vitiligo. We also discuss the implications of our recent findings for the interpretation of patient responses, and the design of next-generation cancer immunotherapies.
Collapse
Affiliation(s)
- Katelyn T Byrne
- Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, NH, USA
| | | |
Collapse
|
180
|
Mace TA, Zhong L, Kilpatrick C, Zynda E, Lee CT, Capitano M, Minderman H, Repasky EA. Differentiation of CD8+ T cells into effector cells is enhanced by physiological range hyperthermia. J Leukoc Biol 2011; 90:951-62. [PMID: 21873456 DOI: 10.1189/jlb.0511229] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In this study, we asked whether exposure to different physiologically relevant temperatures (33°C, 37°C, and 39.5°C) could affect subsequent antigen-specific, activation-related events of naive CD8(+) T cells. We observed that temporary exposure of CD62L(hi)CD44(lo) Pmel-1 CD8(+) cells to 39.5°C prior to their antigen-dependent activation with gp100(25-33) peptide-pulsed C57BL/6 splenocytes resulted in a greater percentage of cells, which eventually differentiated into CD62L(lo)CD44(hi) effector cells compared with cells incubated at 33°C and 37°C. However, the proliferation rate of naive CD8(+) T cells was not affected by mild heating. While exploring these effects further, we observed that mild heating of CD8(+) T cells resulted in the reversible clustering of GM1(+) CD-microdomains in the plasma membrane. This could be attributable to a decrease in line tension in the plasma membrane, as we also observed an increase in membrane fluidity at higher temperatures. Importantly, this same clustering phenomenon was observed in CD8(+) T cells isolated from spleen, LNs, and peripheral blood following mild whole-body heating of mice. Further, we observed that mild heating also resulted in the clustering of TCRβ and the CD8 coreceptor but not CD71R. Finally, we observed an enhanced rate of antigen-specific conjugate formation with APCs following mild heating, which could account for the difference in the extent of differentiation. Overall, these novel findings may help us to further understand the impact of physiologically relevant temperature shifts on the regulation of antigen-specific CD8(+) T cell activation and the subsequent generation of effector cells.
Collapse
Affiliation(s)
- Thomas A Mace
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | | | |
Collapse
|
181
|
Valentino MD, Abdul-Alim CS, Maben ZJ, Skrombolas D, Hensley LL, Kawula TH, Dziejman M, Lord EM, Frelinger JA, Frelinger JG. A broadly applicable approach to T cell epitope identification: application to improving tumor associated epitopes and identifying epitopes in complex pathogens. J Immunol Methods 2011; 373:111-26. [PMID: 21872603 DOI: 10.1016/j.jim.2011.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 08/10/2011] [Accepted: 08/11/2011] [Indexed: 12/22/2022]
Abstract
Epitopes are a hallmark of the antigen specific immune response. The identification and characterization of epitopes is essential for modern immunologic studies, from investigating cellular responses against tumors to understanding host/pathogen interactions especially in the case of bacteria with intracellular residence. Here, we have utilized a novel approach to identify T cell epitopes exploiting the exquisite ability of particulate antigens, in the form of beads, to deliver exogenous antigen to both MHC class I and class II pathways for presentation to T cell hybridomas. In the current study, we coupled this functional assay with two distinct protein expression libraries to develop a methodology for the characterization of T cell epitopes. One set of expression libraries containing single amino acid substitutions in a defined epitope sequence was interrogated to identify epitopes with enhanced T cell stimulation for a MHC class I epitope. The second expression library is comprised of the majority of open reading frames from the intracellular pathogen and potential biowarfare agent, Francisella tularensis. By automating aspects of this technology, we have been able to functionally screen and identify novel T cell epitopes within F. tularensis. We have also expanded upon these studies to generate a novel expression vector that enables immunization of recombinant protein into mice, which has been utilized to facilitate T cell epitope discovery for proteins that are critically linked to Francisella pathogenicity. This methodology should be applicable to a variety of systems and other pathogens.
Collapse
Affiliation(s)
- Michael D Valentino
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Hangalapura BN, Oosterhoff D, de Groot J, Boon L, Tüting T, van den Eertwegh AJ, Gerritsen WR, van Beusechem VW, Pereboev A, Curiel DT, Scheper RJ, de Gruijl TD. Potent antitumor immunity generated by a CD40-targeted adenoviral vaccine. Cancer Res 2011; 71:5827-37. [PMID: 21747119 DOI: 10.1158/0008-5472.can-11-0804] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In situ delivery of tumor-associated antigen (TAA) genes into dendritic cells (DC) has great potential as a generally applicable tumor vaccination approach. Although adenoviruses (Ad) are an attractive vaccine vehicle in this regard, Ad-mediated transduction of DCs is hampered by the lack of expression of the Ad receptor CAR on the DC surface. DC activation also requires interaction of CD40 with its ligand CD40L to generate protective T-cell-mediated tumor immunity. Therefore, to create a strategy to target Ads to DCs in vivo, we constructed a bispecific adaptor molecule with the CAR ectodomain linked to the CD40L extracellular domain via a trimerization motif (CFm40L). By targeting Ad to CD40 with the use of CFm40L, we enhanced both transduction and maturation of cultured bone marrow-derived DCs. Moreover, we improved transduction efficiency of DCs in lymph node and splenic cell suspensions in vitro and in skin and vaccination site-draining lymph nodes in vivo. Furthermore, CD40 targeting improved the induction of specific CD8(+) T cells along with therapeutic efficacy in a mouse model of melanoma. Taken together, our findings support the use of CD40-targeted Ad vectors encoding full-length TAA for in vivo targeting of DCs and high-efficacy induction of antitumor immunity.
Collapse
Affiliation(s)
- Basav N Hangalapura
- Department of Medical Oncology and Pathology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
183
|
Schaer DA, Li Y, Merghoub T, Rizzuto GA, Shemesh A, Cohen AD, Li Y, Avogadri F, Toledo-Crow R, Houghton AN, Wolchok JD. Detection of intra-tumor self antigen recognition during melanoma tumor progression in mice using advanced multimode confocal/two photon microscope. PLoS One 2011; 6:e21214. [PMID: 21731676 PMCID: PMC3120835 DOI: 10.1371/journal.pone.0021214] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 05/23/2011] [Indexed: 11/18/2022] Open
Abstract
Determining how tumor immunity is regulated requires understanding the extent to which the anti-tumor immune response “functions” in vivo without therapeutic intervention. To better understand this question, we developed advanced multimodal reflectance confocal/two photon fluorescence intra-vital imaging techniques to use in combination with traditional ex vivo analysis of tumor specific T cells. By transferring small numbers of melanoma-specific CD8+ T cells (Pmel-1), in an attempt to mimic physiologic conditions, we found that B16 tumor growth alone was sufficient to induce naive Pmel-1 T cell proliferation and acquisition of effector phenotype. Tumor -primed Pmel-1 T cells, are capable of killing target cells in the periphery and secrete IFNγ, but are unable to mediate tumor regression. Within the tumor, Pmel-1 T cells have highly confined mobility, displaying long term interactions with tumor cells. In contrast, adoptively transferred non tumor-specific OT-I T cells show neither confined mobility, nor long term interaction with B16 tumor cells, suggesting that intra-tumor recognition of cognate self antigen by Pmel-1 T cells occurs during tumor growth. Together, these data indicate that lack of anti-tumor efficacy is not solely due to ignorance of self antigen in the tumor microenvironment but rather to active immunosuppressive influences preventing a protective immune response.
Collapse
Affiliation(s)
- David A. Schaer
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Yongbiao Li
- Research Engineering Lab, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Taha Merghoub
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Gabrielle A. Rizzuto
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - Amos Shemesh
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Adam D. Cohen
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Yanyun Li
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Francesca Avogadri
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Ricardo Toledo-Crow
- Research Engineering Lab, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
| | - Alan N. Houghton
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
| | - Jedd D. Wolchok
- Swim Across America Laboratory, Immunology Program, Sloan-Kettering Institute for Cancer Research, New York, New York, United States of America
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
184
|
Zhang Z, Tongchusak S, Mizukami Y, Kang YJ, Ioji T, Touma M, Reinhold B, Keskin DB, Reinherz EL, Sasada T. Induction of anti-tumor cytotoxic T cell responses through PLGA-nanoparticle mediated antigen delivery. Biomaterials 2011; 32:3666-78. [DOI: 10.1016/j.biomaterials.2011.01.067] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 01/26/2011] [Indexed: 12/18/2022]
|
185
|
Wongthida P, Diaz RM, Pulido C, Rommelfanger D, Galivo F, Kaluza K, Kottke T, Thompson J, Melcher A, Vile R. Activating systemic T-cell immunity against self tumor antigens to support oncolytic virotherapy with vesicular stomatitis virus. Hum Gene Ther 2011; 22:1343-53. [PMID: 21366404 DOI: 10.1089/hum.2010.216] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
We have shown that the antitumor activity of vesicular stomatitis virus (VSV) against B16ova tumors in C57BL/6 mice is predominantly due to innate antiviral immune effectors. We have also shown that the innate immune-activating properties of VSV can be harnessed to prime adaptive T-cell responses against a tumor-associated antigen (TAA) if the virus is engineered to express the cDNA of the antigen. Here, we show that the combination of VSV expressing OVA as a model tumor antigen, along with adoptive T-cell therapy targeted against the same antigen, is superior to either treatment alone and induces systemic antitumor activity. In addition, we extend our findings with the OVA model to the therapeutic use of VSV expressing hgp100, a self TAA against which tolerance is well established in C57BL/6 mice. In contrast to VSV-ova, T-cell responses raised by VSV-hgp100 were insufficient to improve therapy against B16ova tumors compared with VSV-GFP alone. However, in combination with adoptive transfer of gp100-specific pmel T cells, intratumoral VSV-hgp100 cured significantly more mice than either virus or T cells alone. Even in an aggressive model of metastatic disease, antitumor therapy was generated at levels similar to those observed in the VSV-ova/OT-I model in which a potently immunogenic, nonself TAA was targeted. Therefore, individual poorly effective virotherapies and T-cell therapies that target self TAA of low immunogenicity, which reflects the situation in patients, can be combined to generate very effective antitumor therapy.
Collapse
|
186
|
Induction of TLR4-dependent CD8+ T cell immunity by murine β-defensin2 fusion protein vaccines. Vaccine 2011; 29:3476-82. [PMID: 21382485 DOI: 10.1016/j.vaccine.2011.02.061] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 01/13/2011] [Accepted: 02/16/2011] [Indexed: 02/02/2023]
Abstract
Our laboratory previously described the strategy of fusing chemokine receptor ligands to antigens in order to generate immunogenic DNA vaccines. In the present study, we produced mouse β-2 defensin (mBD2) fusion proteins using both ovalbumin (OVA) and gp100 as model antigens. Superior cross-presentation by dendritic cells (DC) was observed for mBD2 fused antigens over unfused antigens in vitro. In vivo, we observed significant increases in the expansion of adoptively transferred antigen-specific MHC class I, but not class II-restricted T cells after immunization with mBD2 fused antigen over antigen alone. This enhanced expansion of class I restricted T cells was Toll-like receptor 4 (TLR4) dependent, but CC chemokine receptor 6 (CCR6) independent. Superior tumor resistance was observed for mBD2-fusion protein vaccines, compared to unfused antigen, in both B16-OVA and B16 tumor models. These data suggest that production of mBD2 fusion proteins is feasible and that the vaccines facilitate in vivo expansion of adoptively transferred T cells through a TLR4-dependent mechanism.
Collapse
|
187
|
Dendritic cell-directed lentivector vaccine induces antigen-specific immune responses against murine melanoma. Cancer Gene Ther 2011; 18:370-80. [PMID: 21372855 DOI: 10.1038/cgt.2011.13] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Lentivectors are potential vaccine delivery vehicles because they can efficiently transduce a variety of non-dividing cells, including antigen-presenting cells, and do not cause expression of extra viral proteins. To improve safety while retaining efficiency, a dendritic cell (DC)-specific lentivector was constructed by pseudotyping the vector with an engineered viral glycoprotein derived from Sindbis virus. We assessed the level of anti-tumor immunity conferred by this engineered lentivector encoding the melanoma antigen gp100 in a mouse model. Footpad injection of the engineered lentivectors results in the best antigen-specific immune response as compared with subcutaneous and intraperitoneal injections. A single prime vaccination of the engineered lentivectors can elicit a high frequency (up to 10%) of gp100-specific CD8(+) T cells in peripheral blood 3 weeks after the vaccination and this response will be maintained at around 5% for up to 8 weeks. We found that these engineered lentivectors elicited relatively low levels of anti-vector neutralizing antibody responses. Importantly, direct injection of this engineered lentivector inhibited the growth of aggressive B16 murine melanoma. These data suggest that DC-specific lentivectors can be a novel and alternative vaccine carrier with the potential to deliver effective anti-tumor immunity for cancer immunotherapy.
Collapse
|
188
|
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: 2.1] [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
|
189
|
Côté AL, Zhang P, O’Sullivan JA, Jacobs VL, Clemis CR, Sakaguchi S, Guevara-Patiño JA, Turk MJ. Stimulation of the glucocorticoid-induced TNF receptor family-related receptor on CD8 T cells induces protective and high-avidity T cell responses to tumor-specific antigens. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:275-83. [PMID: 21106849 PMCID: PMC3050990 DOI: 10.4049/jimmunol.1001308] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Treatment of tumor-bearing mice with a stimulatory Ab to glucocorticoid-induced TNFR family-related receptor (GITR) has previously been shown to elicit protective T cell responses against poorly immunogenic tumors. However, the role of GITR stimulation on CD8 T cells and the nature of tumor rejection Ags have yet to be determined. In this study, we show that a stimulatory mAb to GITR (clone DTA-1) acts directly on CD8 T cells, but not on CD4(+)CD25(+) regulatory T (T(reg)) cells, in B16 tumor-bearing mice to induce concomitant immunity against secondary B16 tumors, as well as protective memory following surgical excision of the primary tumor. Melanoma growth itself induced GITR expression on tumor-specific CD8 T cells, providing a mechanism whereby these cells may respond to stimulatory anti-GITR. Unexpectedly, in contrast to T(reg) cell depletion therapy with anti-CD4, GITR stimulation induced very weak CD8 T cell responses to melanocyte differentiation Ags expressed by the tumor, and did not induce autoimmune vitiligo. Accordingly, GITR-stimulated hosts that were primed with B16 melanoma rejected B16, but not the unrelated JBRH melanoma, indicating that tumor rejection Ags are tumor-specific rather than shared. In support of this, we show that GITR stimulation induces CD8 T cell responses to a tumor-specific Ag, and that these responses are of higher functional avidity compared with those induced by T(reg) cell depletion. We conclude that stimulation of GITR on effector CD8 T cells results in high-avidity T cell responses to tumor-specific Ags, thereby inducing potent antitumor immunity in the absence of autoimmunity.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cell Adhesion/immunology
- Cell Line, Tumor
- Epitopes, T-Lymphocyte/immunology
- Female
- Glucocorticoid-Induced TNFR-Related Protein
- Lymphocyte Activation/immunology
- Lymphocyte Depletion
- Male
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Nerve Growth Factor/deficiency
- Receptors, Nerve Growth Factor/immunology
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/deficiency
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Anik L. Côté
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, NH 03756
| | - Peisheng Zhang
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, NH 03756
| | | | - Valerie L. Jacobs
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, NH 03756
| | - Carli R. Clemis
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, NH 03756
| | - Shimon Sakaguchi
- Department of Experimental Pathology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | | | - Mary Jo Turk
- Department of Microbiology and Immunology, Dartmouth Medical School, Norris Cotton Cancer Center, Lebanon, NH 03756
| |
Collapse
|
190
|
Buonaguro L, Petrizzo A, Tornesello ML, Buonaguro FM. Translating tumor antigens into cancer vaccines. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:23-34. [PMID: 21048000 PMCID: PMC3019775 DOI: 10.1128/cvi.00286-10] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Vaccines represent a strategic successful tool used to prevent or contain diseases with high morbidity and/or mortality. However, while vaccines have proven to be effective in combating pathogenic microorganisms, based on the immune recognition of these foreign antigens, vaccines aimed at inducing effective antitumor activity are still unsatisfactory. Nevertheless, the effectiveness of the two licensed cancer-preventive vaccines targeting tumor-associated viral agents (anti-HBV [hepatitis B virus], to prevent HBV-associated hepatocellular carcinoma, and anti-HPV [human papillomavirus], to prevent HPV-associated cervical carcinoma), along with the recent FDA approval of sipuleucel-T (for the therapeutic treatment of prostate cancer), represents a significant advancement in the field of cancer vaccines and a boost for new studies in the field. Specific active immunotherapies based on anticancer vaccines represent, indeed, a field in continuous evolution and expansion. Significant improvements may result from the selection of the appropriate tumor-specific target antigen (to overcome the peripheral immune tolerance) and/or the development of immunization strategies effective at inducing a protective immune response. This review aims to describe the vast spectrum of tumor antigens and strategies to develop cancer vaccines.
Collapse
Affiliation(s)
- Luigi Buonaguro
- Laboratory of Molecular Biology and Viral Oncogenesis & AIDS Reference Center, Istituto Nazionale Tumori Fondazione G. Pascale, Via Mariano Semmola 1, 80131 Naples, Italy.
| | | | | | | |
Collapse
|
191
|
Geng D, Zheng L, Srivastava R, Velasco-Gonzalez C, Riker A, Markovic SN, Davila E. Amplifying TLR-MyD88 signals within tumor-specific T cells enhances antitumor activity to suboptimal levels of weakly immunogenic tumor antigens. Cancer Res 2010; 70:7442-54. [PMID: 20807806 PMCID: PMC3463001 DOI: 10.1158/0008-5472.can-10-0247] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The efficacy of T cell-based immunotherapy to treat cancer patients remains a challenge partly because of the weak activity toward subdominant tumor antigens (TAg) and to tumors expressing suboptimal TAg levels. Recent reports indicate that Toll-like receptor (TLR) stimulation on T cells can lower the activation threshold. In this study, we examined the antitumor activity and survival of TLR2-MyD88-stimulated CD8 T cells derived from melanoma patients and T-cell receptor transgenic pmel mice. TLR2-stimulated pmel CD8 T cells, but not TLR2(-/-)pmel or MyD88(-/-)pmel T cells, responded to significantly lower TAg levels and resulted in increased production of effector molecules and cytotoxicity. Wild-type or MyD88(-/-) mice treated with TLR2 ligand and pmel T cells, but not TLR2(-/-)pmel or MyD88(-/-)pmel T cells, showed tumor regression of an established melanoma tumor. Overexpressing TLR2 in TAg-specific T cells eradicated tumors; four times fewer cells were needed to generate antitumor responses. The enhanced antitumor activity of TLR2-MyD88-stimulated T cells was associated with increased effector function but perhaps more importantly with improved survival of T cells. Activating TLR-MyD88 signals in patient-derived T cells also reduced the activation threshold to several weakly immunogenic TAgs, resulting in increased cytokine production, expansion, and cytotoxicity. These data highlight a previously unappreciated role for activating TLR-MyD88 signals in tumor-reactive T lymphocytes.
Collapse
Affiliation(s)
- Degui Geng
- University of Maryland Greenebaum Cancer Center and Department of Otorhinolaryngology, Baltimore, Maryland, USA
| | - Liqin Zheng
- Louisiana State University Stanley S. Scott Cancer Center, New Orleans, Louisiana, USA
| | - Ratika Srivastava
- University of Maryland Greenebaum Cancer Center and Department of Otorhinolaryngology, Baltimore, Maryland, USA
| | | | - Adam Riker
- Ochsner Cancer Institute, New Orleans, Louisiana, USA
| | - Svetomir N. Markovic
- Division of Hematology/Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Eduardo Davila
- University of Maryland Greenebaum Cancer Center and Department of Otorhinolaryngology, Baltimore, Maryland, USA
| |
Collapse
|
192
|
Zhu X, Fallert-Junecko BA, Fujita M, Ueda R, Kohanbash G, Kastenhuber ER, McDonald HA, Liu Y, Kalinski P, Reinhart TA, Salazar AM, Okada H. Poly-ICLC promotes the infiltration of effector T cells into intracranial gliomas via induction of CXCL10 in IFN-alpha and IFN-gamma dependent manners. Cancer Immunol Immunother 2010; 59:1401-9. [PMID: 20549206 PMCID: PMC2893267 DOI: 10.1007/s00262-010-0876-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 03/17/2010] [Indexed: 01/22/2023]
Abstract
Stimulation of double-stranded (ds)RNA receptors can increase the effectiveness of cancer vaccines, but the underlying mechanisms are not completely elucidated. In this study, we sought to determine critical roles of host IFN-alpha and IFN-gamma pathways in the enhanced therapeutic efficacy mediated by peptide vaccines and polyinosinic-polycytidylic acid [poly(I:C)] stabilized by lysine and carboxymethylcellulose (poly-ICLC) in the murine central nervous system (CNS) GL261 glioma. C57BL/6-background wild type (WT), IFN-alpha receptor-1 (IFN-alphaR1)(-/-) or IFN-gamma(-/-) mice bearing syngeneic CNS GL261 glioma received subcutaneous (s.c.) vaccinations with synthetic peptides encoding CTL epitopes with or without intramuscular (i.m.) injections of poly-ICLC. The combinational treatment induced a robust transcription of CXCL10 in the glioma site. Blockade of CXCL10 with a specific monoclonal antibody (mAb) abrogated the efficient CNS homing of antigen-specific type-1 CTL (Tc1). Both IFN-alphaR(-/-) and IFN-gamma(-/-) hosts failed to up-regulate the CXCL10 mRNA and recruit Tc1 cells to the tumor site, indicating non-redundant roles of type-1 and type-2 IFNs in the effects of poly-ICLC-assisted vaccines. The efficient trafficking of Tc1 also required Tc1-derived IFN-gamma. Our data point to critical roles of the host-IFN-alpha and IFN-gamma pathways in the modulation of CNS glioma microenvironment, and the therapeutic effectiveness of poly-ICLC-assisted glioma vaccines.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/administration & dosage
- Cancer Vaccines
- Carboxymethylcellulose Sodium/administration & dosage
- Carboxymethylcellulose Sodium/analogs & derivatives
- Cell Line, Tumor
- Cell Movement/drug effects
- Cell Movement/genetics
- Cell Movement/immunology
- Central Nervous System Neoplasms/immunology
- Central Nervous System Neoplasms/pathology
- Central Nervous System Neoplasms/therapy
- Chemokine CXCL10/biosynthesis
- Chemokine CXCL10/genetics
- Chemokine CXCL10/immunology
- Epitopes, T-Lymphocyte/chemistry
- Glioma/immunology
- Glioma/pathology
- Glioma/therapy
- Immunization
- Interferon-gamma/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Transplantation
- Peptide Fragments/administration & dosage
- Peptide Fragments/chemistry
- Poly I-C/administration & dosage
- Polylysine/administration & dosage
- Polylysine/analogs & derivatives
- Receptor, Interferon alpha-beta/genetics
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/pathology
Collapse
Affiliation(s)
- Xinmei Zhu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
| | - Beth A. Fallert-Junecko
- Department of Infectious Diseases/Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Mitsugu Fujita
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
| | - Ryo Ueda
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
| | - Gary Kohanbash
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
- Department of Infectious Diseases/Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Edward R. Kastenhuber
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
| | - Heather A. McDonald
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
| | - Yan Liu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Pawel Kalinski
- Department of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Todd A. Reinhart
- Department of Infectious Diseases/Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | | | - Hideho Okada
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213 USA
- Department of Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
- G12a Research Pavilion at Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213 USA
| |
Collapse
|
193
|
Stephan MT, Moon JJ, Um SH, Bershteyn A, Irvine DJ. Therapeutic cell engineering with surface-conjugated synthetic nanoparticles. Nat Med 2010; 16:1035-41. [PMID: 20711198 PMCID: PMC2935928 DOI: 10.1038/nm.2198] [Citation(s) in RCA: 554] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 05/05/2010] [Indexed: 01/10/2023]
Abstract
A major limitation of cell therapies is the rapid decline in viability and function of the transplanted cells. Here we describe a strategy to enhance cell therapy via the conjugation of adjuvant drug-loaded nanoparticles to the surfaces of therapeutic cells. With this method of providing sustained pseudoautocrine stimulation to donor cells, we elicited marked enhancements in tumor elimination in a model of adoptive T cell therapy for cancer. We also increased the in vivo repopulation rate of hematopoietic stem cell grafts with very low doses of adjuvant drugs that were ineffective when given systemically. This approach is a simple and generalizable strategy to augment cytoreagents while minimizing the systemic side effects of adjuvant drugs. In addition, these results suggest therapeutic cells are promising vectors for actively targeted drug delivery.
Collapse
Affiliation(s)
- Matthias T. Stephan
- Department of Material Science and Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - James J. Moon
- Department of Material Science and Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Soong Ho Um
- Department of Material Science and Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of
Technology, Cambridge, MA 02139, USA
| | - Anna Bershteyn
- Department of Material Science and Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
| | - Darrell J. Irvine
- Department of Material Science and Engineering, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of
Technology, Cambridge, MA 02139, USA
- Ragon Institute of MGH, MIT, and Harvard, Boston, MA 02139,
USA
- Howard Hughes Medical Institute, 4000 Jones Bridge Rd., Chevy
Chase, MD 20815, USA
| |
Collapse
|
194
|
Zhang H, Meadows GG. Chronic alcohol consumption enhances myeloid-derived suppressor cells in B16BL6 melanoma-bearing mice. Cancer Immunol Immunother 2010; 59:1151-9. [PMID: 20229084 PMCID: PMC2881944 DOI: 10.1007/s00262-010-0837-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Accepted: 02/18/2010] [Indexed: 12/11/2022]
Abstract
We previously found that chronic alcohol consumption decreases the survival of mice bearing subcutaneous B16BL6 melanoma. The underlying mechanism is still not completely understood. Antitumor T cell immune responses are important to inhibiting tumor progression and extending survival. Therefore, we examined the effects of chronic alcohol consumption on the functionality and regulation of these cells in C57BL/6 mice that chronically consumed 20% (w/v) alcohol and subsequently were inoculated subcutaneously with B16BL6 melanoma cells. Chronic alcohol consumption inhibited melanoma-induced memory T cell expansion and accelerated the decay of interferon (IFN)-gamma producing T cells in the tumor-bearing mice. Foxp3(+)CD4(+)CD25(+) regulatory T cells were not affected; however, the percentage of myeloid-derived suppressor cells (MDSC) was significantly increased in the peripheral blood and spleen. T cell proliferation as determined by carboxyfluorescein succinimidyl ester labeling experiments in vitro was inhibited by alcohol consumption relative to control water-drinking melanoma-bearing mice. Collectively, these data show that chronic alcohol consumption inhibits proliferation of memory T cells, accelerates the decay of IFN-gamma producing CD8(+) T cells, and increases MDSC, all of which could be associated with melanoma progression and reduced survival.
Collapse
MESH Headings
- Alcoholism/complications
- Alcoholism/immunology
- Alcoholism/pathology
- Alcoholism/physiopathology
- Animals
- CD4 Antigens/biosynthesis
- Cell Proliferation/drug effects
- Cells, Cultured
- Ethanol/toxicity
- Forkhead Transcription Factors/biosynthesis
- Immunologic Memory/drug effects
- Immunosuppression Therapy
- Interferon-gamma/metabolism
- Interleukin-2 Receptor alpha Subunit/biosynthesis
- Lymphocyte Activation/drug effects
- Melanoma, Experimental/complications
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/physiopathology
- Mice
- Mice, Inbred C57BL
- Myeloid Progenitor Cells/drug effects
- Myeloid Progenitor Cells/immunology
- Myeloid Progenitor Cells/metabolism
- Myeloid Progenitor Cells/pathology
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Hui Zhang
- Chronic Illness Research Center, Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Box 646534, Pullman, WA 99164-6534 USA
| | - Gary G. Meadows
- Chronic Illness Research Center, Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Box 646534, Pullman, WA 99164-6534 USA
| |
Collapse
|
195
|
Landsberg J, Gaffal E, Cron M, Kohlmeyer J, Renn M, Tüting T. Autochthonous primary and metastatic melanomas in Hgf-Cdk4R24C mice evade T-cell-mediated immune surveillance. Pigment Cell Melanoma Res 2010; 23:649-60. [DOI: 10.1111/j.1755-148x.2010.00744.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
196
|
Tomihari M, Chung JS, Akiyoshi H, Cruz PD, Ariizumi K. DC-HIL/glycoprotein Nmb promotes growth of melanoma in mice by inhibiting the activation of tumor-reactive T cells. Cancer Res 2010; 70:5778-87. [PMID: 20570888 PMCID: PMC2905472 DOI: 10.1158/0008-5472.can-09-2538] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
DC-HIL/glycoprotein nmb (Gpnmb) expressed on antigen-presenting cells attenuates T-cell activation by binding to syndecan-4 (SD-4) on activated T cells. Because DC-HIL/Gpnmb is expressed abundantly by mouse and human melanoma lines, we posited that melanoma-associated DC-HIL/Gpnmb exerts similar inhibitory function on melanoma-reactive T cells. We generated small interfering RNA-transfected B16F10 melanoma cells to completely knock down DC-HIL/Gpnmb expression, with no alteration in cell morphology, melanin synthesis, or MHC class I expression. This knockdown had no effect on B16F10 proliferation in vitro or entry into the cell cycle following growth stimulation, but it markedly reduced the growth of these cells in vivo following their s.c. injection into syngeneic immunocompetent (but not immunodeficient) mice. This reduction in tumor growth was due most likely to an augmented capacity of DC-HIL-knocked down B16F10 cells (compared with controls) to activate melanoma-reactive T cells as documented in vitro and in mice. Whereas DC-HIL knockdown had no effect on susceptibility of melanoma to killing by cytotoxic T cells, blocking SD-4 function enhanced the reactivity of CD8(+) T cells to melanoma-associated antigens on parental B16F10 cells. Using an assay examining the spread to the lung following i.v. injection, DC-HIL-knocked down cells produced lung foci at similar numbers compared with that produced by control cells, but the size of the former foci was significantly smaller than the latter. We conclude that DC-HIL/Gpnmb confers upon melanoma the ability to downregulate the activation of melanoma-reactive T cells, thereby allowing melanoma to evade immunologic recognition and destruction. As such, the DC-HIL/SD-4 pathway is a potentially useful target for antimelanoma immunotherapy.
Collapse
Affiliation(s)
- Mizuki Tomihari
- Department of Dermatology, The University of Texas Southwestern Medical Center and Dermatology Section (Medical Service), Dallas Veterans Affairs Medical Center, Dallas, TX
- Department of Clinical Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Jin-Sung Chung
- Department of Dermatology, The University of Texas Southwestern Medical Center and Dermatology Section (Medical Service), Dallas Veterans Affairs Medical Center, Dallas, TX
| | - Hideo Akiyoshi
- Department of Dermatology, The University of Texas Southwestern Medical Center and Dermatology Section (Medical Service), Dallas Veterans Affairs Medical Center, Dallas, TX
- Laboratory of Advanced Diagnosis and Treatment, Department of Veterinary Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan
| | - Ponciano D. Cruz
- Department of Dermatology, The University of Texas Southwestern Medical Center and Dermatology Section (Medical Service), Dallas Veterans Affairs Medical Center, Dallas, TX
| | - Kiyoshi Ariizumi
- Department of Dermatology, The University of Texas Southwestern Medical Center and Dermatology Section (Medical Service), Dallas Veterans Affairs Medical Center, Dallas, TX
| |
Collapse
|
197
|
Fujita M, Scheurer ME, Decker SA, McDonald HA, Kohanbash G, Kastenhuber ER, Kato H, Bondy ML, Ohlfest JR, Okada H. Role of type 1 IFNs in antiglioma immunosurveillance--using mouse studies to guide examination of novel prognostic markers in humans. Clin Cancer Res 2010; 16:3409-19. [PMID: 20472682 PMCID: PMC2896455 DOI: 10.1158/1078-0432.ccr-10-0644] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We hypothesized that the type 1 IFNs would play a pivotal role in antiglioma immunosurveillance through promotion of type 1 adaptive immunity and suppression of immunoregulatory cells. EXPERIMENTAL DESIGN We induced de novo gliomas in Ifnar1(-/-) (deficient for type 1 IFN receptors) or wild-type mice by intracerebroventricuar transfection of NRas and a short hairpin RNA against P53 using the Sleeping Beauty transposon system. We analyzed the survival of 587 glioma patients for single nucleotide polymorphisms (SNP) in type 1 IFN-related genes. RESULTS Ifnar1(-/-) mice exhibited accelerated tumor growth and death. Analyses of brain tumor-infiltrating lymphocytes in Ifnar1(-/-) mice revealed an increase of cells positive for CD11b(+)Ly6G(+) and CD4(+)FoxP3(+), which represent myeloid-derived suppressor cells and regulatory T cells, respectively, but a decrease of CD8(+) cytotoxic T lymphocytes (CTLs) compared with wild-type mice. Ifnar1(-/-) mouse-derived glioma tissues exhibited a decrease in mRNA for the CTL-attracting chemokine Cxcl10, but an increase of Ccl2 and Ccl22, both of which are known to attract immunoregulatory cell populations. Dendritic cells generated from the bone marrow of Ifnar1(-/-) mice failed to function as effective antigen-presenting cells. Moreover, depletion of Ly6G(+) cells prolonged the survival of mice with developing gliomas. Human epidemiologic studies revealed that SNPs in IFNAR1 and IFNA8 are associated with significantly altered overall survival of patients with WHO grade 2 to 3 gliomas. CONCLUSIONS The novel Sleeping Beauty-induced murine glioma model led us to discover a pivotal role for the type 1 IFN pathway in antiglioma immunosurveillance and relevant human SNPs that may represent novel prognostic markers.
Collapse
Affiliation(s)
- Mitsugu Fujita
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Michael E. Scheurer
- Department of Pediatrics and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Stacy A. Decker
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | - Heather A. McDonald
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Gary Kohanbash
- Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA 15213
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Edward R. Kastenhuber
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Hisashi Kato
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| | - Melissa L. Bondy
- Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030
| | - John R. Ohlfest
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | - Hideho Okada
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213
| |
Collapse
|
198
|
Frasca L, Stonier SW, Overwijk WW, Schluns KS. Differential mechanisms of memory CD8 T cell maintenance by individual myeloid cell types. J Leukoc Biol 2010; 88:69-78. [PMID: 20354106 PMCID: PMC2892527 DOI: 10.1189/jlb.1209816] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Revised: 03/02/2010] [Accepted: 03/05/2010] [Indexed: 12/29/2022] Open
Abstract
This study tested the hypothesis that individual myeloid subsets have a differential ability to maintain memory CD8 T cells via IL-15. Although DCs support IL-15-mediated homeostasis of memory CD8 T cells in vivo, whether various DC subsets and other myeloid cells similarly mediate homeostasis is unknown. Therefore, we studied the ability of different myeloid cells to maintain memory CD8 T cells in vitro. Using an in vitro cocoulture system that recapitulated known roles of DCs and IL-15 on memory CD8 T cells, all in vitro-derived or ex vivo-isolated DCs maintained CD8 T cells better than rIL-15 alone, and FLT-3L-DCs are the most efficient compared with GM-DCs, BM-derived macrophages, or freshly isolated DCs. Although FLT-3L-DCs were the least effective at inducing CD8 T cell proliferation, FLT-3L-DCs promoted better CD8 T cell survival and increased Bcl-2 and MCL-2 expression in CD8 T cells. T cell maintenance correlated only partially with DC expression of IL-15Ralpha and IL-15, suggesting that DCs provided additional support signals. Indeed, in the absence of IL-15 signals, CD70/CD27 further supported CD8 T cell maintenance. IFN-alpha enhanced CD70 expression by DCs, resulting in increased proliferation of CD8 T cells. Overall, this study supports our hypothesis by demonstrating that specific DC subtypes had a greater capacity to support memory CD8 T cell maintenance and did so through different mechanisms. Furthermore, this study shows that IL-15 trans-presentation can work in conjunction with other signals, such as CD70/CD27 interactions, to mediate CD8 T cell homeostasis efficiently.
Collapse
Affiliation(s)
- Loredana Frasca
- Department of Immunology, University of Texas M D Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
199
|
Bridle BW, Stephenson KB, Boudreau JE, Koshy S, Kazdhan N, Pullenayegum E, Brunellière J, Bramson JL, Lichty BD, Wan Y. Potentiating cancer immunotherapy using an oncolytic virus. Mol Ther 2010; 18:1430-9. [PMID: 20551919 DOI: 10.1038/mt.2010.98] [Citation(s) in RCA: 126] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Oncolytic viruses (OVs) are highly immunogenic and this limits their use in immune-competent hosts. Although immunosuppression may improve viral oncolysis, this gain is likely achieved at the cost of antitumoral immunity. We have developed a strategy wherein the immune response against the OV leads to enhanced therapeutic outcomes. We demonstrate that immunization with an adenoviral (Ad) vaccine before treatment with an oncolytic vesicular stomatitis virus (VSV) expressing the same tumor antigen (Ag) leads to significantly enhanced antitumoral immunity. Intratumoral replication of VSV was minimally attenuated in Ad-immunized hosts but extending the interval between treatments reduced the attenuating effect and further increased antitumoral immunity. More importantly, our combination approach shifted the immune response from viral Ags to tumor Ags and further reduced OV replication in normal tissues, leading to enhancements in both efficacy and safety. These studies also highlight the benefits of using a replicating, OV to boost a pre-existing antitumoral immune response as this approach generated larger responses versus tumor Ag in tumor-bearing hosts than could be achieved in tumor-free hosts. This strategy should be applicable to other vector combinations, tumor Ags, and tumor targets.
Collapse
Affiliation(s)
- Byram W Bridle
- Centre for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Xie J, Xiong L, Tao X, Li X, Su Y, Hou X, Shi H. Antitumor effects of murine bone marrow-derived dendritic cells infected with xenogeneic livin alpha recombinant adenoviral vectors against Lewis lung carcinoma. Lung Cancer 2010; 68:338-345. [PMID: 19671483 DOI: 10.1016/j.lungcan.2009.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/27/2009] [Accepted: 07/03/2009] [Indexed: 11/27/2022]
Abstract
Transduction with recombinant, replication-defective adenoviral (rAd) vectors encoding a transgene is an efficient method for gene transfer into dendritic cells (DCs). Livin is a member of the inhibitor of apoptosis protein family. Lung cancer and many other tumors express livin at high levels; whereas, normal fully differentiated cells generally do not. Therefore, livin represents a tumor-specific target for cancer vaccine therapy. Self proteins like livin may not stimulate potent antitumor immune responses due to central immunologic tolerance. Small variations in protein sequence that may exist between homologous proteins of different species can break tolerance to the native antigen. To study immunogenicity of a xenogeneic livin protein, we constructed an recombinant adenoviral vectors containing the human livin alpha genes (rAd-hlivin alpha) and vaccinated C57BL/6 mice with mouse bone marrow dendritic cells (BMDCs) transfected with rAd-hlivin alpha gave rise to potent livin-specific cytotoxic T lymphocyte (CTL) capable of lysing Lewis lung carcinoma (LLC) cells. Moreover, vaccination of mice with rAd-hlivin alpha-transduced DCs (rAd-hlivin alpha DCs) induced a potent protective and therapeutic anti-tumor immunity to LLC in a subcutaneous model along with prolonged survival compared to mice vaccinated with control recombinant adenovirus-transduced DCs(rAd-c DCs) or DCs alone. Therefore, xenogeneic differences between human and murine sequences might be exploited to develop immunogenic tumor vaccines.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Adenoviridae/genetics
- Animals
- Bone Marrow/pathology
- Cancer Vaccines
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/therapy
- Cloning, Molecular
- Cytotoxicity, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Dendritic Cells/virology
- Female
- Genetic Vectors
- Humans
- Inhibitor of Apoptosis Proteins/genetics
- Inhibitor of Apoptosis Proteins/immunology
- Inhibitor of Apoptosis Proteins/metabolism
- Mice
- Mice, Inbred C57BL
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasm Proteins/metabolism
- Neoplasm Transplantation
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/pathology
- Transduction, Genetic
- Transgenes/genetics
Collapse
Affiliation(s)
- Junping Xie
- Department of Respiratory Disease, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | | | | | | | | | | | | |
Collapse
|