1
|
Ninmer EK, Xu F, Slingluff CL. The Landmark Series: Cancer Vaccines for Solid Tumors. Ann Surg Oncol 2025; 32:1443-1452. [PMID: 39704984 PMCID: PMC11811251 DOI: 10.1245/s10434-024-16712-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024]
Abstract
Immunotherapy has become an integral part of the treatment for solid tumors. Cancer vaccines represent a potentially powerful class of immunotherapeutic agents to drive antitumor immunity. Cancer vaccine development involves selecting immunogenic target antigens expressed by tumor cells that can be effectively delivered for uptake by antigen-presenting cells to generate a robust adaptive immune response against tumor. While numerous cancer vaccines have been shown to produce antigen-specific immune responses, translating promising results of immunogenicity from early-phase trials into durable clinical benefit in larger randomized trials has remained elusive. Recent findings support new enthusiasm for several cancer vaccine approaches for solid tumors. This review will discuss landmark historic clinical trials in cancer vaccine development and strategies to optimize cancer vaccines to achieve improved clinical efficacy.
Collapse
Affiliation(s)
- Emily K Ninmer
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Feifan Xu
- Department of Pathology, University of Virginia, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, Charlottesville, VA, USA.
- School of Medicine, Cancer Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
2
|
Willemsen M, Bulgarelli J, Chauhan S, Lereim R, Angeli D, Grisendi G, Krebbers G, Davidson I, Kyte J, Guidoboni M, Luiten R, Bakker W. Changes in AXL and/or MITF melanoma subpopulations in patients receiving immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:101009. [PMID: 39697983 PMCID: PMC11652950 DOI: 10.1016/j.iotech.2024.101009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Background Tumor heterogeneity is a hurdle to effective therapy, as illustrated by the 'mixed responses' frequently seen in immunotherapy-treated patients. Previously, AXL+ tumor cells were identified to be highly resistant to targeted therapy, whereas more differentiated MITF+ tumor cells do respond to RAF and MEK inhibitors. Patients and methods In this study, we analyzed tumor heterogeneity and explored the presence of the previously described AXL+ or MITF+ melanoma subpopulations in metastatic tissues by NanoString gene expression analysis, single-cell RNA sequencing and in situ multiplex immunofluorescence. Furthermore, we analyzed how these subpopulations correlate with immunological pressure and response to immunotherapy by immunomodulating antibodies or autologous tumor lysate-loaded dendritic cell vaccination. Results Our data demonstrate large interpatient variability and variable therapy-induced changes independent of the type of therapy. We identify the presence of previously described AXL+ and MITF+ subpopulations in metastatic tissues both at the mRNA level and in situ at the protein level, and demonstrate that MITF+ melanoma cells are significantly decreased upon immunotherapy, while AXL+ melanoma cell numbers are stable. MITF+ tumor cells showed the most significant inverse correlation with CD8+ T cells. Our patient cohort also shows that immunotherapy-induced changes in the abundance of AXL+ or MITF+ tumor cells did not correlate with improved survival. Conclusions Overall, this study suggests that more differentiated MITF+ tumors are efficiently targeted by immunotherapy, while AXL+ tumor cells may be more resistant, analogous to their response to targeted therapy.
Collapse
Affiliation(s)
- M. Willemsen
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - J. Bulgarelli
- Immunotherapy Cell Therapy and Biobank (ITCB) Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - S.K. Chauhan
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - R.R. Lereim
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - D. Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - G. Grisendi
- Laboratory of Cellular Therapy, Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - G. Krebbers
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - I. Davidson
- Department of Functional Genomics and Cancer, IGBMC, CNRS/INSERM, Illkirch, France
| | - J.A. Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway
| | - M. Guidoboni
- Department of Oncology, Ferrara University Hospital, University of Ferrara, Ferrara, Italy
| | - R.M. Luiten
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - W.J. Bakker
- Department of Dermatology and Netherlands Institute for Pigment Disorders, Amsterdam University Medical Centers, Location AMC, University of Amsterdam, Cancer Center Amsterdam, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Abstract
Gene therapy makes it possible to engineer chimeric antigen receptors (CARs) to create T cells that target specific diseases. However, current approaches require elaborate and expensive protocols to manufacture engineered T cells ex vivo, putting this therapy beyond the reach of many patients who might benefit. A solution could be to program T cells in vivo. Here, we evaluate the clinical need for in situ CAR T cell programming, compare competing technologies, review current progress, and provide a perspective on the long-term impact of this emerging and rapidly flourishing biotechnology field.
Collapse
Affiliation(s)
- Neha N Parayath
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Matthias T Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.,Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, USA;
| |
Collapse
|
4
|
Desai R, Coxon AT, Dunn GP. Therapeutic applications of the cancer immunoediting hypothesis. Semin Cancer Biol 2021; 78:63-77. [PMID: 33711414 DOI: 10.1016/j.semcancer.2021.03.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/15/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022]
Abstract
Since the late 19th century, the immune system has increasingly garnered interest as a novel avenue for cancer therapy, particularly given scientific breakthroughs in recent decades delineating the fundamental role of the immune system in tumorigenesis. The immunoediting hypothesis has articulated this role, describing three phases of the tumor-immune system interaction: Elimination, Equilibrium, and Escape wherein tumors progress from active immunologic surveillance and destruction through dynamic immunologic stasis to unfettered growth. The primary goals of immunotherapy are to restrict and revert progression through these phases, thereby improving the immune system's ability to control tumor growth. In this review, we detail the development and foundation of the cancer immunoediting hypothesis and apply this hypothesis to the dynamic immunotherapy field that includes checkpoint blockade, vaccine therapy, and adoptive cell transfer.
Collapse
Affiliation(s)
- Rupen Desai
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew T Coxon
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Gavin P Dunn
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA; Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
5
|
Mocellin S, Panelli M, Wang E, Rossi CR, Marincola FM. Tumor Microenvironment: What have we Learned Studying the Immune Response in this Puzzling Battlefield? TUMORI JOURNAL 2018; 88:437-44. [PMID: 12597134 DOI: 10.1177/030089160208800601] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recent developments hallmark the progress in the understanding of tumor immunology and related therapeutic strategies. The administration of interleukin-2 (IL-2) to patients with cancer has shown that immune manipulation can mediate the regression of established cancers. The identification of the genes encoding cancer antigens and the development of means for effectively immunizing against these antigens has opened new avenues for the development of active immunization of patients with cancer. However, an efficient immune response against tumor comprises an intricate molecular network still poorly understood. Only when the code governing immune responsiveness of cancer will be deciphered, new therapeutic strategies could be designed to fit biologically defined mechanisms of immune rejection of cancer. In this review, we propose that the mechanisms regulating tumor rejection in response to vaccination will be more efficiently identified by following the evolution of treatment induced events within the tumor microenvironment taking advantage of recently developed technological tools. As a model, we will discuss the observed immune response to tumor antigen -specific immunization and its relationship with the systemic administration of IL-2.
Collapse
Affiliation(s)
- Simone Mocellin
- Immunnogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
6
|
Wirth TC, Kühnel F. Neoantigen Targeting-Dawn of a New Era in Cancer Immunotherapy? Front Immunol 2017; 8:1848. [PMID: 29312332 PMCID: PMC5742119 DOI: 10.3389/fimmu.2017.01848] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/06/2017] [Indexed: 12/30/2022] Open
Abstract
During their development and progression tumors acquire numerous mutations that, when translated into proteins give rise to neoantigens that can be recognized by T cells. Initially, neoantigens were not recognized as preferred targets for cancer immunotherapy due to their enormous diversity and the therefore limited options to develop “one fits all” pharmacologic solutions. In recent years, the experience obtained in clinical trials demonstrating a predictive role of neoantigens in checkpoint inhibition has changed our view on the clinical potential of neoantigens in cancer immunotherapy. Technological advances such as sequencing of whole cancer genomes, the development of reliable algorithms for epitope prediction, and an increasing number of immunotherapeutic options now facilitate the development of personalized tumor therapies directly targeting a patient’s neoantigenic burden. Preclinical studies in mice that support the excellent therapeutic potential of neoantigen-directed immunotherapies have provided blueprints on how this methodology can be translated into clinical applications in humans. Consistently, very recent clinical studies on personalized vaccinations targeting in silico predicted neoepitopes shed a first light on the therapeutic potential of personalized, neoantigen-directed immunotherapies. In our review, we discuss the various subtypes of tumor antigens with a focus on neoantigens and their potential in cancer immunotherapy. We will describe the current methods and techniques of detection as well as the structural requirements for neoantigens that are needed for their recognition by T cells and for tumor destruction. To assess the clinical potential of neoantigens, we will discuss their occurrence and functional relevance in spontaneous and hereditary cancers and their prognostic and predictive value. We will present in detail the existing immunotherapeutic options that exploit the neoantigen burden of tumors encompassing both preclinical efforts that provided convincing technological proof-of-concept and the current clinical studies confirming the potential of neoantigen-directed immunotherapies.
Collapse
Affiliation(s)
- Thomas C Wirth
- Clinic for Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| | - Florian Kühnel
- Clinic for Gastroenterology, Hepatology and Endocrinology, Medical School Hannover, Hannover, Germany
| |
Collapse
|
7
|
Song DG, Ye Q, Poussin M, Chacon JA, Figini M, Powell DJ. Effective adoptive immunotherapy of triple-negative breast cancer by folate receptor-alpha redirected CAR T cells is influenced by surface antigen expression level. J Hematol Oncol 2016; 9:56. [PMID: 27439908 PMCID: PMC4955216 DOI: 10.1186/s13045-016-0285-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/07/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The poor prognosis and the limited efficacy of targeted therapy in patients with triple-negative breast cancer (TNBC) have raised the need for alternative therapies. Recent studies have demonstrated that folate receptor-alpha (FRα) may represent an ideal tumor-associated marker for immunotherapy for TNBC. METHODS The aim of the present study was to apply a chimeric antigen receptor (CAR) approach for the targeting of FRα expressed on TNBC cells and evaluate the antitumor activity of CAR-engineered T cells in vitro and in vivo. RESULTS We found that human T cells expressing a FRα-specific CAR were potent and specific killers of TNBC cells that express moderate levels of FRα in vitro and significantly inhibited tumor outgrowth following infusion into immunodeficient mice bearing an MDA-MB-231 tumor xenograft. However, the antitumor activity of the FRα CAR T cells was modest when compared to the same CAR T cells applied in an ovarian tumor xenograft model where FRα expression is more abundant. Notably, FRα CAR T cells induced superior tumor regression in vivo against MDA-MB-231 that was engineered for overexpression of FRα. CONCLUSIONS Taken together, our results show that FRα CAR T cells can mediate antitumor activity against established TNBC tumor, particularly when FRα is expressed at higher levels. These results have significant implications for the pre-selection of patients with high antigen expression levels when utilizing CAR-based adoptive T cell therapies of cancer in future clinical trials.
Collapse
Affiliation(s)
- De-Gang Song
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Rm 8-103 Smilow CTR, Philadelphia, PA, 19104, USA
- Current address: Intrexon Corporation, 20358 Seneca Meadows Pkwy, Germantown, MD, 20876, USA
| | - Qunrui Ye
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Rm 8-103 Smilow CTR, Philadelphia, PA, 19104, USA
| | - Mathilde Poussin
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Rm 8-103 Smilow CTR, Philadelphia, PA, 19104, USA
| | - Jessica A Chacon
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Rm 8-103 Smilow CTR, Philadelphia, PA, 19104, USA
| | - Mariangela Figini
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Daniel J Powell
- Ovarian Cancer Research Center, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Rm 8-103 Smilow CTR, Philadelphia, PA, 19104, USA.
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, 3400 Civic Center Blvd, Rm 8-103 Smilow CTR, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Abstract
The immune system is an incredibly complex biological network that plays a significant role in almost all disease pathogenesis. With an increased understanding of how this vital system operates, there has been a great emphasis on leveraging, manipulating, and/or supplementing endogenous immunity to better prevent or treat different disease states. More recently, the advent of nanotechnology has ushered in a plethora of new nanoparticle-based platforms that can be used to improve existing immunomodulation modalities. As the ability to engineer at the nanoscale becomes increasingly sophisticated, nanoparticles can be finely tuned to effect the desired immune responses, leading to exciting new avenues for addressing pressing issues in public health. In this review, we give an overview of the different areas in which nanoparticle technology has been applied toward modulating the immune system and highlight the recent advances within each.
Collapse
Affiliation(s)
- Ronnie H Fang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093;
| | - Liangfang Zhang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093;
| |
Collapse
|
9
|
Zah E, Lin MY, Silva-Benedict A, Jensen MC, Chen YY. T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells. Cancer Immunol Res 2016; 4:498-508. [PMID: 27059623 DOI: 10.1158/2326-6066.cir-15-0231] [Citation(s) in RCA: 446] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/06/2016] [Indexed: 01/29/2023]
Abstract
The adoptive transfer of T cells expressing anti-CD19 chimeric antigen receptors (CARs) has shown remarkable curative potential against advanced B-cell malignancies, but multiple trials have also reported patient relapses due to the emergence of CD19-negative leukemic cells. Here, we report the design and optimization of single-chain, bispecific CARs that trigger robust cytotoxicity against target cells expressing either CD19 or CD20, two clinically validated targets for B-cell malignancies. We determined the structural parameters required for efficient dual-antigen recognition, and we demonstrate that optimized bispecific CARs can control both wild-type B-cell lymphoma and CD19(-) mutants with equal efficiency in vivo To our knowledge, this is the first bispecific CAR capable of preventing antigen escape by performing true OR-gate signal computation on a clinically relevant pair of tumor-associated antigens. The CD19-OR-CD20 CAR is fully compatible with existing T-cell manufacturing procedures and implementable by current clinical protocols. These results present an effective solution to the challenge of antigen escape in CD19 CAR T-cell therapy, and they highlight the utility of structure-based rational design in the development of receptors with higher-level complexity. Cancer Immunol Res; 4(6); 498-508. ©2016 AACR
Collapse
Affiliation(s)
- Eugenia Zah
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California
| | - Meng-Yin Lin
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California
| | - Anne Silva-Benedict
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington. Department of Oncology and Hematology, St. Luke's Regional Cancer Center, Duluth, Minnesota
| | - Michael C Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington. Division of Pediatric Hematology-Oncology, University of Washington School of Medicine, Seattle, Washington. Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
10
|
Becht E, Giraldo NA, Germain C, de Reyniès A, Laurent-Puig P, Zucman-Rossi J, Dieu-Nosjean MC, Sautès-Fridman C, Fridman WH. Immune Contexture, Immunoscore, and Malignant Cell Molecular Subgroups for Prognostic and Theranostic Classifications of Cancers. Adv Immunol 2016; 130:95-190. [DOI: 10.1016/bs.ai.2015.12.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
11
|
Fang RH, Kroll AV, Zhang L. Nanoparticle-Based Manipulation of Antigen-Presenting Cells for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:5483-96. [PMID: 26331993 PMCID: PMC4641138 DOI: 10.1002/smll.201501284] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/20/2015] [Indexed: 05/18/2023]
Abstract
Immunotherapeutic approaches for treating cancer overall have been receiving a considerable amount of interest due to the recent approval of several clinical formulations. Among the different modalities, anticancer vaccination acts by training the body to endogenously generate a response against tumor cells. However, despite the large amount of work that has gone into the development of such vaccines, the near absence of clinically approved formulations highlights the many challenges facing those working in the field. The generation of potent endogenous anticancer responses poses unique challenges due to the similarity between cancer cells and normal, healthy cells. As researchers continue to tackle the limited efficacy of vaccine formulations, fresh and novel approaches are being sought after to address many of the underlying problems. Here the application of nanoparticle technology towards the development of anticancer vaccines is discussed. Specifically, there is a focus on the benefits of using such strategies to manipulate antigen presenting cells (APCs), which are essential to the vaccination process, and how nanoparticle-based platforms can be rationally engineered to elicit appropriate downstream immune responses.
Collapse
Affiliation(s)
- Ronnie H. Fang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Ashley V. Kroll
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
12
|
Ophir E, Bobisse S, Coukos G, Harari A, Kandalaft LE. Personalized approaches to active immunotherapy in cancer. Biochim Biophys Acta Rev Cancer 2015; 1865:72-82. [PMID: 26241169 DOI: 10.1016/j.bbcan.2015.07.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 07/14/2015] [Accepted: 07/27/2015] [Indexed: 11/28/2022]
Abstract
Immunotherapy is emerging as a promising anti-cancer curative modality. However, in contrast to recent advances obtained employing checkpoint blockade agents and T cell therapies, clinical efficacy of therapeutic cancer vaccines is still limited. Most vaccination attempts in the clinic represent "off-the shelf" approaches since they target common "self" tumor antigens, shared among different patients. In contrast, personalized approaches of vaccination are tailor-made for each patient and in spite being laborious, hold great potential. Recent technical advancement enabled the first steps in the clinic of personalized vaccines that target patient-specific mutated neo-antigens. Such vaccines could induce enhanced tumor-specific immune response since neo-antigens are mutation-derived antigens that can be recognized by high affinity T cells, not limited by central tolerance. Alternatively, the use of personalized vaccines based on whole autologous tumor cells, overcome the need for the identification of specific tumor antigens. Whole autologous tumor cells could be administered alone, pulsed on dendritic cells as lysate, DNA, RNA or delivered to dendritic cells in-vivo through encapsulation in nanoparticle vehicles. Such vaccines may provide a source for the full repertoire of the patient-specific tumor antigens, including its private neo-antigens. Furthermore, combining next-generation personalized vaccination with other immunotherapy modalities might be the key for achieving significant therapeutic outcome.
Collapse
Affiliation(s)
- Eran Ophir
- Ludwig Center for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Sara Bobisse
- Ludwig Center for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Center for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland; Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexandre Harari
- Ludwig Center for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland; Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland
| | - Lana E Kandalaft
- Ludwig Center for Cancer Research at the University of Lausanne, Department of Oncology, University Hospital of Lausanne, Lausanne, Switzerland; Center of Experimental Therapeutics, Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Lausanne, Switzerland; Ovarian Cancer Research Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
13
|
Identification of DRG-1 As a Melanoma-Associated Antigen Recognized by CD4+ Th1 Cells. PLoS One 2015; 10:e0124094. [PMID: 25993655 PMCID: PMC4439028 DOI: 10.1371/journal.pone.0124094] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/02/2015] [Indexed: 01/08/2023] Open
Abstract
Immunotherapy has emerged as a promising strategy for the treatment of metastatic melanoma. Clinical studies have demonstrated the feasibility of cancer immunotherapy using tumor antigens recognized by CD8+ T cells. However, the overall immune responses induced by these antigens are too weak and transient to induce tumor regression in the majority of patients who received immunization. A growing body of evidence suggests that CD4+ T helper (Th) cells play an important role in antitumor immunity. Therefore, the identification of MHC class II-restricted tumor antigens capable of stimulating CD4+ T cells may provide opportunities for developing effective cancer vaccines. To this end, we describe the identification of developmentally regulated GTP-binding protein 1 (DRG-1) as a melanoma-associated antigen recognized by HLA-DR11-restricted CD4+ Th1 cells. Epitope mapping analysis showed that the DRG1248-268 epitope of DRG-1 was required for T cell recognition. Reverse transcription-polymerase chain reaction revealed that DRG-1 was highly expressed in melanoma cell lines but not in normal tissues. DRG-1 knockdown by lentiviral-based shRNA suppressed melanoma cell proliferation and soft agar colony formation. Taken together, these data suggest that DRG-1 plays an important role in melanoma cell growth and transformation, indicating that DRG1 may represent a novel target for CD4+ T cell-mediated immunotherapy in melanoma.
Collapse
|
14
|
Joshi VB, Geary SM, Gross BP, Wongrakpanich A, Norian LA, Salem AK. Tumor lysate-loaded biodegradable microparticles as cancer vaccines. Expert Rev Vaccines 2014; 13:9-15. [PMID: 24219096 DOI: 10.1586/14760584.2014.851606] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cancer vaccines that use tumor lysate (TL) as a source of tumor-associated antigens (TAAs) have significant potential for generating therapeutic anti-tumor immune responses. Vaccines encompassing TL bypass the limitations of single antigen vaccines by simultaneously stimulating immunity against multiple TAAs, thereby broadening the repertoire of TAA-specific T-cell clones available for activation. Administration of TL in particulate form, such as when encapsulated in biodegradable microparticles, increases its immunostimulatory capacity and produces more robust immune responses than when TL is given in soluble form. These effects can be further enhanced by co-administering TL with adjuvants. A number of recent studies using polymeric microparticle delivery of TL, with or without adjuvants, have produced promising results in preclinical studies. In this review, we will discuss current experimental approaches involving TL being pursued in the oncoimmunology field, and comment on strategies such as combining specific chemotherapeutic agents with TL microparticle delivery that may eventually lead to improved survival outcomes for cancer patients.
Collapse
Affiliation(s)
- Vijaya B Joshi
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | |
Collapse
|
15
|
Rodic S, Mihalcioiu C, Saleh RR. Detection methods of circulating tumor cells in cutaneous melanoma: a systematic review. Crit Rev Oncol Hematol 2014; 91:74-92. [PMID: 24530125 DOI: 10.1016/j.critrevonc.2014.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2013] [Revised: 01/06/2014] [Accepted: 01/14/2014] [Indexed: 12/25/2022] Open
Abstract
The vast majority of melanoma-related deaths are due to disseminated malignancy. Many treated patients who are clinically disease-free will go on to relapse. Therefore, new prognostic tools must be developed to better assess metastatic potential and assist in patient management. Circulating tumor cells are a widely studied metastatic biomarker with promising prognostic utility, as the shedding of cells from the primary tumor into peripheral blood is a necessary step in disease dissemination. An assortment of technologies and techniques has been developed to isolate and detect circulating melanoma cells (CMCs), but a standardized method is yet to be established. It is the aim of this study to systematically review the diverse enrichment and detection methods of circulating tumor cells in cutaneous melanoma. A literature search yielded 351 articles, of which 74 were deemed eligible according to inclusion criteria, the primary requirement being the reporting of patient CMC positivity status stratified by the stage of melanoma. Pertinent studies were used to evaluate the advantages and disadvantages of each method. Additionally, we calculated the sensitivity and specificity of seven common melanoma-associated markers based on the available literature.
Collapse
Affiliation(s)
- Stefan Rodic
- Division of Biology, McGill University, Montreal, Canada
| | - Catalin Mihalcioiu
- Division of Medical Oncology, McGill University Health Centre, Montreal, Canada
| | - Ramy R Saleh
- Division of Medical Oncology, McGill University Health Centre, Montreal, Canada.
| |
Collapse
|
16
|
Kinetics of tumor destruction by chimeric antigen receptor-modified T cells. Mol Ther 2013; 22:623-633. [PMID: 24213558 DOI: 10.1038/mt.2013.262] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 10/30/2013] [Indexed: 01/28/2023] Open
Abstract
The use of chimeric antigen receptor (CAR)-modified T cells as a therapy for hematologic malignancies and solid tumors is becoming more widespread. However, the infusion of a T-cell product targeting a single tumor-associated antigen may lead to target antigen modulation under this selective pressure, with subsequent tumor immune escape. With the purpose of preventing this phenomenon, we have studied the impact of simultaneously targeting two distinct antigens present on tumor cells: namely mucin 1 and prostate stem cell antigen, both of which are expressed in a variety of solid tumors, including pancreatic and prostate cancer. When used individually, CAR T cells directed against either tumor antigen were able to kill target-expressing cancer cells, but tumor heterogeneity led to immune escape. As a combination therapy, we demonstrate superior antitumor effects using both CARs simultaneously, but this was nevertheless insufficient to achieve a complete response. To understand the mechanism of escape, we studied the kinetics of T-cell killing and found that the magnitude of tumor destruction depended not only on the presence of target antigens but also on the intensity of expression-a feature that could be altered by administering epigenetic modulators that upregulated target expression and enhanced CAR T-cell potency.
Collapse
|
17
|
Chan WM, Rahman MM, McFadden G. Oncolytic myxoma virus: the path to clinic. Vaccine 2013; 31:4252-8. [PMID: 23726825 PMCID: PMC3755036 DOI: 10.1016/j.vaccine.2013.05.056] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 05/13/2013] [Indexed: 12/12/2022]
Abstract
Many common neoplasms are still noncurative with current standards of cancer therapy. More therapeutic modalities need to be developed to significantly prolong the lives of patients and eventually cure a wider spectrum of cancers. Oncolytic virotherapy is one of the promising new additions to clinical cancer therapeutics. Successful oncolytic virotherapy in the clinic will be those strategies that best combine tumor cell oncolysis with enhanced immune responses against tumor antigens. The current candidate oncolytic viruses all share the common property that they are relatively nonpathogenic to humans, yet they have the ability to replicate selectively in human cancer cells and induce cancer regression by direct oncolysis and/or induction of improved anti-tumor immune responses. Many candidate oncolytic viruses are in various stages of clinical and preclinical development. One such preclinical candidate is myxoma virus (MYXV), a member of the Poxviridae family that, in its natural setting, exhibits a very restricted host range and is only pathogenic to European rabbits. Despite its narrow host range in nature, MYXV has been shown to productively infect various classes of human cancer cells. Several preclinical in vivo modeling studies have demonstrated that MYXV is an attractive and safe candidate oncolytic virus, and hence, MYXV is currently being developed as a potential therapeutic for several cancers, such as pancreatic cancer, glioblastoma, ovarian cancer, melanoma, and hematologic malignancies. This review highlights the preclinical cancer models that have shown the most promise for translation of MYXV into human clinical trials.
Collapse
Affiliation(s)
- Winnie M. Chan
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Masmudur M. Rahman
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
18
|
Safety and clinical efficacy of rapidly-generated trivirus-directed T cells as treatment for adenovirus, EBV, and CMV infections after allogeneic hematopoietic stem cell transplant. Mol Ther 2013; 21:2113-21. [PMID: 23783429 DOI: 10.1038/mt.2013.151] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 06/13/2013] [Indexed: 11/08/2022] Open
Abstract
Adoptive transfer of virus-specific T cells can prevent and treat serious infections with Epstein-Barr virus (EBV), cytomegalovirus (CMV), and adenovirus (Adv) after allogeneic hematopoietic stem cell transplant. It has, however, proved difficult to make this approach widely available since infectious virus and viral vectors are required for T cell activation, followed by an intensive and prolonged culture period extending over several months. We now show that T cells targeting a range of viral antigens derived from EBV, CMV, and Adv can be reproducibly generated in a single culture over a 2-3-week period, using methods that exclude all viral components and employ a much-simplified culture technology. When administered to recipients of haploidentical (n = 5), matched unrelated (n = 3), mismatched unrelated (n = 1) or matched related (n = 1) transplants with active CMV (n = 3), Adv (n = 1), EBV (n = 2), EBV+Adv (n = 2) or CMV+Adv (n = 2) infections, the cells produced complete virological responses in 80%, including all patients with dual infections. In each case, a decrease in viral load correlated with an increase in the frequency of T cells directed against the infecting virus(es); both immediate and delayed toxicities were absent. This approach should increase both the feasibility and applicability of T cell therapy. The trial was registered at www.clinicaltrials.gov as NCT01070797.
Collapse
|
19
|
Wang M, Yin B, Matsueda S, Deng L, Li Y, Zhao W, Zou J, Li Q, Loo C, Wang RF, Wang HY. Identification of special AT-rich sequence binding protein 1 as a novel tumor antigen recognized by CD8+ T cells: implication for cancer immunotherapy. PLoS One 2013; 8:e56730. [PMID: 23437226 PMCID: PMC3578933 DOI: 10.1371/journal.pone.0056730] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Accepted: 01/14/2013] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A large number of human tumor-associated antigens that are recognized by CD8(+) T cells in a human leukocyte antigen class I (HLA-I)-restricted fashion have been identified. Special AT-rich sequence binding protein 1 (SATB1) is highly expressed in many types of human cancers as part of their neoplastic phenotype, and up-regulation of SATB1 expression is essential for tumor survival and metastasis, thus this protein may serve as a rational target for cancer vaccines. METHODOLOGY/PRINCIPAL FINDINGS Twelve SATB1-derived peptides were predicted by an immuno-informatics approach based on the HLA-A*02 binding motif. These peptides were examined for their ability to induce peptide-specific T cell responses in peripheral blood mononuclear cells (PBMCs) obtained from HLA-A*02(+) healthy donors and/or HLA-A*02(+) cancer patients. The recognition of HLA-A*02(+) SATB1-expressing cancer cells was also tested. Among the twelve SATB1-derived peptides, SATB1(565-574) frequently induced peptide-specific T cell responses in PBMCs from both healthy donors and cancer patients. Importantly, SATB1(565-574)-specific T cells recognized and killed HLA-A*02(+) SATB1(+) cancer cells in an HLA-I-restricted manner. CONCLUSIONS/SIGNIFICANCE We have identified a novel HLA-A*02-restricted SATB1-derived peptide epitope recognized by CD8(+) T cells, which, in turn, recognizes and kills HLA-A*02(+) SATB1(+) tumor cells. The SATB1-derived epitope identified may be used as a diagnostic marker as well as an immune target for development of cancer vaccines.
Collapse
Affiliation(s)
- Mingjun Wang
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
| | - Bingnan Yin
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Satoko Matsueda
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
| | - Lijuan Deng
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Ying Li
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
| | - Wei Zhao
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Jia Zou
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Qingtian Li
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Christopher Loo
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
| | - Rong-Fu Wang
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
| | - Helen Y. Wang
- Center for Inflammation and Epigenetics, The Methodist Hospital Research Institute, Houston, Texas, United States of America
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas, United States of America
- * E-mail:
| |
Collapse
|
20
|
Zhou J, Yuen NK, Zhan Q, Velazquez EF, Murphy GF, Giobbie-Hurder A, Hodi FS. Immunity to the melanoma inhibitor of apoptosis protein (ML-IAP; livin) in patients with malignant melanoma. Cancer Immunol Immunother 2011; 61:655-65. [PMID: 22033581 PMCID: PMC3337996 DOI: 10.1007/s00262-011-1124-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 09/30/2011] [Indexed: 11/29/2022]
Abstract
Therapeutic targeting of melanoma antigens frequently focuses on the melanocyte differentiation or cancer-testis families. Antigen-loss variants can often result, as these antigens are not critical for tumor cell survival. Exploration of functionally relevant targets has been limited. The melanoma inhibitor of apoptosis protein (ML-IAP; livin) is overexpressed in melanoma, contributing to disease progression and treatment resistance. Improved understanding of the significance of ML-IAP immune responses in patients has possible therapeutic applications. We found ML-IAP frequently expressed in melanoma metastases by immunohistochemistry. To assess spontaneous immunity to ML-IAP, an overlapping peptide library representing full-length protein was utilized to screen cellular responses in stage I–IV patients and healthy controls by ELISPOT. A broad array of CD4+ and CD8+ cellular responses against ML-IAP was observed with novel class I and class II epitopes identified. Specific HLA-A*0201 epitopes were analyzed further for frequency of reactivity. The generation of specific CD4+ and cytotoxic T cells revealed potent functional capability including cytokine responsiveness to melanoma cell lines and tumor cell killing. In addition, recombinant ML-IAP protein used in an ELISA demonstrated high titer antibody responses in a subset of patients. Several melanoma patients who received CTLA-4 blockade with ipilimumab developed augmented humoral immune responses to ML-IAP as a function of treatment which was associated with beneficial clinical outcomes. High frequency immune responses in melanoma patients, associations with favorable treatment outcomes, and its essential role in melanoma pathogenesis support the development of ML-IAP as a disease marker and therapeutic target.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Garrido C, Romero I, Berruguilla E, Cancela B, Algarra I, Collado A, García-Lora A, Garrido F. Immunotherapy eradicates metastases with reversible defects in MHC class I expression. Cancer Immunol Immunother 2011; 60:1257-68. [PMID: 21553283 PMCID: PMC11028956 DOI: 10.1007/s00262-011-1027-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2010] [Accepted: 04/21/2011] [Indexed: 01/05/2023]
Abstract
Tumor or metastatic cells lose MHC class I (MHC-I) expression during cancer progression as an escape mechanism from immune surveillance. These defects in MHC-I may be reversible by cytokines or different agents (soft lesions) or irreversible due to structural defects (hard lesions). The nature of these MHC-I alterations might determine the success or failure of immunotherapy treatments. In this study, we have used an MHC-I-positive murine fibrosarcoma tumor clone, GR9-A7, which generates multiple lung and lymph node metastases with reversible MHC-I alterations after treatment with IFN-γ. Four different antitumor treatments were carried out after primary tumor excision to determine their capacity to inhibit spontaneous metastatic colonization of the GR9-A7 tumor clone. We found that 2 different immunotherapy protocols (CpG plus autologous irradiated-GR9-A7 cells and protein-bound polysaccharide K (PSK) and 1 chemoimmunotherapy (docetaxel plus PSK) induced eradication of metastases. In contrast, chemotherapy with docetaxel alone produced only partial reduction in the number of metastases. Flow cytometric analysis of lymphocyte populations showed an immunosuppression in GR9-A7 tumor-bearing host, which could be reverted by immunotherapy treatments. Our results suggest that irreversible or reversible MHC-I alterations in tumor target cells may determine its progression or regression independently of the type of immunotherapy used.
Collapse
Affiliation(s)
- Cristina Garrido
- Departamento De Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Servicio de Análisis Clínicos and Inmunología, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Irene Romero
- Servicio de Análisis Clínicos and Inmunología, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Enrique Berruguilla
- Servicio de Análisis Clínicos and Inmunología, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Bárbara Cancela
- Servicio de Farmacia, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Ignacio Algarra
- Departamento de Ciencias de la Salud, Universidad de Jaén, Jaén, Spain
| | - Antonia Collado
- Unidad de Investigación, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | - Angel García-Lora
- Servicio de Análisis Clínicos and Inmunología, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| | - Federico Garrido
- Departamento De Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
- Servicio de Análisis Clínicos and Inmunología, Hospital Universitario Virgen de las Nieves, Av. Fuerzas Armadas 2, 18014 Granada, Spain
| |
Collapse
|
22
|
Thomas DL, Doty R, Tosic V, Liu J, Kranz DM, McFadden G, Macneill AL, Roy EJ. Myxoma virus combined with rapamycin treatment enhances adoptive T cell therapy for murine melanoma brain tumors. Cancer Immunol Immunother 2011; 60:1461-72. [PMID: 21656158 DOI: 10.1007/s00262-011-1045-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 05/20/2011] [Indexed: 12/11/2022]
Abstract
Adoptive transfer of tumor-specific T cells has shown some success for treating metastatic melanoma. We evaluated a novel strategy to improve adoptive therapy by administering both T cells and oncolytic myxoma virus to mice with syngeneic B16.SIY melanoma brain tumors. Adoptive transfer of activated CD8(+) 2C T cells that recognize SIY peptide doubled survival time, but SIY-negative tumors recurred. Myxoma virus killed B16.SIY cells in vitro, and intratumoral injection of virus led to selective and transient infection of the tumor. Virus treatment recruited innate immune cells to the tumor and induced IFNβ production in the brain, resulting in limited oncolytic effects in vivo. To counter this, we evaluated the safety and efficacy of co-administering 2C T cells, myxoma virus, and either rapamycin or neutralizing antibodies against IFNβ. Mice that received either triple combination therapy survived significantly longer with no apparent side effects, but eventually relapsed. Importantly, rapamycin treatment did not impair T cell-mediated tumor destruction, supporting the feasibility of combining adoptive immunotherapy and rapamycin-enhanced virotherapy. Myxoma virus may be a useful vector for transient delivery of therapeutic genes to a tumor to enhance T cell responses.
Collapse
Affiliation(s)
- Diana L Thomas
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Olson BM, McNeel DG. CD8+ T cells specific for the androgen receptor are common in patients with prostate cancer and are able to lyse prostate tumor cells. Cancer Immunol Immunother 2011; 60:781-92. [PMID: 21350948 PMCID: PMC3319721 DOI: 10.1007/s00262-011-0987-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 02/06/2011] [Indexed: 12/29/2022]
Abstract
The androgen receptor (AR) is a hormone receptor that plays a critical role in prostate cancer, and depletion of its ligand has long been the cornerstone of treatment for metastatic disease. Here, we evaluate the AR ligand-binding domain (LBD) as an immunological target, seeking to identify HLA-A2-restricted epitopes recognized by T cells in prostate cancer patients. Ten AR LBD-derived, HLA-A2-binding peptides were identified and ranked with respect to HLA-A2 affinity and were used to culture peptide-specific T cells from HLA-A2+ prostate cancer patients. These T-cell cultures identified peptide-specific T cells specific for all ten peptides in at least one patient, and T cells specific for peptides AR805 and AR811 were detected in over half of patients. Peptide-specific CD8+ T-cell clones were then isolated and characterized for prostate cancer cytotoxicity and cytokine expression, identifying that AR805 and AR811 CD8+ T-cell clones could lyse prostate cancer cells in an HLA-A2-restricted fashion, but only AR811 CTL had polyfunctional cytokine expression. Epitopes were confirmed using immunization studies in HLA-A2 transgenic mice, in which the AR LBD is an autologous antigen with an identical protein sequence, which showed that mice immunized with AR811 developed peptide-specific CTL that lyse HLA-A2+ prostate cancer cells. These data show that AR805 and AR811 are HLA-A2-restricted epitopes for which CTL can be commonly detected in prostate cancer patients. Moreover, CTL responses specific for AR811 can be elicited by direct immunization of A2/DR1 mice. These findings suggest that it may be possible to elicit an anti-prostate tumor immune response by augmenting CTL populations using AR LBD-based vaccines.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Animals, Genetically Modified
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cytokines/biosynthesis
- Cytokines/immunology
- Epitopes, T-Lymphocyte/immunology
- HLA-A2 Antigen/biosynthesis
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/immunology
- HLA-DR1 Antigen/genetics
- HLA-DR1 Antigen/immunology
- Humans
- Ligands
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Oligopeptides/immunology
- Oligopeptides/pharmacology
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/pathology
- Prostatic Neoplasms/therapy
- Protein Structure, Tertiary
- Receptors, Androgen/immunology
- Receptors, Androgen/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
Collapse
Affiliation(s)
- Brian M. Olson
- Department of Medicine, University of Wisconsin, Madison, WI 53792 USA
| | - Douglas G. McNeel
- Department of Medicine, University of Wisconsin, Madison, WI 53792 USA
- University of Wisconsin Carbone Comprehensive Cancer Center, 7007, Wisconsin Institutes for Medical Research, 1111 Highland Ave., Madison, WI 53705 USA
| |
Collapse
|
24
|
Prasad S, Cody V, Saucier-Sawyer JK, Saltzman WM, Sasaki CT, Edelson RL, Birchall MA, Hanlon DJ. Polymer nanoparticles containing tumor lysates as antigen delivery vehicles for dendritic cell-based antitumor immunotherapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2011; 7:1-10. [PMID: 20692374 PMCID: PMC3073408 DOI: 10.1016/j.nano.2010.07.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 07/19/2010] [Accepted: 07/19/2010] [Indexed: 01/17/2023]
Abstract
Encapsulation of tumor-associated antigens in polymer nanoparticles (NP) is a promising approach to enhance efficiency of antigen delivery for anti-tumor vaccines. Head and neck squamous carcinoma (HNSCC) cell lines were initially used to generate tumor-associated antigens (TAA)-containing poly (lactic-co-glycolic acid) (PLGA) NP; encapsulation efficiency and release kinetics were profiled. Findings were adopted to entrap fresh tumor lysate from five patients with advanced HNSCC. To test the hypothesis that NP enhance antigen presentation, dendritic cell (DC) produced from patient blood monocyte precursors were loaded with either the un-encapsulated or NP-encapsulated versions of tumor lysates. These were used to stimulate freshly-isolated autologous CD8+ T cells. In four of five patients, anti-tumor CD8+ T cells showed significantly increased immunostimulatory IFN-γ (p=0.071) or decreased immmunoinhibitory IL-10 production (p=0.0004) associated with NP-mediated antigen delivery. The observations represent an enabling step in the production of clinically-translatable, inexpensive, highly-efficient, and personalized polymer-based immunotherapy for solid organ malignancies. FROM THE CLINICAL EDITOR Enhancing the antigen presentation may be a viable approach to increase the efficiency of tumor cell directed cytotoxicity via immune mechanisms. This study presents an example for this using head and neck cancer cell lines and nanotechnology-based encapsulated antigen presentation to dendritic cells. The observed CD8+ T-cell response was significantly enhanced. This method may pave the way to a highly efficient cancer cell elimination method with minimal to no toxicity.
Collapse
Affiliation(s)
- Shashi Prasad
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| | - Virginia Cody
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| | | | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06520-8260, USA
| | - Clarence T. Sasaki
- Section of Otolaryngology, Yale University, New Haven, Connecticut 06520-8260, USA
| | - Richard L. Edelson
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| | | | - Douglas J. Hanlon
- Department of Dermatology, Yale University, New Haven, Connecticut 06520-8260, USA
| |
Collapse
|
25
|
Abstract
Survivin, the smallest member of the inhibitors of apoptosis proteins (IAPs), plays an important role in the control of apoptosis, cell division, and cell migration/metastasis. Survivin is expressed and required for normal fetal development but is then generally no longer present in most adult tissues. However, reexpression of survivin is observed in numerous human cancers where presence of the protein is associated with enhanced proliferation, metastasis, poor prognosis, and decreased patient survival. Given the relatively selective expression in cancer cells, but not in normal tissue (tumor-associated antigen), and its importance in tumor cell biology, survivin has emerged as an attractive target for cancer treatment. Here, we discuss some aspects of survivin biology by focusing on why the protein appears to be so important for cancer cells and then discuss strategies that harness this dependence to eradicate tumors and situate survivin as a potential Achilles' heel of cancer.
Collapse
Affiliation(s)
- Alvaro Lladser
- Laboratory of Gene Immunotherapy, Fundacion Ciencia para la Vida, Santiago, Chile
| | | | | | | |
Collapse
|
26
|
Identification of a cyclin B1-derived CTL epitope eliciting spontaneous responses in both cancer patients and healthy donors. Cancer Immunol Immunother 2010; 60:227-34. [PMID: 20981424 PMCID: PMC3024510 DOI: 10.1007/s00262-010-0933-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Accepted: 09/18/2010] [Indexed: 11/03/2022]
Abstract
With the aim to identify cyclin B1-derived peptides with high affinity for HLA-A2, we used three in silico prediction algorithms to screen the protein sequence for possible HLA-A2 binders. One peptide scored highest in all three algorithms, and the high HLA-A2-binding affinity of this peptide was verified in an HLA stabilization assay. By stimulation with peptide-loaded dendritic cells a CTL clone was established, which was able to kill two breast cancer cell lines in an HLA-A2-dependent and peptide-specific manner, demonstrating presentation of the peptide on the surface of cancer cells. Furthermore, blood from cancer patients and healthy donors was screened for spontaneous T-cell reactivity against the peptide in IFN-γ ELISPOT assays. Patients with breast cancer, malignant melanoma, or renal cell carcinoma hosted powerful and high-frequency T-cell responses against the peptide. In addition, when blood from healthy donors was tested, similar responses were observed. Ultimately, serum from cancer patients and healthy donors was analyzed for anti-cyclin B1 antibodies. Humoral responses against cyclin B1 were frequently detected in both cancer patients and healthy donors. In conclusion, a high-affinity cyclin B1-derived HLA-A2-restricted CTL epitope was identified, which was presented on the cell surface of cancer cells, and elicited spontaneous T-cell responses in cancer patients and healthy donors.
Collapse
|
27
|
Kerzerho J, Adotevi O, Castelli FA, Dosset M, Bernardeau K, Szely N, Lang F, Tartour E, Maillere B. The Angiogenic Growth Factor and Biomarker Midkine Is a Tumor-Shared Antigen. THE JOURNAL OF IMMUNOLOGY 2010; 185:418-23. [DOI: 10.4049/jimmunol.0901014] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
28
|
The immune system strikes back: cellular immune responses against indoleamine 2,3-dioxygenase. PLoS One 2009; 4:e6910. [PMID: 19738905 PMCID: PMC2734165 DOI: 10.1371/journal.pone.0006910] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2009] [Accepted: 06/26/2009] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The enzyme indoleamine 2,3-dioxygenase (IDO) exerts an well established immunosuppressive function in cancer. IDO is expressed within the tumor itself as well as in antigen-presenting cells in tumor-draining lymph nodes, where it promotes the establishment of peripheral immune tolerance to tumor antigens. In the present study, we tested the notion whether IDO itself may be subject to immune responses. METHODS AND FINDINGS The presence of naturally occurring IDO-specific CD8 T cells in cancer patients was determined by MHC/peptide stainings as well as ELISPOT. Antigen specific cytotoxic T lymphocytes (CTL) from the peripheral blood of cancer patients were cloned and expanded. The functional capacity of the established CTL clones was examined by chrome release assays. The study unveiled spontaneous cytotoxic T-cell reactivity against IDO in peripheral blood as well as in the tumor microenvironment of different cancer patients. We demonstrate that these IDO reactive T cells are indeed peptide specific, cytotoxic effector cells. Hence, IDO reactive T cells are able to recognize and kill tumor cells including directly isolated AML blasts as well as IDO-expressing dendritic cells, i.e. one of the major immune suppressive cell populations. CONCLUSION IDO may serve as an important and widely applicable target for anti-cancer immunotherapeutic strategies. Furthermore, as emerging evidence suggests that IDO constitutes a significant counter-regulatory mechanism induced by pro-inflammatory signals, IDO-based immunotherapy holds the promise to boost anti-cancer immunotherapy in general.
Collapse
|
29
|
Passoni L, Gambacorti-Passerini C. ALK a Novel Lymphoma-associated Tumor Antigen for Vaccination Strategies. Leuk Lymphoma 2009; 44:1675-81. [PMID: 14692518 DOI: 10.1080/1042819031000099625] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The discovery of Tumor Associated Antigens (TAAs) demonstrated that tumor cells can be specifically recognized by the immune system raising the hypothesis that tumors express antigens that Cytotoxic T Lymphocytes (CTLs) can potentially attack. The identification of immunogenic epitopes led to their use as targets to mediate the specific clearance of neoplastic cells by TAA targeting strategies such as vaccination strategies. One of the critical issues in the development of efficient vaccination protocols is the identification of the appropriate TAAs. The TAA should be effective as a "tumor rejection antigen" able to induce an immune response that will affect tumor growth. A distinct pathologic entity characterized by the expression of the Anaplastic Lymphoma Kinase (ALK) protein and named "ALKoma" has recently emerged within the heterogeneous group of CD30+ Anaplastic Large Cell Lymphoma (ALCL). ALK is a receptor tyrosine kinase whose expression is normally restricted to a few scattered cells in the nervous system. Its pathological expression in lymphoma cells is due to a chromosomal translocation that leads to the formation of an ALK-derived oncogenic fusion proteins. ALK fusion proteins ectopically over-expressed and constitutively activated in lymphoid cells play a key role in the neoplastic transformation by the aberrant phosphorylation of intracellular substrates that likely contributes to the molecular pathogenesis of ALCL. The high level of ALK expression in lymphoma cells and its direct role in lymphomagenesis, combined with the fact that normal ALK is expressed at low levels in the immune privileged nervous system, makes ALK an ideal lymphoma-specific target for immunotherapy of ALK+ALCL.
Collapse
Affiliation(s)
- Lorena Passoni
- Oncogenic Fusion Genes and Proteins Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.
| | | |
Collapse
|
30
|
Zwick C, Held G, Hammermeister V, AlAhmad A, Kubuschok B, Bittenbring J, Ahlgrimm M, Neumann F, Preuss KD, Pfreundschuh M. Spontaneous high-titered IgG antibody responses against BCL-2 in patients with aggressive lymphomas. J Cancer Res Clin Oncol 2009; 135:1207-13. [DOI: 10.1007/s00432-009-0561-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Accepted: 02/02/2009] [Indexed: 11/29/2022]
|
31
|
Deacon DH, Hogan KT, Swanson EM, Chianese-Bullock KA, Denlinger CE, Czarkowski AR, Schrecengost RS, Patterson JW, Teague MW, Slingluff CL. The use of gamma-irradiation and ultraviolet-irradiation in the preparation of human melanoma cells for use in autologous whole-cell vaccines. BMC Cancer 2008; 8:360. [PMID: 19055839 PMCID: PMC2612687 DOI: 10.1186/1471-2407-8-360] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 12/04/2008] [Indexed: 11/10/2022] Open
Abstract
Background Human cancer vaccines incorporating autologous tumor cells carry a risk of implantation and subsequent metastasis of viable tumor cells into the patient who is being treated. Despite the fact that the melanoma cell preparations used in a recent vaccine trial (Mel37) were gamma-irradiated (200 Gy), approximately 25% of the preparations failed quality control release criteria which required that the irradiated cells incorporate 3H-thymidine at no more than 5% the level seen in the non-irradiated cells. We have, therefore, investigated ultraviolet (UV)-irradiation as a possible adjunct to, or replacement for gamma-irradiation. Methods Melanoma cells were gamma- and/or UV-irradiated. 3H-thymidine uptake was used to assess proliferation of the treated and untreated cells. Caspase-3 activity and DNA fragmentation were measured as indicators of apoptosis. Immunohistochemistry and Western blot analysis was used to assess antigen expression. Results UV-irradiation, either alone or in combination with gamma-irradiation, proved to be extremely effective in controlling the proliferation of melanoma cells. In contrast to gamma-irradiation, UV-irradiation was also capable of inducing significant levels of apoptosis. UV-irradiation, but not gamma-irradiation, was associated with the loss of tyrosinase expression. Neither form of radiation affected the expression of gp100, MART-1/MelanA, or S100. Conclusion These results indicate that UV-irradiation may increase the safety of autologous melanoma vaccines, although it may do so at the expense of altering the antigenic profile of the irradiated tumor cells.
Collapse
Affiliation(s)
- Donna H Deacon
- Department of Surgery, University of Virginia, Charlottesville, VA 22908, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Feng X, Hui KM, Younes HM, Brickner AG. Targeting minor histocompatibility antigens in graft versus tumor or graft versus leukemia responses. Trends Immunol 2008; 29:624-32. [PMID: 18952501 PMCID: PMC2593397 DOI: 10.1016/j.it.2008.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2008] [Revised: 08/24/2008] [Accepted: 09/01/2008] [Indexed: 01/25/2023]
Abstract
Allogeneic hematopoietic cell transplantation (alloHCT) represents the only curative therapy for several hematologic malignancies, and shows promise as a nascent treatment modality for select solid tumors. Although the original goal of alloHCT was hematopoietic reconstitution after sub-lethal chemoradiotherapy, recognition of a profound donor lymphocyte-mediated graft-versus-leukemia (GVL) or graft-versus-tumor (GVT) effect has shifted the paradigm from pre-transplant cytoreduction to tumor control via donor lymphocytes. In human leukocyte antigen (HLA)-compatible alloHCT, GVL and GVT reactions are induced primarily by donor T-cell recognition of minor histocompatibility antigens (mHAgs). Here we review the literature regarding mHAg-specific T cells in GVL and GVT reactions, and discuss the prospects of exploiting mHAgs as immunotherapeutic targets.
Collapse
Affiliation(s)
- Xin Feng
- Department of Medicine, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
33
|
Neller MA, López JA, Schmidt CW. Antigens for cancer immunotherapy. Semin Immunol 2008; 20:286-95. [DOI: 10.1016/j.smim.2008.09.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2008] [Accepted: 09/05/2008] [Indexed: 01/19/2023]
|
34
|
Smahel M, Tejklova P, Smahelova J, Polakova I, Mackova J. Mutation in the immunodominant epitope of the HPV16 E7 oncoprotein as a mechanism of tumor escape. Cancer Immunol Immunother 2008; 57:823-31. [PMID: 17962940 PMCID: PMC11030076 DOI: 10.1007/s00262-007-0418-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2007] [Accepted: 10/11/2007] [Indexed: 10/22/2022]
Abstract
Infection with high-risk types of human papillomavirus (HPV) can cause the development of malignant tumors. To study mechanisms responsible for immune escape of tumor cells infected with HPV16, we previously used mouse oncogenic TC-1 cells producing HPV16 E6 and E7 oncoproteins to derive TC-1 clones resistant to immunization against E7. We have found immunoresistance of the clones to correlate with the point mutation in the E7 oncogene, which resulted in the N53S substitution in the immunodominant epitope RAHYNIVTF (aa 49-57). Here, we have shown that this mutation reduced stabilization of H-2D(b) molecules on RMA-S cells and eliminated immunogenicity of E7. The resistance of TC-1 clones was E7-specific as immunization against E6 inhibited tumor growth. Transduction of the TC-1/F9 clone carrying the mutated epitope with the wild-type E7 gene restored susceptibility to immunization against E7. Our results suggest that mutagenesis of tumor antigens can lead to the escape of malignant cells and should be considered in the development and evaluation of cancer immunotherapy.
Collapse
Affiliation(s)
- Michal Smahel
- Department of Experimental Virology, Institute of Hematology and Blood Transfusion, U Nemocnice 1, 128 20 Prague 2, Czech Republic.
| | | | | | | | | |
Collapse
|
35
|
Slingluff CL, Petroni GR, Chianese-Bullock KA, Smolkin ME, Hibbitts S, Murphy C, Johansen N, Grosh WW, Yamshchikov GV, Neese PY, Patterson JW, Fink R, Rehm PK. Immunologic and clinical outcomes of a randomized phase II trial of two multipeptide vaccines for melanoma in the adjuvant setting. Clin Cancer Res 2008; 13:6386-95. [PMID: 17975151 DOI: 10.1158/1078-0432.ccr-07-0486] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Human melanoma cells express shared antigens recognized by CD8(+) T lymphocytes, the most common of which are melanocytic differentiation proteins and cancer-testis antigens. However, peptide vaccines for melanoma usually target only one or two MHC class I-associated peptide antigens. Because melanomas commonly evade immune recognition by selective antigen loss, optimization of melanoma vaccines may require development of more complex multipeptide vaccines. EXPERIMENTAL DESIGN In a prospective randomized clinical trial, we have evaluated the safety and immunogenicity of a vaccine containing a mixture of 12 peptides from melanocytic differentiation proteins and cancer-testis antigens, designed for human leukocyte antigen types that represent 80% of the melanoma patient population. This was compared with a four-peptide vaccine with only melanocytic differentiation peptides. Immune responses were assessed in peripheral blood and in vaccine-draining lymph nodes. RESULTS These data show that (a) the 12-peptide mixture is immunogenic in all treated patients; (b) immunogenicity of individual peptides is maintained despite competition with additional peptides for binding to MHC molecules; (c) a broader and more robust immune response is induced by vaccination with the more complex 12-peptide mixture; and (d) clinical outcome in this peptide vaccine trial correlates with immune responses measured in the peripheral blood lymphocytes. CONCLUSIONS These data support continued investigation of complex multipeptide vaccines for melanoma.
Collapse
Affiliation(s)
- Craig L Slingluff
- Department of Surgery, University of Virginia, Charlottesville, Virginia 22908, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Immunosuppressive mechanisms in human tumors: why we still cannot cure cancer. Immunol Lett 2007; 116:7-14. [PMID: 18164076 DOI: 10.1016/j.imlet.2007.11.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Revised: 11/02/2007] [Accepted: 11/09/2007] [Indexed: 12/29/2022]
Abstract
Tumor cells often evoke specific immune responses that, however, fail to eliminate all the tumor cells. The development of cancer immunotherapies is, therefore, mostly focused on the generation of large numbers of activated anti-tumor effector cells by vaccination or adoptive T cell transfer. These developments are built on an ever-extended list of identified tumor-associated antigens and corresponding T cell epitopes, and a steady flow of reports from proof-of-principle animal model experiments demonstrating cure from disease by immune interventions. However, the promises have not translated into clinical successes for cancer patients. Even where tumor regression or complete responses were achieved there is usually relapse of the disease. Increasing numbers of reports over recent years highlight potential immunosuppressive mechanisms that act in tumors and systemically in cancer patients to block effective anti-tumor immune responses. They account in large parts for the failures of cancer immunotherapy and need to be overcome before progress can be expected. We review here the current state of the research on immunosuppressive networks in human cancer.
Collapse
|
37
|
Sensi M, Anichini A. Unique tumor antigens: evidence for immune control of genome integrity and immunogenic targets for T cell-mediated patient-specific immunotherapy. Clin Cancer Res 2007; 12:5023-32. [PMID: 16951217 DOI: 10.1158/1078-0432.ccr-05-2682] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The molecular identification and characterization of antigenic epitopes recognized by T cells on human cancers has rapidly evolved since the cloning in 1991 of MAGEA1, the first gene reported to encode a CTL-defined human tumor antigen. In the expanding field of human tumor immunology, unique tumor antigens constitute a growing class of T cell-defined epitopes that exhibit strong immunogenicity. Some of these antigens, which often derive from mutation of genes that have relevant biological functions, are less susceptible to immunoselection and may be retained even in advanced tumors. Immunogenicity and constitutive expression of the unique tumor antigens provide a strong rationale for the design of novel, patient-tailored therapies that target such determinants. Here we discuss the immunologic relevance of unique tumor antigens in the light of the prospects for exploiting such epitopes as targets for patient-specific immune intervention strategies.
Collapse
Affiliation(s)
- Marialuisa Sensi
- Human Tumor Immunobiology Unit, Department of Experimental Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | | |
Collapse
|
38
|
Levy A, Pitcovski J, Frankenburg S, Elias O, Altuvia Y, Margalit H, Peretz T, Golenser J, Lotem M. A melanoma multiepitope polypeptide induces specific CD8+ T-cell response. Cell Immunol 2007; 250:24-30. [PMID: 18275944 PMCID: PMC2413004 DOI: 10.1016/j.cellimm.2008.01.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Revised: 01/02/2008] [Accepted: 01/06/2008] [Indexed: 11/17/2022]
Abstract
Strategies using epitope-based vaccination are being considered for melanoma immunotherapy, in an attempt to overcome failure of other modalities. In the present study, we designed and produced a multiepitope polypeptide for melanoma (MEP-mel), which contains three repeats of four antigenic epitopes (gp100: 209-217 (210M); gp100: 280-288 (288V); Mart1: 26-35 (27L); tyrosinase: 368-376 (370D). The peptides were attached to each other by linkers containing sequences recognized by the proteasome, to improve protein cleavage and antigen presentation. The results show that peptide-specific T cells produced IFN-gamma when stimulated with MEP-mel-transfected dendritic cells. The presentation of peptides by MEP-mel-transfected dendritic cells was proteasome-dependent and was more long-lasting than the presentation of exogenously delivered native peptides. When dendritic cells were loaded with MEP-mel protein, weak cross presentation was induced. The production of multiepitope molecules based on several peptides linked by sequences sensitive to proteasomal cleavage represents a promising new tool for the improvement of cancer immunotherapy.
Collapse
|
39
|
Abstract
The genomic revolution has advanced our understanding of breast cancer biology and the molecular basis of antitumor immunity. Trastuzumab, the first monoclonal antibody for breast cancer, is now a routine part of clinical care. Breast cancer vaccines may be more effective by actively recruiting both humoral and cellular immunity to the therapeutic effort. However, immunization alone is unlikely to have significant activity against established breast cancers, where it is limited by potent mechanisms of immune tolerance and the immunobiology of breast cancer itself. The next generation of clinical studies should integrate breast cancer vaccines with standard breast cancer drugs or novel immunotherapeutics in strategic doses and schedules that abrogate immune tolerance and groom the tumor microenvironment for a productive immune response.
Collapse
Affiliation(s)
- Leisha A Emens
- Department of Oncology, The Johns Hopkins University School of Medicine, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Bunting-Blaustein Cancer Research Building, 1650 Orleans Street, Room 4M90, Baltimore, MD 21231-1000, USA.
| |
Collapse
|
40
|
Krüger S, Ola V, Feller AC, Fischer D, Friedrich M. Expression of cancer-testis antigen CT7 (MAGE-C1) in breast cancer: an immunohistochemical study with emphasis on prognostic utility. Pathol Oncol Res 2007; 13:91-6. [PMID: 17607369 DOI: 10.1007/bf02893483] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Accepted: 05/10/2007] [Indexed: 01/03/2023]
Abstract
High expression of the cancer-testis antigen CT7, also referred to as MAGE-C1, has been recently described in a variety of malignant tumors, including breast carcinoma. To our knowledge, no data concerning the prognostic utility of CT7 expression in breast cancer are available. In this retrospective study, we evaluated the relationship between CT7 immunoreactivity and clinicopathological parameters as well as relapse-free survival (RFS) and metastasis-free survival (MFS) of 124 women with invasive breast cancer. A positive CT7 status, defined as immunoreactivity in more than 50% of tumor cells, was found in 18% of cases and correlated significantly with high tumor grade (p=0.004), but with no other clinicopathological parameter. In a univariate analysis, CT7 status showed an association with RFS by trend (p=0.107; relative risk [RR]: 1.85) and a significant association with MFS (p=0.043; RR: 2.02). In a multivariate analysis, tumor grade, stage, nodal status, angioinvasion, HER2 expression as well as estrogen and progesterone receptor expression were identified as significant independent prognostic factors of RFS and/or MFS. In this respect, CT7 expression showed a weak, statistically not significant trend towards an independent prognostic relevance concerning prediction of MFS (p=0.147; RR: 1.95). Our data suggest that estimation of CT7 immunoreactivity is of limited prognostic usefulness in breast cancer. It may provide additional information concerning assessment of MFS in selected cases.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Breast Neoplasms/diagnosis
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/diagnosis
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Disease-Free Survival
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Multivariate Analysis
- Neoplasm Metastasis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Prognosis
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/genetics
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
- Retrospective Studies
Collapse
Affiliation(s)
- Stefan Krüger
- Institute of Pathology, University of Schleswig-Holstein, Lübeck, D-23538, Germany.
| | | | | | | | | |
Collapse
|
41
|
Laust AK, Sur BW, Wang K, Hubby B, Smith JF, Nelson EL. VRP immunotherapy targeting neu: treatment efficacy and evidence for immunoediting in a stringent rat mammary tumor model. Breast Cancer Res Treat 2007; 106:371-82. [PMID: 17351745 DOI: 10.1007/s10549-007-9517-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2007] [Accepted: 01/07/2007] [Indexed: 10/23/2022]
Abstract
The ability to overcome intrinsic tolerance to a strict "self" tumor-associated antigen (TAA) and successfully treat pre-existing tumor is the most stringent test for anti-tumor immunotherapeutic strategies. Although this capacity has been demonstrated in various models using complicated strategies that may not be readily translated into the clinical arena, straightforward antigen-specific immunotherapeutic strategies in the most stringent models of common epithelial cancers have largely failed to meet this standard. We employed an immunotherapeutic strategy using an alphavirus-based, virus-like replicon particle (VRP), which has in vivo tropism for dendritic cells, to elicit immune responses to the non-mutated TAA rat neu in an aggressive rat mammary tumor model. Using this VRP-based immunotherapeutic strategy targeting a single TAA, we generated effective anti-tumor immunity in the setting of pre-existing tumor resulting in the cure of 36% of rats over multiple experiments, P = 0.002. We also observed down-regulation of rat neu expression in tumors that showed initial responses followed by tumor escape with resumption of rapid tumor growth. These responses were accompanied by significant anti-tumor proliferative responses and CD8+ cellular tumor infiltrates, all of which were restricted to animals receiving the anti-neu immunotherapy. Together these data, obtained in a stringent "self" TAA model, indicate that the VRP-based antigen-specific immunotherapy elicits sufficiently potent immune responses to exert immunologic pressure, selection, and editing of the growing tumors, thus supporting the activity of this straightforward immunotherapy and suggesting that it is a promising platform upon which to build even more potent strategies.
Collapse
Affiliation(s)
- Amanda K Laust
- Department of Molecular Biology and Biochemistry, School of Biological Sciences, University of California, Irvine, CA 92697, USA
| | | | | | | | | | | |
Collapse
|
42
|
Zhu X, Nishimura F, Sasaki K, Fujita M, Dusak JE, Eguchi J, Fellows-Mayle W, Storkus WJ, Walker PR, Salazar AM, Okada H. Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. J Transl Med 2007; 5:10. [PMID: 17295916 PMCID: PMC1802742 DOI: 10.1186/1479-5876-5-10] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Accepted: 02/12/2007] [Indexed: 11/13/2022] Open
Abstract
Background Toll-like receptor (TLR)3 ligands serve as natural inducers of pro-inflammatory cytokines capable of promoting Type-1 adaptive immunity, and TLR3 is abundantly expressed by cells within the central nervous system (CNS). To improve the efficacy of vaccine strategies directed against CNS tumors, we evaluated whether administration of a TLR3 ligand, polyinosinic-polycytidylic (poly-IC) stabilized with poly-lysine and carboxymethylcellulose (poly-ICLC) would enhance the anti-CNS tumor effectiveness of tumor peptide-based vaccinations. Methods C57BL/6 mice bearing syngeneic CNS GL261 glioma or M05 melanoma received subcutaneous (s.c.) vaccinations with synthetic peptides encoding CTL epitopes- mEphA2 (671–679), hgp100 (25–33) and mTRP-2 (180–188) for GL261, or ovalbumin (OVA: 257–264) for M05. The mice also received intramuscular (i.m.) injections with poly-ICLC. Results The combination of subcutaneous (s.c.) peptide-based vaccination and i.m. poly-ICLC administration promoted systemic induction of antigen (Ag)-specific Type-1 CTLs expressing very late activation antigen (VLA)-4, which confers efficient CNS-tumor homing of vaccine-induced CTLs based on experiments with monoclonal antibody (mAb)-mediated blockade of VLA-4. In addition, the combination treatment allowed expression of IFN-γ by CNS tumor-infiltrating CTLs, and improved the survival of tumor bearing mice in the absence of detectable autoimmunity. Conclusion These data suggest that poly-ICLC, which has been previously evaluated in clinical trials, can be effectively combined with tumor Ag-specific vaccine strategies, thereby providing a greater index of therapeutic efficacy.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/immunology
- Cancer Vaccines/immunology
- Carboxymethylcellulose Sodium/administration & dosage
- Carboxymethylcellulose Sodium/analogs & derivatives
- Carboxymethylcellulose Sodium/pharmacology
- Cell Line, Tumor
- Combined Modality Therapy
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Ephrin-A2/immunology
- Epitopes/immunology
- Glioma/immunology
- Glioma/prevention & control
- Glioma/therapy
- Humans
- Injections, Intramuscular
- Integrin alpha4beta1/immunology
- Ligands
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred C57BL
- Ovalbumin/immunology
- Peptides/immunology
- Poly I-C/administration & dosage
- Poly I-C/pharmacology
- Polylysine/administration & dosage
- Polylysine/analogs & derivatives
- Polylysine/pharmacology
- Staining and Labeling
- T-Lymphocytes, Cytotoxic/immunology
- Toll-Like Receptor 3/immunology
- Treatment Outcome
- Up-Regulation/drug effects
- Vaccination
Collapse
Affiliation(s)
- Xinmei Zhu
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Fumihiko Nishimura
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Kotaro Sasaki
- Departments of Dermatology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Mitsugu Fujita
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Jill E Dusak
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Junichi Eguchi
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| | - Wendy Fellows-Mayle
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Walter J Storkus
- Departments of Dermatology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Paul R Walker
- Division of Oncology, Geneva University Hospital, Geneva, Switzerland
| | | | - Hideho Okada
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Brain Tumor Program, University of Pittsburgh Cancer Institute, Pittsburgh, USA
| |
Collapse
|
43
|
Wang M, Johansen B, Nissen MH, Thorn M, Kløverpris H, Fomsgaard A, Buus S, Claësson MH. Identification of an HLA-A*0201 restricted Bcl2-derived epitope expressed on tumors. Cancer Lett 2006; 251:86-95. [PMID: 17182178 DOI: 10.1016/j.canlet.2006.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 11/01/2006] [Accepted: 11/06/2006] [Indexed: 11/27/2022]
Abstract
A large number of human tumor-associated antigen-derived peptides have been identified that are recognized by CTLs in a MHC-I restricted fashion. The apoptosis inhibitory protein Bcl2 is overexpressed in many human cancers as part of their neoplastic phenotype. Since inhibition or loss of Bcl2 expression might impair tumor growth and survival, this protein may serve as a rational target for vaccine-induced CTL responses. By Western blot technique, we screened a panel of established human tumor cell lines for proteins involved in the apoptotic process. Two of eight tumor cell lines, a B lymphoma (Loukes) and a colon carcinoma (CCL220) cell line showed increased Bcl2 protein expression whereas the majority of tumor cell lines expressed proapoptotic proteins. Neither fibroblasts nor peripheral blood mononuclear cells showed Bcl2 expression. An HLA-A*0201 restricted CTL epitope was deduced in silica from the amino acid sequence of the Bcl2 protein and its binding affinity for HLA-A*0201 was confirmed using a biochemical binding assay. We here demonstrate that the 9-mer peptide Bcl2(85-93) induces specific CTL reactivity in immunized C57-A2K(b) or -A2D(b) tg mice. These Bcl2(85-93) specific CTLs react with and lyse Bcl2-expressing human colon carcinoma CCL220 cells which have been transfected with a chimeric HLA-A*0201/H2-K(b) DNA construct similar to that expressed in the transgenic mice. Based on these observations, we suggest that Bcl2(85-93) may be a target for immune therapy.
Collapse
Affiliation(s)
- Mingjun Wang
- Laboratory of Cellular Immunology, Department of Medical Anatomy, The Panum Institute, University of Copenhagen, Blegdamsvej 3C, DK-2200 Copenhagen N, Denmark
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Powell DJ, Dudley ME, Hogan KA, Wunderlich JR, Rosenberg SA. Adoptive transfer of vaccine-induced peripheral blood mononuclear cells to patients with metastatic melanoma following lymphodepletion. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:6527-39. [PMID: 17056585 PMCID: PMC2174602 DOI: 10.4049/jimmunol.177.9.6527] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cancer vaccines can induce the in vivo generation of tumor Ag-specific T cells in patients with metastatic melanoma yet seldom elicit objective clinical responses. Alternatively, adoptive transfer of autologous tumor-infiltrating lymphocytes (TIL) can mediate tumor regression in 50% of lymphodepleted patients, but are logistically and technically difficult to generate. In this study, we evaluated the capability of vaccine-induced PBMC to mediate tumor regression after transfer to patients receiving the same chemotherapy-induced lymphodepletion used for TIL transfer therapy. Autologous PBMC from nine gp100-vaccinated patients with metastatic melanoma were stimulated ex vivo with the gp100:209-217(210M) peptide and transferred in combination with high-dose IL-2 and cancer vaccine. Transferred PBMC contained highly avid, gp100:209-217 peptide-reactive CD8(+) T cells. One week after transfer, lymphocyte counts peaked (median of 14.3 x 10(3) cells//microl; range of 0.9-59.7 x 10(3) cells/microl), with 56% of patients experiencing a lymphocytosis. gp100:209-217 peptide-specific CD8(+) T cells persisted at high levels in the blood of all patients and demonstrated significant tumor-specific IFN-gamma secretion in vitro. Melanocyte-directed autoimmunity was noted in two patients; however, no patient experienced an objective clinical response. These studies demonstrate the feasibility and safety of using vaccine-induced PBMC for cell transfer, but suggests that they are not as effective as TIL in adoptive immunotherapy even when transferred into lymphodepleted hosts.
Collapse
Affiliation(s)
- Daniel J Powell
- Surgery Branch, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Rienk Offringa
- Tumor Immunology Group, Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
46
|
Kono M, Dunn IS, Durda PJ, Butera D, Rose LB, Haggerty TJ, Benson EM, Kurnick JT. Role of the Mitogen-Activated Protein Kinase Signaling Pathway in the Regulation of Human Melanocytic Antigen Expression. Mol Cancer Res 2006; 4:779-92. [PMID: 17050671 DOI: 10.1158/1541-7786.mcr-06-0077] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Heterogeneous expression of melanocytic antigens occurs frequently in melanomas and represents a potent barrier to immunotherapy. We previously showed that coordinated losses of several melanocytic antigens are generally attributable to down-regulation of antigen gene expression rather than irreversible mutation. Treatment of melanoma cells with mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitors blocks ERK activation and increases steady-state levels of mRNAs and corresponding protein expression for the melanocytic antigens Melan-A/MART-1, gp100, and tyrosinase. Although the degree of MEK inhibitor enhancement of antigen expression varied among different cell lines irrespective of their antigen expression status, all showed detectable responses. Notably, the antigen-enhancing effects of the MEK inhibitors could not be attributed to the master melanocytic regulator MITF-M. Because MAPK pathway activation via constitutively active mutant forms of BRAF is common in melanomas, correlation between BRAF function and antigen expression was investigated. No simple correlation of endogenous BRAF mutational status and antigen levels was observed, but transient overexpression of V600E BRAF increased ERK activation and reduced Melan-A/MART-1 levels in antigen-positive cell lines. These data indicate that whereas multiple factors may regulate antigen expression in melanomas, enhancement of MAPK signaling can act as a negative influence. Blocking such signaling with MEK inhibitors accordingly augments antigen levels, thereby enhancing Melan-A/MART-1-specific cytotoxic T-cell responses to antigen-negative cells following MEK inhibition treatment. Consequently, MAPK inhibition may assist targeting of melanomas for immunotherapy.
Collapse
Affiliation(s)
- Michihiro Kono
- Department of Pathology, Massachusetts General Hospital, and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Salcedo M, Bercovici N, Taylor R, Vereecken P, Massicard S, Duriau D, Vernel-Pauillac F, Boyer A, Baron-Bodo V, Mallard E, Bartholeyns J, Goxe B, Latour N, Leroy S, Prigent D, Martiat P, Sales F, Laporte M, Bruyns C, Romet-Lemonne JL, Abastado JP, Lehmann F, Velu T. Vaccination of melanoma patients using dendritic cells loaded with an allogeneic tumor cell lysate. Cancer Immunol Immunother 2006; 55:819-29. [PMID: 16187085 PMCID: PMC11030805 DOI: 10.1007/s00262-005-0078-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2005] [Accepted: 08/06/2005] [Indexed: 12/22/2022]
Abstract
The aim of the present phase I/II study was to evaluate the safety, immune responses and clinical activity of a vaccine based on autologous dendritic cells (DC) loaded with an allogeneic tumor cell lysate in advanced melanoma patients. DC derived from monocytes were generated in serum-free medium containing GM-CSF and IL-13 according to Good Manufacturing Practices. Fifteen patients with metastatic melanoma (stage III or IV) received four subcutaneous, intradermal, and intranodal vaccinations of both DC loaded with tumor cell lysate and DC loaded with hepatitis B surface protein (HBs) and/or tetanus toxoid (TT). No grade 3 or 4 adverse events related to the vaccination were observed. Enhanced immunity to the allogeneic tumor cell lysate and to TAA-derived peptides were documented, as well as immune responses to HBs/TT antigens. Four out of nine patients who received the full treatment survived for more than 20 months. Two patients showed signs of clinical response and received 3 additional doses of vaccine: one patient showed regression of in-transit metastases leading to complete remission. Eighteen months later, the patient was still free of disease. The second patient experienced stabilization of lung metastases for approximately 10 months. Overall, our results show that vaccination with DC loaded with an allogeneic melanoma cell lysate was feasible in large-scale and well-tolerated in this group of advanced melanoma patients. Immune responses to tumor-related antigens documented in some treated patients support further investigations to optimize the vaccine formulation.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/therapeutic use
- Cancer Vaccines/adverse effects
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor/chemistry
- Cell Line, Tumor/immunology
- Cells, Cultured/drug effects
- Cells, Cultured/immunology
- Cells, Cultured/transplantation
- Culture Media, Serum-Free
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- HLA-A2 Antigen/immunology
- Hepatitis B Surface Antigens/administration & dosage
- Humans
- Injections
- Injections, Intradermal
- Injections, Subcutaneous
- Interleukin-13/pharmacology
- Isoantigens/administration & dosage
- Isoantigens/therapeutic use
- Liver Neoplasms/immunology
- Liver Neoplasms/secondary
- Liver Neoplasms/therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymph Nodes
- Lymphatic Metastasis
- Male
- Melanoma/immunology
- Melanoma/secondary
- Melanoma/therapy
- Middle Aged
- Skin Neoplasms/immunology
- Skin Neoplasms/therapy
- Tetanus Toxoid/administration & dosage
- Tissue Extracts/administration & dosage
- Tissue Extracts/immunology
- Tissue Extracts/therapeutic use
- Treatment Outcome
- Vaccination/adverse effects
Collapse
Affiliation(s)
- Margarita Salcedo
- IDM Research Laboratory, 15 rue de l'Ecole de Médecine, 75006 Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Sondak VK, Sabel MS, Mulé JJ. Allogeneic and autologous melanoma vaccines: where have we been and where are we going? Clin Cancer Res 2006; 12:2337s-2341s. [PMID: 16609055 DOI: 10.1158/1078-0432.ccr-05-2555] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The past three decades have seen substantial research on vaccines for the treatment of metastatic melanoma and the prevention of recurrence following resection. Despite their enormous promise, the actual results have been disappointing, with several high-profile vaccine clinical trials failing to show a benefit. Nonetheless, enthusiasm for melanoma vaccines remains and has increased with our expanding understanding of the immune response to tumor. Cellular vaccines can be divided into autologous, derived from the patient's own tumor and allogeneic vaccines. Autologous vaccines have the advantage of containing all potentially relevant tumor-associated antigens for that particular patient. However, autologous vaccines are difficult to obtain from most patients with advanced disease and impossible to obtain from patients who present after resection of all clinically evident disease. No consensus exists for how tumors should be processed, preserved, modified, and delivered to serve as an effective vaccine. The amount of autologous tumor available is rarely enough to produce more than two or three vaccination doses, and the time between initial tumor harvest and ultimate availability of the vaccine may result in interval tumor progression that diminishes the likelihood of vaccine efficacy. All these drawbacks of autologous tumor vaccination limit its applicability and also limit the ability to test autologous vaccines in prospective trials. Allogeneic vaccines avoid many of these problems, but may not contain all of the tumor-associated antigens present on the patient's own tumor. In particular, neoantigens created by mutations in the patient's tumor would be unlikely to be represented in an allogeneic vaccine. Although allogeneic vaccines can be manufactured in sufficient quantities to allow large-scale trials, there remain significant limiting issues in the manufacture and standardization of the vaccine product.
Collapse
Affiliation(s)
- Vernon K Sondak
- Division of Cutaneous Oncology, Department of Interdisciplinary Oncology, University of South Florida College of Medicine, Tampa, Florida, USA.
| | | | | |
Collapse
|
49
|
Goto Y, Matsuzaki Y, Kurihara S, Shimizu A, Okada T, Yamamoto K, Murata H, Takata M, Aburatani H, Hoon DSB, Saida T, Kawakami Y. A new melanoma antigen fatty acid-binding protein 7, involved in proliferation and invasion, is a potential target for immunotherapy and molecular target therapy. Cancer Res 2006; 66:4443-9. [PMID: 16618771 DOI: 10.1158/0008-5472.can-05-2505] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The identification of molecules that are preferentially expressed in melanoma cells and involved in their malignant phenotypes is important for understanding melanoma biology and the development of new diagnostic and therapeutic methods. By comparing the expression profile of a melanoma cell line with those of various normal tissues using GeneChip and by confirming the actual expression of the selected genes by reverse transcription-PCR and Northern and Western blot analyses, fatty acid-binding protein 7 (FABP7), which is frequently expressed in melanomas, was identified. Immunohistochemical examination revealed that FABP7 was expressed in 11 of 15 melanoma tissues. By down-regulating the FABP7 expression with FABP7-specific small interfering RNAs, in vitro cell proliferation and Matrigel invasion were suppressed in two of six melanoma cell lines. Overexpression of FABP7 in a FABP7-negative embryonic kidney cell line 293T by transfecting with the FABP7 cDNA resulted in enhanced cell proliferation and Matrigel invasion, indicating that FABP7 plays a role in the malignant phenotype of some melanoma cell lines. IgG antibodies specific for the phage or bacterial recombinant FABP7 protein were detected in 14 of 25 (56%) or in 8 of 31 (26%) sera from melanoma patients, respectively, but not in sera from healthy individuals, indicating that FABP7 is an immunogenic antigen in melanoma patients. These results showed that FABP7 is frequently expressed in melanoma, may be involved in cell proliferation and invasion, and may be a potential target for development of diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Yasufumi Goto
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinanomachi, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Coral S, Sigalotti L, Colizzi F, Spessotto A, Nardi G, Cortini E, Pezzani L, Fratta E, Fonsatti E, Di Giacomo AM, Nicotra MR, Natali PG, Altomonte M, Maio M. Phenotypic and functional changes of human melanoma xenografts induced by DNA hypomethylation: immunotherapeutic implications. J Cell Physiol 2006; 207:58-66. [PMID: 16252259 DOI: 10.1002/jcp.20540] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Emerging in vitro evidence points to an immunomodulatory activity of DNA hypomethylating drugs in human malignancies. We investigated the potential of 5-aza-2'-deoxycytidine (5-AZA-CdR) to modulate the expression of cancer testis antigens (CTA) and of HLA class I antigens by melanoma xenografts, and the resulting modifications in immunogenicity of neoplastic cells. Three primary cultures of melanoma cells, selected for immune phenotype and growth rate, were grafted into BALB/c nu/nu mice that were injected intraperitoneally with different dose- and time-schedules of 5-AZA-CdR. Molecular analyses demonstrated a de novo long-lasting expression of the CTA MAGE-1, -2, -3, -4, -10, GAGE 1-6, NY-ESO-1, and the upregulation of MAGE-1, MAGE-3, and NY-ESO-1 levels in melanoma xenografts from 5-AZA-CdR-treated mice. Serological and biochemical analyses identified a de novo expression of NY-ESO-1 protein and a concomitant and persistent upregulation of HLA class I antigens and of HLA-A1 and -A2 alleles. Immunization of BALB/c mice with 5-AZA-CdR-treated melanoma cells generated high titer circulating anti-NY-ESO-1 antibodies. Altogether, the data obtained identify an immunomodulatory activity of 5-AZA-CdR in vivo and strongly suggest for its clinical use to design novel strategies of CTA-based chemo-immunotherapy for melanoma patients.
Collapse
MESH Headings
- Animals
- Antibody Formation/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Antimetabolites, Antineoplastic/administration & dosage
- Antimetabolites, Antineoplastic/pharmacology
- Antimetabolites, Antineoplastic/therapeutic use
- Azacitidine/administration & dosage
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Azacitidine/therapeutic use
- DNA Methylation/drug effects
- Decitabine
- Female
- Gene Expression/drug effects
- Gene Expression/genetics
- HLA-A1 Antigen/metabolism
- HLA-A2 Antigen/metabolism
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunotherapy/methods
- Melanoma, Experimental/genetics
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Melanoma-Specific Antigens
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neoplasm Proteins/genetics
- Tumor Cells, Cultured
- Vaccination
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Sandra Coral
- Department of Medical Oncology, Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|