301
|
Trippel A, Halling F, Heymann P, Ayna M, Al-Nawas B, Ziebart T. The expression of melanoma-associated antigen A (MAGE-A) in oral squamous cell carcinoma: an evaluation of the significance for tumor prognosis. Oral Maxillofac Surg 2019; 23:343-352. [PMID: 31093793 DOI: 10.1007/s10006-019-00778-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/02/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Melanoma-associated antigens A had been detected repeatedly in oral squamous cell carcinoma, but not in healthy mucosa. Additionally, patients with MAGE-A expressing cancers are regarded to have a worse survival prognosis, so that MAGE-A are supposed to be part of carcinogenesis. Which role these antigens fulfill within OSCC is still, up today, largely unknown. This study examines the hypothesis that MAGE-A is being produced in OSCC but not in mucosa tissue and if MAGE-A has any correlation to clinical patient's parameters like tumor size, lymph node metastasis, distant metastasis, overall survival, and recurrence. MATERIALS AND METHODS For this purpose, 50 tumor samples and 39 mucosa samples were analyzed by means of PCR and immunohistochemical staining with the antibody 6C1. RESULTS Forty of 41 stained tumor samples showed a positive antibody reaction with a maximum staining rate of 53%. Sixteen mucosa samples showed a mild positive reaction. The PCR revealed a linear expression pattern of MAGE-A in which the genes are proportionally expressed in OSCC. We did not find any relationship between MAGE-A and tumor size, overall survival, or recurrence. There was also no connection between MAGE-A and tumor parameters Hif-1 and LDH. Their expression was detected tendentially in tumors with higher staging, advanced lymph node metastasis, and rising age of the patients. The genes MAGE-A3+6 and MAGE-A4 had a statistically significant correlation with lymph node metastasis (p = 0.007 and p = 0.004). Patients got distant metastasis and influence of MAGE-A on metastatic behavior could not be verified. The genes MAGE-A3 and -A4 are consequently qualified as tumor markers in the field of diagnosis and follow-up of OSCC. CONCLUSIONS AND CLINICAL RELEVANCE Two genes have great potential as target proteins in immunotherapy. The genes MAGE-A3+6 and MAGE-A4 had a statistically significant correlation with lymph node metastasis.
Collapse
Affiliation(s)
- Anna Trippel
- Department of Oral and Maxillofacial Surgery University Medical Center Mainz, Mainz, Germany
| | - Frank Halling
- Department of Oral and Maxillofacial Surgery, Baldingerstrasse, Philipps University of Marburg, University Hospital Giessen and Marburg, Campus Marburg, D-35037, Marburg, Germany
| | - Paul Heymann
- Department of Oral and Maxillofacial Surgery, Baldingerstrasse, Philipps University of Marburg, University Hospital Giessen and Marburg, Campus Marburg, D-35037, Marburg, Germany
| | - Mustafa Ayna
- Center for Dental Implantology, 47051, Duisburg, Germany
| | - Bilal Al-Nawas
- Department of Oral and Maxillofacial Surgery University Medical Center Mainz, Mainz, Germany
| | - Thomas Ziebart
- Department of Oral and Maxillofacial Surgery, Baldingerstrasse, Philipps University of Marburg, University Hospital Giessen and Marburg, Campus Marburg, D-35037, Marburg, Germany.
| |
Collapse
|
302
|
Expression dynamics of Mage family genes during self-renewal and differentiation of mouse pluripotent stem and teratocarcinoma cells. Oncotarget 2019; 10:3248-3266. [PMID: 31143371 PMCID: PMC6524934 DOI: 10.18632/oncotarget.26933] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/29/2019] [Indexed: 12/28/2022] Open
Abstract
The biological roles of cancer-testis antigens of the Melanoma antigen (Mage) family in mammalian development, stem cell differentiation and carcinogenesis are largely unknown. In order to understand the involvement of the Mage family genes in maintenance of normal and cancer stem cells, the expression patterns of Mage-a, Mage-b, Mage-d, Mage-e, Mage-h and Mage-l gene subfamilies were analyzed during the self-renewal and differentiation of mouse pluripotent stem and teratocarcinoma cells. Clustering analysis based on the gene expression profiles of undifferentiated and differentiating cell populations revealed strong correlations between Mage expression patterns and differentiation and malignant states. Gene co-expression analysis disclosed the potential contributions of Mage family members in self-renewal and differentiation of pluripotent stem and teratocarcinoma cells. Two gene clusters including Mage-a4 and Mage-a8, Mageb1, Mage-d1, Mage-d2, Mage-e1, Mage-l2 were identified as functional antagonists with opposing roles in the regulation of proliferation and differentiation of mouse pluripotent stem and teratocarcinoma cells. The identified aberrant expression patterns of Mage-a2, Mage-a6, Mage-b4, Mageb-16 and Mage-h1 in teratocarcinoma cells can be considered as specific teratocarcinoma biomarkers promoted the malignant phenotype. Our study first provides a model for the involvement of Mage family members in regulatory networks during the self-renewal and early differentiation of normal and cancerous stem cells for further research of the predicted functional modules and the development of new cancer treatment strategies.
Collapse
|
303
|
Seifi-Alan M, Shamsi R, Ghafouri-Fard S. Application of cancer-testis antigens in immunotherapy of hepatocellular carcinoma. Immunotherapy 2019; 10:411-421. [PMID: 29473472 DOI: 10.2217/imt-2017-0154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a worldwide common malignancy with poor prognosis. Several studies have aimed at identification of appropriate biomarkers for early detection of this cancer. Cancer-testis antigens (CTAs) as a novel group of tumor-associated antigens have been demonstrated to be expressed in HCC samples as well as peripheral blood samples from these patients but not in the corresponding adjacent noncancerous samples. Such pattern of expression has provided them an opportunity to be used as immunotherapeutic targets. The detection of spontaneous immune responses against CTAs in HCC patients has prompted design of CTA-based immunotherapeutic protocols in these patients. The results of some clinical trials have been promising in a subset of patients.
Collapse
Affiliation(s)
- Mahnaz Seifi-Alan
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roshanak Shamsi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
304
|
Magalhaes I, Carvalho-Queiroz C, Hartana CA, Kaiser A, Lukic A, Mints M, Nilsson O, Grönlund H, Mattsson J, Berglund S. Facing the future: challenges and opportunities in adoptive T cell therapy in cancer. Expert Opin Biol Ther 2019; 19:811-827. [PMID: 30986360 DOI: 10.1080/14712598.2019.1608179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION In recent years, immunotherapy for the treatment of solid cancer has emerged as a promising therapeutic alternative. Adoptive cell therapy (ACT), especially T cell-based, has been found to cause tumor regression and even cure in a percentage of treated patients. Checkpoint inhibitors further underscore the potential of the T cell compartment in the treatment of cancer. Not all patients respond to these treatments; however, many challenges remain. AREAS COVERED This review covers the challenges and progress in tumor antigen target identification and selection, and cell product manufacturing for T cell ACT. Tumor immune escape mechanisms and strategies to overcome those in the context of T cell ACT are also discussed. EXPERT OPINION The immunotherapy toolbox is rapidly expanding and improving, and the future promises further breakthroughs in the T cell ACT field. The heterogeneity of the tumor microenvironment and the multiplicity of tumor immune escape mechanisms pose formidable challenges to successful T cell immunotherapy in solid tumors, however. Individualized approaches and strategies combining treatments targeting different immunotherapeutic aspects will be needed in order to expand the applicability and improve the response rates in future.
Collapse
Affiliation(s)
- Isabelle Magalhaes
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden
| | - Claudia Carvalho-Queiroz
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Ciputra Adijaya Hartana
- c Ragon Institute of MGH, MIT and Harvard, Massachusetts General Hospital , Cambridge , MA , USA
| | - Andreas Kaiser
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Ana Lukic
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Michael Mints
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,d Department of Surgical and Perioperative Sciences , Umeå University, Umeå, Sweden.,e Blood and Marrow Transplant Program, Medical Oncology and Hematology , Princess Margaret Cancer Center , Toronto , Canada.,f Department of Medicine , University of Toronto , Toronto , Canada
| | - Ola Nilsson
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Hans Grönlund
- b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| | - Jonas Mattsson
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,f Department of Medicine , University of Toronto , Toronto , Canada
| | - Sofia Berglund
- a Department of Oncology-Pathology , Karolinska Institutet , Stockholm , Sweden.,b Therapeutic Immune Design, Department of Clinical Neuroscience , Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
305
|
Fennemann FL, de Vries IJM, Figdor CG, Verdoes M. Attacking Tumors From All Sides: Personalized Multiplex Vaccines to Tackle Intratumor Heterogeneity. Front Immunol 2019; 10:824. [PMID: 31040852 PMCID: PMC6476980 DOI: 10.3389/fimmu.2019.00824] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/28/2019] [Indexed: 12/23/2022] Open
Abstract
Tumor vaccines are an important asset in the field of cancer immunotherapy. Whether prophylactic or therapeutic, these vaccines aim to enhance the T cell-mediated anti-tumor immune response that is orchestrated by dendritic cells. Although promising preclinical and early-stage clinical results have been obtained, large-scale clinical implementation of cancer vaccination is stagnating due to poor clinical response. The challenges of clinical efficacy of tumor vaccines can be mainly attributed to tumor induced immunosuppression and poor immunogenicity of the chosen tumor antigens. Recently, intratumor heterogeneity and the relation with tumor-specific neoantigen clonality were put in the equation.In this perspective we provide an overview of recent studies showing how personalized tumor vaccines containing multiple neoantigens can broaden and enhance the anti-tumor immune response. Furthermore, we summarize advances in the understanding of the intratumor mutational landscape containing different tumor cell subclones and the temporal and spatial diversity of neoantigen presentation and burden, and the relation between these factors with respect to tumor immunogenicity. Together, the presented knowledge calls for the investment in the characterization of neoantigens in the context of intratumor heterogeneity to improve clinical efficacy of personalized tumor vaccines.
Collapse
Affiliation(s)
- Felix L Fennemann
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| |
Collapse
|
306
|
MAGE-A4 and MAGE-A1 Immunohistochemical Expression in High-grade Endometrial Cancer. Int J Gynecol Pathol 2019; 38:59-65. [PMID: 29140883 DOI: 10.1097/pgp.0000000000000470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The aim was to investigate MAGE-A4 and MAGE-A1 protein expression in high-grade endometrial cancer and determine its correlation with histologic subtype, FIGO stage, presence of vascular invasion, disease free, and overall survival. Immunohistochemical staining was performed by using 77B (MAGE-A1) and 57B (MAGE-A4) monoclonal antibodies on paraffin-embedded sections from high-grade endometrial cancers diagnosed in University Hospital Split between 1998 and 2011 (n=77). Median follow-up time for survivors was 48 mo. MAGE-A4 was found to be expressed in 33% of endometrioid type endometrial cancers grade 3 and in 27% of serous and clear cell carcinomas. MAGE-A1 was found to be expressed in 93% endometrioid endometrial cancer grade 3 and 86% of serous and clear cell carcinomas. Univariate analysis showed that positive immunohistochemical staining for MAGE-A4 was associated with decreased disease free and overall survival in patients with high-grade endometrial cancer. Multivariate analysis showed an association between MAGE-A4 overexpression and decreased disease free but not overall survival in high-grade endometrial cancer. No correlation was found between MAGE-A1 immunohistochemical expression and patient survival. There was no significant correlation between MAGE-A4 and MAGE-A1 expression and histologic subtype, FIGO stage, lymph node metastasis, muscular infiltration, and lymphovascular invasion. MAGE-A4 immunohistochemical expression is associated with decreased disease free and overall survival in patients with high-grade endometrial cancer. Our findings suggest that MAGE-A1 may be expressed in the epithelial cells of the normal endometrium. MAGE-A1 is highly expressed in high-grade endometrial cancer, with no impact on survival.
Collapse
|
307
|
Shimizu Y, Suzuki T, Yoshikawa T, Endo I, Nakatsura T. Next-Generation Cancer Immunotherapy Targeting Glypican-3. Front Oncol 2019; 9:248. [PMID: 31024850 PMCID: PMC6469401 DOI: 10.3389/fonc.2019.00248] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
Glypican-3 (GPC3), a 65 kD protein consisting of 580 amino acids, is a heparan sulfate proteoglycan bound to the cell membrane by glycosylphosphatidylinositol. This protein is expressed in the liver and the kidney of healthy fetuses but is hardly expressed in adults, except in the placenta. Contrarily, GPC3 is specifically expressed in hepatocellular carcinoma (HCC), ovarian clear cell carcinoma, melanoma, squamous cell carcinoma of the lung, hepatoblastoma, nephroblastoma (Wilms tumor), yolk sac tumor, and some pediatric cancers. Although the precise function of GPC3 remains unclear, it has been strongly suggested that it is related to the malignant transformation of HCC. We identified GPC3 as a promising target for cancer immunotherapy and have been working on the development of cancer immunotherapeutic agents targeting it through clinical trials. In some trials, it was revealed that the GPC3 peptide vaccines we developed using human leukocyte antigen-A24- and A2-restricted GPC3-derived peptides could induce GPC3-specific cytotoxic T cells in most vaccinated patients and thereby improve their prognosis. To further improve the clinical efficacy of cancer immunotherapy targeting GPC3, we are also developing next-generation therapeutic strategies using T cells engineered to express antigen-specific T-cell receptor or chimeric antigen receptor. In addition, we have successfully monitored the levels of serum full-length GPC3 protein, which is somehow secreted in the blood. The utility of GPC3 as a biomarker for predicting tumor recurrence and treatment efficacy is now being considered. In this review article, we summarize the results of clinical trials carried out by our team and describe the novel agent targeting the cancer-specific shared antigen, GPC3.
Collapse
Affiliation(s)
- Yasuhiro Shimizu
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
308
|
Kuhn NF, Purdon TJ, van Leeuwen DG, Lopez AV, Curran KJ, Daniyan AF, Brentjens RJ. CD40 Ligand-Modified Chimeric Antigen Receptor T Cells Enhance Antitumor Function by Eliciting an Endogenous Antitumor Response. Cancer Cell 2019; 35:473-488.e6. [PMID: 30889381 PMCID: PMC6428219 DOI: 10.1016/j.ccell.2019.02.006] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 11/16/2018] [Accepted: 02/14/2019] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cells provide great efficacy in B cell malignancies. However, improved CAR T cell therapies are still needed. Here, we engineered tumor-targeted CAR T cells to constitutively express the immune-stimulatory molecule CD40 ligand (CD40L) and explored efficacy in different mouse leukemia/lymphoma models. We observed that CD40L+ CAR T cells circumvent tumor immune escape via antigen loss through CD40/CD40L-mediated cytotoxicity and induction of a sustained, endogenous immune response. After adoptive cell transfer, the CD40L+ CAR T cells displayed superior antitumor efficacy, licensed antigen-presenting cells, enhanced recruitment of immune effectors, and mobilized endogenous tumor-recognizing T cells. These effects were absent in Cd40-/- mice and provide a rationale for the use of CD40L+ CAR T cells in cancer treatment.
Collapse
Affiliation(s)
- Nicholas F Kuhn
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Terence J Purdon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dayenne G van Leeuwen
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea V Lopez
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kevin J Curran
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anthony F Daniyan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
309
|
Jacoberger-Foissac C, Saliba H, Seguin C, Brion A, Kakhi Z, Frisch B, Fournel S, Heurtault B. Optimization of peptide-based cancer vaccine compositions, by sequential screening, using versatile liposomal platform. Int J Pharm 2019; 562:342-350. [PMID: 30880104 DOI: 10.1016/j.ijpharm.2019.03.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/01/2019] [Accepted: 03/02/2019] [Indexed: 12/15/2022]
Abstract
Therapeutic cancer vaccines need thoughtful design to efficiently deliver appropriate antigens and adjuvants to the immune system. In the current study, we took advantage of the versatility of a liposomal platform to conceive and customize vaccines containing three elements needed for the induction of efficient antitumor immunity: i) a CD4 epitope peptide able to activate CD4+ T helper cells, ii) a CD8 tumor-specific epitope peptide recognized by CD8+ T cytotoxic cells and iii) Pattern Recognition Receptor (PRR) agonists which stand as adjuvants. Each type of component, conjugated to liposomes, was evaluated individually by comparing their vaccine efficacy after immunization of naïve mice. These screening steps resulted in the optimization of three liposomal constructs bearing a peptide from HA influenza virus protein as CD4 epitope, a peptide from HPV16 E7 oncoprotein as CD8 epitope and TLR4, TLR2/6 or NOD1 agonists as adjuvant, which displayed antitumor efficiency against a mouse model of disseminated tumors transformed by HPV16. Our results validated the interest of our customizable liposomal platform as delivery system for cancer vaccination. We also demonstrated its interest as tool for vaccine design allowing the strategical selection of components, and the evaluation of epitope-adjuvant association.
Collapse
Affiliation(s)
- Célia Jacoberger-Foissac
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Hanadi Saliba
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Cendrine Seguin
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Anaïs Brion
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Zahra Kakhi
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Benoît Frisch
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Sylvie Fournel
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Béatrice Heurtault
- Université de Strasbourg, CNRS, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| |
Collapse
|
310
|
Som A, Mandaliya R, Alsaadi D, Farshidpour M, Charabaty A, Malhotra N, Mattar MC. Immune checkpoint inhibitor-induced colitis: A comprehensive review. World J Clin Cases 2019; 7:405-418. [PMID: 30842952 PMCID: PMC6397821 DOI: 10.12998/wjcc.v7.i4.405] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/21/2019] [Accepted: 01/26/2019] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that target down-regulators of the anti-cancer immune response: Cytotoxic T-lymphocyte antigen-4, programmed cell death protein-1, and its ligand programmed death-ligand 1. ICIs have revolutionized the treatment of a variety of malignancies. However, many immune-related adverse events have also been described which mainly occurs as the immune system becomes less suppressed, affecting various organs including the gastrointestinal tract and causing diarrhea and colitis. The incidence of immune-mediated colitis (IMC) ranges from 1%-25% depending on the type of ICI and if used in combination. Endoscopically and histologically there is a significant overlap between IMC and inflammatory bowel disease, however more neutrophilic inflammation without chronic inflammation is usually present in IMC. Corticosteroids are recommended for grade 2 or more severe colitis while holding the immunotherapy. About one third to two thirds of patients are steroid refractory and benefit from infliximab. Recently vedolizumab has been found to be efficacious in steroid and infliximab refractory cases. While in grade 4 colitis, the immunotherapy is permanently discontinued, the decision is controversial in grade 3 colitis.
Collapse
Affiliation(s)
- Aniruddh Som
- Department of Internal Medicine, Medstar Washington Hospital Center, Washington, DC 20010, United States
| | - Rohan Mandaliya
- Department of Gastroenterology, MedStar Georgetown University Hospital, Washington, DC 20007, United States
| | - Dana Alsaadi
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC 20007, United States
| | - Maham Farshidpour
- Department of Internal Medicine, MedStar Union Memorial Hospital and Good Samaritan Hospital, Baltimore, MD 21218, United States
| | - Aline Charabaty
- Department of Gastroenterology, Sibley Memorial Hospital, Washington, DC 20007, United States
| | - Nidhi Malhotra
- Department of Gastroenterology, MedStar Washington Hospital Center, Washington, DC 20010, United States
| | - Mark C Mattar
- Department of Gastroenterology, MedStar Georgetown University Hospital, Washington, DC 20007, United States
| |
Collapse
|
311
|
Salama R, Al-Obaidy KI, Perrino CM, Grignon DJ, Ulbright TM, Idrees MT. DOG1 immunohistochemical staining of testicular biopsies is a reliable tool for objective assessment of infertility. Ann Diagn Pathol 2019; 40:18-22. [PMID: 30849695 DOI: 10.1016/j.anndiagpath.2019.02.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 11/30/2022]
Abstract
Testicular biopsy may be a component of the work-up of male infertility. However, no reliable diagnostic tools are available for objective quantitative assessment of spermatogenic cells. It is well known that MAGE-A4 is selectively expressed in spermatogonia and our group has previously demonstrated that DOG1 differentially stains germ cells. Therefore, we performed DOG1 and a double stain cocktail (DOG1 and 57b murine monoclonal anti-MAGE-A4) immunohistochemical stains on 40 testicular infertility biopsies (10 each with active spermatogenesis, Sertoli cell-only, hypospermatogenesis, and maturation arrest), 25 benign seminiferous tubules from radical orchiectomies, and 5 spermatocytic tumors (ST). In biopsies/resections with active spermatogenesis, DOG1 stained spermatocytes and spermatids and was absent in spermatogonia, while MAGE-A4 stained spermatogonia and primary spermatocytes (weak). In hypospermatogenesis, DOG1 highlighted decreased spermatocytes/spermatids and MAGE-A4 highlighted decreased spermatogonia. DOG1 staining confirmed decreased to absent spermatocytes in maturation arrest and MAGE-A4 staining established the presence of preserved spermatogonia in all cases. All STs were negative for DOG1 and positive for MAGE-A4, while all Sertoli cell-only cases were negative for DOG1 and the double stain cocktail. In conclusion, we confirmed that DOG1 is expressed in spermatocytes and spermatids and MAGE-A4 highlights primarily spermatogonia. Usage of these stains facilitates confirmation of maturation arrest, assessment of the percentage of testis involvement in hypospermatogenesis and identification of mixed patterns. Finally, this study supports that the differentiation of STs is more closely related to spermatogonia than the more mature spermatocytes.
Collapse
Affiliation(s)
- Rasha Salama
- Department of Pathology, St. Elizabeth Healthcare, Edgewood, KY, USA
| | - Khaleel I Al-Obaidy
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - David J Grignon
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas M Ulbright
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Muhammad T Idrees
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
312
|
Margolis N, Markovits E, Markel G. Reprogramming lymphocytes for the treatment of melanoma: From biology to therapy. Adv Drug Deliv Rev 2019; 141:104-124. [PMID: 31276707 DOI: 10.1016/j.addr.2019.06.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 05/31/2019] [Accepted: 06/24/2019] [Indexed: 12/15/2022]
Abstract
This decade has introduced drastic changes in melanoma therapy, predominantly due to the materialization of the long promise of immunotherapy. Cytotoxic T cells are the chief component of the immune system, which are targeted by different strategies aimed to increase their capacity against melanoma cells. To this end, reprogramming of T cells occurs by T cell centered manipulation, targeting the immunosuppressive tumor microenvironment or altering the whole patient. These are enabled by delivery of small molecules, functional monoclonal antibodies, different subunit vaccines, as well as living lymphocytes, native or genetically engineered. Current FDA-approved therapies are focused on direct T cell manipulation, such as immune checkpoint inhibitors blocking CTLA-4 and/or PD-1, which paves the way for an effective immunotherapy backbone available for combination with other modalities. Here we review the biology and clinical developments that enable melanoma immunotherapy today and in the future.
Collapse
|
313
|
Jennings JJ, Mandaliya R, Nakshabandi A, Lewis JH. Hepatotoxicity induced by immune checkpoint inhibitors: a comprehensive review including current and alternative management strategies. Expert Opin Drug Metab Toxicol 2019; 15:231-244. [PMID: 30677306 DOI: 10.1080/17425255.2019.1574744] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) block cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1)/PD ligand 1 (PD-L1) receptors that control antitumor activities of lymphocytes. While highly efficacious, these drugs have been associated with several immune-related adverse events (irAEs) due to the disruption of self-tolerance. Immune-mediated hepatitis (IMH) usually presents as mild elevations of liver enzymes though it can rarely be associated with life-threatening hepatic injury. Areas covered: A comprehensive review was performed to define the clinicopathologic forms of liver injury associated with ICIs, comparing the various ICI classes as well as comparing this form of IMH with idiopathic autoimmune hepatitis and drug-induced autoimmune hepatitis. Liver biopsy has proven very useful in selected patients. A specific form of fibrin ring granulomatous hepatitis appears to be associated with IMH. The current societal treatment algorithms and emerging data were reviewed to determine when to utilize corticosteroids. Expert opinion: Monitoring for severe ICI-IMH is recommended although acute liver failure remains rare. Most patients with grade 3-4 hepatotoxicity respond to corticosteroids, but a subset of patients with mild hepatitis on liver biopsy resolve without steroids and need to be carefully selected in concert with the consultation of a hepatologist.
Collapse
Affiliation(s)
- Joseph J Jennings
- a Department of Medicine, Division of Gastroenterology and Hepatology , MedStar Georgetown University Hospital , Washington , DC , USA
| | - Rohan Mandaliya
- a Department of Medicine, Division of Gastroenterology and Hepatology , MedStar Georgetown University Hospital , Washington , DC , USA
| | - Ahmad Nakshabandi
- b Department of Internal Medicine , Mercy Hospital and Medical Center , Chicago , IL , USA
| | - James H Lewis
- a Department of Medicine, Division of Gastroenterology and Hepatology , MedStar Georgetown University Hospital , Washington , DC , USA
| |
Collapse
|
314
|
Han L, Tang M, Xu X, Jiang B, Wei Y, Qian H, Lu X. MiR-143-3p suppresses cell proliferation, migration, and invasion by targeting Melanoma-Associated Antigen A9 in laryngeal squamous cell carcinoma. J Cell Biochem 2019; 120:1245-1257. [PMID: 30302801 DOI: 10.1002/jcb.27084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 04/26/2018] [Indexed: 01/24/2023]
Abstract
Previously we found that melanoma-associated antigen-A9 (MAGE-A9) was a significantly upregulated biomarker in laryngeal squamous cell carcinoma (LSCC). A high expression of MAGE-A9 indicates an unfavorable survival outcome, and the MAGE-A9 expression level is an independent prognostic factor of LSCC. To explore the mechanism of MAGE-A9 upregulation, several predicted regulatory microRNAs were screened and validated in LSCC cells. In the current study, we found that miR-143-3p (MAGE-A9 related miRNAs) expression levels correlated negatively with the MAGE-A9 protein expression in LSCC tissues. Dual-luciferase reporter assays and Western blot analysis revealed MAGE-A9 to be a direct target of miR-143-3p. Furthermore, a series of in vitro gain- and loss-of-function assays revealed that miR-143-3p inhibited LSCC cell proliferation, migration, and invasion. Also, miR-143-3p suppressed LSCC tumorigenesis in vivo. These effects were clinically relevant, as a lower expression of miR-143-3p occurred in severer clinical stages and represented poor overall survival in patients with LSCC. Taken together, these results suggest that downregulation of miR-143-3p contributes to tumor progression through upregulation of MAGE-A9. The expression level of these two key molecules maintained LSCC progression, thus, highlighting the potential of miR-143-3p as a therapeutic target for human LSCC.
Collapse
Affiliation(s)
- Liang Han
- Department of Head and Neck Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mingming Tang
- Department of Head and Neck Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xinjiang Xu
- Department of Head and Neck Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Bin Jiang
- Department of Head and Neck Surgery, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yingze Wei
- Department of Clinical Pathology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hongyan Qian
- Cancer Research Center, Nantong Tumor Hospi tal, Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xueguan Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
315
|
Antigenic Targets for the Immunotherapy of Acute Myeloid Leukaemia. J Clin Med 2019; 8:jcm8020134. [PMID: 30678059 PMCID: PMC6406328 DOI: 10.3390/jcm8020134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 01/10/2019] [Accepted: 01/20/2019] [Indexed: 12/18/2022] Open
Abstract
One of the most promising approaches to preventing relapse is the stimulation of the body’s own immune system to kill residual cancer cells after conventional therapy has destroyed the bulk of the tumour. In acute myeloid leukaemia (AML), the high frequency with which patients achieve first remission, and the diffuse nature of the disease throughout the periphery, makes immunotherapy particularly appealing following induction and consolidation therapy, using chemotherapy, and where possible stem cell transplantation. Immunotherapy could be used to remove residual disease, including leukaemic stem cells from the farthest recesses of the body, reducing, if not eliminating, the prospect of relapse. The identification of novel antigens that exist at disease presentation and can act as targets for immunotherapy have also proved useful in helping us to gain a better understand of the biology that belies AML. It appears that there is an additional function of leukaemia associated antigens as biomarkers of disease state and survival. Here, we discuss these findings.
Collapse
|
316
|
Xu Y, Salazar GT, Zhang N, An Z. T-cell receptor mimic (TCRm) antibody therapeutics against intracellular proteins. Antib Ther 2019; 2:22-32. [PMID: 33928218 PMCID: PMC7990144 DOI: 10.1093/abt/tbz001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 01/03/2019] [Accepted: 01/10/2019] [Indexed: 12/21/2022] Open
Abstract
T-cell receptor mimic (TCRm) antibodies combine the capacity of a T cell to target intracellular antigens with other capacities unique to antibodies. Neoantigens are abnormal proteins that arise as a consequence of somatic mutations. Technological advances promote the development of neoantigen-targeting therapies including TCRm antibody therapies. This review summarizes key characteristics of TCRm antibodies, in particular those targeting neoantigens, and further introduces discussion of obstacles that must be overcome to advance TCRm therapeutics.
Collapse
Affiliation(s)
- Yixiang Xu
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Georgina To'a Salazar
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
317
|
Xie K, Fu C, Wang S, Xu H, Liu S, Shao Y, Gong Z, Wu X, Xu B, Han J, Xu J, Xu P, Jia X, Wu J. Cancer-testis antigens in ovarian cancer: implication for biomarkers and therapeutic targets. J Ovarian Res 2019; 12:1. [PMID: 30609934 PMCID: PMC6318940 DOI: 10.1186/s13048-018-0475-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/11/2018] [Indexed: 12/21/2022] Open
Abstract
Ovarian cancer remains the most fatal gynecologic malignancy worldwide due to delayed diagnosis as well as recurrence and drug resistance. Thus, the development of new tumor-related molecules with high sensitivity and specificity to replace or supplement existing tools is urgently needed. Cancer-testis antigens (CTAs) are exclusively expressed in normal testis tissues but abundantly found in several types of cancers, including ovarian cancer. Numerous novel CTAs have been identified by high-throughput sequencing techniques, and some aberrantly expressed CTAs are associated with ovarian cancer initiation, clinical outcomes and chemotherapy resistance. More importantly, CTAs are immunogenic and may be novel targets for antigen-specific immunotherapy in ovarian cancer. In this review, we attempt to characterize the expression of candidate CTAs in ovarian cancer and their clinical significance as biomarkers, activation mechanisms, function in malignant phenotypes and applications in immunotherapy.
Collapse
Affiliation(s)
- Kaipeng Xie
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China.
| | - Chenyang Fu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Suli Wang
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Hanzi Xu
- Jiangsu Institute of Cancer Research The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Siyu Liu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Yang Shao
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Zhen Gong
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Xiaoli Wu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, 211166, China
| | - Jing Han
- Jiangsu Institute of Cancer Research The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Juan Xu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Pengfei Xu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China
| | - Xuemei Jia
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China.
| | - Jiangping Wu
- The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, 210004, China.
| |
Collapse
|
318
|
Abstract
This chapter focuses on the discovery of the Major Histocompatibility Complex (MHC) in mice (H-2) and in humans (HLA), and on the role played by the International HLA Workshops in the analysis and characterization of this complex genetic system. The early days of Tumour Immunology and the importance of the definition of Tumour Associated Transplantation Antigens (TATA) are also discussed. Today we know that tumour cells can be killed by T lymphocytes by recognizing tumour antigenic peptides presented by MHC molecules and they can also escape this recognition by losing the expression of MHC molecules. This important phenomenon has been profoundly studied for many years both in my lab in Granada and in other laboratories. The results of this research have important implications for the new generation of cancer immunotherapy that boosts T cell responses. A historical perspective of major discoveries is presented in this chapter, with the names of the scientists that have made a significant contribution to the enormous progress made in the field of Tumour Immunology.
Collapse
Affiliation(s)
- Federico Garrido
- Departamento de Analisis Clinicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Facultad de Medicina, Universidad de Granada, Granada, Spain
| |
Collapse
|
319
|
Abstract
Monoclonal antibodies (mAbs) are immunoglobulins designed to target a specific epitope on an antigen. Immunoglobulins of identical amino-acid sequence were originally produced by hybridomas grown in culture and, subsequently, by recombinant DNA technology using mammalian cell expression systems. The antigen-binding region of the mAb is formed by the variable domains of the heavy and light chains and contains the complementarity-determining region that imparts the high specificity for the target antigen. The pharmacokinetics of mAbs involves target-mediated and non-target-related factors that influence their disposition.Preclinical safety evaluation of mAbs differs substantially from that of small molecular (chemical) entities. Immunogenicity of mAbs has implications for their pharmacokinetics and safety. Early studies of mAbs in humans require careful consideration of the most suitable study population, route/s of administration, starting dose, study design and the potential difference in pharmacokinetics in healthy subjects compared to patients expressing the target antigen.Of the ever-increasing diversity of therapeutic indications for mAbs, we have concentrated on two that have proved dramatically successful. The contribution that mAbs have made to the treatment of inflammatory conditions, in particular arthritides and inflammatory bowel disease, has been nothing short of revolutionary. Their benefit has also been striking in the treatment of solid tumours and, most recently, as immunotherapy for a wide variety of cancers. Finally, we speculate on the future with various new approaches to the development of therapeutic antibodies.
Collapse
|
320
|
Haider C, Hnat J, Wagner R, Huber H, Timelthaler G, Grubinger M, Coulouarn C, Schreiner W, Schlangen K, Sieghart W, Peck‐Radosavljevic M, Mikulits W. Transforming Growth Factor-β and Axl Induce CXCL5 and Neutrophil Recruitment in Hepatocellular Carcinoma. Hepatology 2019; 69:222-236. [PMID: 30014484 PMCID: PMC6590451 DOI: 10.1002/hep.30166] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/06/2018] [Indexed: 12/24/2022]
Abstract
Transforming growth factor (TGF)-β suppresses early hepatocellular carcinoma (HCC) development but triggers pro-oncogenic abilities at later stages. Recent data suggest that the receptor tyrosine kinase Axl causes a TGF-β switch toward dedifferentiation and invasion of HCC cells. Here, we analyzed two human cellular HCC models with opposing phenotypes in response to TGF-β. Both HCC models showed reduced proliferation and clonogenic growth behavior following TGF-β stimulation, although they exhibited differences in chemosensitivity and migratory abilities, suggesting that HCC cells evade traits of anti-oncogenic TGF-β. Transcriptome profiling revealed differential regulation of the chemokine CXCL5, which positively correlated with TGF-β expression in HCC patients. The expression and secretion of CXCL5 was dependent on Axl expression, suggesting that CXCL5 is a TGF-β target gene collaborating with Axl signaling. Loss of either TGF-β or Axl signaling abrogated CXCL5-dependent attraction of neutrophils. In mice, tumor formation of transplanted HCC cells relied on CXCL5 expression. In HCC patients, high levels of Axl and CXCL5 correlated with advanced tumor stages, recruitment of neutrophils into HCC tissue, and reduced survival. Conclusion: The synergy of TGF-β and Axl induces CXCL5 secretion, causing the infiltration of neutrophils into HCC tissue. Intervention with TGF-β/Axl/CXCL5 signaling may be an effective therapeutic strategy to combat HCC progression in TGF-β-positive patients.
Collapse
Affiliation(s)
- Christine Haider
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Julia Hnat
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Roland Wagner
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Heidemarie Huber
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Gerald Timelthaler
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Markus Grubinger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| | - Cédric Coulouarn
- INSERM, University of Rennes, INRA, Institute NUMECAN (Nutrition Metabolisms and Cancer)UMR_A 1341, UMR_S 1241RennesFrance
| | - Wolfgang Schreiner
- Division of Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS)Medical University of ViennaViennaAustria
| | - Karin Schlangen
- Division of Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS)Medical University of ViennaViennaAustria
| | - Wolfgang Sieghart
- Department of Internal Medicine III, Division of Gastroenterology and HepatologyMedical University of ViennaViennaAustria
| | - Markus Peck‐Radosavljevic
- Department of Internal Medicine III, Division of Gastroenterology and HepatologyMedical University of ViennaViennaAustria
| | - Wolfgang Mikulits
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center ViennaMedical University of ViennaViennaAustria
| |
Collapse
|
321
|
Abstract
In this chapter I describe Tumour Immune Escape mechanisms associated with MHC/HLA class I loss in human and experimental tumours. Different altered HLA class-I phenotypes can be observed that are produced by different molecular mechanisms. Experimental and histological evidences are summarized indicating that at the early stages of tumour development there is an enormous variety of tumour clones with different MHC class I expression patterns. This phase is followed by a strong T cell mediated immune-selection of MHC/HLA class-I negative tumour cells in the primary tumour lesion. This transition period results in a formation of a tumour composed only of HLA-class I negative cells. An updated description of this process observed in a large variety of human tumors is included. In the second section I focus on MHC/HLA class I alterations observed in mouse and human metastases, and describe the generation of different tumor cell clones with altered MHC class I phenotypes, which could be similar or different from the original tumor clone. The biological and immunological relevance of these observations is discussed. Finally, the interesting phenomenon of metastatic dormancy is analyzed in association with a particular MHC class I negative tumor phenotype.
Collapse
Affiliation(s)
- Federico Garrido
- Departamento de Analisis Clinicos e Inmunologia, Hospital Universitario Virgen de las Nieves, Facultad de Medicina, Universidad de Granada, Granada, Spain
| |
Collapse
|
322
|
Pender A, Jones RL, Pollack S. Optimising Cancer Vaccine Design in Sarcoma. Cancers (Basel) 2018; 11:E1. [PMID: 30577459 PMCID: PMC6356514 DOI: 10.3390/cancers11010001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/15/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022] Open
Abstract
Immunotherapeutics are increasingly recognized as a key tool in the armamentarium against malignancy. The success of immune checkpoint-targeting drugs and adoptive cell therapy has refocused attention on the potential anti-cancer effect of eliciting a tumour-specific immunological response. Sarcomas are a rare and diverse group of tumours with a limited prognosis in advanced disease despite systemic therapeutics. Various vaccine strategies including peptide vaccines against cancer testis antigens, dendritic cell vaccines, and viral vectors have been trialled in sarcoma with growing evidence of efficacy. Here, we review the principles of successful vaccine development and how these have been applied thus far to the treatment of sarcoma.
Collapse
Affiliation(s)
- Alexandra Pender
- Sarcoma Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
| | - Robin L Jones
- Sarcoma Unit, Department of Medicine, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK.
- Division of Clinical Studies, The Institute of Cancer Research, London SW3 6JB, UK.
| | - Seth Pollack
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA 98109, USA.
- Division of Oncology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
323
|
Fu C, Jiang A. Dendritic Cells and CD8 T Cell Immunity in Tumor Microenvironment. Front Immunol 2018; 9:3059. [PMID: 30619378 PMCID: PMC6306491 DOI: 10.3389/fimmu.2018.03059] [Citation(s) in RCA: 397] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/10/2018] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) play a central role in the regulation of the balance between CD8 T cell immunity vs. tolerance to tumor antigens. Cross-priming, a process which DCs activate CD8 T cells by cross-presenting exogenous antigens, plays a critical role in generating anti-tumor CD8 T cell immunity. However, there are compelling evidences now that the tumor microenvironment (TME)-mediated suppression and modulation of tumor-infiltrated DCs (TIDCs) impair their function in initiating potent anti-tumor immunity and even promote tumor progression. Thus, DC-mediated cross-presentation of tumor antigens in tumor-bearing hosts often induces T cell tolerance instead of immunity. As tumor-induced immunosuppression remains one of the major hurdles for cancer immunotherapy, understanding how DCs regulate anti-tumor CD8 T cell immunity in particular within TME has been under intensive investigation. Recent reports on the Batf3-dependent type 1 conventional DCs (cDC1s) in anti-tumor immunity have greatly advanced our understanding on the interplay of DCs and CD8 T cells in the TME, highlighted by the critical role of CD103+ cDC1s in the cross-priming of tumor antigen-specific CD8 T cells. In this review, we will discuss recent advances in anti-tumor CD8 T cell cross-priming by CD103+ cDC1s in TME, and share perspective on future directions including therapeutic applications and memory CD8 T cell responses.
Collapse
Affiliation(s)
- Chunmei Fu
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Aimin Jiang
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
324
|
Phung CD, Nguyen HT, Tran TH, Choi HG, Yong CS, Kim JO. Rational combination immunotherapeutic approaches for effective cancer treatment. J Control Release 2018; 294:114-130. [PMID: 30553850 DOI: 10.1016/j.jconrel.2018.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022]
Abstract
Immunotherapy is an important mode of cancer treatment. Over the past decades, immunotherapy has improved the clinical outcome for cancer patients. However, in many cases, mutations in cancer cells, lack of selectivity, insufficiency of tumor-reactive T cells, and host immunosuppression limit the clinical benefit of immunotherapy. Combination approaches in immunotherapy may overcome these obstacles. Accumulating evidence demonstrates that combination immunotherapy is the future of cancer treatment. However, designing safe and rational combinations of immunotherapy with other treatment modalities is critical. This review will discuss the optimal immunotherapy-based combinations mainly with respect to the mechanisms of action of individual therapeutic agents that target multiple steps in evasion and progression of tumor.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
325
|
Locy H, de Mey S, de Mey W, De Ridder M, Thielemans K, Maenhout SK. Immunomodulation of the Tumor Microenvironment: Turn Foe Into Friend. Front Immunol 2018; 9:2909. [PMID: 30619273 PMCID: PMC6297829 DOI: 10.3389/fimmu.2018.02909] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/27/2018] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy, where the patient's own immune system is exploited to eliminate tumor cells, has become one of the most prominent new cancer treatment options in the last decade. The main hurdle for classical cancer vaccines is the need to identify tumor- and patient specific antigens to include in the vaccine. Therefore, in situ vaccination represents an alternative and promising approach. This type of immunotherapy involves the direct intratumoral administration of different immunomodulatory agents and uses the tumor itself as the source of antigen. The ultimate aim is to convert an immunodormant tumor microenvironment into an immunostimulatory one, enabling the immune system to eradicate all tumor lesions in the body. In this review we will give an overview of different strategies, which can be exploited for the immunomodulation of the tumor microenvironment and their emerging role in the treatment of cancer patients.
Collapse
Affiliation(s)
- Hanne Locy
- Laboratory of Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Sven de Mey
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Wout de Mey
- Laboratory of Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Mark De Ridder
- Department of Radiotherapy, UZ Brussel, Vrije Universiteit Brussel, Brussels, Belgium
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Sarah K. Maenhout
- Laboratory of Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
326
|
Park S, Kim D, Wu G, Jung H, Park JA, Kwon HJ, Lee Y. A peptide-CpG-DNA-liposome complex vaccine targeting TM4SF5 suppresses growth of pancreatic cancer in a mouse allograft model. Onco Targets Ther 2018; 11:8655-8672. [PMID: 30584324 PMCID: PMC6284540 DOI: 10.2147/ott.s186606] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background Patients with pancreatic cancer have a poor prognosis and are usually diagnosed at a late stage. Because TM4SF5 is known to be overexpressed in hepatocellular carcinoma, colon cancer, and pancreatic cancer, it is considered as one of the candidate molecular targets for an anticancer strategies. Purpose The purpose of this study was to evaluate possible utility of TM4SF5 to treat pancreatic cancer using a mouse allograft model. Materials and methods We analyzed expression of TM4SF5 in pancreatic cancer tissues using immunohistochemistry. We established a mouse pancreatic cancer cell line stably expressing TM4SF5 and identified the effect of TM4SF5 expression in vitro. We used the CpG-DNA-peptide-liposome complex as a peptide vaccine and investigated antitumor effects of the vaccine in a mouse model with TM4SF5 expressing pancreatic cells. To investigate the function of produced antibody, we evaluated effects of the anti-TM4SF5 monoclonal antibody in vitro in terms of cell growth and migration properties. Results Immunohistochemical analysis showed that 36.4% of pancreatic cancer tissue samples expressed TM4SF5. Expression of TM4SF5 induced increased cell proliferation and motility in vitro. Injection of the TM4SF5 peptide vaccine induced the production of anti-hTM4SF5 antibodies and reduced the growth of pancreatic tumors in mice established by subcutaneous injection of the TM4SF5-expressing mouse pancreatic cancer cell line. The treatment of TM4SF5-expressing cells with the anti-hTM4SF5 monoclonal antibody reduced cell growth, modulated the expression of the epithelial–mesenchymal transition markers Vimentin and E-cadherin, and decreased cell motility in vitro. Conclusion Our results showed that the TM4SF5 peptide vaccine had a protective effect against pancreatic tumors expressing TM4SF5, and this effect was mediated, at least in part, by the production and suppressive function of the anti-TM4SF5 antibodies. Therefore, we suggest that targeting TM4SF5 could be a novel strategy to prevent or treat pancreatic cancer.
Collapse
Affiliation(s)
- Sangkyu Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea, .,Biotechnology Research Institute, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea,
| | - Dongbum Kim
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea,
| | - Guang Wu
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea, .,School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China,
| | - Harry Jung
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea,
| | - Jeong-A Park
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea, .,Biotechnology Research Institute, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea,
| | - Hyung-Joo Kwon
- Center for Medical Science Research, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea, .,Department of Microbiology, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea, .,Biotechnology Research Institute, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea,
| |
Collapse
|
327
|
Duperret EK, Liu S, Paik M, Trautz A, Stoltz R, Liu X, Ze K, Perales-Puchalt A, Reed C, Yan J, Xu X, Weiner DB. A Designer Cross-reactive DNA Immunotherapeutic Vaccine that Targets Multiple MAGE-A Family Members Simultaneously for Cancer Therapy. Clin Cancer Res 2018; 24:6015-6027. [PMID: 30262507 PMCID: PMC6319943 DOI: 10.1158/1078-0432.ccr-18-1013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 07/13/2018] [Accepted: 08/28/2018] [Indexed: 01/07/2023]
Abstract
PURPOSE Cancer/testis antigens have emerged as attractive targets for cancer immunotherapy. Clinical studies have targeted MAGE-A3, a prototype antigen that is a member of the MAGE-A family of antigens, in melanoma and lung carcinoma. However, these studies have not yet had a significant impact due to poor CD8+ T-cell immunogenicity, platform toxicity, or perhaps limited target antigen availability. In this study, we develop an improved MAGE-A immunogen with cross-reactivity to multiple family members. EXPERIMENTAL DESIGN In this study, we analyzed MAGE-A expression in The Cancer Genome Atlas and observed that many patients express multiple MAGE-A isoforms, not limited to MAGE-A3, simultaneously in diverse tumors. On the basis of this, we designed an optimized consensus MAGE-A DNA vaccine capable of cross-reacting with many MAGE-A isoforms, and tested immunogenicity and antitumor activity of this vaccine in a relevant autochthonous melanoma model. RESULTS Immunization of this MAGE-A vaccine by electroporation in C57Bl/6 mice generated robust IFNγ and TNFα CD8+ T-cell responses as well as cytotoxic CD107a/IFNγ/T-bet triple-positive responses against multiple isoforms. Furthermore, this MAGE-A DNA immunogen generated a cross-reactive immune response in 14 of 15 genetically diverse, outbred mice. We tested the antitumor activity of this MAGE-A DNA vaccine in Tyr::CreER;BRAFCa/+;Ptenlox/lox transgenic mice that develop melanoma upon tamoxifen induction. The MAGE-A DNA therapeutic vaccine significantly slowed tumor growth and doubled median mouse survival. CONCLUSIONS These results support the clinical use of consensus MAGE-A immunogens with the capacity to target multiple MAGE-A family members to prevent tumor immune escape.
Collapse
Affiliation(s)
| | - Shujing Liu
- The University of Pennsylvania Department of Pathology and Laboratory Medicine, Philadelphia PA
| | - Megan Paik
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | - Aspen Trautz
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | - Regina Stoltz
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | - Xiaoming Liu
- The University of Pennsylvania Department of Pathology and Laboratory Medicine, Philadelphia PA
| | - Kan Ze
- The University of Pennsylvania Department of Pathology and Laboratory Medicine, Philadelphia PA
| | | | | | - Jian Yan
- Inovio Pharmaceuticals, Plymouth Meeting, PA
| | - Xiaowei Xu
- The University of Pennsylvania Department of Pathology and Laboratory Medicine, Philadelphia PA
| | - David B. Weiner
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA,Corresponding author: David B. Weiner, Vaccine & Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104,
| |
Collapse
|
328
|
Perspective: cancer vaccines in the era of immune checkpoint blockade. Mamm Genome 2018; 29:703-713. [PMID: 30446791 PMCID: PMC6267701 DOI: 10.1007/s00335-018-9786-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 10/24/2018] [Indexed: 12/30/2022]
Abstract
Current excitement about cancer immunotherapy is the result of unprecedented clinical impact from immune checkpoint inhibitors, particularly those that target programmed death (PD)-1 and PD-ligand (L)-1. Numerous other immunotherapeutics are also finding their way into the clinic either alone or in combination, and these have potential applications in many cancer types. Therapeutic cancer vaccines have been a major focus for many pioneers in the field yet have largely failed to live up to expectations as game-changing immunotherapeutics. This, despite decades of focussed efforts that have identified antigens, optimised adjuvants and refined approaches to pre-clinical modelling and clinical monitoring. If antigen-directed immunotherapeutics are to take a place in the anti-cancer therapeutic armamentarium, it will be crucial to understand the potential niche that could be occupied by cancer vaccines that can specifically induce or modify immune response against cancer antigens.
Collapse
|
329
|
Lu JH, Peng BY, Chang CC, Dubey NK, Lo WC, Cheng HC, Wang JR, Wei HJ, Deng WP. Tumor-Targeted Immunotherapy by Using Primary Adipose-Derived Stem Cells and an Antigen-Specific Protein Vaccine. Cancers (Basel) 2018; 10:E446. [PMID: 30445793 PMCID: PMC6266266 DOI: 10.3390/cancers10110446] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
Cancer is a leading cause of mortality and a major public health problem worldwide. For biological therapy against cancer, we previously developed a unique immunotherapeutic platform by combining mesenchymal stem cells with an antigen-specific protein vaccine. However, this system possesses a few limitations, such as improperly immortalized mesenchymal stem cells (MSCs) along with transfected oncogenic antigens in them. To overcome the limitations of this platform for future clinical application, we freshly prepared primary adipose-derived stem cells (ADSCs) and modified the E7' antigen (E7') as a non-oncogenic protein. Either subcutaneously co-inoculated with cancer cells or systemically administered after tumor growth, ADSC labeled with enhanced green fluorescent protein (eGFP) and combined with modified E7' (ADSC-E7'-eGFP) cells showed significant antitumor activity when combined with the protein vaccine in both colon and lung cancer in mice. Specifically, this combined therapy inhibited tumor through inducing cell apoptosis. The significantly reduced endothelial cell markers, CD31 and vascular endothelial growth factor (VEGF), indicated strongly inhibited tumor angiogenesis. The activated immune system was demonstrated through the response of CD4+ T and natural killer (NK) cells, and a notable antitumor activity might be contributed by CD8+ T cells. Conclusively, these evidences imply that this promising immunotherapeutic platform might be a potential candidate for the future clinical application against cancer.
Collapse
Affiliation(s)
- Jui-Hua Lu
- Graduate Institute of Biomedical Materials and Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan.
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Bou-Yue Peng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Dentistry, Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 110i, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University School of Medicine, Taipei 110, Taiwan.
| | - Navneet Kumar Dubey
- Ceramics and Biomaterials Research Group, Advanced Institute of Materials Science, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam.
| | - Wen-Cheng Lo
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan.
- Division of Neurosurgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsin-Chung Cheng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Joseph R Wang
- Department of Periodontics, College of Dental Medicine, Columbia University, New York 10032, USA.
| | - Hong-Jian Wei
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Win-Ping Deng
- Stem Cell Research Center, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Graduate Institute of Basic Medicine, Fu Jen Catholic University, New Taipei City 110, Taiwan.
| |
Collapse
|
330
|
Fan J, Shang D, Han B, Song J, Chen H, Yang JM. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Am J Cancer Res 2018; 8:5784-5800. [PMID: 30555581 PMCID: PMC6276301 DOI: 10.7150/thno.29035] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed significant advances in the adoptive cell transfer (ACT) technique, which has been appreciated as one of the most promising treatments for patients with cancer. Utilization of ACT can enhance the function of the immune system or improve the specificity and persistence of transferred cells. Various immune cells including T lymphocytes, natural killer cells, dendritic cells, and even stem cells can be used in the ACT despite their different functional mechanisms. Colorectal cancer (CRC) is among the most common malignancies and causes millions of deaths worldwide every year. In this review, we discuss the status and perspective of the ACT in the treatment of CRC.
Collapse
|
331
|
Chakraborty P, Karmakar T, Arora N, Mukherjee G. Immune and genomic signatures in oral (head and neck) cancer. Heliyon 2018; 4:e00880. [PMID: 30417146 PMCID: PMC6218671 DOI: 10.1016/j.heliyon.2018.e00880] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/22/2018] [Accepted: 10/20/2018] [Indexed: 12/25/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is responsible for a large number of deaths each year. Oral cancer is the most frequent subtype of HNSCC. Historically, oral cancer has been associated with an increase in the consumption of tobacco and alcohol products, seen especially in the Asian subcontinent. It has also been associated with infection by the human papilloma virus (HPV), particularly strain HPV16. Treatment usually involves a multidisciplinary approach of surgery combined with chemotherapy and radiation. The advent of immunotherapy has broadened the scope for treatment. A better immune response to the tumour can also elicit the action of other therapeutic approaches. A heightened immune response, on the other hand, can lead to resistant tumour formation through the process of immunoediting. Molecular profiling of the tumour microenvironment (TME) can provide us with better insight into the mechanism and progression of the disease, ultimately opening up new therapeutic options. High-throughput molecular profiling techniques over the past decade have enabled us to appreciate the heterogeneity of the TME. In this review, we will be describing the clinicopathological role of the immune and genomic landscape in oral cancer. This study will update readers on the several immunological and genetic factors that can play an important function as predictive and prognostic biomarkers in various forms of head and neck cancer, with a special emphasis on oral carcinoma.
Collapse
|
332
|
Altmann DM. A Nobel Prize-worthy pursuit: cancer immunology and harnessing immunity to tumour neoantigens. Immunology 2018; 155:283-284. [PMID: 30320408 PMCID: PMC6187215 DOI: 10.1111/imm.13008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The field of cancer immunology stepped into the limelight this year when James P. Allison and Tasuku Honjo received the Nobel Prize in Physiology or Medicine for their discovery of cancer therapy by inhibition of negative immune regulation. Among many exciting advances contributing to the coming of age of tumour immunology as a viable clinical specialty has been the ability to progress from the initial elucidation of tumour antigens, such as the melanoma antigen, MAGE-1, to high-throughput sequencing facilitating identification of T cell epitopes from diverse tumour neoantigens. This has resulted from the convergence of expertise in tumour biology, next-generation sequencing, T cell and structural immunology, and predictive algorithms. Among many examples, immunotherapy for ovarian cancer has been one of the beneficiaries of these advances, leading to a number of recent and ongoing clinical trials.
Collapse
|
333
|
Kissick HT. Is It Possible to Develop Cancer Vaccines to Neoantigens, What Are the Major Challenges, and How Can These Be Overcome? Neoantigens as Vaccine Targets for Cancer. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a033704. [PMID: 29254981 DOI: 10.1101/cshperspect.a033704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent work by several groups has undoubtedly shown that we can produce cancer vaccines targeting neoantigens. However, each vaccine is essentially a single-use, patient-specific product, making this approach resource-intensive. For this reason, it is important to ask whether this approach will be any more successful than what has been attempted during the last 30 years using vaccines targeting self-epitopes. Here, we discuss what might be expected from neoantigen vaccines based on our experience in chronic viral infections, and how this new approach may be applied to cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Departments of Urology and Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
334
|
Sharma P, Debinski W. Receptor-Targeted Glial Brain Tumor Therapies. Int J Mol Sci 2018; 19:E3326. [PMID: 30366424 PMCID: PMC6274942 DOI: 10.3390/ijms19113326] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022] Open
Abstract
Among primary brain tumors, malignant gliomas are notably difficult to manage. The higher-grade tumors represent an unmet need in medicine. There have been extensive efforts to implement receptor-targeted therapeutic approaches directed against gliomas. These approaches include immunotherapies, such as vaccines, adoptive immunotherapy, and passive immunotherapy. Targeted cytotoxic radio energy and pro-drug activation have been designed specifically for brain tumors. The field of targeting through receptors progressed significantly with the discovery of an interleukin 13 receptor alpha 2 (IL-13RA2) as a tumor-associated receptor over-expressed in most patients with glioblastoma (GBM) but not in normal brain. IL-13RA2 has been exploited in novel experimental therapies with very encouraging clinical responses. Other receptors are specifically over-expressed in many patients with GBM, such as EphA2 and EphA3 receptors, among others. These findings are important in view of the heterogeneity of GBM tumors and multiple tumor compartments responsible for tumor progression and resistance to therapies. The combined targeting of multiple receptors in different tumor compartments should be a preferred way to design novel receptor-targeted therapeutic approaches in gliomas.
Collapse
Affiliation(s)
- Puja Sharma
- Brain Tumor Center of Excellence, Department of Cancer Biology, Wake Forest University School of Medicine, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| | - Waldemar Debinski
- Brain Tumor Center of Excellence, Department of Cancer Biology, Wake Forest University School of Medicine, Comprehensive Cancer Center of Wake Forest Baptist Medical Center, 1 Medical Center Boulevard, Winston-Salem, NC 27157, USA.
| |
Collapse
|
335
|
Gao Y, Kardos J, Yang Y, Tamir TY, Mutter-Rottmayer E, Weissman B, Major MB, Kim WY, Vaziri C. The Cancer/Testes (CT) Antigen HORMAD1 promotes Homologous Recombinational DNA Repair and Radioresistance in Lung adenocarcinoma cells. Sci Rep 2018; 8:15304. [PMID: 30333500 PMCID: PMC6192992 DOI: 10.1038/s41598-018-33601-w] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/01/2018] [Indexed: 12/24/2022] Open
Abstract
The Cancer/Testes (CT) Antigen HORMAD1 is germ cell-restricted and plays developmental roles in generation and processing of meiotic DNA Double Strand Breaks (DSB). Many tumors aberrantly overexpress HORMAD1 yet the potential impact of this CT antigen on cancer biology is unclear. We tested a potential role of HORMAD1 in genome maintenance in lung adenocarcinoma cells. We show that HORMAD1 re-distributes to nuclear foci and co-localizes with the DSB marker γH2AX in response to ionizing radiation (IR) and chemotherapeutic agents. The HORMA domain and C-term disordered oligomerization motif are necessary for localization of HORMAD1 to IR-induced foci (IRIF). HORMAD1-depleted cells are sensitive to IR and camptothecin. In reporter assays, Homologous Recombination (HR)-mediated repair of targeted ISce1-induced DSBs is attenuated in HORMAD1-depleted cells. In Non-Homologous End Joining (NHEJ) reporter assays, HORMAD1-depletion does not affect repair of ISce1-induced DSB. Early DSB signaling events (including ATM phosphorylation and formation of γH2AX, 53BP1 and NBS1 foci) are intact in HORMAD1-depleted cells. However, generation of RPA-ssDNA foci and redistribution of RAD51 to DSB are compromised in HORMAD1-depleted cells, suggesting that HORMAD1 promotes DSB resection. HORMAD1-mediated HR is a neomorphic activity that is independent of its meiotic partners (including HORMAD2 and CCDC36. Bioinformatic analysis of TCGA data show that similar to known HR pathway genes HORMAD1 is overexpressed in lung adenocarcinomas. Overexpression of HR genes is associated with specific mutational profiles (including copy number variation). Taken together, we identify HORMAD1-dependent DSB repair as a new mechanism of radioresistance and a probable determinant of mutability in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yanzhe Gao
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA
| | - Jordan Kardos
- Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yang Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA
| | - Tigist Y Tamir
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Elizabeth Mutter-Rottmayer
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA
| | - Bernard Weissman
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA.,Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - William Y Kim
- Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, 614 Brinkhous-Bullitt Building, Chapel Hill, NC, 27599, USA. .,Lineberger Comprehensive Cancer Center, Curriculum in Genetics and Molecular Biology, and Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
336
|
Rao S, Gharib K, Han A. Cancer Immunosurveillance by T Cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 342:149-173. [PMID: 30635090 DOI: 10.1016/bs.ircmb.2018.08.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy is based on the ability of the immune system to recognize tumors as foreign tissue. The idea of cancer immunosurveillance was first conceived over a century ago but remained controversial through much of the 20th century. In the past few decades, however, the field has progressed rapidly, and the concept of tumor immunosurveillance is now well established. With this chapter, we provide a historical background of immunosurveillance, the concept of immunoediting, and the role of different T-cell subsets in the tumor microenvironment. We also discuss the relationship between immune checkpoints, tumor antigens, T cell receptor repertoire, and immunosurveillance. Finally, we comment on the future of immunotherapy as it relates to T cell immunosurveillance.
Collapse
Affiliation(s)
- Samhita Rao
- Departments of Medicine and Microbiology and Immunology, Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| | - Karim Gharib
- Departments of Medicine and Microbiology and Immunology, Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| | - Arnold Han
- Departments of Medicine and Microbiology and Immunology, Columbia Center for Translational Immunology, Columbia University, New York, NY, United States
| |
Collapse
|
337
|
Roguljic A, Spagnoli G, Juretic A, Sarcevic B, Banovic M, Beketic Oreskovic L. Possible predictive role of cancer/testis antigens in breast ductal carcinoma in situ. Oncol Lett 2018; 16:7245-7255. [PMID: 30546463 PMCID: PMC6256292 DOI: 10.3892/ol.2018.9544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/27/2018] [Indexed: 01/10/2023] Open
Abstract
Cancer/testis antigens (CTAs) are a large family of tumor-associated antigens expressed in human tumors of different histological origin, but not in normal tissues, with the exception of the testes and placenta. Numerous immunohistochemical studies have reported associations between CTA expression and a negative estrogen receptor (ER) status in breast tumors, and demonstrated that CTAs are frequently expressed in tumors with higher nuclear grade. The expression of CTAs has not been studied as extensively in ductal carcinoma in situ (DCIS) as it has been in invasive breast cancer. The present retrospective study included archived paraffin-embedded specimens from 83 patients diagnosed with DCIS in the period between January 2007 and December 2014. The follow-up time for local recurrence ranged between 1 and 8 years (mean, 5.02 years). Antigens from the melanoma-associated antigen gene (MAGE) family, namely multi-MAGE-A, MAGE-A1, MAGE-A10 and New York esophageal squamous cell carcinoma 1 (NY-ESO-1) antigen, were evaluated by immunostaining and their subcellular location was investigated. Presence of tumor-infiltrating lymphocytes (TILs) was evaluated on all sections, together with the histopathological variables of DCIS. Specific tested antigens exhibited associations with histopathological parameters for DCIS and all demonstrated statistically significant associations with nuclear staining, simultaneous cytoplasmic and nuclear staining, and local recurrence. Antigen MAGE-A10 demonstrated a significant association with higher expression of ER (P=0.005) and higher tumor nuclear grade (P=0.001), cytoplasmic staining (P=0.029) and antigen NY-ESO-1 with higher tumor size (P=0.001), expression of TILs (P=0.001) and R1 resection (P=0.001). A χ2 test revealed significant associations between simultaneous cytoplasmic and nuclear staining and local recurrence (P=0.005), central necrosis (P=0.016), and the expression of ER (P=0.003) and progesterone receptor (PR) (P=0.010). Additional analysis revealed an association between antigen MAGE-A10 and TILs (P=0.05). Additional analysis of TILs indicated that they were significantly associated with tumor grade (P=0.023), central necrosis (P<0.001), ER (P=0.003) and PR (P=0.029). Overall, CTAs from the MAGE family (MAGE-A1, multi-MAGE-A and MAGE-A10) and NY-ESO-1 associate with histopathological predictive variables of DCIS. The expression of antigens NY-ESO-1 and MAGE-A10 could serve an important role in the treatment of patients with negative histopathological predictive variables, but further analysis is required. Simultaneous cytoplasmic and nuclear protein expression of MAGE-A family and NY-ESO-1 CTAs may represent an independent marker for local recurrence. Taken together, the present data suggest that CTAs are not perfect indicators of invasiveness for DCIS, but could inform treatment strategies for patients when taken in combination with other histopathological predictive variables. However, this was a small study and further larger studies will be necessary to confirm the current findings.
Collapse
Affiliation(s)
- Ana Roguljic
- Department of Radiation and Medical Oncology, Sisters of Mercy University Hospital Center, University Hospital for Tumors, 10000 Zagreb, Croatia
| | - Gulio Spagnoli
- Department of Biomedicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Antonio Juretic
- Department of Oncology, Clinical Hospital Center Zagreb, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Bozena Sarcevic
- Department of Oncology-Pathology, University of Zagreb School of Medicine, Sisters of Mercy University Hospital Center, University Hospital for Tumors, 10000 Zagreb, Croatia
| | - Marija Banovic
- University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Lidija Beketic Oreskovic
- Department of Oncology, University of Zagreb School of Medicine, Sisters of Mercy University Hospital Center, University Hospital for Tumors, 10000 Zagreb, Croatia
| |
Collapse
|
338
|
Addressing cancer immunotherapy research in Iran: adoptive cell therapy on the horizon. Cytotherapy 2018; 20:1227-1237. [DOI: 10.1016/j.jcyt.2018.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/04/2018] [Accepted: 08/14/2018] [Indexed: 12/15/2022]
|
339
|
Want MY, Lugade AA, Battaglia S, Odunsi K. Nature of tumour rejection antigens in ovarian cancer. Immunology 2018; 155:202-210. [PMID: 29772069 PMCID: PMC6142289 DOI: 10.1111/imm.12951] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/12/2018] [Accepted: 04/19/2018] [Indexed: 12/16/2022] Open
Abstract
Major progress in the analysis of human immune responses to cancer has been made through the molecular characterization of human tumour antigens. The development of therapeutic strategies for eliciting immune-mediated rejection of tumours has accelerated due to the elucidation of the molecular basis for tumour cell recognition and destruction by immune cells. Of the various human tumour antigens defined to date in ovarian cancer, the cancer-testis (CT) family of antigens have been studied extensively preclinically and clinically because of their testis-restricted expression in normal tissues and ability to elicit robust immune responses. Recent developments in cancer sequencing technologies offer a unique opportunity to identify tumour mutations with the highest likelihood of being expressed and recognized by the immune system. Such mutations, or neoantigens, could potentially serve as specific immune targets for T-cell-mediated destruction of cancer cells. This review will highlight current work in selecting tumour rejection antigens in ovarian cancer for improving the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Muzamil Y. Want
- Center For ImmunotherapyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | - Amit A. Lugade
- Center For ImmunotherapyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| | | | - Kunle Odunsi
- Center For ImmunotherapyRoswell Park Comprehensive Cancer CenterBuffaloNYUSA
| |
Collapse
|
340
|
Chodon T, Lugade AA, Battaglia S, Odunsi K. Emerging Role and Future Directions of Immunotherapy in Advanced Ovarian Cancer. Hematol Oncol Clin North Am 2018; 32:1025-1039. [PMID: 30390758 DOI: 10.1016/j.hoc.2018.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Clinical progress in cancer immunotherapy has been slow; however, within the last 5 years, breakthrough successes have brought immunotherapy to the forefront in cancer therapy. Promising results have been observed in solid tumors and hematologic malignancies. Most treatment modalities have shown limited efficacy as monotherapy. The complex nature of cancer and the immunosuppressive microenvironment emphasizes the need to personalize immunotherapy by manipulating the patient's own immune system. For successful and long-lasting cure of cancer, a multimodal approach is essential, combining antitumor cell therapy with manipulation of multiple pathways in the tumor microenvironment to ameliorate tumor-induced immunosuppression.
Collapse
Affiliation(s)
- Thinle Chodon
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Amit A Lugade
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sebastiano Battaglia
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Kunle Odunsi
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA; Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
341
|
Akce M, Zaidi MY, Waller EK, El-Rayes BF, Lesinski GB. The Potential of CAR T Cell Therapy in Pancreatic Cancer. Front Immunol 2018; 9:2166. [PMID: 30319627 PMCID: PMC6167429 DOI: 10.3389/fimmu.2018.02166] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 09/03/2018] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer has a dismal prognosis and effective treatment options are limited. It is projected to be the second most common cause of cancer related mortality in the United States by 2030 and there is urgent unmet need for novel systemic treatment options. Immunotherapy with antibodies targeting PD-1, PD-L1, CTLA-4 has not shown clinical activity in unselected pancreatic cancer, emphasizing the need for combination immunotherapy approaches or other therapeutic strategies. As such, chimeric antigen receptor (CAR) T cell therapy represents an emerging therapeutic option for pancreatic cancer. This modality utilizes genetically engineered T cells that are redirected to specific cancer-associated antigens to elicit potent cytotoxic activity. This review summarizes the available preclinical data and highlights early phase clinical trials using CAR T cell approaches in pancreatic cancer, a disease state that is gaining attention as a conduit for cell therapy. Future directions in application of CAR T cell therapy are also considered including its ability to be directed against novel epitopes and combined with other therapeutic regimens.
Collapse
Affiliation(s)
- Mehmet Akce
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Mohammad Y Zaidi
- Department of General Surgery, Indiana University, Bloomington, IN, United States.,Division of Surgical Oncology, Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | - Edmund K Waller
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Bassel F El-Rayes
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| | - Gregory B Lesinski
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
342
|
Quandt J, Schlude C, Bartoschek M, Will R, Cid-Arregui A, Schölch S, Reissfelder C, Weitz J, Schneider M, Wiemann S, Momburg F, Beckhove P. Long-peptide vaccination with driver gene mutations in p53 and Kras induces cancer mutation-specific effector as well as regulatory T cell responses. Oncoimmunology 2018; 7:e1500671. [PMID: 30524892 PMCID: PMC6279329 DOI: 10.1080/2162402x.2018.1500671] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 01/09/2023] Open
Abstract
Mutated proteins arising from somatic mutations in tumors are promising targets for cancer immunotherapy. They represent true tumor-specific antigens (TSAs) as they are exclusively expressed in tumors, reduce the risk of autoimmunity and are more likely to overcome tolerance compared to wild-type (wt) sequences. Hence, we designed a panel of long peptides (LPs, 28–35 aa) comprising driver gene mutations in TP35 and KRAS frequently found in gastrointestinal tumors to test their combined immunotherapeutic potential. We found increased numbers of T cells responsive against respective mutated and wt peptides in colorectal cancer patients that carry the tested mutations in their tumors than patients with other mutations. Further, active immunization of HLA(-A2/DR1)-humanized mice with mixes of the same mutated LPs yielded simultaneous, polyvalent CD8+/CD4+ T cell responses against the majority of peptides. Peptide-specific T cells possessed a multifunctional cytokine profile with CD4+ T cells showing a TH1-like phenotype. Two mutated peptides (Kras[G12V], p53[R248W]) induced significantly higher T cell responses than corresponding wt sequences and comprised HLA-A2/DR1-restricted mutated epitopes. However, vaccination with the same highly immunogenic LPs strongly increased systemic regulatory T cells (Treg) numbers in a syngeneic sarcoma model over-expressing these mutated protein variants and resulted in accelerated tumor outgrowth. In contrast, tumor outgrowth was delayed when vaccination was directed against tumor-intrinsic Kras/Tp53 mutations of lower immunogenicity. Conclusively, we show that LP vaccination targeting multiple mutated TSAs elicits polyvalent, multifunctional, and mutation-specific effector T cells capable of targeting tumors. However, the success of this therapeutic approach can be hampered by vaccination-induced, TSA-specific Tregs.
Collapse
Affiliation(s)
- Jasmin Quandt
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Knapp Research Center, Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Christoph Schlude
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Bartoschek
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Rainer Will
- Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angel Cid-Arregui
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian Schölch
- Department of Visceral Surgery, University Hospital Heidelberg, Heidelberg, Germany.,Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Reissfelder
- Department of Visceral Surgery, University Hospital Heidelberg, Heidelberg, Germany.,Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic, and Vascular Surgery, Medizinische Fakultaet an der TU-Dresden, Dresden, Germany
| | - Martin Schneider
- Department of Visceral Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Wiemann
- Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Momburg
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Antigen Presentation and T/NK Cell Activation Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp Beckhove
- Department of Translational Immunology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany.,Regensburg Center for Interventional Immunology (RCI), University Regensburg and Department of Hematology-Oncology, Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
343
|
Gibbs ZA, Whitehurst AW. Emerging Contributions of Cancer/Testis Antigens to Neoplastic Behaviors. Trends Cancer 2018; 4:701-712. [PMID: 30292353 DOI: 10.1016/j.trecan.2018.08.005] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023]
Abstract
Tumors of nearly every origin activate the expression of genes normally restricted to gametogenic cells. These genes encode proteins termed cancer/testis (CT) antigens, since expression outside of their naturally immune-privileged site can evoke an immune response. Despite extensive efforts to exploit CT antigens as immunotherapeutic targets, investigation of whether these proteins participate in tumorigenic processes has lagged. Here, we discuss emerging evidence that demonstrates that CT antigens can confer a selective advantage to tumor cells by promoting oncogenic processes or permitting evasion of tumor-suppressive mechanisms. These advances indicate the inherent flexibility of tumor cell regulatory networks to engage aberrantly expressed proteins to promote neoplastic behaviors, which could ultimately present novel therapeutic entry points.
Collapse
Affiliation(s)
- Zane A Gibbs
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Angelique W Whitehurst
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA; Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
344
|
Shires K, Van Wyk T. The role of Cancer/Testis Antigens in Multiple Myeloma pathogenesis and their application in disease monitoring and therapy. Crit Rev Oncol Hematol 2018; 132:17-26. [PMID: 30447924 DOI: 10.1016/j.critrevonc.2018.09.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/17/2022] Open
Abstract
A unique group of genes, encoding tumour associated antigens, known as the Cancer/Testis Antigens (CTAs), have been explored as novel markers of disease progression and as targets of immunotherapy in several cancers, including the haematological malignancy Multiple Myeloma (MM). This review aims to update the knowledge of CTA involvement in MM pathogenesis and how their potential as biomarkers for disease monitoring and targets of immunotherapy has been explored in the MM disease arena. Despite the initial promise of these antigens, their use as immunotherapy targets has not been successful, yet with a greater understanding of their role in disease pathogenesis they may still have a significant role to play as biomarkers of disease and therapeutic targets.
Collapse
Affiliation(s)
- Karen Shires
- Division of Haematology, Department of Pathology, University of Cape Town and National Health Laboratory Service/Groote Schuur Hospital, Cape Town, South Africa.
| | - Teagan Van Wyk
- Department of Medicine, University of Cape Town, South Africa
| |
Collapse
|
345
|
Abstract
The identification of critical genes that encode for regulatory proteins has helped elucidate the molecular mechanisms that govern cell proliferation and malignant transformation. Several approaches to gene therapy for cancer have been described. These include the use of fibroblasts expressing suicide genes, restoration of expression of tumor suppressor genes, cytokine gene therapy, genetic modification of T lymphocytes, and protection of normal hemopoiesis in cancer patients. Clinical trials are underway in all these areas.
Collapse
Affiliation(s)
- Julie Y. Djeu
- Immunology Program at H. Lee Moffitt Cancer Center & Research Institute, Tampa, Fla
| |
Collapse
|
346
|
Epigenetic regulation of MAGE family in human cancer progression-DNA methylation, histone modification, and non-coding RNAs. Clin Epigenetics 2018; 10:115. [PMID: 30185218 PMCID: PMC6126015 DOI: 10.1186/s13148-018-0550-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
The melanoma antigen gene (MAGE) proteins are a group of highly conserved family members that contain a common MAGE homology domain. Type I MAGEs are relevant cancer-testis antigens (CTAs), and originally considered as attractive targets for cancer immunotherapy due to their typically high expression in tumor tissues but restricted expression in normal adult tissues. Here, we reviewed the recent discoveries and ideas that illustrate the biological functions of MAGE family in cancer progression. Furthermore, we also highlighted the current understanding of the epigenetic mechanism of MAGE family expression in human cancers.
Collapse
|
347
|
Gordeeva O. Cancer-testis antigens: Unique cancer stem cell biomarkers and targets for cancer therapy. Semin Cancer Biol 2018; 53:75-89. [PMID: 30171980 DOI: 10.1016/j.semcancer.2018.08.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Revised: 08/15/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
Cancer-testis antigens (CTAs) are considered as unique and promising cancer biomarkers and targets for cancer therapy. CTAs are multifunctional protein group with specific expression patterns in normal embryonic and adult cells and various types of cancer cells. CTAs are involved in regulating of the basic cellular processes during development, stem cell differentiation and carcinogenesis though the biological roles and cell functions of CTA families remain largely unclear. Analysis of CTA expression patterns in embryonic germ and somatic cells, pluripotent and multipotent stem cells, cancer stem cells and their cell descendants indicates that rearrangements of characteristic CTA profiles (aberrant expression) could be associated with cancer transformation and failure of the developmental program of cell lineage specification and germ line restriction. Therefore, aberrant CTA profiles can be used as panels of biomarkers for diagnoses and the selection of cancer treatment strategies. Moreover, immunogenic CTAs are prospective targets for cancer immunotherapy. Clinical trials testing broad range of cancer therapeutic vaccines against antigens of MAGEA and NY-ESO-1 families for treating various cancers have shown mixed clinical efficiency, safety and tolerability, suggesting the requirement of in-depth research of CTA expression in normal and cancer stem cells and extensive clinical trials for improving cancer immunotherapy technologies. This review focuses on recent advancement in study of CTAs in normal and cancer cells, particularly in normal and cancer stem cells, and provides a new insight into CTA expression patterns during normal and cancer stem cell lineage development. Additionally, new approaches in development of effective CTA-based therapies exclusively targeting cancer stem cells will be discussed.
Collapse
Affiliation(s)
- Olga Gordeeva
- Laboratory of Cell and Molecular Mechanisms of Histogenesis, Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, Moscow, 119334, Russia.
| |
Collapse
|
348
|
Finn OJ. A Believer's Overview of Cancer Immunosurveillance and Immunotherapy. THE JOURNAL OF IMMUNOLOGY 2018; 200:385-391. [PMID: 29311379 DOI: 10.4049/jimmunol.1701302] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/19/2017] [Indexed: 12/20/2022]
Abstract
The field of tumor immunology has grown around the idea that one of the important roles of the immune system is to eliminate cancer. This idea was difficult to reconcile with the accepted notion that the immune system evolved to distinguish self from nonself and therefore tumors derived from self-tissues would not be recognized. Lack of appropriate animal models prevented experimental testing of cancer immunosurveillance. This changed with the realization that the immune system evolved to recognize danger and with the advent of mouse models deficient in one or more immune function, which showed predicted increases in susceptibility to cancer. Simultaneously, technical advances that enabled the study of the human immune system provided data for the existence of tumor-specific T cells and Abs and led to molecular identification of tumor Ags, fully validating the cancer immunosurveillance hypothesis. Immunotherapy designed to strengthen cancer immunosurveillance has achieved unprecedented clinical successes.
Collapse
Affiliation(s)
- Olivera J Finn
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15232
| |
Collapse
|
349
|
Zamora AE, Crawford JC, Thomas PG. Hitting the Target: How T Cells Detect and Eliminate Tumors. THE JOURNAL OF IMMUNOLOGY 2018; 200:392-399. [PMID: 29311380 DOI: 10.4049/jimmunol.1701413] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/16/2017] [Indexed: 12/21/2022]
Abstract
The successes of antitumor immuno-based therapies and the application of next-generation sequencing to mutation profiling have produced insights into the specific targets of antitumor T cells. Mutated proteins have tremendous potential as targets for interventions using autologous T cells or engineered cell therapies and may serve as important correlates of efficacy for immunoregulatory interventions including immune checkpoint blockade. As mutated self, tumors present an exceptional case for host immunity, which has primarily evolved in response to foreign pathogens. Tumor Ags' resemblance to self may limit immune recognition, but key features appear to be the same between antipathogen and antitumor responses. Determining which targets will make efficacious Ags and which responses might be elicited therapeutically are key questions for the field. Here we discuss current knowledge on antitumor specificity, the mutations that provide immunogenic targets, and how cross-reactivity and immunodominance may contribute to variation in immune responses among tumor types.
Collapse
Affiliation(s)
- Anthony E Zamora
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| |
Collapse
|
350
|
Hutchison S, Pritchard AL. Identifying neoantigens for use in immunotherapy. Mamm Genome 2018; 29:714-730. [PMID: 30167844 PMCID: PMC6267674 DOI: 10.1007/s00335-018-9771-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/08/2018] [Indexed: 12/14/2022]
Abstract
This review focuses on the types of cancer antigens that can be recognised by the immune system and form due to alterations in the cancer genome, including cancer testis, overexpressed and neoantigens. Specifically, neoantigens can form when cancer cell-specific mutations occur that result in alterations of the protein from ‘self’. This type of antigen can result in an immune response sufficient to clear tumour cells when activated. Furthermore, studies have reported that the likelihood of successful immunotherapeutic targeting of cancer by many different methods was reliant on immune response to neoantigens. The recent resurgence of interest in the immune response to tumour cells, in conjunction with technological advances, has resulted in a large increase in the predicted, identified and functionally confirmed neoantigens. This growth in identified neoantigen sequences has increased the contents of training sets for algorithms, which in turn improves the prediction of which genetic mutations may form neoantigens. Additionally, algorithms predicting how proteins will be processed into peptide epitopes by the proteasome and which peptides bind to the transporter complex are also improving with this research. Now that large screens of all the tumour-specific protein altering mutations are possible, the emerging data from assessment of the immunogenicity of neoantigens suggest that only a minority of variants will form targetable epitopes. The potential for immunotherapeutic targeting of neoantigens will therefore be greater in cancers with a higher frequency of protein altering somatic variants. There is considerable potential in the use of neoantigens to treat patients, either alone or in combination with other immunotherapies and with continued advancements, these potentials will be realised.
Collapse
Affiliation(s)
- Sharon Hutchison
- Genetics and Immunology Research Group, University of the Highlands and Islands, An Lòchran, 10 Inverness Campus, Inverness, IV2 5NA, Scotland, UK
| | - Antonia L Pritchard
- Genetics and Immunology Research Group, University of the Highlands and Islands, An Lòchran, 10 Inverness Campus, Inverness, IV2 5NA, Scotland, UK.
| |
Collapse
|