101
|
Soliman HH, Jackson E, Neuger T, Dees EC, Harvey RD, Han H, Ismail-Khan R, Minton S, Vahanian NN, Link C, Sullivan DM, Antonia S. A first in man phase I trial of the oral immunomodulator, indoximod, combined with docetaxel in patients with metastatic solid tumors. Oncotarget 2015; 5:8136-46. [PMID: 25327557 PMCID: PMC4226672 DOI: 10.18632/oncotarget.2357] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) is an enzyme that tumors use to create a state of immunosuppression. Indoximod is an IDO pathway inhibitor. Preclinical studies demonstrated that indoximod combined with chemotherapy was synergistic in a mouse model of breast cancer. A phase I 3+3 trial was designed to study the combination of docetaxel and indoximod. METHODS Docetaxel was administered at 60 mg/m2 intravenously every 3 weeks dose levels 1-4 and 75 mg/m2 for dose level 5. Indoximod was given at 300, 600, 1000, 2000, and 1200 mg PO twice daily continuously for levels 1-5, respectively. Serum drug levels were measured. RESULTS Twenty-seven patients were treated, with 22 evaluable for response. DLTs included grade 3 dehydration (level 1), hypotension(level 4), mucositis (level 4) and grade 5 enterocolitis (level 2). Dose level 5 is the recommended phase II dose. The most frequent adverse events were fatigue (58.6%), anemia (51.7%), hyperglycemia (48.3%), infection (44.8%), and nausea (41.4%). There were 4 partial responses (2 breast, 1 NSCLC, 1 thymic tumor). No drug-drug interactions were noted. CONCLUSIONS Docetaxel plus indoximod was well tolerated with no increase in expected toxicities or pharmacokinetic interactions. It was active in a pretreated population of patients with metastatic solid tumors.
Collapse
Affiliation(s)
- Hatem H Soliman
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Tony Neuger
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - E Claire Dees
- University of North Carolina/Lineberger Cancer Center, Chapel Hill, NC
| | | | - Hyo Han
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Susan Minton
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | | | | | - Scott Antonia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
102
|
Abstract
The pathogenesis of Alzheimer's disease (AD) is a critical unsolved question; and although recent studies have demonstrated a strong association between altered brain immune responses and disease progression, the mechanistic cause of neuronal dysfunction and death is unknown. We have previously described the unique CVN-AD mouse model of AD, in which immune-mediated nitric oxide is lowered to mimic human levels, resulting in a mouse model that demonstrates the cardinal features of AD, including amyloid deposition, hyperphosphorylated and aggregated tau, behavioral changes, and age-dependent hippocampal neuronal loss. Using this mouse model, we studied longitudinal changes in brain immunity in relation to neuronal loss and, contrary to the predominant view that AD pathology is driven by proinflammatory factors, we find that the pathology in CVN-AD mice is driven by local immune suppression. Areas of hippocampal neuronal death are associated with the presence of immunosuppressive CD11c(+) microglia and extracellular arginase, resulting in arginine catabolism and reduced levels of total brain arginine. Pharmacologic disruption of the arginine utilization pathway by an inhibitor of arginase and ornithine decarboxylase protected the mice from AD-like pathology and significantly decreased CD11c expression. Our findings strongly implicate local immune-mediated amino acid catabolism as a novel and potentially critical mechanism mediating the age-dependent and regional loss of neurons in humans with AD.
Collapse
|
103
|
Bhushan S, Tchatalbachev S, Lu Y, Fröhlich S, Fijak M, Vijayan V, Chakraborty T, Meinhardt A. Differential activation of inflammatory pathways in testicular macrophages provides a rationale for their subdued inflammatory capacity. THE JOURNAL OF IMMUNOLOGY 2015; 194:5455-64. [PMID: 25917085 DOI: 10.4049/jimmunol.1401132] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 04/01/2015] [Indexed: 01/11/2023]
Abstract
Spermatogenic cells express cell-specific molecules with the potential to be seen as "foreign" by the immune system. Owing to the time difference between their appearance in puberty and the editing of the lymphocyte repertoire around birth, local adaptations of the immune system coined immune privilege are required to confer protection from autoattack. Testicular macrophages (TM) play an important role in maintaining testicular immune privilege and display reduced proinflammatory capacity compared with other macrophages. However, the molecular mechanism underlying this macrophage phenotype remained elusive. We demonstrate that TM have a lower constitutive expression of TLR pathway-specific genes compared with peritoneal macrophages. Moreover, in TM stimulated with LPS, the NF-κB signaling pathway is blocked due to lack of IκBα ubiquitination and, hence, degradation. Instead, challenge of TM with LPS or polyinosinic-polycytidylic acid induces MAPK, AP-1, and CREB signaling pathways, which leads to production of proinflammatory cytokines such as TNF-α, although at much lower levels than in peritoneal macrophages. Pretreatment of TM with inhibitors for MAPKs p38 and ERK1/2 suppresses activation of AP-1 and CREB signaling pathways and attenuates LPS-induced TNF-α and IL-10 secretion. High levels of IL-10 production and activation of STAT3 by LPS stimulation in TM indicate a regulatory macrophage phenotype. Our results suggest that TM maintain testicular immune privilege by inhibiting NF-κB signaling through impairment of IκBα ubiquitination and a general reduction of TLR cascade gene expression. However, TM do maintain some capacity for innate immune responses through AP-1 and CREB signaling pathways.
Collapse
Affiliation(s)
- Sudhanshu Bhushan
- Department of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; and
| | - Svetlin Tchatalbachev
- Department of Medical Microbiology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Yongning Lu
- Department of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; and
| | - Suada Fröhlich
- Department of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; and
| | - Monika Fijak
- Department of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; and
| | - Vijith Vijayan
- Department of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; and
| | - Trinad Chakraborty
- Department of Medical Microbiology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Andreas Meinhardt
- Department of Anatomy and Cell Biology, Justus Liebig University Giessen, 35392 Giessen, Germany; and
| |
Collapse
|
104
|
Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway. Med Oncol 2015; 32:352. [DOI: 10.1007/s12032-014-0352-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 11/12/2014] [Indexed: 02/07/2023]
|
105
|
Fafournoux P, Averous J, Bruhat A, Carraro V, Jousse C, Maurin AC, Mesclon F, Parry L. [Adaptation to the availability of essential amino-acids: role of GCN2/eIF2α/ATF4 pathway]. Biol Aujourdhui 2015; 209:317-23. [PMID: 27021050 DOI: 10.1051/jbio/2016005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Indexed: 11/14/2022]
Abstract
In mammals, metabolic adaptations are required to overcome nutritional deprivation in amino-acids/proteins as well as episodes of malnutrition. GCN2 protein kinase, which phosphorylates the α subunit of the translation initiation factor eIF2, is a sensor of amino-acid(s) deficiencies. On one hand, this review briefly describes the main features of amino-acid metabolism. On the other hand, it describes the role of GCN2 in regulating numerous physiological functions.
Collapse
|
106
|
Abstract
Immunotherapy has demonstrated impressive outcomes for some patients with cancer. However, selecting patients who are most likely to respond to immunotherapy remains a clinical challenge. Here, we discuss immune escape mechanisms exploited by cancer and present strategies for applying this knowledge to improving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Gregory L Beatty
- Abramson Cancer Center, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| | - Whitney L Gladney
- Abramson Cancer Center, Department of Medicine, Division of Hematology-Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
107
|
Redgrove KA, McLaughlin EA. The Role of the Immune Response in Chlamydia trachomatis Infection of the Male Genital Tract: A Double-Edged Sword. Front Immunol 2014; 5:534. [PMID: 25386180 PMCID: PMC4209867 DOI: 10.3389/fimmu.2014.00534] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/09/2014] [Indexed: 01/16/2023] Open
Abstract
Chlamydia trachomatis (CT) is the most prevalent bacterial sexually transmitted infection in the world, with more than 100 million cases reported annually. While there have been extensive studies into the adverse effects that CT infection has on the female genital tract, and on the subsequent ability of these women to conceive, studies into the consequences on male fertility have been limited and controversial. This is in part due to the asymptomatic nature of the infection, where it is estimated that 50% of men with Chlamydia fail to show any symptoms. It is accepted, however, that acute and/or persistent CT infection is the causative agent for conditions such as urethritis, epididymitis, epididymo-orchitis, and potentially prostatitis. As with most infections, the immune system plays a fundamental role in the body’s attempts to eradicate the infection. The first and most important immune response to Chlamydia infection is a local one, whereby immune cells such as leukocytes are recruited to the site of infections, and subsequently secrete pro-inflammatory cytokines and chemokines such as interferon gamma. Immune cells also work to initiate and potentiate chronic inflammation through the production of reactive oxygen species (ROS), and the release of molecules with degradative properties including defensins, elastase, collagenase, cathespins, and lysozyme. This long-term inflammation can lead to cell proliferation (a possible precursor to cancer), tissue remodeling, and scarring, as well as being linked to the onset of autoimmune responses in genetically disposed individuals. This review will focus on the ability of the immune system to recognize and clear acute and persistent chlamydial infections in the male genital tract, and on the paradoxical damage that chronic inflammation resulting from the infection can cause on the reproductive health of the individual.
Collapse
Affiliation(s)
- Kate A Redgrove
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| | - Eileen A McLaughlin
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| |
Collapse
|
108
|
Norström MM, Rådestad E, Stikvoort A, Egevad L, Bergqvist M, Henningsohn L, Mattsson J, Levitsky V, Uhlin M. Novel method to characterize immune cells from human prostate tissue. Prostate 2014; 74:1391-9. [PMID: 25111297 DOI: 10.1002/pros.22854] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 06/17/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND Benign prostatic hyperplasia (BPH) is the most common benign adenoma and prostate cancer is the most frequent malignancy in men over 50 years of age in the Western world, where it remains a significant health problem. Prostate lesions are known to contain immune cells, which may contribute to the immune control of tumor progression. However, due to their low numbers and restricted access to necessary material it is difficult to isolate immune cells from prostate tissue to characterize their immunological features. METHODS An efficient and robust method was developed to process prostate tissue and isolate immune cells for phenotypic analysis by multicolor flow cytometry as downstream application. Fresh prostate tissue from 11 patients undergoing surgery for bladder outlet obstruction due to BPH was processed to evaluate the number, viability, yield, and frequency of various immune cell types. RESULTS The presented method does not include enzymatic digestion nor incubation steps at 37 °C, increasing cellular viability and avoiding possible phenotypic modification. Various immune cell populations were detected in all patient samples and the median cellular viability was 90%. The number of detected events of individual cell populations varied between patients. The median frequency of different immune cell populations also varied, being 87% for the CD3- and 15% for the CD3+ cell population. CONCLUSIONS This novel method will allow the phenotypic characterization of immune cell populations present in tumor tissue of prostate cancer patients and promote development of novel approaches to immunotherapy of the disease.
Collapse
Affiliation(s)
- Melissa M Norström
- Centre for Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge, Stockholm, Sweden; Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Dai M, Yip YY, Hellstrom I, Hellstrom KE. Curing mice with large tumors by locally delivering combinations of immunomodulatory antibodies. Clin Cancer Res 2014; 21:1127-38. [PMID: 25142145 DOI: 10.1158/1078-0432.ccr-14-1339] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Immunomodulatory mAbs can treat cancer, but cures are rare except for small tumors. Our objective was to explore whether the therapeutic window increases by combining mAbs with different modes of action and injecting them into tumors. EXPERIMENTAL DESIGN Combinations of mAbs to CD137/PD-1/CTLA-4 or CD137/PD-1/CTLA-4/CD19 were administrated intratumorally to mice with syngeneic tumors (B16 and SW1 melanoma, TC1 lung carcinoma), including tumors with a mean surface of approximately 80 mm(2). Survival and tumor growth were assessed. Immunologic responses were evaluated using flow cytometry and qRT-PCR. RESULTS More than 50% of tumor-bearing mice had complete regression and long-term survival after tumor injection with mAbs recognizing CD137/PD-1/CTLA-4/CD19 with similar responses in three models. Intratumoral injection was more efficacious than intraperitoneal injection in causing rejection also of untreated tumors in the same mice. The three-mAb combination could also induce regression, but was less efficacious. There were few side effects, and therapy-resistant tumors were not observed. Transplanted tumor cells rapidly caused a Th2 response with increased CD19 cells. Successful therapy shifted this response to the Th1 phenotype with decreased CD19 cells and increased numbers of long-term memory CD8 effector cells and T cells making IFNγ and TNFα. CONCLUSIONS Intratumoral injection of mAbs recognizing CD137/PD-1/CTLA-4/CD19 can eradicate established tumors and reverse a Th2 response with tumor-associated CD19 cells to Th1 immunity, whereas a combination lacking anti-CD19 is less effective. There are several human cancers for which a similar approach may provide clinical benefit.
Collapse
Affiliation(s)
- Min Dai
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington
| | - Yuen Yee Yip
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington
| | - Ingegerd Hellstrom
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington
| | - Karl Erik Hellstrom
- University of Washington Harborview Medical Center, Department of Pathology, Seattle, Washington.
| |
Collapse
|
110
|
Yanagihara S, Goto H, Hirota T, Fukuda S, Ohno H, Yamamoto N. Lactobacillus acidophilus L-92 Cells Activate Expression of Immunomodulatory Genes in THP-1 Cells. BIOSCIENCE OF MICROBIOTA FOOD AND HEALTH 2014; 33:157-64. [PMID: 25379363 PMCID: PMC4219982 DOI: 10.12938/bmfh.33.157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 07/03/2014] [Indexed: 12/24/2022]
Abstract
To understand the immunomodulatory effects of Lactobacillus acidophilus L-92 cells suggested from our previous
study of in vivo anti-allergy and anti-virus effects, host immune responses in macrophage-like THP-1 cells after
4 h (the early phase) and 24 h (the late phase) of cocultivation with L-92 cells were investigated by transcriptome analysis. In
the early phase of L-92 treatment, various transcription regulator genes, such as, NFkB1, NFkB2, JUN, HIVEP2 and
RELB, and genes encoding chemokines and cytokines, such as CCL4, CXCL11, CCL3 and
TNF, were upregulated. Two transmembrane receptor genes, TLR7 and ICAM1, were
also upregulated in the early phase of treatment. In contrast, many transmembrane receptor genes, such as IL7R, CD80,
CRLF2, CD86, CD5, HLA-DQA1, IL2RA, IL15RA and CSF2RA, and some cytokine genes, including IL6,
IL23A and CCL22, were significantly upregulated in the late phase after L-92 exposure. Some genes
encoding cytokines, such as IL1A, IL1B and IL8, and the enzyme IDO1 were
upregulated at both the early and the late phases of treatment. These results suggest that probiotic L-92 might promote Th1 and
regulatory T-cell responses by activation of the MAPK signaling pathway, followed by the NOD-like receptor signaling pathway in
THP-1 cells.
Collapse
Affiliation(s)
- Sae Yanagihara
- Microbiology and Fermentation Laboratory, Calpis Co. Ltd., 5-11-10 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa 252-0206, Japan
| | - Hiroaki Goto
- Microbiology and Fermentation Laboratory, Calpis Co. Ltd., 5-11-10 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa 252-0206, Japan
| | - Tatsuhiko Hirota
- Microbiology and Fermentation Laboratory, Calpis Co. Ltd., 5-11-10 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa 252-0206, Japan
| | - Shinji Fukuda
- Intestinal Microbe Symbiosis Laboratory, RIKEN, Wako, Saitama 351-0198, Japan ; Laboratory for Intestinal Ecosystem, RCAI, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan ; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa 230-0045, Japan ; Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0052, Japan
| | - Hiroshi Ohno
- Intestinal Microbe Symbiosis Laboratory, RIKEN, Wako, Saitama 351-0198, Japan ; Laboratory for Intestinal Ecosystem, RCAI, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa 230-0045, Japan ; Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Kanagawa 230-0045, Japan
| | - Naoyuki Yamamoto
- Intestinal Microbe Symbiosis Laboratory, RIKEN, Wako, Saitama 351-0198, Japan ; Research and Development Planning Department, Calpis Co. Ltd., 5-11-10 Fuchinobe, Chuo-ku, Sagamihara-shi, Kanagawa 252-0206, Japan
| |
Collapse
|
111
|
Thomas S, DuHadaway J, Prendergast GC, Laury-Kleintop L. Specific in situ detection of murine indoleamine 2, 3-dioxygenase. J Cell Biochem 2014; 115:391-6. [PMID: 24123235 DOI: 10.1002/jcb.24674] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Accepted: 09/10/2013] [Indexed: 11/09/2022]
Abstract
Indoleamine 2,3-dioxygenase-1 (IDO1) catabolizes the essential amino acid tryptophan, acting as a modifier of inflammation and immune tolerance. Recent work has implicated IDO1 in many human diseases, including in cancer, chronic infection, autoimmune disorders, and neurodegenerative disease, stimulating a major surge in preclinical and clinical studies of its pathogenic functions. In the mouse, IDO1 is expressed widely but in situ detection of the enzyme in murine tissues has been unreliable due to the lack of specific antibodies that do not also react with tissues from animals that are genetically deficient in IDO1. Such probes are crucial to establish cellular mechanisms since IDO1 appears to act in different cell types depending on disease context, but reliable probes have been elusive in the field. In this report, we address this issue with the development of IDO1 monoclonal antibody 4B7 which specifically recognizes the murine enzyme in tissue sections, offering a reliable tool for immunohistology in preclinical disease models.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, 100 Lancaster Ave, Wynnewood, 19096, Pennsylvania
| | | | | | | |
Collapse
|
112
|
Ohta A, Sitkovsky M. Extracellular adenosine-mediated modulation of regulatory T cells. Front Immunol 2014; 5:304. [PMID: 25071765 PMCID: PMC4091046 DOI: 10.3389/fimmu.2014.00304] [Citation(s) in RCA: 224] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 06/15/2014] [Indexed: 12/20/2022] Open
Abstract
Extracellular adenosine-dependent suppression and redirection of pro-inflammatory activities are mediated by the signaling through adenosine receptors on the surface of most immune cells. The immunosuppression by endogenously-produced adenosine is pathophysiologically significant since inactivation of A2A/A2B adenosine receptor (A2AR/A2BR) and adenosine-producing ecto-enzymes CD39/CD73 results in the higher intensity of immune response and exaggeration of inflammatory damage. Regulatory T cells (Treg) can generate extracellular adenosine, which is implicated in the immunoregulatory activity of Tregs. Interestingly, adenosine has been shown to increase the numbers of Tregs and further promotes their immunoregulatory activity. A2AR-deficiency in Tregs reduces their immunosuppressive efficacy in vivo. Thus, adenosine is not only directly and instantly inhibiting to the immune response through interaction with A2AR/A2BR on the effector cells, but also adenosine signaling can recruit other immunoregulatory mechanisms, including Tregs. Such interaction between adenosine and Tregs suggests the presence of a positive feedback mechanism, which further promotes negative regulation of immune system through the establishment of immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Akio Ohta
- New England Inflammation and Tissue Protection Institute, Northeastern University , Boston, MA , USA
| | - Michail Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University , Boston, MA , USA
| |
Collapse
|
113
|
Liu J, Zhang S, Li H, Cheng Y. [Advances of immunotherapy in small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:474-80. [PMID: 24949688 PMCID: PMC6000108 DOI: 10.3779/j.issn.1009-3419.2014.06.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
小细胞肺癌(small cell lung cancer, SCLC)具有复杂的异质性,由于细胞起源、发病机制和驱动基因尚不明确,SCLC的诊治进展缓慢,鲜有突破,迫切需要新的治疗策略提高SCLC疗效。肿瘤免疫治疗可提高免疫系统识别和排除肿瘤细胞的能力,且对正常组织影响轻微。目前已经开展了肿瘤疫苗、过继细胞免疫治疗、细胞因子、checkpoint抑制剂等治疗SCLC的临床研究,ipilimumab是最有前景的药物。免疫治疗有望为SCLC治疗带来新的希望,未来还需要对SCLC的异质性、免疫治疗靶点不明确、免疫治疗耐受等影响免疫治疗疗效的问题开展进一步研究。
Collapse
Affiliation(s)
- Jingjing Liu
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | - Shuang Zhang
- Department of Thoracic Oncology, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | - Hui Li
- Hematological Oncology Laboratory, Jilin Provincial Cancer Hospital, Changchun 130012, China
| | | |
Collapse
|
114
|
Mockler MB, Conroy MJ, Lysaght J. Targeting T cell immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol 2014; 4:107. [PMID: 24904823 PMCID: PMC4032940 DOI: 10.3389/fonc.2014.00107] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/28/2014] [Indexed: 12/20/2022] Open
Abstract
The immune system has a key role to play in controlling cancer initiation and progression. T cell activation, which is central to anti-tumor immune responses, coincides with changes in cellular metabolism. Naïve T cells predominantly require an ATP generating metabolic profile, whereas proliferating effector T cells require anabolic metabolic profiles that promote rapid growth and proliferation. Furthermore, specific T cell subsets require distinct energetic and biosynthetic pathways to match their functional requirements. The often hostile tumor microenvironment can affect T cell immune responses by altering the resulting cellular metabolism. Tailoring immune responses by manipulating cellular metabolic pathways may provide an exciting new option for cancer immunotherapy. T cell responses might also be skewed via metabolic manipulation to treat the complications of obesity-associated inflammation, which is a rapidly growing global health problem and a major risk factor for many malignancies. In this review, the diverse metabolic requirements of T cells in anti-tumor immunity are discussed, as well as the profound influence of the tumor microenvironment and the possible avenues for manipulation to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Mary B Mockler
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| | - Melissa J Conroy
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
115
|
Prendergast GC, Smith C, Thomas S, Mandik-Nayak L, Laury-Kleintop L, Metz R, Muller AJ. Indoleamine 2,3-dioxygenase pathways of pathogenic inflammation and immune escape in cancer. Cancer Immunol Immunother 2014; 63:721-35. [PMID: 24711084 DOI: 10.1007/s00262-014-1549-4] [Citation(s) in RCA: 366] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 03/26/2014] [Indexed: 12/15/2022]
Abstract
Genetic and pharmacological studies of indoleamine 2,3-dioxygenase (IDO) have established this tryptophan catabolic enzyme as a central driver of malignant development and progression. IDO acts in tumor, stromal and immune cells to support pathogenic inflammatory processes that engender immune tolerance to tumor antigens. The multifaceted effects of IDO activation in cancer include the suppression of T and NK cells, the generation and activation of T regulatory cells and myeloid-derived suppressor cells, and the promotion of tumor angiogenesis. Mechanistic investigations have defined the aryl hydrocarbon receptor, the master metabolic regulator mTORC1 and the stress kinase Gcn2 as key effector signaling elements for IDO, which also exerts a non-catalytic role in TGF-β signaling. Small-molecule inhibitors of IDO exhibit anticancer activity and cooperate with immunotherapy, radiotherapy or chemotherapy to trigger rapid regression of aggressive tumors otherwise resistant to treatment. Notably, the dramatic antitumor activity of certain targeted therapeutics such as imatinib (Gleevec) in gastrointestinal stromal tumors has been traced in part to IDO downregulation. Further, antitumor responses to immune checkpoint inhibitors can be heightened safely by a clinical lead inhibitor of the IDO pathway that relieves IDO-mediated suppression of mTORC1 in T cells. In this personal perspective on IDO as a nodal mediator of pathogenic inflammation and immune escape in cancer, we provide a conceptual foundation for the clinical development of IDO inhibitors as a novel class of immunomodulators with broad application in the treatment of advanced human cancer.
Collapse
Affiliation(s)
- George C Prendergast
- Lankenau Institute for Medical Research (LIMR), 100 Lancaster Avenue, Wynnewood, PA, 19096, USA,
| | | | | | | | | | | | | |
Collapse
|
116
|
Barreda DR, Konowalchuk JD, Rieger AM, Wong ME, Havixbeck JJ. Triennial Growth Symposium--Novel roles for vitamin D in animal immunity and health. J Anim Sci 2014; 92:930-8. [PMID: 24665105 DOI: 10.2527/jas.2013-7341] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent years have seen significant advances in the generation, validation, and implementation of nutritional supplements for food production animals. Examination of their impact on animal performance and health requires collaboration among animal scientists, nutritionists, biochemists, immunologists, veterinarians, and others. Each provides a unique perspective on the mechanisms of action, short and long-term impacts, and most effective strategies for implementation into continuously evolving industrial practices. In this review we provide a comparative immunology perspective on the impact of vitamin D on animal performance and health, describe the differential contributions of vitamin D3 and of a commercial hydroxylated version of vitamin D3, 25-hydroxyvitamin D3 (25(OH)D3 or HyD) to swine immunity, and highlight recent advances in the technologies that can be used to dissect the cellular and molecular mechanisms that impact production animal immunity and health. Among others, we pay particular attention to how these novel approaches help decrease the variability often observed in immune-associated datasets. From a practical perspective, this is critical for evaluation of in vivo effects for this nutritional supplement as small but meaningful changes to specific immune responses are typical under normal physiological conditions. Furthermore, as the range of reagents and technologies expands for comparative animal models, it is imperative that continued efforts are placed on the capacity to compare results across different experimental platforms.
Collapse
Affiliation(s)
- D R Barreda
- Department of Agricultural, Food and Nutritional Science
| | | | | | | | | |
Collapse
|
117
|
Corona G, Polesel J, Fratino L, Miolo G, Rizzolio F, Crivellari D, Addobbati R, Cervo S, Toffoli G. Metabolomics Biomarkers of Frailty in Elderly Breast Cancer Patients. J Cell Physiol 2014; 229:898-902. [DOI: 10.1002/jcp.24520] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/20/2013] [Indexed: 12/21/2022]
Affiliation(s)
- Giuseppe Corona
- Experimental and Clinical Pharmacology Division, Department of Translational Research; IRCCS-National Cancer Institute; Aviano PN Italy
| | - Jerry Polesel
- Epidemiology and Biostatistics; IRCCS-National Cancer Institute; Aviano PN Italy
| | - Lucia Fratino
- Medical Oncology Department; IRCCS-National Cancer Institute; Aviano PN Italy
| | - Gianmaria Miolo
- Medical Oncology Department; IRCCS-National Cancer Institute; Aviano PN Italy
| | - Flavio Rizzolio
- Experimental and Clinical Pharmacology Division, Department of Translational Research; IRCCS-National Cancer Institute; Aviano PN Italy
- Sbarro Institute for Cancer Research and Molecular Medicine; Center for Biotechnology; College of Science and Technology; Temple University; Philadelphia Pennsylvania
| | - Diana Crivellari
- Medical Oncology Department; IRCCS-National Cancer Institute; Aviano PN Italy
| | - Riccardo Addobbati
- Metabolic Disease Laboratory; IRCCS-Burlo Garofolo; Children's Hospital; Trieste TS Italy
| | - Silvia Cervo
- Clinical Pathology Laboratory; IRCCS-National Cancer Institute; Aviano PN Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Division, Department of Translational Research; IRCCS-National Cancer Institute; Aviano PN Italy
| |
Collapse
|
118
|
Toomer KH, Chen Z. Autoimmunity as a double agent in tumor killing and cancer promotion. Front Immunol 2014; 5:116. [PMID: 24672527 PMCID: PMC3957029 DOI: 10.3389/fimmu.2014.00116] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Accepted: 03/05/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy through manipulation of the immune system holds great potential for the treatment of human cancers. However, recent trials targeting the negative immune regulators cytotoxic T-lymphocyte antigen 4, programed death 1 (PD-1), and PD-1 receptor ligand (PD-L1) demonstrated that clinically significant antitumor responses were often associated with the induction of autoimmune toxicity. This finding suggests that the same immune mechanisms that elicit autoimmunity may also contribute to the destruction of tumors. Given the fact that the immunological identity of tumors might be largely an immunoprivileged self, autoimmunity may not represent a wholly undesirable outcome in the context of cancer immunotherapy. Rather, targeted killing of cancer cells and autoimmune damage to healthy tissues may be intricately linked through molecular mechanisms, in particular inflammatory cytokine signaling. On the other hand, since chronic inflammation is a well-recognized condition that promotes tumor development, it appears that autoimmunity can be a "double agent" in mediating either pro-tumor or antitumor effects. This review surveys the tumor-promoting and tumoricidal activities of several prominent cytokines: IFN-γ, TNF-α, TGF-β, IL-17, IL-23, IL-4, and IL-13, produced by three major subsets of T helper cells that interact with innate immune cells. Many of these cytokines exert divergent and seemingly contradictory effects on cancer development in different human and animal models, suggesting a high degree of context dependence in their functions. We hypothesize that these inflammatory cytokines could mediate a feedback loop of autoimmunity, antitumor immunity, and tumorigenesis. Understanding the diverse and paradoxical roles of cytokines from autoimmune responses in the setting of cancer will advance the long-term goal of improving cancer immunotherapy, while minimizing the hazards of immune-mediated tissue damage and the possibility of de novo tumorigenesis, through proper monitoring and preventive measures.
Collapse
Affiliation(s)
- Kevin H Toomer
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine , Miami, FL , USA
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine , Miami, FL , USA ; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine , Miami, FL , USA
| |
Collapse
|
119
|
Bertolini M, Meyer KC, Slominski R, Kobayashi K, Ludwig RJ, Paus R. The immune system of mouse vibrissae follicles: cellular composition and indications of immune privilege. Exp Dermatol 2014; 22:593-8. [PMID: 23947674 DOI: 10.1111/exd.12205] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2013] [Indexed: 12/22/2022]
Abstract
Although vibrissae hair follicles (VHFs) have long been a key research model in the life sciences, their immune system (IS) is essentially unknown. Therefore, we have characterized basic parameters of the VHF-IS of C57BL/6J mice by quantitative (immuno-)histomorphometry. Murine anagen VHF harbour few CD4+ and CD8+ T cells in the distal mesenchyme and sinuses but hardly any gamma-delta T cells in their distal epithelium. MHC class II+ Langerhans cells are seeded in the VHF infundibulum, which is also surrounded by MHC class II+ and CD11b+ cells (macrophages). The number of Langerhans cells then declines sharply in the VHF bulge, and the VHF bulb lacks MHC class II+ cells. Mast cells densely populate the VHF connective tissue sheath, where they strikingly cluster around the bulge. Both the bulge and the bulb of VHF display signs of immune privilege, that is, low MHC class I and MHC class II expression and local immunoinhibitor expression (CD200, TGFβ1). This immunophenotyping study fills an important gap in the immunobiology of murine skin and identifies differences between the IS of VHF, mouse pelage and human terminal HFs. This facilitates utilizing murine VHF as a versatile organ culture model for general immunology and immune privilege research in situ.
Collapse
Affiliation(s)
- Marta Bertolini
- Department of Dermatology, University of Lübeck, Lübeck, Germany
| | | | | | | | | | | |
Collapse
|
120
|
Mittendorf EA, Sharma P. Mechanisms of T-cell inhibition: implications for cancer immunotherapy. Expert Rev Vaccines 2014; 9:89-105. [DOI: 10.1586/erv.09.144] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
121
|
Metz R, Smith C, DuHadaway JB, Chandler P, Baban B, Merlo LMF, Pigott E, Keough MP, Rust S, Mellor AL, Mandik-Nayak L, Muller AJ, Prendergast GC. IDO2 is critical for IDO1-mediated T-cell regulation and exerts a non-redundant function in inflammation. Int Immunol 2014; 26:357-67. [PMID: 24402311 DOI: 10.1093/intimm/dxt073] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IDO2 is implicated in tryptophan catabolism and immunity but its physiological functions are not well established. Here we report the characterization of mice genetically deficient in IDO2, which develop normally but exhibit defects in IDO-mediated T-cell regulation and inflammatory responses. Construction of this strain was prompted in part by our discovery that IDO2 function is attenuated in macrophages from Ido1 (-/-) mice due to altered message splicing, generating a functional mosaic with implications for interpreting findings in Ido1 (-/-) mice. No apparent defects were observed in Ido2 (-/-) mice in embryonic development or hematopoietic differentiation, with wild-type profiles documented for kynurenine in blood serum and for immune cells in spleen, lymph nodes, peritoneum, thymus and bone marrow of naive mice. In contrast, upon immune stimulation we determined that IDO1-dependent T regulatory cell generation was defective in Ido2 (-/-) mice, supporting Ido1-Ido2 genetic interaction and establishing a functional role for Ido2 in immune modulation. Pathophysiologically, both Ido1 (-/-) and Ido2 (-/-) mice displayed reduced skin contact hypersensitivity responses, but mechanistic distinctions were apparent, with only Ido2 deficiency associated with a suppression of immune regulatory cytokines that included GM-CSF, G-CSF, IFN-γ, TNF-α, IL-6 and MCP-1/CCL2. Different contributions to inflammation were likewise indicated by the finding that Ido2 (-/-) mice did not phenocopy Ido1 (-/-) mice in the reduced susceptibility of the latter to inflammatory skin cancer. Taken together, our results offer an initial glimpse into immune modulation by IDO2, revealing its genetic interaction with IDO1 and distinguishing its non-redundant contributions to inflammation.
Collapse
Affiliation(s)
- Richard Metz
- New Link Genetics Corporation, Ames, IA 50010, USA
| | - Courtney Smith
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | | | - Phillip Chandler
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Babak Baban
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Lauren M F Merlo
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Elizabeth Pigott
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Martin P Keough
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA
| | - Sonja Rust
- New Link Genetics Corporation, Ames, IA 50010, USA
| | - Andrew L Mellor
- Immunotherapy Center, Georgia Regents University, Augusta, GA 30912, USA
| | - Laura Mandik-Nayak
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Alexander J Muller
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - George C Prendergast
- Lankenau Institute for Medical Research, Wynnewood, PA 19096, USA Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA Department of Pathology, Anatomy and Cell Biology, Jefferson Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
122
|
Trabanelli S, Očadlíková D, Ciciarello M, Salvestrini V, Lecciso M, Jandus C, Metz R, Evangelisti C, Laury-Kleintop L, Romero P, Prendergast GC, Curti A, Lemoli RM. The SOCS3-independent expression of IDO2 supports the homeostatic generation of T regulatory cells by human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2014; 192:1231-40. [PMID: 24391212 DOI: 10.4049/jimmunol.1300720] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Dendritic cells (DCs) are professional APCs that have a role in the initiation of adaptive immune responses and tolerance. Among the tolerogenic mechanisms, the expression of the enzyme IDO1 represents an effective tool to generate T regulatory cells. In humans, different DC subsets express IDO1, but less is known about the IDO1-related enzyme IDO2. In this study, we found a different pattern of expression and regulation between IDO1 and IDO2 in human circulating DCs. At the protein level, IDO1 is expressed only in circulating myeloid DCs (mDCs) and is modulated by PGE2, whereas IDO2 is expressed in both mDCs and plasmacytoid DCs and is not modulated by PGE2. In healthy subjects, IDO1 expression requires the presence of PGE2 and needs continuous transcription and translation, whereas IDO2 expression is constitutive, independent from suppressor of cytokine signaling 3 activity. Conversely, in patients suffering from inflammatory arthritis, circulating DCs express both IDO1 and IDO2. At the functional level, both mDCs and plasmacytoid DCs generate T regulatory cells through an IDO1/IDO2-dependent mechanism. We conclude that, in humans, whereas IDO1 provides an additional mechanism of tolerance induced by proinflammatory mediators, IDO2 is stably expressed in steady-state conditions and may contribute to the homeostatic tolerogenic capacity of DCs.
Collapse
Affiliation(s)
- Sara Trabanelli
- Department of Specialistic, Diagnostic, and Experimental Medicine, Institute of Hematology "Seràgnoli," University of Bologna, 40138 Bologna, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Naegele M, Martin R. The good and the bad of neuroinflammation in multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2014; 122:59-87. [PMID: 24507513 DOI: 10.1016/b978-0-444-52001-2.00003-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Multiple sclerosis (MS) is the most common inflammatory, demyelinating, neurodegenerative disorder of the central nervous system (CNS). It is widely considered a T-cell mediated autoimmune disease that develops in genetically susceptible individuals, possibly under the influence of certain environmental trigger factors. The invasion of autoreactive CD4+ T-cells into the CNS is thought to be a central step that initiates the disease. Several other cell types, including CD8+ T-cells, B-cells and phagocytes appear to be involved in causing inflammation and eventually neurodegeneration. But inflammation is not entirely deleterious in MS. Evidence has accumulated in the recent years that show the importance of regulatory immune mechanisms which restrain tissue damage and initiate regeneration. More insight into the beneficial aspects of neuroinflammation might allow us to develop new treatment strategies for this enigmatic disease.
Collapse
Affiliation(s)
- Matthias Naegele
- Institute for Neuroimmunology and Clinical Multiple Sclerosis Research, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Roland Martin
- Neuroimmunology and MS Research, Neurology Clinic, University Hospital, Zurich, Switzerland.
| |
Collapse
|
124
|
Hou Y, Yin M, Sun F, Zhang T, Zhou X, Li H, Zheng J, Chen X, Li C, Ning X, Lou G, Li K. A metabolomics approach for predicting the response to neoadjuvant chemotherapy in cervical cancer patients. ACTA ACUST UNITED AC 2014; 10:2126-33. [PMID: 24865370 DOI: 10.1039/c4mb00054d] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cervical cancer is a clinical and pathological heterogeneity disease, which requires different types of treatments and leads to a variety of outcomes.
Collapse
Affiliation(s)
- Yan Hou
- Department of Epidemiology and Biostatistics
- Harbin Medical University
- Harbin 150081, China
| | - Mingzhu Yin
- Department of Gynecology Oncology
- The Tumor Hospital of Harbin Medical University
- Harbin 150086, China
| | - Fengyu Sun
- Department of Cardiology
- The First Affiliated Hospital of Harbin Medical University
- Harbin, China
| | - Tao Zhang
- Department of Epidemiology and Biostatistics
- Public Health School
- Shandong University
- Jinan, P. R. China
| | - Xiaohua Zhou
- Department of Biostatistics
- University of Washington
- Seattle, USA
| | - Huiyan Li
- Department of Radiotherapy Oncology
- The Tumor Hospital of Harbin Medical University
- Harbin, China
| | - Jian Zheng
- Department of Radiotherapy Oncology
- The Tumor Hospital of Harbin Medical University
- Harbin, China
| | - Xiuwei Chen
- Department of Gynecology Oncology
- The Tumor Hospital of Harbin Medical University
- Harbin 150086, China
| | - Cong Li
- Department of Pathology
- The Tumor Hospital of Harbin Medical University
- Harbin, China
| | - Xiaoming Ning
- Department of Pathology
- The Tumor Hospital of Harbin Medical University
- Harbin, China
| | - Ge Lou
- Department of Gynecology Oncology
- The Tumor Hospital of Harbin Medical University
- Harbin 150086, China
| | - Kang Li
- Department of Epidemiology and Biostatistics
- Harbin Medical University
- Harbin 150081, China
| |
Collapse
|
125
|
Lavoué V, Foucher F, Henno S, Bauville E, Catros V, Cabillic F, Levêque J. [Immunotherapy in epithelial ovarian carcinoma: hope and reality]. ACTA ACUST UNITED AC 2013; 43:198-210. [PMID: 24230482 DOI: 10.1016/j.jgyn.2013.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 10/14/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Epithelial ovarian carcinoma (EOC) has a worst prognosis with little progress in terms of survival for the last two decades. Immunology received little interest in EOC in the past, but now appears very important in the natural history of this cancer. This review is an EOC immunology state of art and focuses on the place of immunotherapy in future. MATERIAL AND METHODS A systematic review of published studies was performed. Medline baseline interrogation was performed with the following keywords: "Ovarian carinoma, immunotherapy, T-lymphocyte, regulator T-lymphocyte, dendritic cells, macrophage, antigen, chemotherapy, surgery, clinical trials". Identified publications (English or French) were assessed for the understanding of EOC immunology and the place of conventional treatment and immunotherapy strategy. RESULTS Intratumoral infiltration by immune cells is a strong prognotic factor in EOC. Surgery and chemotherapy in EOC decrease imunosuppression in patients. The antitumoral immunity is a part of the therapeutic action of surgery and chemotherapy. Until now, immunotherapy gave some disappointing results, but the new drugs that target the tolerogenic tumoral microenvironnement rise and give a new hope in the treatment of cancer. CONCLUSION Immunology controls the EOC natural history. The modulation of immunosuppressive microenvironment associated with the stimulation of antitumoral immunity could be the next revolution in the treatment of cancer.
Collapse
Affiliation(s)
- V Lavoué
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France.
| | - F Foucher
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - S Henno
- Service d'anatomo-pathologie, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - E Bauville
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - V Catros
- Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France; Service de biologie cellulaire, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - F Cabillic
- Faculté de médecine, université de Rennes 1, 35000 Rennes, France; Inserm, UMR991, 35000 Rennes, France; Service de biologie cellulaire, centre hospitalier universitaire de Rennes, 35000 Rennes, France
| | - J Levêque
- Service de chirurgie gynécologique, centre hospitalier universitaire de Rennes, 35000 Rennes, France; Faculté de médecine, université de Rennes 1, 35000 Rennes, France
| |
Collapse
|
126
|
Ohta A, Madasu M, Subramanian M, Kini R, Jones G, Choukèr A, Ohta A, Sitkovsky M. Hypoxia-induced and A2A adenosine receptor-independent T-cell suppression is short lived and easily reversible. Int Immunol 2013; 26:83-91. [PMID: 24150242 DOI: 10.1093/intimm/dxt045] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tissue hypoxia plays a key role in establishing an immunosuppressive environment in vivo by, among other effects, increasing the level of extracellular adenosine, which then signals through A2A adenosine receptor (A2AR) to elicit its immunosuppressive effect. Although the important role of the adenosine--A2AR interaction in limiting inflammation has been established, the current study revisited this issue by asking whether hypoxia can also exert its T-cell inhibitory effects even without A2AR. A similar degree of hypoxia-triggered inhibition was observed in wild-type and A2AR-deficient T cells both in vitro and, after exposure of mice to a hypoxic atmosphere, in vivo. This A2AR-independent hypoxic T-cell suppression was qualitatively and mechanistically different from immunosuppression by A2AR stimulation. The A2AR-independent hypoxic immunosuppression strongly reduced T-cell proliferation, while IFN-γ-producing activity was more susceptible to the A2AR-dependent inhibition. In contrast to the sustained functional impairment after A2AR-mediated T-cell inhibition, the A2AR-independent inhibition under hypoxia was short lived, as evidenced by the quick recovery of IFN-γ-producing activity upon re-stimulation. These data support the view that T-cell inhibition by hypoxia can be mediated by multiple mechanisms and that both A2AR and key molecules in the A2AR-independent T-cell inhibition should be targeted to overcome the hypoxia-related immunosuppression in infected tissues and tumors.
Collapse
Affiliation(s)
- Akio Ohta
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
127
|
Bellone M, Calcinotto A. Ways to enhance lymphocyte trafficking into tumors and fitness of tumor infiltrating lymphocytes. Front Oncol 2013; 3:231. [PMID: 24062984 PMCID: PMC3769630 DOI: 10.3389/fonc.2013.00231] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 08/23/2013] [Indexed: 12/26/2022] Open
Abstract
The tumor is a hostile microenvironment for T lymphocytes. Indeed, irregular blood flow, and endothelial cell (EC) anergy that characterize most solid tumors hamper leukocyte adhesion, extravasation, and infiltration. In addition, hypoxia and reprograming of energy metabolism within cancer cells transform the tumor mass in a harsh environment that limits survival and effector functions of T cells, regardless of being induced in vivo by vaccination or adoptively transferred. In this review, we will summarize on recent advances in our understanding of the characteristics of tumor-associated neo-angiogenic vessels as well as of the tumor metabolism that may impact on T cell trafficking and fitness of tumor infiltrating lymphocytes. In particular, we will focus on how advances in knowledge of the characteristics of tumor ECs have enabled identifying strategies to normalize the tumor-vasculature and/or overcome EC anergy, thus increasing leukocyte-vessel wall interactions and lymphocyte infiltration in tumors. We will also focus on drugs acting on cells and their released molecules to transiently render the tumor microenvironment more suitable for tumor infiltrating T lymphocytes, thus increasing the therapeutic effectiveness of both active and adoptive immunotherapies.
Collapse
Affiliation(s)
- Matteo Bellone
- Cellular Immunology Unit, Department of Immunology, Infectious Diseases and Transplantation, San Raffaele Scientific Institute , Milan , Italy
| | | |
Collapse
|
128
|
McElwee KJ, Gilhar A, Tobin DJ, Ramot Y, Sundberg JP, Nakamura M, Bertolini M, Inui S, Tokura Y, Jr LEK, Duque-Estrada B, Tosti A, Keren A, Itami S, Shoenfeld Y, Zlotogorski A, Paus R. What causes alopecia areata? Exp Dermatol 2013; 22:609-26. [PMID: 23947678 PMCID: PMC4094373 DOI: 10.1111/exd.12209] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The pathobiology of alopecia areata (AA), one of the most frequent autoimmune diseases and a major unsolved clinical problem, has intrigued dermatologists, hair biologists and immunologists for decades. Simultaneously, both affected patients and the physicians who take care of them are increasingly frustrated that there is still no fully satisfactory treatment. Much of this frustration results from the fact that the pathobiology of AA remains unclear, and no single AA pathogenesis concept can claim to be universally accepted. In fact, some investigators still harbour doubts whether this even is an autoimmune disease, and the relative importance of CD8(+) T cells, CD4(+) T cells and NKGD2(+) NK or NKT cells and the exact role of genetic factors in AA pathogenesis remain bones of contention. Also, is AA one disease, a spectrum of distinct disease entities or only a response pattern of normal hair follicles to immunologically mediated damage? During the past decade, substantial progress has been made in basic AA-related research, in the development of new models for translationally relevant AA research and in the identification of new therapeutic agents and targets for future AA management. This calls for a re-evaluation and public debate of currently prevalent AA pathobiology concepts. The present Controversies feature takes on this challenge, hoping to attract more skin biologists, immunologists and professional autoimmunity experts to this biologically fascinating and clinically important model disease.
Collapse
Affiliation(s)
- K. J. McElwee
- Department of Dermatology and Skin Science, University of British Columbia, Vancouver, BC, Canada
| | - A. Gilhar
- Laboratory for Skin, Research, Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel Marta Bertolini
| | - D. J. Tobin
- Centre for Skin Sciences, School of Life Sciences, University of Bradford, Bradford, West Yorkshire, BD7 1DP, UK
| | - Y. Ramot
- Department of Dermatology, Hadassah- Hebrew University Medical Center, Jerusalem 91120, Israel
| | - J. P. Sundberg
- The Jackson Laboratory, Bar Harbor, ME, USA; Division of Dermatology, Skin Disease Research Center, Vanderbilt University, Nashville, TN, USA
| | - M. Nakamura
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan Yoshiki Tokura
| | - M. Bertolini
- Department of Dermatology, University of Lübeck, Germany Yehuda Shoenfeld
| | - S. Inui
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y. Tokura
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - L. E. King Jr
- The Jackson Laboratory, Bar Harbor, ME, USA; Division of Dermatology, Skin Disease Research Center, Vanderbilt University, Nashville, TN, USA
| | - B. Duque-Estrada
- Instituto de Dermatologia Prof. Rubem David Azulay, Rio de Janeiro, Brazil Antonella Tosti
| | - A Tosti
- Department of Dermatology, University of Miami, Miami, FL, USA
| | - A. Keren
- Laboratory for Skin, Research, Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel Marta Bertolini
| | - S. Itami
- Department of Regenerative Dermatology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Y. Shoenfeld
- Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer and Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Israel
| | - A. Zlotogorski
- Department of Dermatology, Hadassah- Hebrew University Medical Center, Jerusalem 91120, Israel
| | - R. Paus
- Department of Dermatology, University of Lübeck, Germany; Institute of Inflammation and Repair, University of Manchester, Manchester, UK ,
| |
Collapse
|
129
|
Huang L, Li L, Klonowski KD, Tompkins SM, Tripp RA, Mellor AL. Induction and role of indoleamine 2,3 dioxygenase in mouse models of influenza a virus infection. PLoS One 2013; 8:e66546. [PMID: 23785507 PMCID: PMC3681773 DOI: 10.1371/journal.pone.0066546] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 05/07/2013] [Indexed: 11/18/2022] Open
Abstract
Influenza infection stimulates protective host immune responses but paradoxically enhances lung indoleamine 2,3 dioxygenase (IDO) activity, an enzyme that suppresses helper/effector T cells and activates Foxp3-lineage regulatory CD4 T cells (Tregs). Influenza A/PR/8/34 (PR8) infection stimulated rapid elevation of IDO activity in lungs and lung-draining mediastinal lymph nodes (msLNs). Mice lacking intact IDO1 genes (IDO1-KO mice) exhibited significantly lower morbidity after sub-lethal PR8 infection, and genetic or pharmacologic IDO ablation led to much faster recovery after virus clearance. More robust influenza-specific effector CD8 T cell responses manifested in lungs of PR8-infected IDO1-KO mice, though virus clearance rates were unaffected by IDO ablation. Similar outcomes manifested in mice infected with a less virulent influenza A strain (X31). IDO induction in X31-infected lungs was dependent on IFN type II (IFNγ) signaling and was restricted to non-hematopoietic cells, while redundant IFN type 1 or type II signaling induced IDO exclusively in hematopoietic cells from msLNs. Memory T cells generated in X31-primed IDO1-KO mice protected mice from subsequent challenge with lethal doses of PR8 (100×LD50). However recall T cell responses were less robust in lung interstitial tissues, and classic dominance of TCR Vβ8.3 chain usage amongst memory CD8(+) T cells specific for influenza nucleoprotein (NP366) did not manifest in IDO1-KO mice. Thus, influenza induced IDO activity in lungs enhanced morbidity, slowed recovery, restrained effector T cell responses in lungs and shaped memory T cell repertoire generation, but did not attenuate virus clearance during primary influenza A infection.
Collapse
Affiliation(s)
- Lei Huang
- Cancer Immunology, Inflammation and Tolerance Program, Cancer Center, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Radiology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Lingqian Li
- Cancer Immunology, Inflammation and Tolerance Program, Cancer Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - Kim D. Klonowski
- Department of Cell Biology, College of Arts and Sciences, University of Georgia, Athens, Georgia, United States of America
| | - S. Mark Tompkins
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia Athens, Georgia, United States of America
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia Athens, Georgia, United States of America
| | - Andrew L. Mellor
- Cancer Immunology, Inflammation and Tolerance Program, Cancer Center, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Medicine, Georgia Regents University, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
130
|
Lavoué V, Thédrez A, Levêque J, Foucher F, Henno S, Jauffret V, Belaud-Rotureau MA, Catros V, Cabillic F. Immunity of human epithelial ovarian carcinoma: the paradigm of immune suppression in cancer. J Transl Med 2013; 11:147. [PMID: 23763830 PMCID: PMC3683338 DOI: 10.1186/1479-5876-11-147] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 05/27/2013] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is a significant cause of cancer-related mortality in women, and there has been no substantial decrease in the death rates due to EOC in the last three decades. Thus, basic knowledge regarding ovarian tumor cell biology is urgently needed to allow the development of innovative treatments for EOC. Traditionally, EOC has not been considered an immunogenic tumor, but there is evidence of an immune response to EOC in patients. Clinical data demonstrate that an antitumor immune response and immune evasion mechanisms are correlated with a better and lower survival, respectively, providing evidence for the immunoediting hypothesis in EOC. This review focuses on the immune response and immune suppression in EOC. The immunological roles of chemotherapy and surgery in EOC are also described. Finally, we detail pilot data supporting the efficiency of immunotherapy in the treatment of EOC and the emerging concept that immunomodulation aimed at counteracting the immunosuppressive microenvironment must be associated with immunotherapy strategies.
Collapse
Affiliation(s)
- Vincent Lavoué
- Lady Davis Institut, Jewish General Hospital, McGill University, Montreal QC H3T 1E2, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Tüting T. T cell immunotherapy for melanoma from bedside to bench to barn and back: how conceptual advances in experimental mouse models can be translated into clinical benefit for patients. Pigment Cell Melanoma Res 2013; 26:441-56. [PMID: 23617831 DOI: 10.1111/pcmr.12111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 04/18/2013] [Indexed: 12/27/2022]
Abstract
A solid scientific basis now supports the concept that cytotoxic T lymphocytes can specifically recognize and destroy melanoma cells. Over the last decades, clinicians and basic scientists have joined forces to advance our concepts of melanoma immunobiology. This has catalyzed the rational development of therapeutic approaches to enforce melanoma-specific T cell responses. Preclinical studies in experimental mouse models paved the way for their successful translation into clinical benefit for patients with metastatic melanoma. A more thorough understanding of how melanomas develop resistance to T cell immunotherapy is necessary to extend this success. This requires a continued interdisciplinary effort of melanoma biologists and immunologists that closely connects clinical observations with in vitro investigations and appropriate in vivo mouse models: From bedside to bench to barn and back.
Collapse
Affiliation(s)
- Thomas Tüting
- Laboratory of Experimental Dermatology, Department of Dermatology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
132
|
Hölzel M, Bovier A, Tüting T. Plasticity of tumour and immune cells: a source of heterogeneity and a cause for therapy resistance? Nat Rev Cancer 2013; 13:365-76. [PMID: 23535846 DOI: 10.1038/nrc3498] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunotherapies, signal transduction inhibitors and chemotherapies can successfully achieve remissions in advanced stage cancer patients, but durable responses are rare. Using malignant melanoma as a paradigm, we propose that therapy-induced injury to tumour tissue and the resultant inflammation can activate protective and regenerative responses that represent a shared resistance mechanism to different treatments. Inflammation-driven phenotypic plasticity alters the antigenic landscape of tumour cells, rewires oncogenic signalling networks, protects against cell death and reprogrammes immune cell functions. We propose that the successful combination of cancer treatments to tackle resistance requires an interdisciplinary understanding of these resistance mechanisms, supported by mathematical models.
Collapse
Affiliation(s)
- Michael Hölzel
- Unit for RNA Biology, Department of Clinical Chemistry and Clinical Pharmacology, University of Bonn, 53105 Bonn, Germany
| | | | | |
Collapse
|
133
|
Graham JG, Zhang X, Goodman A, Pothoven K, Houlihan J, Wang S, Gower RM, Luo X, Shea LD. PLG scaffold delivered antigen-specific regulatory T cells induce systemic tolerance in autoimmune diabetes. Tissue Eng Part A 2013; 19:1465-75. [PMID: 23432371 DOI: 10.1089/ten.tea.2012.0643] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Islet transplantation is a promising treatment for human type 1 diabetes mellitus. Transplantation requires systemic immunosuppression, which has numerous deleterious side effects. Islet antigen-specific regulatory T cells (Tregs) have been shown to protect islet grafts from autoimmune destruction in the nonobese diabetic (NOD) model when co-localized in the kidney capsule. An extra-hepatic transplant site was established by transplanting islet-loaded microporous poly (lactide-co-glycolide) (PLG) scaffolds into abdominal fat. This study examined an autoimmune transplantation model and determined whether co-localized Tregs could protect islet grafts in an extra-hepatic and extra-renal transplant site. Normoglycemia was restored, and co-transplanted Tregs extended graft survival, including several instances of indefinite protection. Transplanted Tregs were replaced by recipient-derived Tregs over time, indicating that islet antigen-specific Tregs induce tolerance to islet grafts through host-derived Tregs. Thus, Tregs provided protection against a diverse repertoire of autoreactive T-cell-receptor specificities mediating diabetes in the NOD model, possibly through a phenomenon previously described as infectious tolerance. Interestingly, the infiltration by Tregs protected a second islet transplant, indicating systemic tolerance to islet antigens. In summary, PLG scaffolds can serve as an alternative delivery system for islet transplantation that allows for the co-localization of immunomodulatory cells within islet grafts and induces long-term graft survival in an autoimmune diabetes model. This method of co-localizing immunomodulatory cells with islets in a clinically translatable transplant site to affect the immune system on a local and systemic level has potential therapeutic implications for human islet transplantation.
Collapse
Affiliation(s)
- John G Graham
- Department of Chemical and Biological Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
134
|
Ricupito A, Grioni M, Calcinotto A, Hess Michelini R, Longhi R, Mondino A, Bellone M. Booster vaccinations against cancer are critical in prophylactic but detrimental in therapeutic settings. Cancer Res 2013; 73:3545-54. [PMID: 23539449 DOI: 10.1158/0008-5472.can-12-2449] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although cancer vaccines are in the clinic, several issues remain to be addressed to increase vaccine efficacy. In particular, whether how and how frequently a patient should be boosted remains to be defined. Here, we have assessed the ability of dendritic cell (DC)-based vaccines to induce a long-lasting tumor-specific CTL response in either prophylactic or therapeutic settings by taking advantage of transplantable and spontaneous mouse tumor models. Implementing a 24-hour ex vivo intracellular cytokine production assay, we have found that priming with a DC-based vaccine induced a long-lasting CTL response in wild-type mice, and homologous boosting better sustained the pool of central memory T cells, which associated with potent protection against B16F1 melanoma challenge. Appropriate timing of booster vaccination was also critical, as a tight boosting schedule hindered persistence of IFN-γ-competent memory CD8(+) T cells and mice survival in prophylactic settings. Conversely, prime/boost vaccination proved to be of no advantage or even detrimental in therapeutic settings in B16F1 and transgenic adenocarcinoma of the mouse prostate (TRAMP) models, respectively. Although DC priming was indeed needed for tumor shrinkage, restoration of immune competence, and prolonged survival of TRAMP mice, repeated boosting did not sustain the pool of central memory CTLs and was detrimental for mice overall survival. Thus, our results indicate that booster vaccinations impact antitumor immunity to different extents, depending on their prophylactic or therapeutic administration, and suggest evaluating the need for boosting in any given patient with cancer depending on the state of the disease.
Collapse
Affiliation(s)
- Alessia Ricupito
- Cellular Immunology Unit; Program of Immunology, Gene Therapy and Bio-Immunotherapy of Cancer (PIBIC, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
135
|
Dobrzanski MJ. Expanding roles for CD4 T cells and their subpopulations in tumor immunity and therapy. Front Oncol 2013; 3:63. [PMID: 23533029 PMCID: PMC3607796 DOI: 10.3389/fonc.2013.00063] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/07/2013] [Indexed: 12/18/2022] Open
Abstract
The importance of CD4 T cells in orchestrating the immune system and their role in inducing effective T cell-mediated therapies for the treatment of patients with select established malignancies are undisputable. Through a complex and balanced array of direct and indirect mechanisms of cellular activation and regulation, this functionally diverse family of lymphocytes can potentially promote tumor eradication, long-term tumor immunity, and aid in establishing and/or rebalancing immune cell homeostasis through interaction with other immune cell populations within the highly dynamic tumor environment. However, recent studies have uncovered additional functions and roles for CD4 T cells, some of which are independent of other lymphocytes, that can not only influence and contribute to tumor immunity but paradoxically promote tumor growth and progression. Here, we review the recent advances in our understanding of the various CD4 T cell lineages and their signature cytokines in disease progression and/or regression. We discuss their direct and indirect mechanistic interplay among themselves and with other responding cells of the antitumor response, their potential roles and abilities for "plasticity" and memory cell generation within the hostile tumor environment, and their potentials in cancer treatment and immunotherapy.
Collapse
Affiliation(s)
- Mark J. Dobrzanski
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of MedicineAmarillo, TX, USA
| |
Collapse
|
136
|
Regateiro FS, Cobbold SP, Waldmann H. CD73 and adenosine generation in the creation of regulatory microenvironments. Clin Exp Immunol 2013. [PMID: 23199317 DOI: 10.1111/j.1365-2249.2012.04623.x] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Extracellular adenosine 5'-triphosphate (ATP) acts on many immune cells to promote inflammation. Conversely, the ATP metabolite adenosine is mainly an anti-inflammatory molecule. The ecto-enzymes CD39 and CD73 can dephosphorylate extracellular ATP to adenosine, thereby controlling this important pathway of immune modulation. Despite their established roles in the immune system, little is known of how CD39 and CD73 are themselves regulated. Recent data have shown that CD73 expression and adenosine generation are up-regulated by transforming growth factor-β, depending on the cytokine content of the local microenvironment. We review here these recent findings and discuss their implications in disease.
Collapse
Affiliation(s)
- F S Regateiro
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
137
|
Mast cell interleukin-10 drives localized tolerance in chronic bladder infection. Immunity 2013; 38:349-59. [PMID: 23415912 DOI: 10.1016/j.immuni.2012.10.019] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 10/09/2012] [Indexed: 01/13/2023]
Abstract
The lower urinary tract's virtually inevitable exposure to external microbial pathogens warrants efficient tissue-specialized defenses to maintain sterility. The observation that the bladder can become chronically infected in combination with clinical observations that antibody responses after bladder infections are not detectable suggest defects in the formation of adaptive immunity and immunological memory. We have identified a broadly immunosuppressive transcriptional program specific to the bladder, but not the kidney, during infection of the urinary tract that is dependent on tissue-resident mast cells (MCs). This involves localized production of interleukin-10 and results in suppressed humoral and cell-mediated responses and bacterial persistence. Therefore, in addition to the previously described role of MCs orchestrating the early innate immunity during bladder infection, they subsequently play a tissue-specific immunosuppressive role. These findings may explain the prevalent recurrence of bladder infections and suggest the bladder as a site exhibiting an intrinsic degree of MC-maintained immune privilege.
Collapse
|
138
|
Abstract
Upon activation, quiescent naive T cells undergo a growth phase followed by massive clonal expansion and differentiation that are essential for appropriate immune defense and regulation. Accumulation of cell biomass during the initial growth and rapid proliferation during the expansion phase is associated with dramatically increased bioenergetic and biosynthetic demands. This not only requires a metabolic rewiring during the transition between resting and activation but also 'addicts' active T cells to certain metabolic pathways in ways that naive and memory T cells are not. We consider such addiction in terms of the biological effects of deprivation of metabolic substrates or inhibition of specific pathways in T cells. In this review, we illustrate the relevant metabolic pathways revealed by recent metabolic flux analysis and discuss the consequences of metabolic intervention on specific metabolic pathways in T lymphocytes.
Collapse
Affiliation(s)
- Ruoning Wang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | | |
Collapse
|
139
|
Indoleamine 2,3-dioxygenase expression is associated with chronic rhinosinusitis. Curr Opin Allergy Clin Immunol 2013; 13:37-44. [DOI: 10.1097/aci.0b013e32835b350e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
140
|
Sunay M, Marincola F, Khleif SN, Silverstein SC, Fox BA, Galon J, Emens LA. Focus on the target: the tumor microenvironment, Society for Immunotherapy of Cancer Annual Meeting Workshop, October 24th-25th 2012. J Immunother Cancer 2013. [PMCID: PMC4019899 DOI: 10.1186/2051-1426-1-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Workshop associated with the 27th Annual Meeting of the Society for Immunotherapy of Cancer (SITC), North Bethesda, MD, October 24-25, 2012 focused on targeting the tumor microenvironment as part of an integrative approach to immune-based cancer therapy.
Collapse
|
141
|
Abstract
Immune privilege provides protection to vital tissues or cells of the body when foreign antigens are introduced into these sites. The modern concept of relative immune privilege applies to a variety of tissues and anatomical structures, including the hair follicles and mucosal surfaces. Even sites of chronic inflammation and developing tumors may acquire immune privilege by recruiting immunoregulatory effector cells. Adult stem cells are no exception. For their importance and vitality, many adult stem cell populations are believed to be immune privileged. A preimplantation-stage embryo that derives from a totipotent stem cell (i.e., a fertilized oocyte) must be protected from maternal allo-rejection for successful implantation and development to occur. Embryonic stem cells, laboratory-derived cell lines of preimplantation blastocyst-origin, may, therefore, retain some of the immunological properties of the developing embryo. However, embryonic stem cells and their differentiated tissue derivatives transplanted into a recipient do not necessarily have an ability to subvert immune responses to the extent required to exploit their pluripotency for regenerative medicine. In this review, an extended definition of immune privilege is developed and the capacity of adult and embryonic stem cells to display both relative and acquired immune privilege is discussed. Furthermore, we explore how these intrinsic properties of stem cells may one day be harnessed for therapeutic gain.
Collapse
Affiliation(s)
- Naoki Ichiryu
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | |
Collapse
|
142
|
Tardito S, Negrini S, Conteduca G, Ferrera F, Parodi A, Battaglia F, Kalli F, Fenoglio D, Cutolo M, Filaci G. Indoleamine 2,3 dioxygenase gene polymorphisms correlate with CD8+ Treg impairment in systemic sclerosis. Hum Immunol 2012. [PMID: 23200754 DOI: 10.1016/j.humimm.2012.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Systemic sclerosis (SSc) is characterized by tissue fibrosis, vasculopathy and autoimmunity. Indoleamine 2,3 dioxygenase (IDO) plays a pivotal role in immunological tolerance modulating regulatory T cell (Treg) generation and function. Single nucleotide polymorphisms (SNPs) of IDO gene could impact on Treg function and predispose to autoimmunity. Here, the existence of an association between specific IDO SNPs and SSc was analyzed. Five specific SNPs in IDO gene were searched in 31 SSc patients and 37 healthy controls by gene sequencing or restriction fragment length polymorphism. The function of both CD4+CD25+ and CD8+ Treg from SSc patients was analyzed by proliferation suppression assay. SNP rs7820268 was statistically more frequent in SSc patients than in controls. Notably, SSc patients bearing the T allelic variant of the rs7820268 SNP showed impaired CD8+ Treg function. Our unprecedented data show that a specific IDO gene SNP is associated with an autoimmune disease such as SSc.
Collapse
Affiliation(s)
- Samuele Tardito
- Centre of Excellence for Biomedical Research, University of Genoa, Genoa 16132, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Forrester JV, Xu H. Good news-bad news: the Yin and Yang of immune privilege in the eye. Front Immunol 2012; 3:338. [PMID: 23230433 PMCID: PMC3515883 DOI: 10.3389/fimmu.2012.00338] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Accepted: 10/23/2012] [Indexed: 12/27/2022] Open
Abstract
The eye and the brain are prototypical tissues manifesting immune privilege (IP) in which immune responses to foreign antigens, particularly alloantigens are suppressed, and even completely inhibited. Explanations for this phenomenon are numerous and mostly reflect our evolving understanding of the molecular and cellular processes underpinning immunological responses generally. IP is now viewed as a property of many tissues and the level of expression of IP varies not only with the tissue but with the nature of the foreign antigen and changes in the limited conditions under which privilege can operate as a mechanism of immunological tolerance. As a result, IP functions normally as a homeostatic mechanism preserving normal function in tissues, particularly those with highly specialized function and limited capacity for renewal such as the eye and brain. However, IP is relatively easily bypassed in the face of a sufficiently strong immunological response, and the privileged tissues may be at greater risk of collateral damage because its natural defenses are more easily breached than in a fully immunocompetent tissue which rapidly rejects foreign antigen and restores integrity. This two-edged sword cuts its swathe through the eye: under most circumstances, IP mechanisms such as blood-ocular barriers, intraocular immune modulators, induction of T regulatory cells, lack of lymphatics, and other properties maintain tissue integrity; however, when these are breached, various degrees of tissue damage occur from severe tissue destruction in retinal viral infections and other forms of uveoretinal inflammation, to less severe inflammatory responses in conditions such as macular degeneration. Conversely, ocular IP and tumor-related IP can combine to permit extensive tumor growth and increased risk of metastasis thus threatening the survival of the host.
Collapse
Affiliation(s)
- John V. Forrester
- Laboratory of Immunology, Lion’s Eye Institute, University of Western AustraliaPerth, WA, Australia
- Ocular Immunology Laboratory, Section of Immunology and Infection, Institute of Medical Sciences, University of AberdeenAberdeen, UK
| | - Heping Xu
- Laboratory of Immunology, Lion’s Eye Institute, University of Western AustraliaPerth, WA, Australia
| |
Collapse
|
144
|
Qian F, Liao J, Villella J, Edwards R, Kalinski P, Lele S, Shrikant P, Odunsi K. Effects of 1-methyltryptophan stereoisomers on IDO2 enzyme activity and IDO2-mediated arrest of human T cell proliferation. Cancer Immunol Immunother 2012; 61:2013-20. [PMID: 22527253 PMCID: PMC11028567 DOI: 10.1007/s00262-012-1265-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 04/10/2012] [Indexed: 12/17/2022]
Abstract
IDO2 is a newly discovered enzyme with 43 % similarity to classical IDO (IDO1) protein and shares the same critical catalytic residues. IDO1 catalyzes the initial and rate-limiting step in the degradation of tryptophan and is a key enzyme in mediating tumor immune tolerance via arrest of T cell proliferation. The role of IDO2 in human T cell immunity remains controversial. Here, we demonstrate that similar to IDO1, IDO2 also degrades tryptophan into kynurenine and is inhibited more efficiently by Levo-1-methyl tryptophan (L-1MT), an IDO1 competitive inhibitor, than by dextro-methyl tryptophan (D-1MT). Although IDO2 enzyme activity is weaker than IDO1, it is less sensitive to 1-MT inhibition than IDO1. Moreover, our results indicate that human CD4(+) and CD8(+) T cell proliferation was inhibited by IDO2, but both L-1MT and D-1MT could not reverse IDO2-mediated arrest of cell proliferation, even at high concentrations. These data indicate that IDO2 is an inhibitory mechanism in human T cell proliferation and support efforts to develop more effective IDO1 and IDO2 inhibitors in order to overcome IDO-mediated immune tolerance.
Collapse
Affiliation(s)
- Feng Qian
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY USA
- Roswell Park Cancer Institute, Center for Immunotherapy, Elm & Carlton Streets, Buffalo, NY 14263 USA
| | - Jianqun Liao
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY USA
| | - Jeannine Villella
- Division of Gynecologic Oncology, Winthrop-University Hospital, New York, NY USA
| | - Robert Edwards
- Department of Obstetrics and Gynecology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Pawel Kalinski
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Shashikant Lele
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY USA
| | - Protul Shrikant
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY USA
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY USA
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY USA
- Roswell Park Cancer Institute, Center for Immunotherapy, Elm & Carlton Streets, Buffalo, NY 14263 USA
| |
Collapse
|
145
|
Colton CA. Immune heterogeneity in neuroinflammation: dendritic cells in the brain. J Neuroimmune Pharmacol 2012; 8:145-62. [PMID: 23114889 PMCID: PMC4279719 DOI: 10.1007/s11481-012-9414-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 10/22/2012] [Indexed: 12/20/2022]
Abstract
Dendritic cells (DC) are critical to an integrated immune response and serve as the key link between the innate and adaptive arms of the immune system. Under steady state conditions, brain DC’s act as sentinels, continually sampling their local environment. They share this function with macrophages derived from the same basic hemopoietic (bone marrow-derived) precursor and with parenchymal microglia that arise from a unique non-hemopoietic origin. While multiple cells may serve as antigen presenting cells (APCs), dendritic cells present both foreign and self-proteins to naïve T cells that, in turn, carry out effector functions that serve to protect or destroy. The resulting activation of the adaptive response is a critical step to resolution of injury or infection and is key to survival. In this review we will explore the critical roles that DCs play in the brain’s response to neuroinflammatory disease with emphasis on how the brain’s microenvironment impacts these actions.
Collapse
Affiliation(s)
- Carol A Colton
- Neurology, Duke University Medical Center, Box 2900, Durham, NC 27710, USA.
| |
Collapse
|
146
|
Platten M, Wick W, Van den Eynde BJ. Tryptophan Catabolism in Cancer: Beyond IDO and Tryptophan Depletion. Cancer Res 2012; 72:5435-40. [DOI: 10.1158/0008-5472.can-12-0569] [Citation(s) in RCA: 482] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
147
|
Johnson TS, Munn DH. Host Indoleamine 2,3-Dioxygenase: Contribution to Systemic Acquired Tumor Tolerance. Immunol Invest 2012; 41:765-97. [DOI: 10.3109/08820139.2012.689405] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
148
|
Cawood R, Hills T, Wong SL, Alamoudi AA, Beadle S, Fisher KD, Seymour LW. Recombinant viral vaccines for cancer. Trends Mol Med 2012; 18:564-74. [PMID: 22917663 DOI: 10.1016/j.molmed.2012.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 07/14/2012] [Accepted: 07/18/2012] [Indexed: 01/21/2023]
Abstract
Cancer arises from 'self' in a series of steps that are all subject to immunoediting. Therefore, therapeutic cancer vaccines must stimulate an immune response against tumour antigens that have already evaded the body's immune defences. Vaccines presenting a tumour antigen in the context of obvious danger signals seem more likely to stimulate a response. This approach can be facilitated by genetic engineering using recombinant viral vectors expressing tumour antigens, cytokines, or both, from an immunogenic virus particle. We overview clinical attempts to use these agents for systemic immunisation and contrast the results with strategies employing direct intratumoural administration. We focus on the challenge of producing an effective response within the immune-suppressive tumour microenvironment, and discuss how the technology can overcome these obstacles.
Collapse
Affiliation(s)
- Ryan Cawood
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | | | | | | | | | | | | |
Collapse
|
149
|
Eleftheriadis T, Yiannaki E, Antoniadi G, Liakopoulos V, Pissas G, Galaktidou G, Stefanidis I. Plasma indoleamine 2,3-dioxygenase and arginase type I may contribute to decreased blood T-cell count in hemodialysis patients. Ren Fail 2012; 34:1118-22. [PMID: 22897519 DOI: 10.3109/0886022x.2012.713297] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Acquired immunity is impaired in hemodialysis (HD) patients, and decreased T-cell number may contribute. Indoleamine 2,3-dioxygenase (IDO) and arginase type I (ARG) catabolize tryptophane and arginine, respectively, and exert proapoptotic and antiproliferative effects on T-cells. Plasma levels of IDO and ARG and their relation to blood T-cell number were evaluated in HD patients. METHODS Thirty-two HD patients and 20 healthy controls participated in the study. Plasma IDO and ARG were measured by means of enzyme-linked immunosorbent assay. T-cell number was assessed by means of flow cytometry. RESULTS IDO concentration was significantly higher in HD patients than in healthy volunteers (44.30 ± 31.83 ng/mL vs. 21.28 ± 26.21 ng/mL, p = 0.009). There was a trend for higher ARG concentration in HD patients (13.43 ± 11.91 ng/mL) than in healthy volunteers (9.56 ± 4.03 ng/mL), which, however, did not reach statistic significance (p = 0.099). Absolute T-cell count was significantly lower in HD patients than in healthy controls (1176.99 ± 567.71 cells/mm3 vs. 1519.85 ± 594.96 cells/mm3, p = 0.040). Absolute blood T-cell number was inversely related to plasma IDO (r = -0.490, p = 0.004) and to plasma ARG (r = -0.387, p = 0.029) concentrations. CONCLUSIONS Plasma IDO and ARG may contribute to decreased blood T-cell count in HD patients.
Collapse
|
150
|
Pre-administration of L-tryptophan improved ADR-induced early renal failure in mice. Life Sci 2012; 91:100-6. [DOI: 10.1016/j.lfs.2012.06.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 05/11/2012] [Accepted: 06/15/2012] [Indexed: 11/18/2022]
|