101
|
Minami S, Ihara S, Komuta K. Pretreatment Lymphocyte to Monocyte Ratio as a Prognostic Marker for Advanced Pulmonary Squamous Cell Carcinoma Treated With Chemotherapy. J Clin Med Res 2018; 10:657-664. [PMID: 29977424 PMCID: PMC6031246 DOI: 10.14740/jocmr3490w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/29/2018] [Indexed: 01/14/2023] Open
Abstract
Background Lower lymphocyte to monocyte ratio (LMR), higher neutrophil to lymphocyte ratio (NLR) and modified Glasgow prognostic score (mGPS) 2 have been demonstrated as independent prognostic markers for poor prognosis of advanced non-small cell lung cancer (NSCLC). However, little is known about these three markers as prognostic markers for a specific histological subset of NSCLC, squamous cell carcinoma (SCC). This study aimed to evaluate the prognostic significance of LMR, NLR and mGPS for advanced SCC. Methods We retrospectively collected 107 patients who met the following criteria: pathologically confirmed SCC, chemo-naive patients who had initiated first-line cytotoxic chemotherapy between September 2007 and February 2017 at our institution, and c-stage IIIB, IV or recurrence after curative-intent surgery or thoracic radiotherapy. In order to demonstrate these three markers as significant prognostic factors, we compared overall survival (OS) between two groups divided by LMR, NLR and mGPS 0 - 1 versus 2, and performed univariate and multivariate Cox proportional hazard analyses. Results Groups with low LMR (< 2.07) and high NLR (≥ 5.28) experienced shorter OS (LMR: 6.5 versus 15.6 months in median, P < 0.01; NLR: 8.2 versus 15.6 months, P < 0.01) than groups with high LMR (≥ 2.07) and low NLR (< 5.28). However, no significant difference was detected in OS between mGPS 0 - 1 and 2 (13.0 versus 13.7 months, P = 0.61). As significant poor prognostic factors, our multivariate Cox hazard analysis detected ECOG PS 2 - 4 (hazard ration (HR): 3.09, 95% confidence interval (CI): 1.77 - 5.40; P < 0.01) and LMR < 2.07 (HR: 0.39, 95% CI: 0.21 - 0.79; P < 0.01). However, NLR was not selected in the multivariate analysis. Conclusion LMR is an independent prognostic factor for advanced pulmonary SCC. Neither NLR nor mGPS is useful as prognostic factor for this histology. The optimal prognostic markers may differ from each subset of NSCLC.
Collapse
Affiliation(s)
- Seigo Minami
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Shouichi Ihara
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Kiyoshi Komuta
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| |
Collapse
|
102
|
Yuan XH, Yang J, Wang XY, Zhang XL, Qin TT, Li K. Association between EGFR/KRAS mutation and expression of VEGFA, VEGFR and VEGFR2 in lung adenocarcinoma. Oncol Lett 2018; 16:2105-2112. [PMID: 30008907 PMCID: PMC6036498 DOI: 10.3892/ol.2018.8901] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 11/10/2017] [Indexed: 02/06/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) and Kirsten rat sarcoma viral oncogene homolog (KRAS) are two of the most notable driver genes in lung cancer, whilst vascular endothelial growth factor (VEGF) signaling serves a critical function in tumor angiogenesis. However, few studies have focused on the potential connection between EGFR/KRAS mutational status, and VEGFA, VEGF receptor (VEGFR)1 and VEGFR2 expression in lung adenocarcinoma. EGFR (exon 19, 20 and 21) and KRAS (exon 2) mutations were detected using an amplification refractory mutation system technique, and the expression of VEGFA, VEGFR1 and VEGFR2 was analyzed using immunohistochemistry in 204 patients with lung adenocarcinoma. Associations between EGFR/KRAS mutational status and VEGFA, VEGFR1, and VEGFR2 expression was analyzed using Pearson χ2 tests. It was revealed that EGFR 21 exon (P=0.033) and EGFR 20 exon (P=0.002) mutated tumors exhibited a significantly higher level of expression of VEGFA. EGFR 21 exon mutant tumors additionally demonstrated a significantly higher level of co-expression of VEGFA and VEGFR1 (P<0.001). EGFR 19 exon mutation was significantly associated with low levels of VEGFR1 (P=0.008). KRAS mutation was significantly associated with a high level of co-expression of VEGFA, VEGFR1 and VEGFR2 (P=0.035), but no such association with the individual expression of VEGFA, VEGFR1 or VEGFR2 was identified. However, neither KRAS or EGFR mutations exhibited an association with the expression of VEGFR2. The present study may help in the treatment of various patients with KRAS or subtype of EGFR mutation with anti-angiogenesis therapy.
Collapse
Affiliation(s)
- Xiao-Han Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Jie Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xin-Yue Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xiao-Ling Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Ting-Ting Qin
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Kai Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Henan, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Tianjin Clinical Research Center for Cancer, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
103
|
Grizzi F, Basso G, Borroni EM, Cavalleri T, Bianchi P, Stifter S, Chiriva-Internati M, Malesci A, Laghi L. Evolving notions on immune response in colorectal cancer and their implications for biomarker development. Inflamm Res 2018; 67:375-389. [PMID: 29322204 DOI: 10.1007/s00011-017-1128-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 12/27/2017] [Accepted: 12/29/2017] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Colorectal cancer (CRC) still represents the third most commonly diagnosed type of cancer in men and women worldwide. CRC is acknowledged as a heterogeneous disease that develops through a multi-step sequence of events driven by clonal selections; this observation is sustained by the fact that histologically similar tumors may have completely different outcomes, including a varied response to therapy. METHODS In "early" and "intermediate" stage of CRC (stages II and III, respectively) there is a compelling need for new biomarkers fit to assess the metastatic potential of their disease, selecting patients with aggressive disease that might benefit from adjuvant and targeted therapies. Therefore, we review the actual notions on immune response in colorectal cancer and their implications for biomarker development. RESULTS The recognition of the key role of immune cells in human cancer progression has recently drawn attention on the tumor immune microenvironment, as a source of new indicators of tumor outcome and response to therapy. Thus, beside consolidated histopathological biomarkers, immune endpoints are now emerging as potential biomarkers. CONCLUSIONS The introduction of immune signatures and cellular and molecular components of the immune system as biomarkers is particularly important considering the increasing use of immune-based cancer therapies as therapeutic strategies for cancer patients.
Collapse
Affiliation(s)
- Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| | - Gianluca Basso
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Elena Monica Borroni
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Tommaso Cavalleri
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Paolo Bianchi
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Sanja Stifter
- Department of Pathology, School of Medicine, University of Rijeka, Rijeka, Croatia
| | | | - Alberto Malesci
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Gastroenterology, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Hereditary Cancer Genetics Clinic, Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| |
Collapse
|
104
|
Bae JY, Lee SW, Shin YH, Lee JH, Jahng JW, Park K. P2X7 receptor and NLRP3 inflammasome activation in head and neck cancer. Oncotarget 2018; 8:48972-48982. [PMID: 28430665 PMCID: PMC5564741 DOI: 10.18632/oncotarget.16903] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/29/2017] [Indexed: 12/24/2022] Open
Abstract
In this study, we investigated purinergic receptor P2X7 and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome expressions, and their role in head and neck cancer. We found upregulation of purinergic receptor P2X7 and all NLRP3 inflammasome components in biopsied head and neck squamous cell carcinoma tissues. Similarly, the expression of purinergic receptor P2X7, apoptosis-associated speck-like protein containing CARD, and pro-form caspase 1 in A253 cells derived from epidermoid carcinoma were highly upregulated in comparison to normal Human Salivary Gland cell line. Active caspase-1 and its final product, active interleukin-1β, both increased in primed A253 cells stimulated with purinergic receptor P2X7 agonists, while this elevated NLRP3 inflammasome activity was suppressed by purinergic receptor P2X7 antagonists. However, we observed none of these effects in Human Salivary Gland cells. Inhibition of both NLRP3 inflammasome and purinergic receptor P2X7 led to the significant cell death of primed A253 cells, but had no effect on the viability of primed HSG cells or the primary cultured human fibroblast cells. Furthermore, inhibition of either purinergic receptor P2X7 or NLRP3 inflammasome decreased invasiveness of A253, and this effect became more evident when both purinergic receptor P2X7 and NLRP3 inflammasome were simultaneously blocked. Therefore, it is concluded that the purinergic receptor P2X7 and the activation of NLRP3 inflammasome play important roles in the survival and invasiveness of head and neck squamous cell carcinoma in humans.
Collapse
Affiliation(s)
- Ju Young Bae
- Department of Physiology, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Sang-Woo Lee
- Department of Physiology, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Yong-Hwan Shin
- Department of Physiology, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | - Jong-Ho Lee
- Oral and Maxillofacial Surgery, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| | | | - Kyungpyo Park
- Department of Physiology, School of Dentistry, Seoul National University, Seoul 110-749, Korea
| |
Collapse
|
105
|
Frampton D, Schwenzer H, Marino G, Butcher LM, Pollara G, Kriston-Vizi J, Venturini C, Austin R, de Castro KF, Ketteler R, Chain B, Goldstein RA, Weiss RA, Beck S, Fassati A. Molecular Signatures of Regression of the Canine Transmissible Venereal Tumor. Cancer Cell 2018; 33:620-633.e6. [PMID: 29634949 PMCID: PMC5896242 DOI: 10.1016/j.ccell.2018.03.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 12/08/2017] [Accepted: 03/01/2018] [Indexed: 01/16/2023]
Abstract
The canine transmissible venereal tumor (CTVT) is a clonally transmissible cancer that regresses spontaneously or after treatment with vincristine, but we know little about the regression mechanisms. We performed global transcriptional, methylation, and functional pathway analyses on serial biopsies of vincristine-treated CTVTs and found that regression occurs in sequential steps; activation of the innate immune system and host epithelial tissue remodeling followed by immune infiltration of the tumor, arrest in the cell cycle, and repair of tissue damage. We identified CCL5 as a possible driver of CTVT regression. Changes in gene expression are associated with methylation changes at specific intragenic sites. Our results underscore the critical role of host innate immunity in triggering cancer regression.
Collapse
Affiliation(s)
- Dan Frampton
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Hagen Schwenzer
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Gabriele Marino
- Department of Veterinary Sciences, Polo Universitario dell'Annunziata, University of Messina, Messina 98168, Italy
| | - Lee M Butcher
- Department of Cancer Biology, Cancer Institute, UCL, 72 Huntley Street, London WC1E 6BT, UK
| | - Gabriele Pollara
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Janos Kriston-Vizi
- MRC Laboratory for Molecular Cell Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Cristina Venturini
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Rachel Austin
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Karina Ferreira de Castro
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - Benjamin Chain
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Richard A Goldstein
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Robin A Weiss
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK
| | - Stephan Beck
- Department of Cancer Biology, Cancer Institute, UCL, 72 Huntley Street, London WC1E 6BT, UK
| | - Ariberto Fassati
- Department of Infection, Division of Infection & Immunity, University College London (UCL), Cruciform Building, 90 Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
106
|
Narasimhan PB, Akabas L, Tariq S, Huda N, Bennuru S, Sabzevari H, Hofmeister R, Nutman TB, Tolouei Semnani R. Similarities and differences between helminth parasites and cancer cell lines in shaping human monocytes: Insights into parallel mechanisms of immune evasion. PLoS Negl Trop Dis 2018; 12:e0006404. [PMID: 29668679 PMCID: PMC5927465 DOI: 10.1371/journal.pntd.0006404] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/30/2018] [Accepted: 03/22/2018] [Indexed: 12/24/2022] Open
Abstract
A number of features at the host-parasite interface are reminiscent of those that are also observed at the host-tumor interface. Both cancer cells and parasites establish a tissue microenvironment that allows for immune evasion and may reflect functional alterations of various innate cells. Here, we investigated how the phenotype and function of human monocytes is altered by exposure to cancer cell lines and if these functional and phenotypic alterations parallel those induced by exposure to helminth parasites. Thus, human monocytes were exposed to three different cancer cell lines (breast, ovarian, or glioblastoma) or to live microfilariae (mf) of Brugia malayi-a causative agent of lymphatic filariasis. After 2 days of co-culture, monocytes exposed to cancer cell lines showed markedly upregulated expression of M1-associated (TNF-α, IL-1β), M2-associated (CCL13, CD206), Mreg-associated (IL-10, TGF-β), and angiogenesis associated (MMP9, VEGF) genes. Similar to cancer cell lines, but less dramatically, mf altered the mRNA expression of IL-1β, CCL13, TGM2 and MMP9. When surface expression of the inhibitory ligands PDL1 and PDL2 was assessed, monocytes exposed to both cancer cell lines and to live mf significantly upregulated PDL1 and PDL2 expression. In contrast to exposure to mf, exposure to cancer cell lines increased the phagocytic ability of monocytes and reduced their ability to induce T cell proliferation and to expand Granzyme A+ CD8+ T cells. Our data suggest that despite the fact that helminth parasites and cancer cell lines are extraordinarily disparate, they share the ability to alter the phenotype of human monocytes.
Collapse
Affiliation(s)
- Prakash Babu Narasimhan
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Leor Akabas
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Sameha Tariq
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Naureen Huda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Sasisekhar Bennuru
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Helen Sabzevari
- EMD Serono Research and Development Institute, Billerica, MA, United States of America
| | - Robert Hofmeister
- EMD Serono Research and Development Institute, Billerica, MA, United States of America
| | - Thomas B. Nutman
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| | - Roshanak Tolouei Semnani
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States of America
| |
Collapse
|
107
|
Soldati L, Di Renzo L, Jirillo E, Ascierto PA, Marincola FM, De Lorenzo A. The influence of diet on anti-cancer immune responsiveness. J Transl Med 2018; 16:75. [PMID: 29558948 PMCID: PMC5859494 DOI: 10.1186/s12967-018-1448-0] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/12/2018] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has matured into standard treatment for several cancers, but much remains to be done to extend the reach of its effectiveness particularly to cancers that are resistant within each indication. This review proposes that nutrition can affect and potentially enhance the immune response against cancer. The general mechanisms that link nutritional principles to immune function and may influence the effectiveness of anticancer immunotherapy are examined. This represents also the premise for a research project aimed at identifying the best diet for immunotherapy enhancement against tumours (D.I.E.T project). Particular attention is turned to the gut microbiota and the impact of its composition on the immune system. Also, the dietary patterns effecting immune function are discussed including the value of adhering to a healthy diets such as the Mediterranean, Veg, Japanese, or a Microbiota-regulating diet, the very low ketogenic diet, which have been demonstrated to lower the risk of developing several cancers and reduce the mortality associated with them. Finally, supplements, as omega-3 and polyphenols, are discussed as potential approaches that could benefit healthy dietary and lifestyle habits in the context of immunotherapy.
Collapse
Affiliation(s)
- Laura Soldati
- Department of Health Sciences, Università degli Studi di Milano, Via A di Rudinì 8, 20124, Milan, Italy.
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neuroscience and Sensory Organs, University of Bari, 70124, Bari, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Via Mariano Semmola snc, 80131, Naples, Italy
| | | | - Antonino De Lorenzo
- Section of Clinical Nutrition and Nutrigenomics, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| |
Collapse
|
108
|
Minami S, Ihara S, Kim SH, Yamamoto S, Komuta K. Lymphocyte to Monocyte Ratio and Modified Glasgow Prognostic Score Predict Prognosis of Lung Adenocarcinoma Without Driver Mutation. World J Oncol 2018; 9:13-20. [PMID: 29581811 PMCID: PMC5862078 DOI: 10.14740/wjon1084w] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 02/06/2018] [Indexed: 01/18/2023] Open
Abstract
Background Neutrophil to lymphocyte ratio (NLR), lymphocyte to monocyte ratio (LMR) and modified Glasgow prognostic score (mGPS) are useful prognostic markers based on host-related systemic inflammatory response. They have been shown as independent prognostic biomarkers in various cancers, including non-small cell lung cancer. However, there has been little evidence for a specific population of pulmonary adenocarcinoma without active epidermal growth factor receptor (EGFR) mutation. Methods We retrospectively reviewed 159 patients who met the following criteria: histologically or cytologically diagnosed adenocarcinoma, confirmed wild-type EGFR, started first-line cytotoxic chemotherapy between July 2007 and March 2017 at our hospital, and c-stage IIIB or IV. We compared overall survival (OS) between dichotomized groups by the optimal cut-off points of NLR and LMR, and mGPS 0 - 1 vs. 2. Univariate and multivariate Cox proportional hazard analyses also detected prognostic factors for OS. Results As favorable prognostic factors for OS, multivariate analysis detected Eastern Cooperative Oncology Group performance status (ECOG PS) 0 - 1 (hazard ratio (HR) 3.43, 95% confidence interval (CI): 2.12 - 5.53; P < 0.01), LMR ≥ 1.97 (HR 0.39, 95% CI: 0.21 - 0.72; P < 0.01) and mGPS 0 - 1 (HR 1.95, 95% CI: 1.20 - 3.16; P < 0.01). The OS of LMR ≥ 1.97 and mGPS 0 - 1 groups were significantly longer than those of LMR < 1.97 and mGPS 2 groups, respectively. We divided 159 patients into three groups, both LMR ≥ 1.97 and mGPS 0 - 1, either LMR ≥ 1.97 or mGPS 0 - 1 and both LMR < 1.97 and mGPS 2. The OS of both LMR < 1.97 and mGPS 2 was significantly shorter than the other two groups. After adjustment for age, sex, ECOG PS, sodium, alkaline phosphatase and NLR, multivariate analysis found both LMR < 1.97 and mGPS 2 as an independent poor prognostic combination in comparison with both LMR ≥ 1.97 and mGPS0-1 (HR 5.98, 95% CI: 2.64 - 13.5; P < 0.01). Conclusions LMR and mGPS are independent prognostic markers for pulmonary adenocarcinoma with wild-type EGFR. Combination of LMR and mGPS can stratify patients according to prognosis.
Collapse
Affiliation(s)
- Seigo Minami
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Shouichi Ihara
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Sung-Ho Kim
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Suguru Yamamoto
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Kiyoshi Komuta
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| |
Collapse
|
109
|
Garcia-Gomez A, Rodríguez-Ubreva J, Ballestar E. Epigenetic interplay between immune, stromal and cancer cells in the tumor microenvironment. Clin Immunol 2018; 196:64-71. [PMID: 29501540 DOI: 10.1016/j.clim.2018.02.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 02/26/2018] [Indexed: 12/14/2022]
Abstract
Compelling evidences highlight the critical role of the tumor microenvironment as mediator of tumor progression and immunosuppression in several types of cancer. The reciprocal interplay between neoplastic and non-tumoral host cells is mediated by direct cell-to-cell contact, soluble factors and exosomes that result in differential gene expression patterns that are driven by epigenetic mechanisms. In this regard, extensive literature has described the abnormalities in the DNA methylation status and histone modification profiles in tumor cells. However, little is known about the mechanisms of epigenetic dysregulation that participate as a consequence of the intricate crosstalk among the cells within the tumor niche. This review summarizes the current knowledge on epigenetic changes that result from the interactions between myeloid, stromal and cancer cells in the tumor microenvironment and its functional impact in both tumorigenesis and tumor progression. We also discuss potential niche-specific epigenetic biomarkers to improve the prognosis and clinical treatment of cancer patients.
Collapse
Affiliation(s)
- Antonio Garcia-Gomez
- Chromatin and Disease Group, Cancer Epigenetics and Biology Programme (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Javier Rodríguez-Ubreva
- Chromatin and Disease Group, Cancer Epigenetics and Biology Programme (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Esteban Ballestar
- Chromatin and Disease Group, Cancer Epigenetics and Biology Programme (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
110
|
Wang Z, Wang C, Abudukeremu A, Rui X, Liu S, Zhang X, Zhang M, Zhang J, Dong L. Engineering a Tumor Microenvironment-Mimetic Niche for Tissue Regeneration with Xenogeneic Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1700666. [PMID: 29593968 PMCID: PMC5867037 DOI: 10.1002/advs.201700666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 11/22/2017] [Indexed: 05/05/2023]
Abstract
The insufficient number of cells suitable for transplantation is a long-standing problem to cell-based therapies aimed at tissue regeneration. Xenogeneic cancer cells (XCC) may be an alternative source of therapeutic cells, but their transplantation risks both immune rejection and unwanted spreading. In this study, a strategy to facilitate XCC transplantation is reported and their spreading in vivo is confined by constructing an engineering matrix that mimics the characteristics of tumor microenvironment. The data show that this matrix, a tumor homogenate-containing hydrogel (THAG), successfully creates an immunosuppressive enclave after transplantation into immunocompetent mice. XCC of different species and tissue origins seeded into THAG survive well, integrated with the host and developed the intrinsic morphology of the native tissue, without being eliminated or spreading out of the enclave. Most strikingly, immortalized human hepatocyte cells and rat β-cells loaded into THAG exert the physiological functions of the human liver and rat pancreas islets, respectively, in the mouse body. This study demonstrates a novel and feasible approach to harness the unique features of tumor development for tissue transplantation and regenerative medicine.
Collapse
Affiliation(s)
- Zhenzhen Wang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical SciencesUniversity of MacauTaipa999078Macau SAR
| | - Ayipaxia Abudukeremu
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| | - Xiaying Rui
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| | - Shang Liu
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| | - Xiaoyi Zhang
- Department of ChemistryEmory University1515 Dickey DriveAtlantaGA30322USA
| | - Min Zhang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210093China
| |
Collapse
|
111
|
Zini C, Venneri MA, Miglietta S, Caruso D, Porta N, Isidori AM, Fiore D, Gianfrilli D, Petrozza V, Laghi A. USPIO‐labeling in M1 and M2‐polarized macrophages: An in vitro study using a clinical magnetic resonance scanner. J Cell Physiol 2018; 233:5823-5828. [DOI: 10.1002/jcp.26360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/29/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Chiara Zini
- Faculty of Medicine and DentistryDepartment of Radiological SciencesOncology and PathologyUniversity of Rome “Sapienza”LatinaItaly
| | - Mary A. Venneri
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Selenia Miglietta
- Department of Anatomy, Histology, Forensic Medicine and OrthopaedicsElectron Microscopy Unit, Laboratory “Pietro M. Motta”University of Rome “Sapienza”RomeItaly
| | - Damiano Caruso
- Faculty of Medicine and DentistryDepartment of Radiological SciencesOncology and PathologyUniversity of Rome “Sapienza”LatinaItaly
| | - Natale Porta
- Faculty of Pharmacy and MedicineDepartment of Medico‐Surgical Sciences and BiotechnologiesUniversity of Rome “Sapienza”LatinaItaly
| | - Andrea M. Isidori
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Daniela Fiore
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Daniele Gianfrilli
- Faculty of Medicine and DentistryDepartment of Experimental MedicineUniversity of Rome “Sapienza”RomeItaly
| | - Vincenzo Petrozza
- Faculty of Pharmacy and MedicineDepartment of Medico‐Surgical Sciences and BiotechnologiesUniversity of Rome “Sapienza”LatinaItaly
| | - Andrea Laghi
- Faculty of Medicine and DentistryDepartment of Radiological SciencesOncology and PathologyUniversity of Rome “Sapienza”LatinaItaly
| |
Collapse
|
112
|
Hong AM, Vilain RE, Romanes S, Yang J, Smith E, Jones D, Scolyer RA, Lee CS, Zhang M, Rose B. PD-L1 expression in tonsillar cancer is associated with human papillomavirus positivity and improved survival: implications for anti-PD1 clinical trials. Oncotarget 2018; 7:77010-77020. [PMID: 27776338 PMCID: PMC5363566 DOI: 10.18632/oncotarget.12776] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 10/13/2016] [Indexed: 12/16/2022] Open
Abstract
In this study, we examined PD-L1 expression by immunohistochemistry in 99 patients with tonsillar cancer and known human papillomavirus (HPV) status to assess its clinical significance. We showed that the pattern of PD-L1 expression is strongly related to HPV status. The PD-L1 positivity rate was 83.3% in HPV-positive cases and 56.9% in HPV-negative cases (p < 0.05). Patients with HPV-positive/PD-L1-positive cancer had significantly better event free survival and overall survival compared with patients with HPV-negative/PD-L1-negative cancer. Relative to those patients with HPV-negative/PD-L1-negative disease who had the highest risk of death, patients with HPV-positive/PD-L1-positive cancers had a 2.85 fold lower risk of developing an event (HR 0.35, 95% CI: 0.16–0.79) and a 4.5 fold lower risk of death (HR =0.22, 95% CI: 0.09–0.53). Our findings will help to guide future clinical trial design in immunotherapy based on PD-L1 expression in tonsillar cancer.
Collapse
Affiliation(s)
- Angela M Hong
- Sydney Medical School, The University of Sydney, NSW, Australia
| | - Ricardo E Vilain
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia.,Pathology North (Hunter), John Hunter Hospital, Newcastle, NSW, Australia
| | - Sarah Romanes
- School of Mathematics and Statistics, The University of Sydney, NSW, Australia
| | - Jean Yang
- School of Mathematics and Statistics, The University of Sydney, NSW, Australia
| | - Elizabeth Smith
- Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia
| | - Deanna Jones
- Sydney Medical School, The University of Sydney, NSW, Australia
| | - Richard A Scolyer
- Sydney Medical School, The University of Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia
| | - C Soon Lee
- Sydney Medical School, The University of Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, NSW, Australia.,Discipline of Pathology, School of Medicine, Western Sydney University, NSW, Australia
| | - Mei Zhang
- Sydney Medical School, The University of Sydney, NSW, Australia
| | - Barbara Rose
- Sydney Medical School, The University of Sydney, NSW, Australia
| |
Collapse
|
113
|
Yan R, Chen X, Wang W, Liang L, Zhou C, Wei W, Yi H, Wu X, Liu G, Zhong M, Yu Y. [Association between HLA-A and HLA-DRB1 allele polymorphisms and susceptibility to tuberculosis in southern Chinese population]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:101-105. [PMID: 33177033 DOI: 10.3969/j.issn.1673-4254.2018.01.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the correlation between tumor-associated macrophages (TAMs) and the development of high risk human papilloma virus (hr-HPV)-related cervical cancer. METHODS A total of 112 cases of cervical tissue were collected, including 16 normal cervical tissues, 55 cervical intraepithelial neoplasia (CIN) tissues and 41 squamous cervical cancer (SCC) tissues. The expression of CD163+ macrophages in the cervical tissues was detected by immunohistochemical method, and the results were analyzed in relation with the clinical data of the patients. RESULTS Immunohistochemical analysis showed that the cell density of CD163+ macrophages increased progressively with the increase in the tissue malignancy, in the order of normal cervical tissue, CIN Ⅰ, CIN Ⅱ-Ⅲ, and SCC. Correlation analysis revealed a positive relationship between CD163+ macrophage density and tissue malignancy (P=0.000). The density of CD163+ macrophages was significantly upregulated in HR-HPV-positive SCC tissue (P < 0.05). CD163+ macrophages were positively correlated with cervical lymph node metastasis (P=0.005) and FIGO stage (P=0.004) of SCC. CONCLUSIONS The expression of CD163+ macrophages is positively correlated with malignant transformation of cervical tissues, and hr-HPV infection is significantly correlated with CD163 expression level in the macrophages. CD163+ macrophages can be used as predictors of the occurrence and progression of cervical cancer caused by hr-HPV infection.
Collapse
Affiliation(s)
- Ruiming Yan
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Xiaojing Chen
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Wei Wang
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Li Liang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China
| | - Chenfei Zhou
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Wenfei Wei
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Hongyan Yi
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Xiangguang Wu
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Guobing Liu
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Mei Zhong
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| | - Yanhong Yu
- Department of Internal Medicine, Guangzhou Chest Hospital, Guangzhou 510095, China
| |
Collapse
|
114
|
Chen J, Pan Y, He B, Ying H, Sun H, Deng Q, Liu X, Wang S. Meta-analysis of prognostic value of inflammation parameter in breast cancer. J Cancer Res Ther 2018; 14:S85-S89. [PMID: 29578155 DOI: 10.4103/0973-1482.160917] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
CONTEXT Recently, increasing studies investigated the association between inflammation parameter such as neutrophil to lymphocyte ratio (NLR) and the prognosis of cancers. However, the clinical and prognostic significance of NLR in breast cancer remains controversial. AIM This meta-analysis was conducted to establish the overall accuracy of the NLR test in the diagnosis of breast cancer. MATERIALS AND METHODS A comprehensive search of the literature was conducted using PubMed and Web of Science. Six studies dating up to July 2014 with 2267 patients were enrolled in the present study. STATA 11.0 software (STATA Corporation, College Station, TX, USA) was selected for data analysis. In order to evaluate the association between NLR and overall survival (OS), disease-free survival (DFS), recurrence-free survival or cancer-specific survival, the hazard ratios (HRs), and their 95% confidence intervals (CIs) were extracted. RESULTS Subgroup analyses showed that NLR was a strong prognostic factor for OS in multivariate analysis (HR = 2.81, 95% CI = 2.13-3.71, PH = 0.992) and without metastasis (HR = 1.45, 95% CI = 0.37-5.66, PH < 0.001). Elevated NLR was associated with a high risk for DFS in subgroups of multivariate analysis (HR = 2.16, 95% CI = 1.67-2.80, PH = 0.977) and mixed metastasis (HR = 2.13, 95% CI = 1.38-3.30, PH = 0.84). CONCLUSION In summary, NLR may be considered as a predictive factor for patients with breast cancer.
Collapse
Affiliation(s)
- Jie Chen
- Department of Life Sciences, Nanjing Normal University; Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuqin Pan
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bangshun He
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Houqun Ying
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University; Medical College, Southeast University, Nanjing, Jiangsu, China
| | - Huiling Sun
- Department of Life Sciences, Nanjing Normal University; Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiwen Deng
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xian Liu
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shukui Wang
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
115
|
Minami S, Ogata Y, Ihara S, Yamamoto S, Komuta K. Neutrophil-to-Lymphocyte Ratio Predicts Overall Survival of Advanced Non-Small Cell Lung Cancer Harboring Mutant Epidermal Growth Factor Receptor. World J Oncol 2017; 8:180-187. [PMID: 29317963 PMCID: PMC5755625 DOI: 10.14740/wjon1069w] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Background Neutrophil-to-lymphocyte ratio (NLR) and lymphocyte-to-monocyte ratio (LMR) have been demonstrated to be prognostic biomarkers in various cancers, including non-small cell lung cancer (NSCLC). However, little has been known about these two ratios for a specific population of NSCLC harboring active epidermal growth factor receptor (EGFR) mutation. Methods We retrospectively reviewed electrical medical records of 152 patients who met the following criteria: NSCLC harboring mutant EGFR, EGFR-tyrosine kinase inhibitor (EGFR-TKI) monotherapy initiated between October 2007 and February 2017 at our hospital, stage III-IV or post-surgical recurrence. We compared overall survival (OS) and progression-free survival (PFS) between dichotomized groups by the optimal cut-off points of the two biomarkers. Univariate and multivariate Cox hazard analyses also searched for prognostic factors of survival time. Results OSs of NLR < 2.11 (median 38.6 vs. 24.1 months, P < 0.01) and LMR ≥ 5.09 (median 39.4 vs. 26.4 months, P < 0.01) were significantly longer than those of NLR ≥ 2.11 and LMR < 5.09. Multivariate analyses found lower NLR (hazard ratio (HR) 1.07, 95% CI: 1.01 - 1.14; P = 0.03) as an independent prognostic factor for longer OS, in addition to Eastern Cooperative Oncology Group performance status 0 - 1, first-line EGFR-TKI, higher serum sodium concentration and lower lactate dehydrogenase. However, LMR was not detected as a significant prognostic factor for OS. None of these two biomarkers was selected as an independent prognostic factor for PFS. Conclusions This study demonstrated that elevated NLR is an independent prognostic factor for poor survival of patients with EGFR mutant NSCLC. NLR is a useful and simple biomarker for these patients.
Collapse
Affiliation(s)
- Seigo Minami
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Yoshitaka Ogata
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Shouichi Ihara
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Suguru Yamamoto
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| | - Kiyoshi Komuta
- Department of Respiratory Medicine, Osaka Police Hospital, 10-31 Kitayama-cho, Tennoji-ku, Osaka 543-0035, Japan
| |
Collapse
|
116
|
Bedognetti D, Roelands J, Decock J, Wang E, Hendrickx W. The MAPK hypothesis: immune-regulatory effects of MAPK-pathway genetic dysregulations and implications for breast cancer immunotherapy. Emerg Top Life Sci 2017; 1:429-445. [PMID: 33525803 PMCID: PMC7289005 DOI: 10.1042/etls20170142] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/08/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022]
Abstract
With the advent of checkpoint inhibition, immunotherapy has revolutionized the clinical management of several cancers, but has demonstrated limited efficacy in mammary carcinoma. Transcriptomic profiling of cancer samples defined distinct immunophenotypic categories characterized by different prognostic and predictive connotations. In breast cancer, genomic alterations leading to the dysregulation of mitogen-activated protein kinase (MAPK) pathways have been linked to an immune-silent phenotype associated with poor outcome and treatment resistance. These aberrations include mutations of MAP3K1 and MAP2K4, amplification of KRAS, BRAF, and RAF1, and truncations of NF1. Anticancer therapies targeting MAPK signaling by BRAF and MEK inhibitors have demonstrated clear immunologic effects. These off-target properties could be exploited to convert the immune-silent tumor phenotype into an immune-active one. Preclinical evidence supports that MAPK-pathway inhibition can dramatically increase the efficacy of immunotherapy. In this review, we provide a detailed overview of the immunomodulatory impact of MAPK-pathway blockade through BRAF and MEK inhibitions. While BRAF inhibition might be relevant in melanoma only, MEK inhibition is potentially applicable to a wide range of tumors. Context-dependent similarities and differences of MAPK modulation will be dissected, in light of the complexity of the MAPK pathways. Therapeutic strategies combining the favorable effects of MAPK-oriented interventions on the tumor microenvironment while maintaining T-cell function will be presented. Finally, we will discuss recent studies highlighting the rationale for the implementation of MAPK-interference approaches in combination with checkpoint inhibitors and immune agonists in breast cancer.
Collapse
Affiliation(s)
- Davide Bedognetti
- Tumor Biology, Immunology, and Therapy Section, Department of Immunology, Inflammation and Metabolism, Division of Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
- College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Jessica Roelands
- Tumor Biology, Immunology, and Therapy Section, Department of Immunology, Inflammation and Metabolism, Division of Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Julie Decock
- Cancer Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Ena Wang
- Division of Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Wouter Hendrickx
- Tumor Biology, Immunology, and Therapy Section, Department of Immunology, Inflammation and Metabolism, Division of Translational Medicine, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| |
Collapse
|
117
|
The role of the systemic inflammatory response in predicting outcomes in patients with operable cancer: Systematic review and meta-analysis. Sci Rep 2017; 7:16717. [PMID: 29196718 PMCID: PMC5711862 DOI: 10.1038/s41598-017-16955-5] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023] Open
Abstract
Cancer remains a leading causes of death worldwide and an elevated systemic inflammatory response (SIR) is associated with reduced survival in patients with operable cancer. This review aims to examine the evidence for the role of systemic inflammation based prognostic scores in patients with operable cancers. A wide-ranging literature review using targeted medical subject headings for human studies in English was carried out in the MEDLINE, EMBASE, and CDSR databases until the end of 2016. The SIR has independent prognostic value, across tumour types and geographical locations. In particular neutrophil lymphocyte ratio (NLR) (n = 158), platelet lymphocyte ratio (PLR) (n = 68), lymphocyte monocyte ratio (LMR) (n = 21) and Glasgow Prognostic Score/ modified Glasgow Prognostic Score (GPS/mGPS) (n = 60) were consistently validated. On meta-analysis there was a significant relationship between elevated NLR and overall survival (OS) (p < 0.00001)/ cancer specific survival (CSS) (p < 0.00001), between elevated LMR and OS (p < 0.00001)/CSS (p < 0.00001), and elevated PLR and OS (p < 0.00001)/CSS (p = 0.005). There was also a significant relationship between elevated GPS/mGPS and OS (p < 0.00001)/CSS (p < 0.00001). These results consolidate the prognostic value of the NLR, PLR, LMR and GPS/mGPS in patients with resectable cancers. This is particularly true for the NLR/GPS/mGPS which should form part of the routine preoperative and postoperative workup.
Collapse
|
118
|
Varol C, Sagi I. Phagocyte-extracellular matrix crosstalk empowers tumor development and dissemination. FEBS J 2017; 285:734-751. [DOI: 10.1111/febs.14317] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/01/2017] [Accepted: 10/31/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Chen Varol
- The Research Center for Digestive Tract and Liver Diseases; Tel-Aviv Sourasky Medical Center; Sackler Faculty of Medicine; Tel-Aviv University; Israel
- Department of Clinical Microbiology and Immunology; Sackler Faculty of Medicine; Tel Aviv University; Israel
| | - Irit Sagi
- Department of Biological Regulation; Weizmann Institute of Science; Rehovot Israel
| |
Collapse
|
119
|
Zhao QT, Zhang XP, Zhang H, Duan GC. Prognostic role of platelet to lymphocyte ratio in esophageal cancer: A meta-analysis. Oncotarget 2017; 8:112085-112093. [PMID: 29340113 PMCID: PMC5762381 DOI: 10.18632/oncotarget.22557] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 10/28/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose The prognostic role of inflammation index like platelet to lymphocyte ratio (PLR) in esophageal cancer remains controversial. We evaluated the prognostic significance of PLR in esophageal cancer patients. Methods We searched databases to identify relevant literatures. Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated. A meta-analysis was performed to evaluate the prognostic value of PLR in patients with esophageal cancer. Results A total of 6,699 patients from 16 studies (17 cohorts) were finally enrolled in the meta-analysis. The results demonstrate that the elevated PLR predicted poorer overall survival (OS) (HR: 1.389, 95% CI: 1.161-1.663) and disease-free survival (DFS) (HR: 1.404, 95% CI: 1.169-1.687) and cancer-specific survival (CSS) (HR: 1.686, 95% CI: 1.146-2.480) in patients with esophageal cancer. Subgroup analysis revealed that the elevated PLR was also associated with poor OS in esophageal cancer treated by surgery (HR: 1.492, 95%CI: 1.149-1.938, P<0.05) and mixed treatment (HR: 1.222, 95%CI: 1.009-1.479, P<0.05). In addition, PLR Cut-off value≤160 (HR: 1.484, 95%CI: 1.088-2.024, P<0.05) and PLR Cut-off value>160 (HR: 1.391, 95%CI: 1.161-1.666, P<0.05). Conclusion This meta-analysis result suggested that PLR might be a significant predicative biomarker of poor prognosis for esophageal cancer patients.
Collapse
Affiliation(s)
- Qing-Tao Zhao
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang 050051, Hebei, PR China
| | - Xiao-Peng Zhang
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang 050051, Hebei, PR China
| | - Hua Zhang
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang 050051, Hebei, PR China
| | - Guo-Chen Duan
- Department of Thoracic Surgery, Hebei General Hospital, Shijiazhuang 050051, Hebei, PR China
| |
Collapse
|
120
|
Zhang D, Li Z, Xu X, Zhou D, Tang S, Yin X, Xu F, Li H, Zhou Y, Zhu T, Deng H, Zhang S, Huang Q, Wang J, Yin W, Zhu Y, Lai M. Deletions at SLC18A1 increased the risk of CRC and lower SLC18A1 expression associated with poor CRC outcome. Carcinogenesis 2017; 38:1057-1062. [PMID: 28968818 DOI: 10.1093/carcin/bgx088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/24/2017] [Indexed: 12/26/2022] Open
Abstract
Copy number variations (CNVs) contribute to the development of colorectal cancer (CRC). We conducted a two-stage association study to identify CNV risk loci for CRC. We performed a gene-based rare CNV study on 694 sporadic CRC and 1641 controls using Illumina Human-OmniExpress-12v1.0 BeadChips, and further replicated in 934 CRC cases and 2680 controls for risk CNVs by using TaqMan Copy Number Assay. Tumor buddings, cancer cells in the center of primary tumor and normal intestinal epithelial cells were captured using laser capture microdissection (LCM) and were assayed using AffymetrixGeneChip® Human Genome U133 Plus 2.0 Array. In addition, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus data were assessed for the effects of risk CNVs. We found that germline deletions affecting the last six exons of SLC18A1 significantly associated with CRC with a combined P value of 6.4 × 10-5 by a two-stage analysis. Both in TCGA CRC RNA seq dataset and GDS4382, SLC18A1 was significantly down regulated in CRC tissues than in paired normal tissues (N = 32 and 17 pairs, P = 0.004 and 0.009, respectively). In LCM samples, similar observations were obtained that the expression levels of SLC18A1 in the tumor buddings, cancer cells in the center of primary tumor, and stroma of both tumor budding and cancer cells were lower than normal intestinal epithelial and stromal cells (fold change = 0.17-0.62, 0.12-0.57 and 0.37-0.68, respectively). In summary, the germline deletions at SLC18A1 contributed to the development of CRC. The role of SLC18A1 required further exploration.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Zhenli Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Xiaohong Xu
- Clinical laboratory, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou 310022, China
| | - Dan Zhou
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Shunli Tang
- The Second Department of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoyang Yin
- The Second Department of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fangying Xu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Hui Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Yuan Zhou
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Tao Zhu
- Departments of Pathology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Hong Deng
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Shuai Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Qiong Huang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Jing Wang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Wei Yin
- The Core Facilities, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yimin Zhu
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou 310058, China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
121
|
Schernberg A, Blanchard P, Chargari C, Deutsch E. Neutrophils, a candidate biomarker and target for radiation therapy? Acta Oncol 2017; 56:1522-1530. [PMID: 28835188 DOI: 10.1080/0284186x.2017.1348623] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Neutrophils are the most abundant blood-circulating white blood cells, continuously generated in the bone marrow. Growing evidence suggests they regulate the innate and adaptive immune system during tumor evolution. This review will first summarize the recent findings on neutrophils as a key player in cancer evolution, then as a potential biomarker, and finally as therapeutic targets, with respective focuses on the interplay with radiation therapy. A complex interplay: Neutrophils have been associated with tumor progression through multiple pathways. Ionizing radiation has cytotoxic effects on cancer cells, but the sensitivity to radiation therapy in vivo differ from isolated cancer cells in vitro, partially due to the tumor microenvironment. Different microenvironmental states, whether baseline or induced, can modulate or even attenuate the effects of radiation, with consequences for therapeutic efficacy. Inflammatory biomarkers: Inflammation-based scores have been widely studied as prognostic biomarkers in cancer patients. We have performed a large retrospective cohort of patients undergoing radiation therapy (1233 patients), with robust relationship between baseline blood neutrophil count and 3-year's patient's overall survival in patients with different cancer histologies. (Pearson's correlation test: p = .001, r = -.93). Therapeutic approaches: Neutrophil-targeting agents are being developed for the treatment of inflammatory and autoimmune diseases. Neutrophils either can exert antitumoral (N1 phenotype) or protumoral (N2 phenotype) activity, depending on the Tumor Micro Environment. Tumor associated N2 neutrophils are characterized by high expression of CXCR4, VEGF, and gelatinase B/MMP9. TGF-β within the tumor microenvironment induces a population of TAN with a protumor N2 phenotype. TGF-β blockade slows tumor growth through activation of CD8 + T cells, macrophages, and tumor associated neutrophils with an antitumor N1 phenotype. CONCLUSIONS This supports the need for prospective neutrophils evaluation in clinical trials, making neutrophils a predictive biomarker with potential specific therapies.
Collapse
Affiliation(s)
- Antoine Schernberg
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Pierre Blanchard
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM U1018, CESP, Université Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Cyrus Chargari
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM 1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine du Kremlin-Bicetre, Université Paris Sud, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Eric Deutsch
- Radiation Oncology Department, SIRIC SOCRATES, Gustave Roussy Cancer Campus, Villejuif, France
- INSERM 1030, Molecular Radiotherapy, Gustave Roussy Cancer Campus, Villejuif, France
- Faculté de Médecine du Kremlin-Bicetre, Université Paris Sud, Université Paris Saclay, Le Kremlin-Bicetre, France
| |
Collapse
|
122
|
Wandering pathways in the regulation of innate immunity and inflammation. J Autoimmun 2017; 85:1-5. [PMID: 29079064 DOI: 10.1016/j.jaut.2017.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 10/11/2017] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) have served as a paradigm of cancer-related inflammation. Moreover, investigations on TAM have led to the dissection of macrophage plasticity and polarization and to the discovery and analysis of molecular pathways of innate immunity, in particular cytokines, chemokines and PTX3 as a prototypic fluid phase pattern recognition molecule. Mechanisms of negative regulation are complex and include decoy receptors, receptor antagonists, anti-inflammatory cytokines and the signalling regulator IL-1R8. In this review, topics and open issues in relation to regulation of innate immunity and inflammation are discussed: 1) how macrophage and neutrophil plasticity and polarization underlie diverse pathological conditions ranging from autoimmunity to cancer and may pave the way to innovative diagnostic and therapeutic approaches; 2) the key role of decoy receptors and negative regulators (e.g. IL-1R2, ACKR2, IL-1R8) in striking a balance between amplification of immunity and resolution versus uncontrolled inflammation and tissue damage; 3) role of humoral innate immunity, illustrated by PTX3, in resistance against selected microbes, regulation of inflammation and immunity and tissue repair, with implications for diagnostic and therapeutic translation.
Collapse
|
123
|
Single-cell mass cytometry and transcriptome profiling reveal the impact of graphene on human immune cells. Nat Commun 2017; 8:1109. [PMID: 29061960 PMCID: PMC5653675 DOI: 10.1038/s41467-017-01015-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 07/25/2017] [Indexed: 12/12/2022] Open
Abstract
Understanding the biomolecular interactions between graphene and human immune cells is a prerequisite for its utilization as a diagnostic or therapeutic tool. To characterize the complex interactions between graphene and immune cells, we propose an integrative analytical pipeline encompassing the evaluation of molecular and cellular parameters. Herein, we use single-cell mass cytometry to dissect the effects of graphene oxide (GO) and GO functionalized with amino groups (GONH2) on 15 immune cell populations, interrogating 30 markers at the single-cell level. Next, the integration of single-cell mass cytometry with genome-wide transcriptome analysis shows that the amine groups reduce the perturbations caused by GO on cell metabolism and increase biocompatibility. Moreover, GONH2 polarizes T-cell and monocyte activation toward a T helper-1/M1 immune response. This study describes an innovative approach for the analysis of the effects of nanomaterials on distinct immune cells, laying the foundation for the incorporation of single-cell mass cytometry on the experimental pipeline. Understanding the interaction of nanomaterials and immune cells at the biomolecular level is of great significance in therapeutic applications. Here, the authors investigated the interaction of graphene oxide nanomaterials and several immune cell subpopulations using single-cell mass cytometry and genome-wide transcriptome analysis.
Collapse
|
124
|
Chen XJ, Han LF, Wu XG, Wei WF, Wu LF, Yi HY, Yan RM, Bai XY, Zhong M, Yu YH, Liang L, Wang W. Clinical Significance of CD163+ and CD68+ Tumor-associated Macrophages in High-risk HPV-related Cervical Cancer. J Cancer 2017; 8:3868-3875. [PMID: 29151975 PMCID: PMC5688941 DOI: 10.7150/jca.21444] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022] Open
Abstract
Objective. To explore the influence of M2-polarized tumor-associated macrophages (TAMs) on high-risk human papillomavirus (hr-HPV)-related cervical carcinogenesis and metastasis. Methods. CD68+ and CD163+ macrophages were examined immunohistochemically in a series of 130 samples, including 26 cases of normal cervical tissues, 59 cases of cervical intraepithelial neoplasia (CIN), and 45 cases of squamous cell carcinoma (SCC), and the results were statistically analyzed. The macrophage count was corrected for the epithelial and stromal compartments respectively. Clinical data were also obtained. Results. High counts of CD68+ and CD163+ macrophages were associated with hr-HPV infection (both p < 0.05) and positively correlated with cervical carcinogenesis (Spearman's rho = 0.478, p = 0.000; Spearman's rho = 0.676, p =0.000, respectively). The immunostaining pattern of CD163 exhibited clearer background than that of CD68. CD163+ macrophages showed a more obviously increasing migration into the epithelium along with the progression of CIN to invasive cancer. Notably, a high index of CD163+ macrophages was significantly associated with higher FIGO stages (p = 0.009) and lymph node metastasis (p = 0.012), but a similar finding was not found for CD68+ macrophages (p = 0.067, p = 0.079, respectively). Conclusions. Our study supported a critical role of TAMs as a prospective predictor for hr-HPV-related cervical carcinogenesis. CD163, as a promising TAMs marker, is superior to CD68 for predicting the malignant transformation and metastatic potential of cervical cancer.
Collapse
Affiliation(s)
- Xiao-Jing Chen
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Ling-Fei Han
- Department of Minimally Invasive Gynecologic Surgery, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiang-Guang Wu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Wen-Fei Wei
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Lan-Fang Wu
- Department of Obstetrics and Gynecology, the Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Hong-Yan Yi
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Rui-Ming Yan
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xiang-Yang Bai
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yan-Hong Yu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Wei Wang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China.,Department of Obstetrics and Gynecology, the First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
125
|
Pu WL, Sun LK, Gao XM, Rüegg C, Cuendet M, Hottiger MO, Zhou K, Miao L, Zhang YS, Gebauer M. Targeting tumor-associated macrophages by anti-tumor Chinese materia medica. Chin J Integr Med 2017; 23:723-732. [PMID: 28988387 DOI: 10.1007/s11655-017-2974-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Indexed: 12/23/2022]
Abstract
Tumor-associated macrophages (TAMs) play a key role in all stages of tumorigenesis and tumor progression. TAMs secrete different kinds of cytokines, chemokines, and enzymes to affect the progression, metastasis, and resistance to therapy depending on their state of reprogramming. Therapeutic benefit in targeting TAMs suggests that macrophages are attractive targets for cancer treatment. Chinese materia medica (CMM) is an important approach for treating cancer in China and in the Asian region. According to the theory of Chinese medicine (CM) and its practice, some prescriptions of CM regulate the body's internal environment possibly including the remodeling the tumor microenvironment (TME). Here we briefly summarize the pivotal effects of TAMs in shaping the TME and promoting tumorigenesis, invasion, metastasis and immunosuppression. Furthermore, we illustrate the effects and mechanisms of CMM targeting TAMs in antitumor therapy. Finally, we reveal the CMM's dual-regulatory and multi-targeting functions on regulating TAMs, and hopefully, provide the theoretical basis for CMM clinical practice related to cancer therapy.
Collapse
Affiliation(s)
- Wei-Ling Pu
- Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Li-Kang Sun
- Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Xiu-Mei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Curzio Rüegg
- Pathology Unit, Department of Medicine, Faculty of Sciences, University of Fribourg, Fribourg, Switzerland
| | - Muriel Cuendet
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Genève, Switzerland
| | - Micheal O Hottiger
- Department of Molecular Mechanisms of Disease, University of Zurich-Irchel, Zurich, Switzerland
| | - Kun Zhou
- Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lin Miao
- Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- Department of Molecular Mechanisms of Disease, University of Zurich-Irchel, Zurich, Switzerland
| | - Yun-Sha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- Pathology Unit, Department of Medicine, Faculty of Sciences, University of Fribourg, Fribourg, Switzerland
| | - Margaret Gebauer
- Chingcura, Center for Traditional Chinese Medicine, Zurich, Switzerland
| |
Collapse
|
126
|
Pistollato F, Calderón Iglesias R, Ruiz R, Aparicio S, Crespo J, Dzul Lopez L, Giampieri F, Battino M. The use of natural compounds for the targeting and chemoprevention of ovarian cancer. Cancer Lett 2017; 411:191-200. [PMID: 29017913 DOI: 10.1016/j.canlet.2017.09.050] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/25/2017] [Accepted: 09/25/2017] [Indexed: 12/21/2022]
Abstract
Among gynaecological cancers, ovarian cancer represents the leading cause of death in women. Current treatment for ovarian cancer entails surgery followed by combined chemotherapy with platinum and taxane, which are associated, particularly cisplatin, with severe side effects. While this treatment approach appears to be initially effective in a high number of patients, nearly 70% of them suffer a relapse within a few months after initial treatment. Therefore, more effective and better-tolerated treatment options are clearly needed. In recent years, several natural compounds (such as curcumin, epigallocatechin 3-gallate (EGCG), resveratrol, sulforaphane and Withaferin-A), characterized by long-term safety and negligible and/or inexistent side effects, have been proposed as possible adjuvants of traditional chemotherapy. Indeed, several in vitro and in vivo studies have shown that phytocompounds can effectively inhibit tumor cell proliferation, stimulate autophagy, induce apoptosis, and specifically target ovarian cancer stem cells (CSCs), which are generally considered to be responsible for tumor recurrence in several types of cancer. Here we review current literature on the role of natural products in ovarian cancer chemoprevention, highlighting their effects particularly on the regulation of inflammation, autophagy, proliferation and apoptosis, chemotherapy resistance, and ovarian CSC growth.
Collapse
Affiliation(s)
- Francesca Pistollato
- Centre for Nutrition and Health, Universidad Europea Del Atlántico (UEA), Santander, Spain
| | | | - Roberto Ruiz
- Centre for Nutrition and Health, Universidad Europea Del Atlántico (UEA), Santander, Spain
| | - Silvia Aparicio
- Centre for Nutrition and Health, Universidad Europea Del Atlántico (UEA), Santander, Spain
| | - Jorge Crespo
- Centre for Nutrition and Health, Universidad Europea Del Atlántico (UEA), Santander, Spain
| | - Luis Dzul Lopez
- Universidad Internacional Iberoamericana (UNINI), Campeche, Mexico
| | - Francesca Giampieri
- Dipartimento di Scienze Cliniche Specialistiche Ed Odontostomatologiche, Sez. Biochimica, Università Politecnica Delle Marche, Ancona, Italy.
| | - Maurizio Battino
- Centre for Nutrition and Health, Universidad Europea Del Atlántico (UEA), Santander, Spain; Dipartimento di Scienze Cliniche Specialistiche Ed Odontostomatologiche, Sez. Biochimica, Università Politecnica Delle Marche, Ancona, Italy.
| |
Collapse
|
127
|
Gao Y, Yang Y, Yuan F, Huang J, Xu W, Mao B, Yuan Z, Bi W. TNFα-YAP/p65-HK2 axis mediates breast cancer cell migration. Oncogenesis 2017; 6:e383. [PMID: 28945218 PMCID: PMC5623908 DOI: 10.1038/oncsis.2017.83] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 07/30/2017] [Accepted: 08/19/2017] [Indexed: 12/16/2022] Open
Abstract
Clinical and experimental evidence indicates that macrophages could promote solid-tumor progression and metastasis. However, the mechanisms underlying this process remain unclear. Here we show that yes-associated protein 1 (YAP1), a transcriptional regulator that controls tissue growth and regeneration, has an important role in tumor necrosis factor α (TNF α)-induced breast cancer migration. Mechanistically, macrophage conditioned medium (CM) or TNFα triggers IκB kinases (IKKs)-mediated YAP phosphorylation and activation in breast cancer cells. We further found that TNFα or macrophage CM treatment increases the interaction between p65 and YAP. Chromatin immunoprecipitation (ChIP) assay shows that YAP/TEAD (TEA domain family member) and p65 proteins synergistically regulate the transcription of hexokinase 2 (HK2), a speed-limiting enzyme in glycolysis, and promotes TNFα-induced or macrophage CM-induced cell migration. Together, our findings indicate an important role of TNFα-IKK-YAP/p65-HK2 signaling axis in the process of inflammation-driven migration in breast cancer cells, which reveals a new molecular link between inflammation and breast cancer metastasis.
Collapse
Affiliation(s)
- Y Gao
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.,The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Y Yang
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - F Yuan
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - J Huang
- Department of Orthopedics, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing, China
| | - W Xu
- General Surgery Center, Chinese PLA General Hospital, Beijing, China
| | - B Mao
- State Key Laboratory of Brain and Cognitive Sciences, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Z Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China.,Center of Alzheimer's Disease, Beijing Institute for Brain Disorders, Beijing, China
| | - W Bi
- Department of Orthopedics, Clinical Division of Surgery, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
128
|
Fougère B, Boulanger E, Nourhashémi F, Guyonnet S, Cesari M. Chronic Inflammation: Accelerator of Biological Aging. J Gerontol A Biol Sci Med Sci 2017; 72:1218-1225. [PMID: 28003373 DOI: 10.1093/gerona/glw240] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 11/09/2016] [Indexed: 12/13/2022] Open
Abstract
Biological aging is characterized by a chronic low-grade inflammation level. This chronic phenomenon has been named "inflamm-aging" and is a highly significant risk factor for morbidity and mortality in the older persons. The most common theories of inflamm-aging include redox stress, mitochondrial dysfunction, glycation, deregulation of the immune system, hormonal changes, epigenetic modifications, and dysfunction telomere attrition. Inflamm-aging plays a role in the initiation and progression of age-related diseases such as type II diabetes, Alzheimer's disease, cardiovascular disease, frailty, sarcopenia, osteoporosis, and cancer. This review will cover the identification of pathways that control age-related inflammation across multiple systems and its potential causal role in contributing to adverse health outcomes.
Collapse
Affiliation(s)
- Bertrand Fougère
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| | | | - Fati Nourhashémi
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Sophie Guyonnet
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| | - Matteo Cesari
- Gérontopôle, Centre Hospitalier Universitaire de Toulouse, Toulouse, France.,Inserm UMR1027, Université de Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
129
|
The prognostic significance of postoperative neutrophil-to-lymphocyte ratio after radical prostatectomy for localized prostate cancer. Oncotarget 2017; 8:11778-11787. [PMID: 28052031 PMCID: PMC5355303 DOI: 10.18632/oncotarget.14349] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/15/2016] [Indexed: 02/07/2023] Open
Abstract
Background The pretreatment neutrophil-to-lymphocyte ratio has prognostic value after radical prostatectomy for treating localized prostate cancer. However, the use of postoperative neutrophil-to-lymphocyte ratio has not been evaluated in this population. We investigated the prognostic significance of early postoperative neutrophil-to-lymphocyte ratio after radical prostatectomy for prostate cancer. Methods We retrospectively reviewed clinical data from 2,302 patients with localized prostate cancer who underwent radical prostatectomy at our institution between years 2000 and 2010. Only patients with pre- and postoperative complete blood counts with differential results were included. Patients who received neoadjuvant or postoperative adjuvant treatment and those without adequate medical records were excluded. Kaplan-Meier analyses were performed to analyze biochemical recurrence-free survival and overall survival rates. Univariate and multivariate Cox regression models were used for each endpoint. Results Kaplan-Meier curves showed that high postoperative neutrophil-to-lymphocyte ratio (>3.5) was significantly associated with decreased biochemical recurrence-free survival (p = 0.009) and overall survival (p = 0.010). In the univariate and multivariate Cox regression analyses, high postoperative neutrophil-to-lymphocyte ratio was a significant predictor of biochemical recurrence (hazard ratio 1.270, p = 0.008) and overall survival (hazard ratio 1.437, p = 0.033). Conclusions Our results demonstrate that postoperative neutrophil-to-lymphocyte ratio is an independent factor for biochemical recurrence and overall survival in patients who underwent radical prostatectomy for prostate cancer. These findings suggest that neutrophil-to-lymphocyte ratio can be a potentially valuable tool for stratifying high-risk patients and facilitating choices of postoperative therapy in patients with prostate cancer.
Collapse
|
130
|
Hanna M, Dumas I, Orain M, Jacob S, Têtu B, Sanschagrin F, Bureau A, Poirier B, Diorio C. Association between local inflammation and breast tissue age-related lobular involution among premenopausal and postmenopausal breast cancer patients. PLoS One 2017; 12:e0183579. [PMID: 28846716 PMCID: PMC5573208 DOI: 10.1371/journal.pone.0183579] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 08/07/2017] [Indexed: 12/28/2022] Open
Abstract
Increased levels of pro-inflammatory markers and decreased levels of anti-inflammatory markers in the breast tissue can result in local inflammation. We aimed to investigate whether local inflammation in the breast tissue is associated with age-related lobular involution, a process inversely related to breast cancer risk. Levels of eleven pro- and anti-inflammatory markers were assessed by immunohistochemistry in normal breast tissue obtained from 164 pre- and postmenopausal breast cancer patients. Involution status of the breast (degree of lobular involution and the predominant lobule type) was microscopically assessed in normal breast tissue on hematoxylin-eosin stained mastectomy slides. Multivariate generalized linear models were used to assess the associations. In age-adjusted analyses, higher levels of pro-inflammatory markers IL-6, TNF-α, CRP, COX-2, leptin, SAA1 and IL-8; and anti-inflammatory marker IL-10, were inversely associated with the prevalence of complete lobular involution (all P≤0.04). Higher levels of the pro-inflammatory marker COX-2 were also associated with lower prevalence of predominant type 1/no type 3 lobules in the breast, an indicator of complete involution, in age-adjusted analysis (P = 0.017). Higher tissue levels of inflammatory markers, mainly the pro-inflammatory ones, are associated with less involuted breasts and may consequently be associated with an increased risk of developing breast cancer.
Collapse
Affiliation(s)
- Mirette Hanna
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
- Department of Social and Preventive Medicine, Cancer Research Center, Université Laval, Québec, Québec, Canada
| | - Isabelle Dumas
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
| | - Michèle Orain
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
| | - Simon Jacob
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
- Department of Molecular Biology, Medical Chemistry and Pathology, Cancer Research Center, Université Laval, Québec, Québec, Canada
- Service of Molecular Biology, Medical Chemistry and Pathology, Hôpital Saint-Sacrement, CHU de Québec, Université Laval, Québec, Québec, Canada
- Centre des Maladies du Sein Deschênes-Fabia, Hôpital du Saint-Sacrement, Québec, Québec, Canada
| | - Bernard Têtu
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
- Department of Molecular Biology, Medical Chemistry and Pathology, Cancer Research Center, Université Laval, Québec, Québec, Canada
- Service of Molecular Biology, Medical Chemistry and Pathology, Hôpital Saint-Sacrement, CHU de Québec, Université Laval, Québec, Québec, Canada
- Centre des Maladies du Sein Deschênes-Fabia, Hôpital du Saint-Sacrement, Québec, Québec, Canada
| | - François Sanschagrin
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
- Centre des Maladies du Sein Deschênes-Fabia, Hôpital du Saint-Sacrement, Québec, Québec, Canada
| | - Alexandre Bureau
- Department of Social and Preventive Medicine, Cancer Research Center, Université Laval, Québec, Québec, Canada
- Centre de Recherche de l’Institut Universitaire en Santé Mentale de Québec, Université Laval, Québec, Québec, Canada
| | - Brigitte Poirier
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
- Centre des Maladies du Sein Deschênes-Fabia, Hôpital du Saint-Sacrement, Québec, Québec, Canada
- Department of Surgery, Cancer Research Center, Université Laval, Québec, Québec, Canada
| | - Caroline Diorio
- Oncology Research Unit, CHU de Québec Research Center, Université Laval, Québec, Québec, Canada
- Department of Social and Preventive Medicine, Cancer Research Center, Université Laval, Québec, Québec, Canada
- Centre des Maladies du Sein Deschênes-Fabia, Hôpital du Saint-Sacrement, Québec, Québec, Canada
- * E-mail:
| |
Collapse
|
131
|
Bianco TM, Abdalla DR, Desidério CS, Thys S, Simoens C, Bogers JP, Murta EFC, Michelin MA. The influence of physical activity in the anti-tumor immune response in experimental breast tumor. Immunol Lett 2017; 190:148-158. [PMID: 28818640 DOI: 10.1016/j.imlet.2017.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/03/2017] [Accepted: 08/09/2017] [Indexed: 12/31/2022]
Abstract
This study aimed to investigate the influence of physical activity in innate immunity to conduce to an effective antitumoral immune response analyzing the phenotype and activation status of infiltrating cells. We analysed the intracellular cytokines and the transcription factors of tumor infiltrating lymphocytes (TILS) and spleen leukocytes. The Nos2 gene expression was evaluated in spleen cells and futhermore the ROS production was measured and spleen cells; another cell evaluated was dendritic cells (TIDCs), their cytokines expression and membrane molecules; finally to understood the results obtained, we analysed the dendritic cells obtained from bone marrow. Were used female Balb/c mice divided into 4 groups: two controls without tumor, sedentary (GI) and trained (GII) and two groups with tumor, sedentary (GIII) or trained (GIV). The physical activity (PA) was realized acoording swimming protocol. Tumor was induced by injection of 4T1 cells. All experiments were performed in biological triplicate. After the experimental period, the tumor was removed and the cells were identified by flow cytometry with labeling to CD4, CD8, CD11c, CD11b, CD80, CD86 and Ia, and intracelular staining IL-10, IL-12, TNF-α, IFN-γ, IL-17, Tbet, GATA3, RORγt and FoxP3. The bone marrow of the animals was obtained to analyse the derivated DCs by flow cytometry and culture cells to obtain the supernatant to measure the cytokines. Our results demonstrated that the PA inhibit the tumoral growth although not to change the number of TILS, but reduced expression of GATA-3, ROR-γT, related with poor prognosis, and TNF-α intracellular; however occur one significantly reduction in TIDCS, but these cells expressed more co-stimulatory and presentation molecules. Furthermore, we observed that the induced PA stimulated the gene expression of Tbet and the production of inflammatory cytokines suggesting an increase of Th1 systemic response. The results evaluating the systemic influence in DCs showed that the PA improve significantly the number of those cells in bone marrow as well the number of co-stimulatory molecules. Therefore, we could conclude that PA influence the innate immunity by interfering to promote in process of maturation of DCs both in tumor and systemically, that by its turn promote a modification in acquired immune cells, representing by T helper to induce an important alteration transcription factors that are responsible to maintain a suppressive microenviroment, and thereby, allowing the latter cells can thus activate antitumor immune response. The PA was able improve the Th1 systemic response by enhance to Tbet gene expression, promote a slightly increased of Th1-type cytokines and decrease Gata3 and Foxp3 gene expression in which can inhibit the Th1 immune response.
Collapse
Affiliation(s)
- Thiago M Bianco
- Oncology Research Institute (Instituto de Pesquisa em Oncologia-IPON), Federal University of the Triângulo Mineiro (UFTM), Brazil
| | - Douglas R Abdalla
- Oncology Research Institute (Instituto de Pesquisa em Oncologia-IPON), Federal University of the Triângulo Mineiro (UFTM), Brazil
| | - Chamberttan S Desidério
- Oncology Research Institute (Instituto de Pesquisa em Oncologia-IPON), Federal University of the Triângulo Mineiro (UFTM), Brazil
| | - Sofie Thys
- Laboratory of Cell Biology and Histology, University of Antwerp, Belgium
| | - Cindy Simoens
- Laboratory of Cell Biology and Histology, University of Antwerp, Belgium
| | - John-Paul Bogers
- Laboratory of Cell Biology and Histology, University of Antwerp, Belgium
| | - Eddie F C Murta
- Oncology Research Institute (IPON)/Discipline of Gynecology and Obstetrics, UFTM, Brazil
| | - Márcia A Michelin
- Oncology Research Institute (IPON)/Discipline of Immunology, UFTM, Uberaba, Minas Gerais, Brazil.
| |
Collapse
|
132
|
Terry S, Savagner P, Ortiz-Cuaran S, Mahjoubi L, Saintigny P, Thiery JP, Chouaib S. New insights into the role of EMT in tumor immune escape. Mol Oncol 2017; 11:824-846. [PMID: 28614624 PMCID: PMC5496499 DOI: 10.1002/1878-0261.12093] [Citation(s) in RCA: 296] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/27/2017] [Accepted: 06/02/2017] [Indexed: 01/02/2023] Open
Abstract
Novel immunotherapy approaches have provided durable remission in a significant number of cancer patients with cancers previously considered rapidly lethal. Nonetheless, the high degree of nonresponders, and in some cases the emergence of resistance in patients who do initially respond, represents a significant challenge in the field of cancer immunotherapy. These issues prompt much more extensive studies to better understand how cancer cells escape immune surveillance and resist immune attacks. Here, we review the current knowledge of how cellular heterogeneity and plasticity could be involved in shaping the tumor microenvironment (TME) and in controlling antitumor immunity. Indeed, recent findings have led to increased interest in the mechanisms by which cancer cells undergoing epithelial‐mesenchymal transition (EMT), or oscillating within the EMT spectrum, might contribute to immune escape through multiple routes. This includes shaping of the TME and decreased susceptibility to immune effector cells. Although much remains to be learned on the mechanisms at play, cancer cell clones with mesenchymal features emerging from the TME seem to be primed to face immune attacks by specialized killer cells of the immune system, the natural killer cells, and the cytotoxic T lymphocytes. Recent studies investigating patient tumors have suggested EMT as a candidate predictive marker to be explored for immunotherapy outcome. Promising data also exist on the potential utility of targeting these cancer cell populations to at least partly overcome such resistance. Research is now underway which may lead to considerable progress in optimization of treatments.
Collapse
Affiliation(s)
- Stéphane Terry
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine - Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| | - Pierre Savagner
- Institut de Recherche en Cancérologie de Montpellier, France.,U1194, INSERM, Montpellier, France.,Université Montpellier, France.,Institut du Cancer Montpellier, France
| | - Sandra Ortiz-Cuaran
- INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon, France.,Université de Lyon, France.,Centre Léon Bérard, Lyon, France.,Faculté de Pharmacie de Lyon, ISPB, Université Lyon 1, France.,LabEx DEVweCAN, Université de Lyon, France
| | - Linda Mahjoubi
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine - Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| | - Pierre Saintigny
- INSERM U1052, CNRS UMR 5286, Cancer Research Center of Lyon, France.,Université de Lyon, France.,Centre Léon Bérard, Lyon, France.,Faculté de Pharmacie de Lyon, ISPB, Université Lyon 1, France.,LabEx DEVweCAN, Université de Lyon, France
| | - Jean-Paul Thiery
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine - Univ. Paris-Sud, University Paris-Saclay, Villejuif, France.,CNRS UMR 7057, Matter and Complex Systems, Paris, France.,Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Genetic Oncology, Gustave Roussy, EPHE, Fac. de médecine - Univ. Paris-Sud, University Paris-Saclay, Villejuif, France
| |
Collapse
|
133
|
Wilkie KP, Hahnfeldt P. Modeling the Dichotomy of the Immune Response to Cancer: Cytotoxic Effects and Tumor-Promoting Inflammation. Bull Math Biol 2017; 79:1426-1448. [PMID: 28585066 DOI: 10.1007/s11538-017-0291-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/04/2017] [Indexed: 12/18/2022]
Abstract
Although the immune response is often regarded as acting to suppress tumor growth, it is now clear that it can be both stimulatory and inhibitory. The interplay between these competing influences has complex implications for tumor development, cancer dormancy, and immunotherapies. In fact, early immunotherapy failures were partly due to a lack in understanding of the nonlinear growth dynamics these competing immune actions may cause. To study this biological phenomenon theoretically, we construct a minimally parameterized framework that incorporates all aspects of the immune response. We combine the effects of all immune cell types, general principles of self-limited logistic growth, and the physical process of inflammation into one quantitative setting. Simulations suggest that while there are pro-tumor or antitumor immunogenic responses characterized by larger or smaller final tumor volumes, respectively, each response involves an initial period where tumor growth is stimulated beyond that of growth without an immune response. The mathematical description is non-identifiable which allows an ensemble of parameter sets to capture inherent biological variability in tumor growth that can significantly alter tumor-immune dynamics and thus treatment success rates. The ability of this model to predict non-intuitive yet clinically observed patterns of immunomodulated tumor growth suggests that it may provide a means to help classify patient response dynamics to aid identification of appropriate treatments exploiting immune response to improve tumor suppression, including the potential attainment of an immune-induced dormant state.
Collapse
Affiliation(s)
- Kathleen P Wilkie
- Center of Cancer Systems Biology, Boston, MA, USA.
- Department of Mathematics, Ryerson University, Toronto, ON, Canada.
| | | |
Collapse
|
134
|
Abstract
OBJECTIVE To examine the survival outcomes in women with endometrial cancer who were taking low-dose aspirin (81-100 mg/d). METHODS A multicenter retrospective study was conducted examining patients with stage I-IV endometrial cancer who underwent hysterectomy-based surgical staging between January 2000 and December 2013 (N=1,687). Patient demographics, medical comorbidities, medication types, tumor characteristics, and treatment patterns were correlated to survival outcomes. A Cox proportional hazard regression model was used to estimate adjusted hazard ratio for disease-free and disease-specific overall survival. RESULTS One hundred fifty-eight patients (9.4%, 95% confidence interval [CI] 8.8-11.9) were taking low-dose aspirin. Median follow-up time for the study cohort was 31.5 months. One hundred twenty-seven patients (7.5%) died of endometrial cancer. Low-dose aspirin use was significantly correlated with concurrent obesity, hypertension, diabetes mellitus, and hypercholesterolemia (all P<.001). Low-dose aspirin users were more likely to take other antihypertensive, antiglycemic, and anticholesterol agents (all P<.05). Low-dose aspirin use was not associated with histologic subtype, tumor grade, nodal metastasis, or cancer stage (all P>.05). On multivariable analysis, low-dose aspirin use remained an independent prognostic factor associated with an improved 5-year disease-free survival rate (90.6% compared with 80.9%, adjusted hazard ratio 0.46, 95% CI 0.25-0.86, P=.014) and disease-specific overall survival rate (96.4% compared with 87.3%, adjusted hazard ratio 0.23, 95% CI 0.08-0.64, P=.005). The increased survival effect noted with low-dose aspirin use was greatest in patients whose age was younger than 60 years (5-year disease-free survival rates, 93.9% compared with 84.0%, P=.013), body mass index was 30 or greater (92.2% compared with 81.4%, P=.027), who had type I cancer (96.5% compared with 88.6%, P=.029), and who received postoperative whole pelvic radiotherapy (88.2% compared with 61.5%, P=.014). These four factors remained significant for disease-specific overall survival (all P<.05). CONCLUSION Our results suggest that low-dose aspirin use is associated with improved survival outcomes in women with endometrial cancer, especially in those who are young, obese, with low-grade disease, and who receive postoperative radiotherapy.
Collapse
|
135
|
Modelling and investigation of theCD4+T cells – Macrophages paradox in melanoma immunotherapies. J Theor Biol 2017; 420:82-104. [DOI: 10.1016/j.jtbi.2017.02.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2016] [Revised: 02/12/2017] [Accepted: 02/16/2017] [Indexed: 12/18/2022]
|
136
|
Yan H, Zhang P, Kong X, Hou X, Zhao L, Li T, Yuan X, Fu H. Primary Tr1 cells from metastatic melanoma eliminate tumor-promoting macrophages through granzyme B- and perforin-dependent mechanisms. Tumour Biol 2017; 39:1010428317697554. [PMID: 28378637 DOI: 10.1177/1010428317697554] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In malignant melanoma, tumor-associated macrophages play multiple roles in promoting tumor growth, such as inducing the transformation of melanocytes under ultraviolet irradiation, increasing angiogenesis in melanomas, and suppressing antitumor immunity. Because granzyme B- and perforin-expressing Tr1 cells could specifically eliminate antigen-presenting cells of myeloid origin, we examined whether Tr1 cells in melanoma could eliminate tumor-promoting macrophages and how the interaction between Tr1 cells and macrophages could affect the growth of melanoma cells. Tr1 cells were characterized by high interleukin 10 secretion and low Foxp3 expression and were enriched in the CD4+CD49b+LAG-3+ T-cell fraction. Macrophages derived from peripheral blood monocytes in the presence of modified melanoma-conditioned media demonstrated tumor-promoting capacity, exemplified by improving the proliferation of cocultured A375 malignant melanoma cells. But when primary Tr1 cells were present in the macrophage-A375 coculture, the growth of A375 cells was abrogated. The conventional CD25+ Treg cells, however, were unable to inhibit macrophage-mediated increase in tumor cell growth. Further analyses showed that Tr1 cells did not directly eliminate A375 cells, but mediated the killing of tumor-promoting macrophages through the secretion of granzyme B and perforin. The tumor-infiltrating interleukin 10+Foxp3-CD4+ T cells expressed very low levels of granzyme B and perforin, possibly suggested the downregulation of Tr1 cytotoxic capacity in melanoma tumors. Together, these data demonstrated an antitumor function of Tr1 cells through the elimination of tumor-promoting macrophages, which was not shared by conventional Tregs.
Collapse
Affiliation(s)
- Hongxia Yan
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| | - Ping Zhang
- 2 Jining Maternity and Child Health Care Hospital, Jining, China
| | - Xue Kong
- 3 Department of Integrated Traditional Chinese and Western Medicine, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xianglian Hou
- 4 Department of Supply and Services, Jiaxiang County People's Hospital, Jining, China
| | - Li Zhao
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| | - Tianhang Li
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| | - Xiaozhou Yuan
- 5 DICAT Biomedical Computation Centre, Vancouver, BC, Canada
| | - Hongjun Fu
- 1 Department of Dermatology, The First People's Hospital of Jining City, Jining, China
| |
Collapse
|
137
|
Hendrickx W, Simeone I, Anjum S, Mokrab Y, Bertucci F, Finetti P, Curigliano G, Seliger B, Cerulo L, Tomei S, Delogu LG, Maccalli C, Wang E, Miller LD, Marincola FM, Ceccarelli M, Bedognetti D. Identification of genetic determinants of breast cancer immune phenotypes by integrative genome-scale analysis. Oncoimmunology 2017; 6:e1253654. [PMID: 28344865 PMCID: PMC5353940 DOI: 10.1080/2162402x.2016.1253654] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/20/2016] [Accepted: 10/22/2016] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy is revolutionizing the clinical management of several tumors, but has demonstrated limited activity in breast cancer. The development of more effective treatments is hindered by incomplete knowledge of the genetic determinant of immune responsiveness. To fill this gap, we mined copy number alteration, somatic mutation, and expression data from The Cancer Genome Atlas (TCGA). By using RNA-sequencing data from 1,004 breast cancers, we defined distinct immune phenotypes characterized by progressive expression of transcripts previously associated with immune-mediated rejection. The T helper 1 (Th-1) phenotype (ICR4), which also displays upregulation of immune-regulatory transcripts such as PDL1, PD1, FOXP3, IDO1, and CTLA4, was associated with prolonged patients' survival. We validated these findings in an independent meta-cohort of 1,954 breast cancer gene expression data. Chromosome segment 4q21, which includes genes encoding for the Th-1 chemokines CXCL9-11, was significantly amplified only in the immune favorable phenotype (ICR4). The mutation and neoantigen load progressively decreased from ICR4 to ICR1 but could not fully explain immune phenotypic differences. Mutations of TP53 were enriched in the immune favorable phenotype (ICR4). Conversely, the presence of MAP3K1 and MAP2K4 mutations were tightly associated with an immune-unfavorable phenotype (ICR1). Using both the TCGA and the validation dataset, the degree of MAPK deregulation segregates breast tumors according to their immune disposition. These findings suggest that mutation-driven perturbations of MAPK pathways are linked to the negative regulation of intratumoral immune response in breast cancer. Modulations of MAPK pathways could be experimentally tested to enhance breast cancer immune sensitivity.
Collapse
Affiliation(s)
- Wouter Hendrickx
- Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Research Branch, Sidra Medical and Research Center , Doha, Qatar
| | - Ines Simeone
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar; Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Samreen Anjum
- Qatar Computing Research Institute, Hamad Bin Khalifa University , Doha, Qatar
| | - Younes Mokrab
- Division of Biomedical Informatics, Research Branch, Sidra Medical and Research Center , Doha, Qatar
| | - François Bertucci
- Département d'Oncologie Moléculaire, Center de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France; Département d'Oncologie Médicale, CRCM, Institut Paoli-Calmettes, Marseille, France; Faculté de Médecine, Aix-Marseille Université, Marseille, France
| | - Pascal Finetti
- Département d'Oncologie Moléculaire, Center de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes , INSERM UMR1068, CNRS UMR725 , Marseille, France
| | - Giuseppe Curigliano
- Division of Experimental Therapeutics, Division of Medical Oncology, European Institute of Oncology , Milan, Italy
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg , Halle, Germany
| | - Luigi Cerulo
- Department of Science and Technology, University of Sannio, Benevento, Italy; BIOGEM Research Center, Ariano Irpino, Italy
| | - Sara Tomei
- Division of Translational Medicine, Research Branch, Sidra Medical and Research Center , Doha, Qatar
| | - Lucia Gemma Delogu
- Department of Chemistry and Pharmacy, University of Sassari , Sassari, Italy
| | - Cristina Maccalli
- Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Research Branch, Sidra Medical and Research Center , Doha, Qatar
| | - Ena Wang
- Division of Translational Medicine, Research Branch, Sidra Medical and Research Center , Doha, Qatar
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest School of Medicine , Winston-Salem, NC, USA
| | - Francesco M Marincola
- Office of the Chief Research Officer (CRO), Research Branch, Sidra Medical and Research Center , Doha, Qatar
| | - Michele Ceccarelli
- Qatar Computing Research Institute, Hamad Bin Khalifa University , Doha, Qatar
| | - Davide Bedognetti
- Tumor Biology, Immunology, and Therapy Section, Division of Translational Medicine, Research Branch, Sidra Medical and Research Center , Doha, Qatar
| |
Collapse
|
138
|
Laohawiriyakamol S, Mahattanobon S, Laohawiriyakamol S, Puttawibul P. The Pre-Treatment Neutrophil-Lymphocyte Ratio: a Useful Tool in Predicting Non-Sentinel Lymph Node Metastasis in Breast Cancer Cases. Asian Pac J Cancer Prev 2017; 18:557-562. [PMID: 28345845 PMCID: PMC5454758 DOI: 10.22034/apjcp.2017.18.2.557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: The sentinel lymph node (SLN) biopsy is a highly accurate predictor of overall axillary nodal status in early breast cancer patients. There is however, still a debate on which patients with a positive SLN can benefit from axillary lymph node dissection (ALND). Numerous studies have been designed to identify variables that are predictive of non-SLN metastasis to avoid a complete ALND. The aim of this study was to determine whether the pre-treatment neutrophil-lymphocyte ratio (NLR) can be a predictive factor of non-SLN metastasis in early breast cancer patients. Materials and Methods: The records of 214 consecutive patients with cT1-3N0 invasive breast cancer who had undergone intraoperative SLN evaluation at Songklanagarind Hospital between the 1st of March 2011 and the 30th of May 2016 were examined. Data on patient demographics, tumor variables and NLR were collected and factors for non-SLN metastasis were analyzed using multivariate logistic regression. The power of the NLR was quantified with receiver operating characteristics (ROC) curves as measured by the areas under curves (AUC). Results: Multivariate analysis established presence of lymphovascular invasion (OR 8.4, 95%CI 2.3-31.3, p=0.002), macrometastasis (OR 6.6, 95%CI 1.8-24.7, p=0.005), and NLR (OR 2.3, 95%CI 1.1-4.8, p=0.033) as predictive factors of non-SLN metastasis with statistical significance. The AUC for NLR was 0.7 (95%CI 0.6-0.8) with an optimal cut-off of 2.6 giving a sensitivity of 62%, a specificity of 83.8%, a positive predictive value of 77.3% and a negative predictive value of 70.5%. Conclusion: Pre-treatment NLR is a useful diagnostic aid for predicting additional non-SLN metastasis.
Collapse
Affiliation(s)
- Suphawat Laohawiriyakamol
- Division of General Surgery, Department of Surgery, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla,
Thailand.
| | | | | | | |
Collapse
|
139
|
Dmitrieva OS, Shilovskiy IP, Khaitov MR, Grivennikov SI. Interleukins 1 and 6 as Main Mediators of Inflammation and Cancer. BIOCHEMISTRY (MOSCOW) 2017; 81:80-90. [PMID: 27260388 DOI: 10.1134/s0006297916020024] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The idea of a potential link between cancer and inflammation was first proposed by R. Virchow in the nineteenth century. However, clear evidence regarding a key role of inflammation in oncogenesis appeared only during the last decade. Now the tumor microenvironment is commonly considered as an obligatory and significant component of almost all types of cancer, and the cells infiltrating such microenvironment are a source of inflammatory cytokines. Such cytokines play a key role in regulating inflammation during both normal immune response and developing cancer. In this review, we explore the role of two inflammatory cytokines interleukin 1 and interleukin 6 in cancer development. These cytokines have pleiotropic effects on various cell types in the tumor microenvironment, particularly being able to regulate pro-oncogenic transcription factors NF-κB and STAT3. For this reason, such cytokines influence key parameters of oncogenesis, increasing cell resistance to apoptosis, proliferation of cancer cells, angiogenesis, invasion and malignancy as well as the ability of tumor cells to respond to anticancer therapy. Here we summarize novel experimental data regarding mechanisms underlying the interaction between chronic inflammation and malignant neoplasms.
Collapse
Affiliation(s)
- O S Dmitrieva
- Fox Chase Cancer Center, Cancer Prevention and Control program, Philadelphia, PA, USA.
| | | | | | | |
Collapse
|
140
|
Berger T, Saunders ME, Mak TW. Beyond the Oncogene Revolution: Four New Ways to Combat Cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 81:85-92. [PMID: 28057846 DOI: 10.1101/sqb.2016.81.031161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
It has become clear that tumorigenesis results from much more than just the activation of an oncogene and/or the inactivation of a tumor-suppressor gene, and that the cancer cell genome contains many more alterations than can be specifically targeted at once. This observation has led our group to a search for alternative ways to kill cancer cells (while sparing normal cells) by focusing on properties unique to the former. We have identified four approaches with the potential to generate new anticancer therapies: combatting the tactics by which cancers evade antitumor immune responses, targeting metabolic adaptations that tumor cells use to survive conditions that would kill normal cells, manipulating a cancer cell's response to excessive oxidative stress, and exploiting aneuploidy. This review describes our progress to date on these fronts.
Collapse
Affiliation(s)
- Thorsten Berger
- The Campbell Family Institute for Breast Cancer Research and Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Mary E Saunders
- The Campbell Family Institute for Breast Cancer Research and Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research and Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| |
Collapse
|
141
|
Jiang C, Yuan F, Wang J, Wu L. Oral squamous cell carcinoma suppressed antitumor immunity through induction of PD-L1 expression on tumor-associated macrophages. Immunobiology 2016; 222:651-657. [PMID: 28017495 DOI: 10.1016/j.imbio.2016.12.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 11/03/2016] [Accepted: 12/13/2016] [Indexed: 12/22/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common solid tumor in the oral cavity. Development and progression of OSCC is associated with the elevated presence of inhibitory M2 type tumor-associated macrophages (TAMs). However, the underlying mechanism leading to the enrichment of M2 TAMs and the pathway through which TAMs foster tumor progression are still unclear. In this study, we harvested TAMs and tumor cells from primary OSCC resections of stage II and stage III patients. We showed that compared to peritumoral macrophages, TAMs presented upregulated expression of PD-L1 and elevated capacity in inducing T cell apoptosis. The level of PD-L1 expression directly correlated with the level of T cell apoptosis. Interestingly, peripheral blood monocytes with low initial PD-L1 level had upregulated PD-L1 expression and acquired the ability to induce T cell apoptosis, after incubation with primary tumor cells from OSCC patients. The PD-L1 expression by monocytes depended on interleukin 10 (IL-10), since blockade of IL-10 in the tumor-monocyte coculture abrogated PD-L1 upregulation. IL-10 mRNA expression in tumor cells and monocytes also preceded PD-L1 mRNA expression in monocytes. Furthermore, the IL-10 concentration in the tumor microenvironment directly correlated with the PD-L1 level on TAMs. Together, these results suggest that OSCC could directly suppress antitumor T cell immunity through conditioning TAMs.
Collapse
Affiliation(s)
- Canhua Jiang
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Fulai Yuan
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Changsha, 410078, China.
| | - Limeng Wu
- Department of Oral and Maxillofacial Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| |
Collapse
|
142
|
Shi X, Lu L, Jin X, Liu B, Sun X, Lu L, Jiang Y. GRIN3A and MAPT stimulate nerve overgrowth in macrodactyly. Mol Med Rep 2016; 14:5637-5643. [PMID: 27840953 DOI: 10.3892/mmr.2016.5923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 10/12/2016] [Indexed: 11/06/2022] Open
|
143
|
Takakura K, Ito Z, Suka M, Kanai T, Matsumoto Y, Odahara S, Matsudaira H, Haruki K, Fujiwara Y, Saito R, Gocho T, Nakashiro KI, Hamakawa H, Okamoto M, Kajihara M, Misawa T, Ohkusa T, Koido S. Comprehensive assessment of the prognosis of pancreatic cancer: peripheral blood neutrophil-lymphocyte ratio and immunohistochemical analyses of the tumour site. Scand J Gastroenterol 2016; 51:610-7. [PMID: 26679084 DOI: 10.3109/00365521.2015.1121515] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Several studies have suggested that an elevated neutrophil-lymphocyte ratio (NLR) is associated with a poorer prognosis in patients with pancreatic cancer (PC). The correlations between the NLR and immunohistochemical (IHC) analysis with regard to the prognosis of patients with PC remain to be elucidated. By using IHC findings, we determined the value of the NLR as a prognostic factor in patients with PC. MATERIAL AND METHODS We collected the clinico-pathological data of 28 consecutive patients who underwent surgical resection for PC between January 2008 and December 2012 at The Jikei University Kashiwa Hospital. We investigated whether the NLR and IHC results were related and ensured the consistency of the prognosis of patients with PC. RESULTS The Kaplan-Meier curves for the disease-free survival (DFS) and the overall survival (OS) revealed that an NLR ≥ 5 is an implicit factor for decreased DFS and OS in patients with PC (p = 0.003, p < 0.001, log-rank test). The density of CD163(+) macrophages and CD66b(+) neutrophils was significantly higher in the high NLR group; on the contrary, the density of CD20(+) lymphocytes was significantly higher in the low NLR group. Moreover, a Mann-Whitney U test showed that the NLR was significantly correlated with a high density of CD20(+) lymphocytes (p = 0.031) and CD163(+) macrophages (p = 0.023), while the NLR was not significantly correlated with CD66b(+) neutrophils (p = 0.397). CONCLUSIONS Our results demonstrated the validity of the NLR by IHC analyses and we determined that a higher value of NLR is a trustworthy prognostic factor for patients with PC.
Collapse
Affiliation(s)
- Kazuki Takakura
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Zensho Ito
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Machi Suka
- b Department of Public Health and Environmental Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Tomoya Kanai
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Yoshihiro Matsumoto
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Shunichi Odahara
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Hiroshi Matsudaira
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Koichiro Haruki
- c Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Yuki Fujiwara
- c Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Ryota Saito
- c Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Takeshi Gocho
- c Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Koh-Ichi Nakashiro
- d Department of Oral and Maxillofacial Surgery , Ehime University Graduate School of Medicine , Ehime , Japan
| | - Hiroyuki Hamakawa
- d Department of Oral and Maxillofacial Surgery , Ehime University Graduate School of Medicine , Ehime , Japan
| | - Masato Okamoto
- e Department of Advanced Immunotherapeutics , Kitasato University School of Pharmacy , Tokyo , Japan
| | - Mikio Kajihara
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Takeyuki Misawa
- c Department of Surgery , Jikei University School of Medicine , Tokyo , Japan
| | - Toshifumi Ohkusa
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| | - Shigeo Koido
- a Division of Gastroenterology and Hepatology, Department of Internal Medicine , Jikei University School of Medicine , Tokyo , Japan
| |
Collapse
|
144
|
Borges VF, Elder AM, Lyons TR. Deciphering Pro-Lymphangiogenic Programs during Mammary Involution and Postpartum Breast Cancer. Front Oncol 2016; 6:227. [PMID: 27853703 PMCID: PMC5090124 DOI: 10.3389/fonc.2016.00227] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022] Open
Abstract
Postpartum breast cancers are a highly metastatic subset of young women’s breast cancers defined as breast cancers diagnosed in the postpartum period or within 5 years of last child birth. Women diagnosed with postpartum breast cancer are nearly twice as likely to develop metastasis and to die from breast cancer when compared with nulliparous women. Additionally, epidemiological studies utilizing multiple cohorts also suggest that nearly half of all breast cancers in women aged <45 qualify as postpartum cases. Understanding the biology that underlies this increased risk for metastasis and death may lead to identification of targeted interventions that will benefit the large number of young women with breast cancer who fall into this subset. Preclinical mouse models of postpartum breast cancer have revealed that breast tumor cells become more aggressive if they are present during the normal physiologic process of postpartum mammary gland involution in mice. As involution appears to be a period of lymphatic growth and remodeling, and human postpartum breast cancers have high peritumor lymphatic vessel density (LVD) and increased incidence of lymph node metastasis (1, 2), we propose that novel insight into is to be gained through the study of the biological mechanisms driving normal postpartum mammary lymphangiogenesis as well as in the microenvironment of postpartum tumors.
Collapse
Affiliation(s)
- Virginia F Borges
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO, USA; Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alan M Elder
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO, USA; Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Traci R Lyons
- Young Women's Breast Cancer Translational Program, University of Colorado Cancer Center, Aurora, CO, USA; Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
145
|
Patterns and prognostic relevance of PD-1 and PD-L1 expression in colorectal carcinoma. Mod Pathol 2016; 29:1433-1442. [PMID: 27443512 PMCID: PMC5083129 DOI: 10.1038/modpathol.2016.139] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/18/2016] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
Abstract
Immune checkpoint blockade targeting the programmed death-1 (PD-1) pathway has shown efficacy in several types of cancers including mismatch-repair-deficient colorectal carcinoma. In some tumor types, programmed death-ligand 1 (PD-L1) expression detected by immunohistochemistry has shown utility as a predictive marker for response to anti-PD-1 therapies. This utility, however, remains to be determined in colorectal carcinoma. In addition, although tumor-infiltrating lymphocytes have been associated with better prognosis in colorectal carcinoma, the prognostic value of PD-1 expression in these lymphocytes and its interaction with PD-L1 expression still await investigation. To address these questions, we performed a pilot study to evaluate the patterns of PD-L1 and PD-1 immunohistochemical expression on colorectal carcinoma cells and their tumor-infiltrating lymphocytes, respectively. Using tissue microarray, we found that 5% (19/394) of colorectal carcinomas exhibited high tumor PD-L1 expression, and 19% (76/392) had elevated numbers of PD-1-positive tumor-infiltrating lymphocytes. PD-L1 levels correlated with PD-1 levels (P<0.001), and mismatch-repair-deficient tumors had significantly higher rates of high PD-L1 and PD-1 expression when compared with mismatch-repair-proficient tumors (18% vs 2% and 50% vs 13%, respectively; P<0.001 for both). Staining intensity was also stronger for both markers in mismatch-repair-deficient tumors. Furthermore, we observed that among patients with mismatch-repair-deficient colorectal carcinoma, PD-1/PD-L1 expression stratified recurrence-free survival in an inter-dependent manner: an association between high PD-1-positive tumor-infiltrating lymphocytes and improved recurrence-free survival (P=0.041) was maintained only when the tumors had low-level PD-L1 expression (P=0.006); patients whose tumors had both high PD-1-positive tumor-infiltrating lymphocytes and high PD-L1 expression had a significantly worse recurrence-free survival (P<0.001). Thus, our results not only provide a foundation for further assessment of PD-L1 immunohistochemistry as a predictive marker for anti-PD-1 therapy in colorectal carcinoma, they also shed light on the prognostic impact of tumor-infiltrating lymphocytes in different subsets of mismatch-repair-deficient colorectal carcinomas.
Collapse
|
146
|
Long H, Xiang T, Luo J, Li F, Lin R, Liu S, Jiang S, Hu C, Chen G, Wong E, Wan Y, Li QJ, Zhu B. The tumor microenvironment disarms CD8 + T lymphocyte function via a miR-26a-EZH2 axis. Oncoimmunology 2016; 5:e1245267. [PMID: 28123882 DOI: 10.1080/2162402x.2016.1245267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 09/29/2016] [Accepted: 10/01/2016] [Indexed: 01/05/2023] Open
Abstract
One of the most important factors that limit the potency of CD8+ cytotoxic T lymphocyte (CTL) responses is the tumor microenvironment (TME). Here, we provide evidence that miR-26a is a negative regulator of CTL function in the TME. Specifically, we identified miR-26a as a crucial suppressor gene in CTLs from the TME, as we found that, miR-26a expression was elevated in CTLs to respond to TME secretome stimulation. CTLs from miR-26a-transgenic mice showed impaired IFNγ and granzyme B production in response to their cognate antigen. Conversely, we found that miR-26a inhibition in CTLs could effectively increase the cytotoxicity and suppress tumor growth. Mechanically, we identified EZH2 as a direct target of miR-26a. miR-26a and EZH2 expression were found to be inversely correlated in CTLs, and the inhibition of EZH2 in CTLs impairs CTL function. These functional correlations were validated in a cohort of non-small cell lung cancer patients, indicating that the miR-26a-EZH2 axis is clinically relevant. Our findings suggested that miR-26a silencing as a novel strategy to improve the efficacy of CTL-based cancer immunotherapy.
Collapse
Affiliation(s)
- Haixia Long
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Tong Xiang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Jing Luo
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University , Chongqing, China
| | - Fei Li
- Biomedical Analysis Center, Third Military Medical University , Chongqing, China
| | - Regina Lin
- Department of Immunology, Duke University Medical Center , Durham, NC, USA
| | - Siqi Liu
- Department of Immunology, Duke University Medical Center , Durham, NC, USA
| | - Shan Jiang
- Department of Immunology, Duke University Medical Center , Durham, NC, USA
| | - Chunyan Hu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University , Chongqing, China
| | - Gang Chen
- Biomedical Analysis Center, Third Military Medical University , Chongqing, China
| | - Elizabeth Wong
- Department of Immunology, Duke University Medical Center , Durham, NC, USA
| | - Ying Wan
- Biomedical Analysis Center, Third Military Medical University , Chongqing, China
| | - Qi-Jing Li
- Department of Immunology, Duke University Medical Center , Durham, NC, USA
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Chongqing Key Laboratory of tumor immunotherapy, Chongqing, China
| |
Collapse
|
147
|
Prognostic value of platelet to lymphocyte ratio in hepatocellular carcinoma: a meta-analysis. Sci Rep 2016; 6:35378. [PMID: 27739490 PMCID: PMC5064312 DOI: 10.1038/srep35378] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/26/2016] [Indexed: 01/21/2023] Open
Abstract
This study was designed to evaluate the prognostic value of platelet to lymphocyte ratio (PLR) in hepatocellular carcinoma (HCC). A comprehensive literature search for relevant studies was performed in Web of science, Embase and Pubmed. A total of nine studies with 2017 patients were included in this meta-analysis, and combined hazard ratio (HR) or odds ratio (OR) and 95% confidence intervals (95%CIs) were served as effect measures. Pooled results showed that elevated PLR was associated with poor overall survival (OS) (HR = 1.63, 95%CI: 1.42–1.88, p = 0.000; I2 = 0.0%, Ph = 0.637) and poor disease-free survival (DFS)/recurrence-free survival (RFS) (HR=1.32, 95%CI: 1.15–1.52, p = 0.000; I2 = 19.3%, Ph = 0.287) in HCC patients. In addition, high PLR was not significantly correlated with the presence of vascular invasion, tumor multifocality, poor tumor grade or high level of serum AFP (>400 ng/ml). In conclusion, elevated PLR indicated a poor prognosis for patients with HCC. PLR may be a reliable, easily-obtained, and low cost biomarker with prognostic potential for HCC.
Collapse
|
148
|
Zhu S, Miao CW, Wang ZT, Peng L, Li B. Sensitivity value of hematological markers in patients receiving chemoradiotherapy for esophageal squamous cell carcinoma. Onco Targets Ther 2016; 9:6187-6193. [PMID: 27789959 PMCID: PMC5072515 DOI: 10.2147/ott.s115011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Hematological markers of the systemic inflammatory response (SIR) including the neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), and the combination of NLR with PLR (CNP) are associated with prognosis of patients with esophageal squamous cell carcinoma (ESCC). However, their value in predicting the sensitivity to chemoradiotherapy in patients with ESCC is unclear. The aim of this study was to investigate whether these markers can be used as sensitivity predictors for chemoradiotherapy in patients with ESCC. Patients and methods A total of 114 patients with newly diagnosed ESCC were retrospectively evaluated. They were treated with curative intent by primary radiotherapy only or concurrent chemoradiotherapy. These patients were grouped for further analysis according to the optimum cutoff values of NLR, PLR, and CNP. A univariate analysis was conducted to compare the ability of each of the hematological markers of SIR and clinicopathological characteristics. Multivariate analysis was performed to identify whether the markers were associated with the sensitivity to chemoradiotherapy. The relationship between clinicopathological characteristics and hematological markers was assessed. Results NLR, CNP, T stage, M stage, and clinical stage were significantly associated with the sensitivity to chemoradiotherapy. In multivariate analysis, CNP and clinical stage were the independent risk factors predicting a poorer sensitivity. Conclusion This study validated novel, easy-to-use hematological markers and found that CNP, an SIR score, is an independent hematological marker of poor sensitivity to chemoradiotherapy in patients with ESCC. This may help guide the planning of follow-up regimens.
Collapse
Affiliation(s)
- Shan Zhu
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan; Department of Radiotherapy
| | - Chuan-Wang Miao
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan; Department of Radiotherapy
| | | | - Li Peng
- Department of Clinical Laboratory, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, People's Republic of China
| | | |
Collapse
|
149
|
Mirzaei HR, Mirzaei H, Lee SY, Hadjati J, Till BG. Prospects for chimeric antigen receptor (CAR) γδ T cells: A potential game changer for adoptive T cell cancer immunotherapy. Cancer Lett 2016; 380:413-423. [PMID: 27392648 PMCID: PMC5003697 DOI: 10.1016/j.canlet.2016.07.001] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 12/20/2022]
Abstract
Excitement is growing for therapies that harness the power of patients' immune systems to combat their diseases. One approach to immunotherapy involves engineering patients' own T cells to express a chimeric antigen receptor (CAR) to treat advanced cancers, particularly those refractory to conventional therapeutic agents. Although these engineered immune cells have made remarkable strides in the treatment of patients with certain hematologic malignancies, success with solid tumors has been limited, probably due to immunosuppressive mechanisms in the tumor niche. In nearly all studies to date, T cells bearing αβ receptors have been used to generate CAR T cells. In this review, we highlight biological characteristics of γδ T cells that are distinct from those of αβ T cells, including homing to epithelial and mucosal tissues and unique functions such as direct antigen recognition, lack of alloreactivity, and ability to present antigens. We offer our perspective that these features make γδ T cells promising for use in cellular therapy against several types of solid tumors, including melanoma and gastrointestinal cancers. Engineered γδ T cells should be considered as a new platform for adoptive T cell cancer therapy for mucosal tumors.
Collapse
MESH Headings
- Animals
- Genes, T-Cell Receptor delta
- Genes, T-Cell Receptor gamma
- Genetic Therapy/methods
- Humans
- Immunotherapy, Adoptive/methods
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/transplantation
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Neoplasms/therapy
- Phenotype
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Tumor Microenvironment
Collapse
Affiliation(s)
- Hamid Reza Mirzaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sang Yun Lee
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jamshid Hadjati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Biology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
150
|
Huang JL, LaRocca CJ, Yamamoto M. Showing the Way: Oncolytic Adenoviruses as Chaperones of Immunostimulatory Adjuncts. Biomedicines 2016; 4:E23. [PMID: 28536390 PMCID: PMC5344254 DOI: 10.3390/biomedicines4030023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022] Open
Abstract
Oncolytic adenoviruses (OAds) are increasingly recognized as vectors for immunotherapy in the treatment of various solid tumors. The myriads of advantages of using adenovirus include targeted specificity upon infection and selective replication, which lead to localized viral burst, exponential spread of OAds, and antitumor effect. OAds can also induce a strong immune reaction due to the massive release of tumor antigens upon cytolysis and the presence of viral antigens. This review will highlight recent advances in adenoviral vectors expressing immunostimulatory effectors, such as GM-CSF (granulocyte macrophage colony-stimulating factor), interferon-α, interleukin-12, and CD40L. We will also discuss the combination of OAds with other immunotherapeutic strategies and describe the current understanding of how adenoviral vectors interact with the immune system to eliminate cancer cells.
Collapse
Affiliation(s)
- Jing Li Huang
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|