101
|
Stewart RA, Pilié PG, Yap TA. Development of PARP and Immune-Checkpoint Inhibitor Combinations. Cancer Res 2018; 78:6717-6725. [DOI: 10.1158/0008-5472.can-18-2652] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 09/25/2018] [Accepted: 10/10/2018] [Indexed: 11/16/2022]
|
102
|
The clinical role of the TME in solid cancer. Br J Cancer 2018; 120:45-53. [PMID: 30413828 PMCID: PMC6325164 DOI: 10.1038/s41416-018-0327-z] [Citation(s) in RCA: 397] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
The highly complex and heterogenous ecosystem of a tumour not only contains malignant cells, but also interacting cells from the host such as endothelial cells, stromal fibroblasts, and a variety of immune cells that control tumour growth and invasion. It is well established that anti-tumour immunity is a critical hurdle that must be overcome for tumours to initiate, grow and spread and that anti-tumour immunity can be modulated using current immunotherapies to achieve meaningful anti-tumour clinical responses. Pioneering studies in melanoma, ovarian and colorectal cancer have demonstrated that certain features of the tumour immune microenvironment (TME)-in particular, the degree of tumour infiltration by cytotoxic T cells-can predict a patient's clinical outcome. More recently, studies in renal cell cancer have highlighted the importance of assessing the phenotype of the infiltrating T cells to predict early relapse. Furthermore, intricate interactions with non-immune cellular players such as endothelial cells and fibroblasts modulate the clinical impact of immune cells in the TME. Here, we review the critical components of the TME in solid tumours and how they shape the immune cell contexture, and we summarise numerous studies evaluating its clinical significance from a prognostic and theranostic perspective.
Collapse
|
103
|
IDO expression in breast cancer: an assessment of 281 primary and metastatic cases with comparison to PD-L1. Mod Pathol 2018; 31:1513-1522. [PMID: 29802358 DOI: 10.1038/s41379-018-0061-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/30/2018] [Accepted: 03/30/2018] [Indexed: 02/06/2023]
Abstract
The immune inhibitory enzyme indoleamine 2,3-dioxygenase (IDO) has been associated with immune evasion in numerous malignancies and may mark these cancers as susceptible to anti-IDO therapies. We herein address IDO expression in breast cancers, examine the relationship between IDO and PD-L1, and investigate IDO fidelity across breast cancer primaries and metastases. IDO and PD-L1 expression was assessed in tissue microarrays containing 242 invasive primary breast cancers, 20 nodal metastases, and 19 distant metastases. IDO and PD-L1 were scored by extent in the tumor cells and immune infiltrate. Tumor IDO staining was seen in 14% of primaries including 38% of triple-negative cancers. IDO immune cell staining was seen in 14% of primaries and 29% of triple-negative cancers. Tumoral IDO and PD-L1 co-expression was seen in 8% of primaries, including 70% of tumoral PD-L1-positive cases. Immune IDO and PD-L1 co-expression was identified in 14% of primaries, including 48% of immune PD-L1-positive cases. Tumoral and immune cell IDO was conserved in 94% of matched primary/metastasis. In summary, IDO expression is common among high-grade, triple-negative breast cancers and is frequently associated with PD-L1 co-expression, suggesting that IDO might be a mechanism of anti-PD-1/PD-L1 immunotherapy resistance and that dual therapy may be of utility. Tumoral and immune cell IDO expression shows fidelity between primary and metastatic sites in treatment-naïve cancers, arguing against IDO as an independent driver for metastatic spread. Clinical trials are needed to assess the efficacy of IDO inhibition relative to IDO expression, as well as its possible role in combination with anti-PD-1/PD-L1 immunotherapy.
Collapse
|
104
|
Kather JN, Suarez-Carmona M, Charoentong P, Weis CA, Hirsch D, Bankhead P, Horning M, Ferber D, Kel I, Herpel E, Schott S, Zörnig I, Utikal J, Marx A, Gaiser T, Brenner H, Chang-Claude J, Hoffmeister M, Jäger D, Halama N. Topography of cancer-associated immune cells in human solid tumors. eLife 2018; 7:36967. [PMID: 30179157 PMCID: PMC6133554 DOI: 10.7554/elife.36967] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/30/2018] [Indexed: 12/14/2022] Open
Abstract
Lymphoid and myeloid cells are abundant in the tumor microenvironment, can be quantified by immunohistochemistry and shape the disease course of human solid tumors. Yet, there is no comprehensive understanding of spatial immune infiltration patterns (‘topography’) across cancer entities and across various immune cell types. In this study, we systematically measure the topography of multiple immune cell types in 965 histological tissue slides from N = 177 patients in a pan-cancer cohort. We provide a definition of inflamed (‘hot’), non-inflamed (‘cold’) and immune excluded patterns and investigate how these patterns differ between immune cell types and between cancer types. In an independent cohort of N = 287 colorectal cancer patients, we show that hot, cold and excluded topographies for effector lymphocytes (CD8) and tumor-associated macrophages (CD163) alone are not prognostic, but that a bivariate classification system can stratify patients. Our study adds evidence to consider immune topographies as biomarkers for patients with solid tumors.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany.,Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Aachen, Germany
| | - Meggy Suarez-Carmona
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Pornpimol Charoentong
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Cleo-Aron Weis
- Department of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniela Hirsch
- Department of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Peter Bankhead
- Northern Ireland Molecular Pathology Laboratory, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Northern Ireland, United Kingdom
| | - Marcel Horning
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Dyke Ferber
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Ivan Kel
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Tissue Bank of the National Center for Tumor Diseases, Heidelberg, Germany
| | - Sarah Schott
- Department of Gynecology, University Hospital Heidelberg, Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexander Marx
- Department of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Timo Gaiser
- Department of Pathology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Herrmann Brenner
- German Cancer Consortium, Heidelberg, Germany.,Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany.,Division of Preventive Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Centre, Heidelberg, Germany.,Cancer Epidemiology Group, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.,German Cancer Consortium, Heidelberg, Germany.,Applied Tumor Immunity, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
105
|
Liu Q, Tong D, Liu G, Gao J, Wang LA, Xu J, Yang X, Xie Q, Huang Y, Pang J, Wang L, He Y, Zhang D, Ma Q, Lan W, Jiang J. Metformin Inhibits Prostate Cancer Progression by Targeting Tumor-Associated Inflammatory Infiltration. Clin Cancer Res 2018; 24:5622-5634. [PMID: 30012567 DOI: 10.1158/1078-0432.ccr-18-0420] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/21/2018] [Accepted: 07/10/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Inflammatory infiltration plays important roles in both carcinogenesis and metastasis. We are interested in understanding the inhibitory mechanism of metformin on tumor-associated inflammation in prostate cancer.Experimental Design: By using a transgenic adenocarcinoma of the mouse prostate (TRAMP) mouse model, in vitro macrophage migration assays, and patient samples, we examined the effect of metformin on tumor-associated inflammation during the initiation and after androgen deprivation therapy of prostate cancer.Results: Treating TRAMP mice with metformin delays prostate cancer progression from low-grade prostatic intraepithelial neoplasia to high-grade PIN, undifferentiated to well-differentiated, and PIN to adenocarcinoma with concurrent inhibition of inflammatory infiltration evidenced by reduced recruitment of macrophages. Furthermore, metformin is capable of inhibiting the following processes: inflammatory infiltration after androgen deprivation therapy (ADT) induced by surgically castration in mice, bicalutamide treatment in patients, and hormone deprivation in LNCaP cells. Mechanistically, metformin represses inflammatory infiltration by downregulating both COX2 and PGE2 in tumor cells.Conclusions: Metformin is capable of repressing prostate cancer progression by inhibiting infiltration of tumor-associated macrophages, especially those induced by ADT, by inhibiting the COX2/PGE2 axis, suggesting that a combination of ADT with metformin could be a more efficient therapeutic strategy for prostate cancer treatment. Clin Cancer Res; 24(22); 5622-34. ©2018 AACR.
Collapse
Affiliation(s)
- Qiuli Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Dali Tong
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Gaolei Liu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jie Gao
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Lin-Ang Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jing Xu
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Xingxia Yang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Qiubo Xie
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Yiqiang Huang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Jian Pang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Luofu Wang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Yong He
- Department of Respiratory Medicine, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Qiang Ma
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China
| | - Weihua Lan
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China.
| | - Jun Jiang
- Department of Urology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, P. R. China.
| |
Collapse
|
106
|
Zinovkin DA, Pranjol MZI, Bilsky IA, Zmushko VA. Tumor-Associated T-Lymphocytes and Macrophages are Decreased in Endometrioid Endometrial Carcinoma with MELF-Pattern Stromal Changes. CANCER MICROENVIRONMENT 2018; 11:107-114. [PMID: 30008066 DOI: 10.1007/s12307-018-0213-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/12/2018] [Indexed: 11/26/2022]
Abstract
Microcystic, elongated, fragmented (MELF)-pattern is an unusual morphology of myometrial invasive front in endometrioid endometrial carcinoma (EA). The aim of the study was to investigate potential correlation between MELF-pattern and peritumoral inflammatory immune response. A total of 96 out of 368 patients were included in this study. CD3, CD20, CD57. CD68 and S100 markers were used for the detection of tumor-associated T-lymphocytes (TAT), tumor-associated B-lymphocytes (TAB), tumor-associated NK-lymphocytes (NK), tumor-associated macrophages and dendritic cells respectively. Mann-Whitney tests, receiver operating characteristic (ROC) curve analysis, and Spearman correlation were used as methods for statistical analyses. Odds ratio with 95% confidence interval (95% CI) was determined with the use of a logistic regression model. A p < 0.05 was considered statistically significant. Our results suggested that the number of CD3 and CD68 cells were significantly lower (p < 0.001) in cases of endometrioid carcinoma with MELF-pattern. A significant correlation between the presence of MELF-pattern and decrease of CD3 positive T-lymphocytes (r = 0.691; p < 0.001) was also observed. Additionally, we found an inverse correlation between the presence of MELF-pattern and TAM (r = 0.568; p = 0.001). Therefore, our data suggest that MELF-pattern may be associated with EA stroma fibrosis that contains immune cells infiltration and demonstrated a decrease in the number of TAT and TAM cells. This may indicate the poor clinical prognosis of this disease.
Collapse
Affiliation(s)
| | - Md Zahidul Islam Pranjol
- William Harvey Research Institute, Barts & The London School of Medicine & Dentistry Queen Mary University of London, London, UK
| | - Il'ya Andreevich Bilsky
- Department of Pathology, Gomel State Medical University, Lange str. 5, Gomel, 246029, Republic of Belarus
| | | |
Collapse
|
107
|
Lim SY, Rizos H. Immune cell profiling in the age of immune checkpoint inhibitors: implications for biomarker discovery and understanding of resistance mechanisms. Mamm Genome 2018; 29:866-878. [PMID: 29968076 PMCID: PMC6267680 DOI: 10.1007/s00335-018-9757-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/28/2018] [Indexed: 12/27/2022]
Abstract
Immunotherapy has changed the landscape of cancer treatment. The introduction of immune checkpoint inhibitors has seen tremendous success in improving overall survival of patients with advanced metastatic cancers and has now become the standard of care for multiple tumor types. However, efficacy of immune checkpoint blockade appears to be limited to immunogenic cancers, and even amongst immune-reactive cancers, response rates are low and variable between patients. Recent data have also demonstrated the rapid emergence of resistance to immune checkpoint inhibitors, with some patients progressing on treatment within one year. Significant research efforts are now directed at identifying predictive biomarkers and mechanisms of resistance to immune checkpoint blockade. These studies are underpinned by comprehensive and detailed profiling of the immune milieu. In this review, we discuss the utility and efficacy of immune cell profiling to uncover biomarkers of response and mechanisms of resistance to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Su Yin Lim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia. .,Melanoma Institute Australia, Sydney, NSW, Australia.
| | - Helen Rizos
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Melanoma Institute Australia, Sydney, NSW, Australia
| |
Collapse
|
108
|
Zou MX, Liu FS, Lv GH, Wang XB, Li J. Letter: Cranial Chordoma: A New Preoperative Grading System. Neurosurgery 2018; 83:E50-E51. [PMID: 29672759 DOI: 10.1093/neuros/nyy134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Ming-Xiang Zou
- Department of Spine Surgery The Second Xiangya Hospital Central South University Changsha, China
| | - Fu-Sheng Liu
- Department of Spine Surgery The Second Xiangya Hospital Central South University Changsha, China
| | - Guo-Hua Lv
- Department of Spine Surgery The Second Xiangya Hospital Central South University Changsha, China
| | - Xiao-Bin Wang
- Department of Spine Surgery The Second Xiangya Hospital Central South University Changsha, China
| | - Jing Li
- Department of Spine Surgery The Second Xiangya Hospital Central South University Changsha, China
| |
Collapse
|
109
|
Petitprez F, Vano YA, Becht E, Giraldo NA, de Reyniès A, Sautès-Fridman C, Fridman WH. Transcriptomic analysis of the tumor microenvironment to guide prognosis and immunotherapies. Cancer Immunol Immunother 2018; 67:981-988. [PMID: 28884365 PMCID: PMC11028160 DOI: 10.1007/s00262-017-2058-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 08/29/2017] [Indexed: 12/19/2022]
Abstract
Tumors are highly heterogeneous tissues where malignant cells are surrounded by and interact with a complex tumor microenvironment (TME), notably composed of a wide variety of immune cells, as well as vessels and fibroblasts. As the dialectical influence between tumor cells and their TME is known to be clinically crucial, we need tools that allow us to study the cellular composition of the microenvironment. In this focused research review, we report MCP-counter, a methodology based on transcriptomic markers that assesses the proportion of several immune and stromal cell populations in the TME from transcriptomic data, and we highlight how it can provide a way to decipher the complex mechanisms at play in tumors. In several malignancies, MCP-counter scores have been used to show various prognostic impacts of the TME, which we also show to be linked with the mutational burden of tumors. We also compared established molecular classifications of colorectal cancer and clear-cell renal cell carcinoma with the output of MCP-counter, and show that molecular subgroups have different TME profiles, and that these profiles are consistent within a given subgroup. Finally, we provide insights as to how knowing the TME composition may shape patient care in the near future.
Collapse
Affiliation(s)
- Florent Petitprez
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, 75006, Paris, France
- University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Eq. 13 escalier E 3e étage, 15 rue de l'école de médecine, 75006, Paris, France
- University UPMC Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Yann A Vano
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, 75006, Paris, France
- University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Eq. 13 escalier E 3e étage, 15 rue de l'école de médecine, 75006, Paris, France
- University UPMC Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Department of Medical Oncology, Georges Pompidou European Hospital, University Paris Descartes Paris 5, Paris, France
| | - Etienne Becht
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, 75006, Paris, France
- University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Eq. 13 escalier E 3e étage, 15 rue de l'école de médecine, 75006, Paris, France
- University UPMC Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Nicolas A Giraldo
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, 75006, Paris, France
- University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Eq. 13 escalier E 3e étage, 15 rue de l'école de médecine, 75006, Paris, France
- University UPMC Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, 75006, Paris, France
- University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Eq. 13 escalier E 3e étage, 15 rue de l'école de médecine, 75006, Paris, France
- University UPMC Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France
| | - Wolf H Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, 75006, Paris, France.
- University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Eq. 13 escalier E 3e étage, 15 rue de l'école de médecine, 75006, Paris, France.
- University UPMC Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, 75006, Paris, France.
| |
Collapse
|
110
|
Frey B, Rückert M, Deloch L, Rühle PF, Derer A, Fietkau R, Gaipl US. Immunomodulation by ionizing radiation-impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol Rev 2018; 280:231-248. [PMID: 29027224 DOI: 10.1111/imr.12572] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ionizing radiation is often regarded as an element of danger. But, danger responses on the cellular and molecular level are often beneficial with regard to the induction of anti-tumor immunity and for amelioration of inflammation. We outline how in dependence of radiation dose and fraction, radiation itself-and especially in combination with immune modulators-impacts on the innate and adaptive immune system. Focus is set on radiation-induced changes of the tumor cell phenotype and the cellular microenvironment including immunogenic cancer cell death. Mechanisms how anti-tumor immune responses are triggered by radiotherapy in combination with hyperthermia, inhibition of apoptosis, the adjuvant AnnexinA5, or vaccination with high hydrostatic pressure-killed autologous tumor cells are discussed. Building on this, feasible multimodal radio-immunotherapy concepts are reviewed including overcoming immune suppression by immune checkpoint inhibitors and by targeting TGF-β. Since radiation-induced tissue damage, inflammation, and anti-tumor immune responses are interconnected, the impact of lower doses of radiation on amelioration of inflammation is outlined. Closely meshed immune monitoring concepts based on the liquid biopsy blood are suggested for prognosis and prediction of cancer and non-cancer inflammatory diseases. Finally, challenges and visions for the design of cancer radio-immunotherapies and for treatment of benign inflammatory diseases are given.
Collapse
Affiliation(s)
- Benjamin Frey
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Michael Rückert
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa Deloch
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Paul F Rühle
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Anja Derer
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
111
|
Bloy N, Garcia P, Laumont CM, Pitt JM, Sistigu A, Stoll G, Yamazaki T, Bonneil E, Buqué A, Humeau J, Drijfhout JW, Meurice G, Walter S, Fritsche J, Weinschenk T, Rammensee HG, Melief C, Thibault P, Perreault C, Pol J, Zitvogel L, Senovilla L, Kroemer G. Immunogenic stress and death of cancer cells: Contribution of antigenicity vs adjuvanticity to immunosurveillance. Immunol Rev 2018; 280:165-174. [PMID: 29027230 DOI: 10.1111/imr.12582] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cancer cells are subjected to constant selection by the immune system, meaning that tumors that become clinically manifest have managed to subvert or hide from immunosurveillance. Immune control can be facilitated by induction of autophagy, as well as by polyploidization of cancer cells. While autophagy causes the release of ATP, a chemotactic signal for myeloid cells, polyploidization can trigger endoplasmic reticulum stress with consequent exposure of the "eat-me" signal calreticulin on the cell surface, thereby facilitating the transfer of tumor antigens into dendritic cells. Hence, both autophagy and polyploidization cause the emission of adjuvant signals that ultimately elicit immune control by CD8+ T lymphocytes. We investigated the possibility that autophagy and polyploidization might also affect the antigenicity of cancer cells by altering the immunopeptidome. Mass spectrometry led to the identification of peptides that were presented on major histocompatibility complex (MHC) class I molecules in an autophagy-dependent fashion or that were specifically exposed on the surface of polyploid cells, yet lost upon passage of such cells through immunocompetent (but not immunodeficient) mice. However, the preferential recognition of autophagy-competent and polyploid cells by the innate and cellular immune systems did not correlate with the preferential recognition of such peptides in vivo. Moreover, vaccination with such peptides was unable to elicit tumor growth-inhibitory responses in vivo. We conclude that autophagy and polyploidy increase the immunogenicity of cancer cells mostly by affecting their adjuvanticity rather than their antigenicity.
Collapse
Affiliation(s)
- Norma Bloy
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Institut Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Pauline Garcia
- Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Institut Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Céline M Laumont
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Jonathan M Pitt
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France
| | - Antonella Sistigu
- Unit of Tumor Immunology and Immunotherapy, Department of Research, Advanced Diagnostics and Technological Innovation, Regina Elena National Cancer Institute, Rome, Italy
| | - Gautier Stoll
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Takahiro Yamazaki
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France
| | - Eric Bonneil
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec, Canada
| | - Aitziber Buqué
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Institut Gustave Roussy Cancer Campus, Villejuif, France
| | - Juliette Humeau
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Institut Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Medicine, University of Paris Sud, Kremlin-Bicêtre, France
| | - Jan W Drijfhout
- Department of Immunohematology and Blood Transfusion, Leiden University, Leiden, The Netherlands.,Medical Center, Leiden, The Netherlands
| | - Guillaume Meurice
- Bioinformatic Core Facility, UMS AMMICA, INSERM US23, CNRS UMS3665, Gustave Roussy, Villejuif, France
| | | | | | - Toni Weinschenk
- Immatics US, Houston, TX, USA.,Immatics Biotechnologies, Tübingen, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | | | - Pierre Thibault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec, Canada.,Department of Chemistry, Faculty of Arts and Sciences, Université de Montréal, Montreal, Quebec, Canada
| | - Claude Perreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, Quebec, Canada.,Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Division of Hematology, Hôpital Maisonneuve-Rosemont, Montreal, Quebec, Canada
| | - Jonathan Pol
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Institut Gustave Roussy Cancer Campus, Villejuif, France
| | - Laurence Zitvogel
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1015, Equipe Labellisée Ligue Nationale Contre le Cancer, Gustave Roussy Cancer Campus, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Laura Senovilla
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Guido Kroemer
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale, U1138, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
112
|
Shi Y, Deng L, Song Y, Lin D, Lai Y, Zhou L, Yang L, Li X. CD3+/CD8+ T-cell density and tumoral PD-L1 predict survival irrespective of rituximab treatment in Chinese diffuse large B-cell lymphoma patients. Int J Hematol 2018; 108:254-266. [DOI: 10.1007/s12185-018-2466-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/25/2018] [Accepted: 04/25/2018] [Indexed: 10/16/2022]
|
113
|
Huang SY, Lin HH, Lin CW, Li CC, Yao M, Tang JL, Hou HA, Tsay W, Chou SJ, Cheng CL, Tien HF. Soluble PD-L1: A biomarker to predict progression of autologous transplantation in patients with multiple myeloma. Oncotarget 2018; 7:62490-62502. [PMID: 27566569 PMCID: PMC5308741 DOI: 10.18632/oncotarget.11519] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/11/2016] [Indexed: 12/31/2022] Open
Abstract
Autologous hematopoietic stem cell transplantation (AuHSCT) is standard in treating eligible multiple myeloma (MM) patients. However, the outcome after treatment is highly variable. We used ELISA to analyze the levels of soluble PD-L1 (suPD-L1) in bone marrow (BM) plasma from 61 patients with MM at 100 days after AuHSCT. Patients were classified into high (H) and normal-to-low (NL) groups depending on their suPD-L1 levels. Among patients who had a very good partial response (VGPR) or better after AuHSCT, those in the H-group had a shorter response period (RpSCT) as well as shorter overall survival (OS) than those in the NL-group. Multivariate analyses confirmed that a high suPD-L1 level and high-risk cytogenetic abnormalities are independent factors for RpSCT. Our data suggest that suPD-L1 in the BM plasma of MM patients who have VGPR or better after AuHSCT could be used as a biomarker to predict outcome.
Collapse
Affiliation(s)
- Shang-Yi Huang
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Hsiu-Hsia Lin
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Chung-Wu Lin
- Department of Pathology, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Chi-Cheng Li
- Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Ming Yao
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Jih-Luh Tang
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan.,Tai-Cheng Stem Cell Therapy Center, National Taiwan University, Taipei, Taiwan
| | - Hsin-An Hou
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Woei Tsay
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Sheng-Je Chou
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Chieh-Lung Cheng
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| | - Hwei-Fang Tien
- Department of Internal Medicine, National Taiwan University, Medical College and Hospital, Taipei, Taiwan
| |
Collapse
|
114
|
Immune-based identification of cancer patients at high risk of progression. Curr Opin Immunol 2018; 51:97-102. [PMID: 29554496 DOI: 10.1016/j.coi.2018.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 01/29/2018] [Accepted: 03/01/2018] [Indexed: 12/20/2022]
Abstract
Tumors are highly heterogeneous structures where malignant cells interact with a large variety of cell populations, including a clinically-relevant immune component. We review and compare the most recent methods designed to analyze and quantify the composition of immune and stromal microenvironment of tumors and discuss their use in identification of patients for high risk of progression. If the impact of the various immune components on patient's relapse share common rules in most malignancies, clear cell renal cell tumors behave differently with regards to immunity. We focus on this specific pathology to show how the tumor interacts with the host's immune system and how this intricate relationship shapes the clinical outcome.
Collapse
|
115
|
Kather JN, Halama N, Jaeger D. Genomics and emerging biomarkers for immunotherapy of colorectal cancer. Semin Cancer Biol 2018; 52:189-197. [PMID: 29501787 DOI: 10.1016/j.semcancer.2018.02.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/19/2018] [Accepted: 02/28/2018] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is a common and lethal disease with a high therapeutic need. For most patients with metastatic CRC, chemotherapy is the only viable option. Currently, immunotherapy is restricted to the particular genetic subgroup of mismatch-repair deficient (MMRd)/microsatellite instable (MSI) CRC. Anti-PD1 therapy was recently FDA-approved as a second-line treatment in this subgroup. However, in a metastatic setting, these MMRd/MSI tumors are vastly outnumbered by mismatch-repair proficient (MMRp)/microsatellite stable (MSS) tumors. These MMRp/MSS tumors do not meaningfully respond to any traditional immunotherapy approach including checkpoint blockade, adoptive cell transfer and vaccination. This resistance to immunotherapy is due to a complex tumor microenvironment that counteracts antitumor immunity through a combination of poorly antigenic tumor cells and an immunosuppressive tumor microenvironment. To find ways of overcoming immunotherapy resistance in the majority of CRC patients, it is necessary to analyze the immunological makeup in an in-depth and personalized way and in the context of their tumor genetic makeup. Flexible, biomarker-guided early-phase immunotherapy trials are needed to optimize this workflow. In this review, we detail key mechanisms for immune evasion and emerging immune biomarkers for personalized immunotherapy in CRC. Also, we present a template for biomarker-guided clinical trials that are needed to move new immunotherapy approaches closer to clinical application.
Collapse
Affiliation(s)
- Jakob Nikolas Kather
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Niels Halama
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Dirk Jaeger
- Department of Medical Oncology and Internal Medicine VI, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
116
|
Elia AR, Grioni M, Basso V, Curnis F, Freschi M, Corti A, Mondino A, Bellone M. Targeting Tumor Vasculature with TNF Leads Effector T Cells to the Tumor and Enhances Therapeutic Efficacy of Immune Checkpoint Blockers in Combination with Adoptive Cell Therapy. Clin Cancer Res 2018; 24:2171-2181. [DOI: 10.1158/1078-0432.ccr-17-2210] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 01/16/2018] [Accepted: 02/23/2018] [Indexed: 11/16/2022]
|
117
|
Berinstein NL, McNamara M, Nguyen A, Egan J, Wolf GT. Increased immune infiltration and chemokine receptor expression in head and neck epithelial tumors after neoadjuvant immunotherapy with the IRX-2 regimen. Oncoimmunology 2018; 7:e1423173. [PMID: 29721379 PMCID: PMC5927542 DOI: 10.1080/2162402x.2017.1423173] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/21/2017] [Accepted: 12/22/2017] [Indexed: 01/07/2023] Open
Abstract
IRX-2 is an injectable cancer immunotherapy composed of cytokines purified from stimulated normal-donor peripheral blood mononuclear cells. In a phase 2a trial (n = 27), neoadjuvant IRX-2 significantly increased lymphocyte infiltration (LI) into resected head and neck tumors and was associated with changes in fibrosis and necrosis. Event-free survival was 65% at 2 years, and overall survival 65% at 5 years. Overall survival was longer for patients with LI greater versus lower than the median. This substudy of the mechanisms responsible for the increase in LI with neoadjuvant IRX-2 employed multiplex immunohistochemistry (IHC) and transcriptome analysis to interrogate matched pre- and post-treatment tumor specimens from 7 available phase 2a trial patients. Multiplex IHC showed substantial increases in CD68-expressing cells (5 patients), T-cell density (4 patients), and PDL1 mean fluorescent intensity (4 patients). Consistent with IRX-2 activation of multiple immune cells, transcriptome analysis showed mean increases in expression of genes associated with NK cells, B cells, CD4+ T cells, CD8+ T cells, and dendritic cells, but not of genes associated with neutrophils. There were increases in mean expression of genes for most immune subsets, most markedly (2- to 3-fold) for B cells and dendritic cells. Mean increases in gene expression for chemokines suggest that tumor LI may be driven in part by IRX-2-induced production of chemo-attractants. Upregulation of checkpoint genes including PDL1 and CTLA4 along with increased T-cell infiltration suggests a functional antitumor immune response such that the efficacy of IRX-2 may be enhanced by combination with immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Michael McNamara
- Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon, USA
| | - Ariane Nguyen
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - James Egan
- IRX Therapeutics, New York, New York, USA
| | - Gregory T Wolf
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
118
|
Abstract
Harnessing the power of the human immune system to treat cancer is the essence of immunotherapy. Monoclonal antibodies engage the innate immune system to destroy targeted cells. For the last 30years, antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity have been the main mechanisms of anti-tumor action of unconjugated antibody drugs. Efforts to exploit the potentials of other immune cells, in particular T cells, culminated in the recent approval of two T cell engaging bispecific antibody (T-BsAb) drugs, thereby stimulating new efforts to accelerate similar platforms through preclinical and clinical trials. In this review, we have compiled the worldwide effort in exploring T cell engaging bispecific antibodies. Our special emphasis is on the lessons learned, with the hope to derive insights in this fast evolving field with tremendous clinical potential.
Collapse
Affiliation(s)
- Z Wu
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - N V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States.
| |
Collapse
|
119
|
Chen M, Zheng SH, Yang M, Chen ZH, Li ST. The diagnostic value of preoperative inflammatory markers in craniopharyngioma: a multicenter cohort study. J Neurooncol 2018; 138:113-122. [PMID: 29388032 DOI: 10.1007/s11060-018-2776-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/22/2018] [Indexed: 02/06/2023]
Abstract
To compare the different levels of preoperative inflammatory markers in peripheral blood samples between craniopharyngioma (CP) and other sellar region tumors so as to explore their differential diagnostic value. The level of white blood cell (WBC), neutrophil, lymphocyte, monocyte, platelet, albumin, neutrophil lymphocyte ratio (NLR), derived NLR (dNLR), platelet lymphocyte ratio (PLR), monocyte lymphocyte ratio (MLR) and prognostic nutritional index (PNI) were compared between the CP and other sellar region tumors. A receiver operating characteristics (ROC) curve analysis was performed to evaluate the diagnostic significance of the peripheral blood inflammatory markers and their paired combinations for CP including its pathological types. Patients with CP had higher levels of pre-operative WBC, lymphocyte and PNI. The papillary craniopharyngioma (PCP) group had higher neutrophil count and NLR than the adamantinomatous craniopharyngioma (ACP) and healthy control groups whereas the ACP group had higher platelet count and PNI than the PCP and healthy control groups. There were not any significant differences in preoperative inflammatory markers between the primary and recurrent CP groups. The AUC values of WBC, neutrophil, NLR + PLR and dNLR + PLR in PCP were all higher than 0.7. Inflammation seems to be closely correlated with CP's development. The preoperative inflammatory markers including WBC, neutrophil, NLR + PLR and dNLR + PLR may differentially diagnose PCP, pituitary tumor (PT) and Rathke cleft cyst (RCC). In addition, some statistical results in this study indirectly proved previous experimental conclusions and strictly matched CP's biological features.
Collapse
Affiliation(s)
- Ming Chen
- Department of Neurosurgery, XinHua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Shi-Hao Zheng
- Department of Neurosurgery, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Min Yang
- Department of Neurosurgery, XinHua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China
| | - Zhi-Hua Chen
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Shi-Ting Li
- Department of Neurosurgery, XinHua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200092, China.
| |
Collapse
|
120
|
Newell EW, Becht E. High-Dimensional Profiling of Tumor-Specific Immune Responses: Asking T Cells about What They “See” in Cancer. Cancer Immunol Res 2018; 6:2-9. [DOI: 10.1158/2326-6066.cir-17-0519] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/18/2017] [Accepted: 12/04/2017] [Indexed: 11/16/2022]
|
121
|
|
122
|
|
123
|
Diagnostic Efficiency in Digital Pathology: A Comparison of Optical Versus Digital Assessment in 510 Surgical Pathology Cases. Am J Surg Pathol 2017; 42:53-59. [PMID: 28877052 DOI: 10.1097/pas.0000000000000930] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prior work has shown that digital images and microscopic slides can be interpreted with comparable diagnostic accuracy. Although accuracy has been well-validated, the interpretative time for digital images has scarcely been studied and concerns about efficiency remain a major barrier to adoption. We investigated the efficiency of digital pathology when compared with glass slide interpretation in the diagnosis of surgical pathology biopsy and resection specimens. Slides were pulled from 510 surgical pathology cases from 5 organ systems (gastrointestinal, gynecologic, liver, bladder, and brain). Original diagnoses were independently confirmed by 2 validating pathologists. Diagnostic slides were scanned using the Philips IntelliSite Pathology Solution. Each case was assessed independently on digital and optical by 3 reading pathologists, with a ≥6 week washout period between modalities. Reading pathologists recorded assessment times for each modality; digital times included time to load the case. Diagnostic accuracy was determined based on whether a rendered diagnosis differed significantly from the original diagnosis. Statistical analysis was performed to assess for differences in interpretative times across modalities. All 3 reading pathologists showed comparable diagnostic accuracy across optical and digital modalities (mean major discordance rates with original diagnosis: 4.8% vs. 4.4%, respectively). Mean assessment times ranged from 1.2 to 9.1 seconds slower on digital versus optical. The slowest reader showed a significant learning effect during the course of the study so that digital assessment times decreased over time and were comparable with optical times by the end of the series. Organ site and specimen type did not significantly influence differences in interpretative times. In summary, digital image reading times compare favorably relative to glass slides across a variety of organ systems and specimen types. Mean increase in assessment time is 4 seconds/case. This time can be minimized with experience and may be further balanced by the improved ease of electronic chart access allowed by digital slide viewing, as well as quantitative assessments which can be expedited on digital images.
Collapse
|
124
|
Mismatch repair status and PD-L1 expression in clear cell carcinomas of the ovary and endometrium. Mod Pathol 2017; 30:1622-1632. [PMID: 28752845 DOI: 10.1038/modpathol.2017.67] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/02/2017] [Accepted: 05/06/2017] [Indexed: 12/17/2022]
Abstract
Clear cell carcinoma represents a distinct histologic type of müllerian carcinoma that is resistant to conventional chemotherapy. Expression of programmed cell death ligand (PD-L1) has been associated with immune evasion in numerous tumor types and may be used to identify patients who will benefit from targeted immunotherapy, particularly in the setting of mismatch repair defects. We evaluated PD-L1 expression in 23 ovarian clear cell carcinomas and 21 endometrial clear cell carcinomas, and correlated expression with mismatch repair status. Tumor PD-L1 staining was seen in 43% of ovarian tumors and 76% of endometrial tumors, including 71% of cases (67% of ovarian and 75% of endometrial) with mismatch repair defects. Extensive tumoral staining (>50%) was seen in only one case (an endometrial case with MSH6 loss). However, tumoral PD-L1 expression remained common in mismatch repair-intact tumors and mismatch repair status was not significantly correlated with PD-L1 expression. The increased incidence of PD-L1 positivity in tumor cells (P=0.04) in endometrial vs ovarian clear cell carcinomas suggests differences in the tumor microenvironment of these histologically and molecularly similar tumors that may inform treatment options. These results suggest that clear cell histology may be a useful susceptibility marker for immunotherapy targeting the PD-1/PD-L1 axis irrespective of mismatch repair status, particularly in endometrial carcinomas.
Collapse
|
125
|
Welters MJP, Ma W, Santegoets SJAM, Goedemans R, Ehsan I, Jordanova ES, van Ham VJ, van Unen V, Koning F, van Egmond SI, Charoentong P, Trajanoski Z, van der Velden LA, van der Burg SH. Intratumoral HPV16-Specific T Cells Constitute a Type I-Oriented Tumor Microenvironment to Improve Survival in HPV16-Driven Oropharyngeal Cancer. Clin Cancer Res 2017; 24:634-647. [PMID: 29018052 DOI: 10.1158/1078-0432.ccr-17-2140] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Human papillomavirus (HPV)-associated oropharyngeal squamous cell cancer (OPSCC) has a much better prognosis than HPV-negative OPSCC, and this is linked to dense tumor immune infiltration. As the viral antigens may trigger potent immunity, we studied the relationship between the presence of intratumoral HPV-specific T-cell responses, the immune contexture in the tumor microenvironment, and clinical outcome.Experimental Design: To this purpose, an in-depth analysis of tumor-infiltrating immune cells in a prospective cohort of 97 patients with HPV16-positive and HPV16-negative OPSCC was performed using functional T-cell assays, mass cytometry (CyTOF), flow cytometry, and fluorescent immunostaining of tumor tissues. Key findings were validated in a cohort of 75 patients with HPV16-positive OPSCC present in the publicly available The Cancer Genome Atlas database.Results: In 64% of the HPV16-positive tumors, type I HPV16-specific T cells were present. Their presence was not only strongly related to a better overall survival, a smaller tumor size, and less lymph node metastases but also to a type I-oriented tumor microenvironment, including high numbers of activated CD161+ T cells, CD103+ tissue-resident T cells, dendritic cells (DC), and DC-like macrophages.Conclusions: The viral antigens trigger a tumor-specific T-cell response that shapes a favorable immune contexture for the response to standard therapy. Hence, reinforcement of HPV16-specific T-cell reactivity is expected to boost this process. Clin Cancer Res; 24(3); 634-47. ©2017 AACRSee related commentary by Laban and Hoffmann, p. 505.
Collapse
Affiliation(s)
- Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Wenbo Ma
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Renske Goedemans
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ilina Ehsan
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Vanessa J van Ham
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Vincent van Unen
- Department of Immunohematology and Blood Bank, Leiden University Medical Center, Leiden, the Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Bank, Leiden University Medical Center, Leiden, the Netherlands
| | - Sylvia I van Egmond
- Department of Otorhinolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Zlatko Trajanoski
- Division for Bioinformatics, Innsbruck Medical University, Innsbruck, Austria
| | - Lilly-Ann van der Velden
- Department of Otorhinolaryngology and Head and Neck Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
126
|
Zhang X, Liu Q, Liao Q, Zhao Y. Potential Roles of Peripheral Dopamine in Tumor Immunity. J Cancer 2017; 8:2966-2973. [PMID: 28928888 PMCID: PMC5604448 DOI: 10.7150/jca.20850] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Recent years, immunotherapy has turned out to be a promising strategy against tumors. Peripheral dopamine (DA) has important roles in immune system among tumor patients. Accumulated reports demonstrate variable expression and distribution of DA receptors (DRs) in diverse immune cells. Interestingly, peripheral DA also involves in tumor progression and it exerts anticancer effects on immunomodulation, which includes inflammasomes in cancer, function of immune effector cells, such as T lymphocytes, myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) and natural killer (NK) cells. Given the specific immunologic status, DA medication may be a valuable candidate in pancreatic cancer treatment. The major purpose of this review is to discuss the novel potential interactions between peripheral dopamine and tumor immunity.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Qiaofei Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Quan Liao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
127
|
Ding G, Li W, Liu J, Zeng Y, Mao C, Kang Y, Shang J. LncRNA GHET1 activated by H3K27 acetylation promotes cell tumorigenesis through regulating ATF1 in hepatocellular carcinoma. Biomed Pharmacother 2017; 94:326-331. [PMID: 28772210 DOI: 10.1016/j.biopha.2017.07.046] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/26/2017] [Accepted: 07/09/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND GHET1 is one of tumor-related lncRNAs. We aimed to explore the functional involvement of GHET1 in hepatocellular carcinoma (HCC). METHODS In this study, HCC tissues and the paired normal tissues were collected for the detection of target molecules. The expression level of target molecules in HCC tissues or cell lines was determined by qRT-PCR and western blot, respectively. The expression of endogenous GHET1 and ATF1 was modulated by using cell transfection. RNA pull down assay was performed to examine the interaction between GHET1 and ATF1. ChIP assay was conducted to determine the H3K27Ac acetylation of GHET1 promoter. RESULTS H3K27 acetylation activated-GHET1 was upregulated in HCC tissues and cell lines. Moreover, GHET1 silencing could inhibit the proliferation, migration, invasion and EMT of HCC cells in vitro. GHET1 could regulate the expression of ATF1 mRNA and protein; RNA pull-down assays supported that GHET1 could bind to ATF1 protein. Furthermore, overexpression of ATF1 almost completely reversed the GHET1 knockdown mediated inhibition on the proliferation, migration, invasion and EMT of HCC cells. CONCLUSION LncRNA GHET1 was intimately involved in the occurrence and development of HCC through regulating ATF1.
Collapse
Affiliation(s)
- Gangqiang Ding
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China.
| | - Wei Li
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Junping Liu
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Yanli Zeng
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Chongshan Mao
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Yi Kang
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Jia Shang
- Department of Infectious Diseases, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| |
Collapse
|
128
|
Abstract
Immunotherapy is currently the most rapidly advancing area of clinical oncology, and provides the unprecedented opportunity to effectively treat, and even cure, several previously untreatable malignancies. A growing awareness exists of the fact that the success of chemotherapy and radiotherapy, in which the patient's disease can be stabilized well beyond discontinuation of treatment (and occasionally is cured), also relies on the induction of a durable anticancer immune response. Indeed, the local immune infiltrate undergoes dynamic changes that accompany a shift from a pre-existing immune response to a therapy-induced immune response. As a result, the immune contexture, which is determined by the density, composition, functional state and organization of the leukocyte infiltrate of the tumour, can yield information that is relevant to prognosis, prediction of a treatment response and various other pharmacodynamic parameters. Several complementary technologies can be used to explore the immune contexture of tumours, and to derive biomarkers that could enable the adaptation of individual treatment approaches for each patient, as well as monitoring a response to anticancer therapies.
Collapse
|
129
|
PD-L1 Expression in Mismatch Repair-deficient Endometrial Carcinomas, Including Lynch Syndrome-associated and MLH1 Promoter Hypermethylated Tumors. Am J Surg Pathol 2017; 41:326-333. [PMID: 27984238 DOI: 10.1097/pas.0000000000000783] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mismatch repair (MMR)-deficient endometrial carcinomas (ECs) bearing Lynch syndrome (LS)-associated germline mutations or sporadic MLH1 promoter hypermethylation (MLH1hm) are highly immunogenic and may represent excellent candidates for therapies targeting the programmed cell death (PD)/programmed cell death ligand-1 (PD-L1) immune checkpoint pathway. This study evaluates PD-L1 expression in MMR-deficient ECs including LS-associated and MLH1hm cases, in comparison with MMR-intact tumors. Immunohistochemistry for PD-L1/CD274 was performed on 38 MMR-deficient and 29 MMR-intact ECs. Staining was scored in the tumor and the peritumoral immune compartment. The majority of MMR-deficient tumors were PD-L1 positive (53%) in at least a subset of tumor cells. LS-associated tumors were more likely to be PD-L1 positive relative to MLH1hm tumors (70% vs. 33%, P=0.05). Only 10% of MMR-intact ECs demonstrated any tumoral PD-L1 expression; this was significantly lower than was observed in MMR-deficient tumors (P=0.0005). When reviewed by histologic grade, PD-L1 expression remained highest in LS-associated ECs followed by MLH1hm and MMR-intact carcinomas, respectively. The MMR immunohistochemical pattern most uniformly associated with PD-L1 expression was MSH6 loss. Immune PD-L1 expression was seen in 100% of MMR-deficient and 66% of MMR-intact cases. This study represents the first to characterize differences in PD-L1 expression between LS-associated and MLH1hm endometrial cancers. It demonstrates that tumoral PD-L1 expression is more common in LS-associated endometrial cancers relative to MLH1hm and MMR-intact tumors, although sporadic cancers often show PD-L1 positive immune staining. These data suggest that MMR deficiency may be a better predictor of response to PD-1/PD-L1 inhibitor therapy than tumor grade in EC, and that potential benefit may vary based on the molecular mechanism of MMR defects.
Collapse
|
130
|
Suarez‐Carmona M, Lesage J, Cataldo D, Gilles C. EMT and inflammation: inseparable actors of cancer progression. Mol Oncol 2017; 11:805-823. [PMID: 28599100 PMCID: PMC5496491 DOI: 10.1002/1878-0261.12095] [Citation(s) in RCA: 385] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/14/2022] Open
Abstract
Tumors can be depicted as wounds that never heal, and are infiltrated by a large array of inflammatory and immune cells. Tumor-associated chronic inflammation is a hallmark of cancer that fosters progression to a metastatic stage, as has been extensively reviewed lately. Indeed, inflammatory cells persisting in the tumor establish a cross-talk with tumor cells that may result in a phenotype switch into tumor-supporting cells. This has been particularly well described for macrophages and is referred to as tumor-associated 'M2' polarization. Epithelial-to-mesenchymal transition (EMT), the embryonic program that loosens cell-cell adherence complexes and endows cells with enhanced migratory and invasive properties, can be co-opted by cancer cells during metastatic progression. Cancer cells that have undergone EMT are more aggressive, displaying increased invasiveness, stem-like features, and resistance to apoptosis. EMT programs can also stimulate the production of proinflammatory factors by cancer cells. Conversely, inflammation is a potent inducer of EMT in tumors. Therefore, the two phenomena may sustain each other, in an alliance for metastasis. This is the focus of this review, where the interconnections between EMT programs and cellular and molecular actors of inflammation are described. We also recapitulate data linking the EMT/inflammation axis to metastasis.
Collapse
Affiliation(s)
- Meggy Suarez‐Carmona
- National Center for Tumor Diseases (NCT) – University Hospital HeidelbergGermany
| | - Julien Lesage
- Laboratory of Tumor and Development BiologyGIGA‐Cancer University of LiègeBelgium
| | - Didier Cataldo
- Inserm UMR‐S 903SFR CAP‐SantéUniversity of Reims Champagne‐Ardenne (URCA)France
| | - Christine Gilles
- Inserm UMR‐S 903SFR CAP‐SantéUniversity of Reims Champagne‐Ardenne (URCA)France
| |
Collapse
|
131
|
Petitprez F, Fossati N, Vano Y, Freschi M, Becht E, Lucianò R, Calderaro J, Guédet T, Lacroix L, Rancoita PMV, Montorsi F, Fridman WH, Sautès-Fridman C, Briganti A, Doglioni C, Bellone M. PD-L1 Expression and CD8 + T-cell Infiltrate are Associated with Clinical Progression in Patients with Node-positive Prostate Cancer. Eur Urol Focus 2017; 5:192-196. [PMID: 28753812 DOI: 10.1016/j.euf.2017.05.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 05/25/2017] [Indexed: 10/19/2022]
Abstract
Prostate cancer (PCa) patients with lymph node invasion at radical prostatectomy are at higher risk of tumor recurrence and receive immediate androgen deprivation therapy (ADT). While approximately 30% of these patients do not experience recurrence, others experience disease recurrence despite ADT, and currently no biomarkers can accurately identify them. We analyzed tumors from 51 patients with node-positive prostate cancer using immunohistochemistry to investigate whether expression of the immune checkpoint ligand PD-L1 by tumor cells or the density of CD8+ or CD20+ cells are associated with clinical progression. Patients with at least 1% PD-L1+ tumor cells had shorter metastasis-free survival than those with PD-L1- tumors (p=0.008, log-rank test). Univariate Cox regression showed that patients with PD-L1+ tumors had almost four times the risk of experiencing distant metastases than those with PD-L1- tumors (hazard ratio 3.90). In addition, we found that PD-L1 expression was significantly associated with CD8+ T-cell density, but not with CD20+ B-cell density. While these results need to be confirmed in larger studies, they show that PD-L1 and CD8 may be used as biomarkers for node-positive patients at high risk of progression. The study also provides a rationale for selecting patients with node-positive PCa who might benefit the most from adjuvant immunotherapies. PATIENT SUMMARY: None of the available biomarkers can identify node-positive prostate cancer that will recur after surgery. We found that expression of PD-L1 by tumor cells and a high density of CD8+ T cells in tumor are associated with a higher risk of clinical progression in men with node-positive prostate cancer.
Collapse
Affiliation(s)
- Florent Petitprez
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Nicola Fossati
- Unit of Urology and URI, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Yann Vano
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; Department of Medical Oncology, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Massimo Freschi
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Unit of Pathology, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Etienne Becht
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Roberta Lucianò
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Unit of Pathology, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Julien Calderaro
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Tiffany Guédet
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Laetitia Lacroix
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Paola M V Rancoita
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Montorsi
- Unit of Urology and URI, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Paris Descartes Paris 5, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France; UPMC University Paris 6, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Alberto Briganti
- Unit of Urology and URI, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Claudio Doglioni
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Unit of Pathology, Division of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Matteo Bellone
- NET-IMPACT, IRCCS Ospedale San Raffaele, Milan, Italy; Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS Ospedale San Raffaele, Milan, Italy.
| |
Collapse
|
132
|
Bellone M, Elia AR. Constitutive and acquired mechanisms of resistance to immune checkpoint blockade in human cancer. Cytokine Growth Factor Rev 2017; 36:17-24. [PMID: 28606732 DOI: 10.1016/j.cytogfr.2017.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 06/01/2017] [Indexed: 12/16/2022]
Abstract
Cancer immunotherapy with monoclonal antibodies directed against regulatory pathways in T lymphocytes has been revolutionizing medical oncology, and the clinical success of monoclonal antibodies targeting either cytotoxic T lymphocyte antigen-4 (CTLA-4) or program death-1 (PD-1) in patients affected by melanoma, Hodgkin's lymphoma, Merkel cell carcinoma, and head and neck, bladder, renal cell or non-small cell lung cancer is way beyond the most optimistic expectation. However, immune checkpoint blockade (ICB) has failed to arrest progression in a consistent amount of patients affected by those tumors, and various histological types, including breast, colon and prostate cancer, are less sensitive to this therapeutic approach. Such clinical findings have fueled massive research efforts in the attempt to identify pre-existing and acquired mechanisms of resistance to ICB. Here we focus on evidences emerging from studies in humans on how tumor cells and the tumor microenvironment contribute to the heterogeneous clinical responses, and we propose strategies stemming from pre-clinical models that might improve clinical outcomes for patients.
Collapse
|
133
|
Gross S, Erdmann M, Haendle I, Voland S, Berger T, Schultz E, Strasser E, Dankerl P, Janka R, Schliep S, Heinzerling L, Sotlar K, Coulie P, Schuler G, Schuler-Thurner B. Twelve-year survival and immune correlates in dendritic cell-vaccinated melanoma patients. JCI Insight 2017; 2:91438. [PMID: 28422751 DOI: 10.1172/jci.insight.91438] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 03/02/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Reports on long-term (≥10 years) effects of cancer vaccines are missing. Therefore, in 2002, we initiated a phase I/II trial in cutaneous melanoma patients to further explore the immunogenicity of our DC vaccine and to establish its long-term toxicity and clinical benefit after a planned 10-year followup. METHODS Monocyte-derived DCs matured by TNFα, IL-1β, IL-6, and PGE2 and then loaded with 4 HLA class I and 6 class II-restricted tumor peptides were injected intradermally in high doses over 2 years. We performed serial immunomonitoring in all 53 evaluable patients. RESULTS Vaccine-specific immune responses including high-affinity, IFNγ-producing CD4+ and lytic polyfunctional CD8+ T cells were de novo induced or boosted in most patients. Exposure of mature DCs to trimeric soluble CD40 ligand, unexpectedly, did not further enhance such immune responses, while keyhole limpet hemocyanin (KLH) pulsing to provide unspecific CD4+ help promoted CD8+ T cell responses - notably, their longevity. An unexpected 19% of nonresectable metastatic melanoma patients are still alive after 11 years, a survival rate similar to that observed in ipilimumab-treated patients and achieved without any major (>grade 2) toxicity. Survival correlated significantly with the development of intense vaccine injection site reactions, and with blood eosinophilia after the first series of vaccinations, suggesting that prolonged survival was a consequence of DC vaccination. CONCLUSIONS Long-term survival in advanced melanoma patients undergoing DC vaccination is similar to ipilimumab-treated patients and occurs upon induction of tumor-specific T cells, blood eosinophilia, and strong vaccine injection site reactions occurring after the initial vaccinations. TRIAL REGISTRATION ClinicalTrials.gov NCT00053391. FUNDING European Community, Sixth Framework Programme (Cancerimmunotherapy LSHC-CT-2006-518234; DC-THERA LSHB-CT-2004-512074), and German Research Foundation (CRC 643, C1, Z2).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Peter Dankerl
- Department of Radiology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - Rolf Janka
- Department of Radiology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | | | | | - Karl Sotlar
- Institute of Pathology, Ludwig-Maximilians-University, Munich, Germany
| | - Pierre Coulie
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | |
Collapse
|
134
|
Fortis SP, Sofopoulos M, Sotiriadou NN, Haritos C, Vaxevanis CK, Anastasopoulou EA, Janssen N, Arnogiannaki N, Ardavanis A, Pawelec G, Perez SA, Baxevanis CN. Differential intratumoral distributions of CD8 and CD163 immune cells as prognostic biomarkers in breast cancer. J Immunother Cancer 2017; 5:39. [PMID: 28428887 PMCID: PMC5395775 DOI: 10.1186/s40425-017-0240-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 04/06/2017] [Indexed: 12/21/2022] Open
Abstract
Background Tumor immune cell infiltrates are essential in hindering cancer progression and may complement the TNM classification. CD8+ and CD163+ cells have prognostic impact in breast cancer but their spatial heterogeneity has not been extensively explored in this type of cancer. Here, their potential as prognostic biomarkers was evaluated, depending on their combined densities in the tumor center (TC) and the tumor invasive margin (IM). Methods CD8+ and CD163+ cells were quantified by immunohistochemistry of formalin-fixed, paraffin-embedded (FFPE) tumor tissue samples from a cohort totaling 162 patients with histologically-confirmed primary invasive non-metastatic ductal breast cancer diagnosed between 2000 and 2015. Clinical follow-up (median 6.9 years) was available for 97 of these patients. Results Differential densities of CD8+ and CD163+ cells in the combined TC and IM compartments (i.e., high(H)/low(L), respectively for CD8+ cells and the reverse L/H combination for CD163+ cells) were found to have significant prognostic value for survival, and allowed better patient stratification than TNM stage, tumor size, lymph node invasion and histological grade. The combined evaluation of CD8+ and CD163+ cell densities jointly in TC and IM further improves prediction of clinical outcomes based on disease-free and overall survival. Patients having the favorable immune signatures had favorable clinical outcomes despite poor clinicopathological parameters. Conclusions Given the important roles of CD8+ and CD163+ cells in regulating opposing immune circuits, adding an assessment of their differential densities to the prognostic biomarker armamentarium in breast cancer would be valuable. Larger validation studies are necessary to confirm these findings. Trial registrations Study code: IRB-ID 6079/448/10-6-13 Date of approval: 10/06/2013 Retrospective study (2000–2010) First patient prospectively enrolled 14/2/2014 Electronic supplementary material The online version of this article (doi:10.1186/s40425-017-0240-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sotirios P Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | | | | | - Christoforos Haritos
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | | | | | - Nicole Janssen
- Center for Medical Research, Eberhard-Karls Universität, Tübingen, Germany
| | | | | | - Graham Pawelec
- Center for Medical Research, Eberhard-Karls Universität, Tübingen, Germany
| | - Sonia A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | | |
Collapse
|
135
|
Basile D, Garattini SK, Bonotto M, Ongaro E, Casagrande M, Cattaneo M, Fanotto V, De Carlo E, Loupakis F, Urbano F, Negri FV, Pella N, Russano M, Brunetti O, Scartozzi M, Santini D, Silvestris N, Casadei Gardini A, Puzzoni M, Calvetti L, Cardarelli N, Aprile G. Immunotherapy for colorectal cancer: where are we heading? Expert Opin Biol Ther 2017; 17:709-721. [PMID: 28375039 DOI: 10.1080/14712598.2017.1315405] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In the last few years, significant advances in molecular biology have provided new therapeutic options for colorectal cancer (CRC). The development of new drugs that target the immune response to cancer cells seems very promising and has already been established for other tumor types. In particular, the use of immune checkpoint inhibitors seems to be an encouraging immunotherapeutic strategy. Areas covered: In this review, the authors provide an update of the current evidence related to this topic, though most immunotherapies are still in early-phase clinical trials for CRC. To understand the key role of immunotherapy in CRC, the authors discuss the delicate balance between immune-stimulating and immune-suppressive networks that occur in the tumor microenvironment. Expert opinion: Modulation of the immune system through checkpoint inhibition is an emerging approach in CRC therapy. Nevertheless, selection criteria that could enable the identification of patients who may benefit from these agents are necessary. Furthermore, potential prognostic and predictive immune biomarkers based on immune and molecular classifications have been proposed. As expected, additional studies are required to develop biomarkers, effective therapeutic strategies and novel combinations to overcome immune escape resistance and enhance effector response.
Collapse
Affiliation(s)
- Debora Basile
- a Department of Oncology , University and General Hospital , Udine , Italy
| | | | - Marta Bonotto
- a Department of Oncology , University and General Hospital , Udine , Italy
| | - Elena Ongaro
- a Department of Oncology , University and General Hospital , Udine , Italy
| | | | - Monica Cattaneo
- a Department of Oncology , University and General Hospital , Udine , Italy
| | - Valentina Fanotto
- a Department of Oncology , University and General Hospital , Udine , Italy
| | - Elisa De Carlo
- a Department of Oncology , University and General Hospital , Udine , Italy
| | - Fotios Loupakis
- c Medical Oncology 1, Department of Medical and Experimental Oncology , IOV - IRCCS , Padova , Italy
| | - Federica Urbano
- b Medical Oncology B, Department of Dermatological Sciences, Radiology and Pathology , La Sapienza University , Rome , Italy.,c Medical Oncology 1, Department of Medical and Experimental Oncology , IOV - IRCCS , Padova , Italy
| | | | - Nicoletta Pella
- a Department of Oncology , University and General Hospital , Udine , Italy
| | - Marco Russano
- e Medical Oncology , Campus Biomedico University , Roma , Italy
| | - Oronzo Brunetti
- f Medical Oncology Unit , National Cancer Institute IRCCS "Giovanni Paolo II" , Bari , Italy
| | - Mario Scartozzi
- g Medical Oncology , University Hospital and University of Cagliari , Cagliari , Italy
| | - Daniele Santini
- e Medical Oncology , Campus Biomedico University , Roma , Italy
| | - Nicola Silvestris
- f Medical Oncology Unit , National Cancer Institute IRCCS "Giovanni Paolo II" , Bari , Italy
| | | | - Marco Puzzoni
- g Medical Oncology , University Hospital and University of Cagliari , Cagliari , Italy
| | - Lorenzo Calvetti
- i Department of Oncology , San Bortolo General Hospital , Vicenza , Italy
| | - Nadia Cardarelli
- i Department of Oncology , San Bortolo General Hospital , Vicenza , Italy
| | - Giuseppe Aprile
- a Department of Oncology , University and General Hospital , Udine , Italy.,i Department of Oncology , San Bortolo General Hospital , Vicenza , Italy
| |
Collapse
|
136
|
Kapetanakis NI, Baloche V, Busson P. Tumor exosomal microRNAs thwarting anti-tumor immune responses in nasopharyngeal carcinomas. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:164. [PMID: 28480200 DOI: 10.21037/atm.2017.03.57] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | - Valentin Baloche
- CNRS UMR8126, Gustave Roussy and Université Paris-Sud/Paris-Saclay, 39 rue Camille Desmoulins, Villejuif, France
| | - Pierre Busson
- CNRS UMR8126, Gustave Roussy and Université Paris-Sud/Paris-Saclay, 39 rue Camille Desmoulins, Villejuif, France
| |
Collapse
|
137
|
Gulley JL, Berzofsky JA, Butler MO, Cesano A, Fox BA, Gnjatic S, Janetzki S, Kalavar S, Karanikas V, Khleif SN, Kirsch I, Lee PP, Maccalli C, Maecker H, Schlom J, Seliger B, Siebert J, Stroncek DF, Thurin M, Yuan J, Butterfield LH. Immunotherapy biomarkers 2016: overcoming the barriers. J Immunother Cancer 2017; 5:29. [PMID: 28653584 PMCID: PMC5359902 DOI: 10.1186/s40425-017-0225-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 02/13/2017] [Indexed: 12/18/2022] Open
Abstract
This report summarizes the symposium, 'Immunotherapy Biomarkers 2016: Overcoming the Barriers', which was held on April 1, 2016 at the National Institutes of Health in Bethesda, Maryland. The symposium, cosponsored by the Society for Immunotherapy of Cancer (SITC) and the National Cancer Institute (NCI), focused on emerging immunotherapy biomarkers, new technologies, current hurdles to further progress, and recommendations for advancing the field of biomarker development.
Collapse
Affiliation(s)
- James L Gulley
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, 10 Center Dr., 13 N240, Bethesda, MD, 20892, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, 41 Medlars Dr, Bldg 41 Rm D702D, Bethesda, MD, 20892, USA
| | - Marcus O Butler
- Princess Margaret Cancer Center/Ontario Cancer Institute, RM 9-622, 610 University Ave, Toronto, ON, Canada
| | - Alessandra Cesano
- NanoString, Inc., 500 Fairview Avenue North, Seattle, WA, 98109, USA
| | - Bernard A Fox
- Earle A. Chiles Research Institute, Providence Cancer Center, 4805 NE Glisan Street, Portland, OR, 97213, USA
| | - Sacha Gnjatic
- Department of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, S5-105, 1470 Madison Avenue, Box 1128, New York, NY, 10029, USA
| | - Sylvia Janetzki
- ZellNet Consulting, Inc., 555 North Avenue, Fort Lee, NJ, 07024, USA
| | - Shyam Kalavar
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, 1401 Rockville Pike, Rockville, MD, 20852, USA
| | - Vaios Karanikas
- Roche Innovation Center Zurich, Wagistrasse 18, Schlieren, Switzerland
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, 1120 15th Street, CN-2101A, Augusta, GA, 30912, USA
| | - Ilan Kirsch
- Adaptive Biotechnologies, Inc., 1551 Eastlake Ave. E., Seattle, WA, 98102, USA
| | - Peter P Lee
- Department of Immuno-oncology, City of Hope, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Cristina Maccalli
- Department of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Holden Maecker
- Stanford University Medical Center, 299 Campus Drive, Stanford, CA, 94303, USA
| | - Jeffrey Schlom
- National Cancer Institute, National Institutes of Health, 10 Center Drive, Bldg. 10, Room 8B09, Bethesda, MD, 20892, USA
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, Halle, Germany
| | - Janet Siebert
- CytoAnalytics, 3500 South Albion Street, Cherry Hills Village, CO, 80113, USA
| | - David F Stroncek
- Department of Transfusion Medicine, National Institutes of Health, 10 Center Drive, Building 10, Room 3C720, Bethesda, MD, 20892, USA
| | - Magdalena Thurin
- National Cancer Institute, Cancer Diagnosis Program, DCTD, National Institutes of Health, 9609 Medical Center Drive, Bethesda, 20892, MD, USA
| | - Jianda Yuan
- Early Clinical Oncology Development, Merck Research Laboratories, Rahway, NJ, 07065, USA
| | - Lisa H Butterfield
- Department of Medicine, Surgery and Immunology, University of Pittsburgh Cancer Institute, 5117 Centre Avenue, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
138
|
Giraldo NA, Becht E, Vano Y, Petitprez F, Lacroix L, Validire P, Sanchez-Salas R, Ingels A, Oudard S, Moatti A, Buttard B, Bourass S, Germain C, Cathelineau X, Fridman WH, Sautès-Fridman C. Tumor-Infiltrating and Peripheral Blood T-cell Immunophenotypes Predict Early Relapse in Localized Clear Cell Renal Cell Carcinoma. Clin Cancer Res 2017; 23:4416-4428. [PMID: 28213366 DOI: 10.1158/1078-0432.ccr-16-2848] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 12/12/2016] [Accepted: 02/06/2017] [Indexed: 11/16/2022]
Abstract
Purpose: The efficacy of PD-1 checkpoint blockade as adjuvant therapy in localized clear cell renal cell carcinoma (ccRCC) is currently unknown. The identification of tumor microenvironment (TME) prognostic biomarkers in this setting may help define which patients could benefit from checkpoint blockade and uncover new therapeutic targets.Experimental Design: We performed multiparametric flow cytometric immunophenotypic analysis of T cells isolated from tumor tissue [tumor-infiltrating lymphocytes (TIL)], adjacent non-malignant renal tissue [renal-infiltrating lymphocytes (RIL)], and peripheral blood lymphocytes (PBL), in a cohort of patients (n = 40) with localized ccRCC. Immunophenotypic data were integrated with prognostic and histopathologic variables, T-cell receptor (TCR) repertoire analysis of sorted CD8+PD-1+ TILs, tumor mRNA expression, and digital quantitative immunohistochemistry.Results: On the basis of TIL phenotypic characterization, we identified three dominant immune profiles in localized ccRCC: (i) immune-regulated, characterized by polyclonal/poorly cytotoxic CD8+PD-1+Tim-3+Lag-3+ TILs and CD4+ICOS+ cells with a Treg phenotype (CD25+CD127-Foxp3+/Helios+GITR+), that developed in inflamed tumors with prominent infiltrations by dysfunctional dendritic cells and high PD-L1 expression; (ii) immune-activated, enriched in oligoclonal/cytotoxic CD8+PD-1+Tim-3+ TILs, that represented 22% of the tumors; and (iii) immune-silent, enriched in TILs exhibiting RIL-like phenotype, that represented 56% of patients in the cohort. Only immune-regulated tumors displayed aggressive histologic features, high risk of disease progression in the year following nephrectomy, and a CD8+PD-1+Tim-3+ and CD4+ICOS+ PBL phenotypic signature.Conclusions: In localized ccRCC, the infiltration with CD8+PD-1+Tim-3+Lag-3+ exhausted TILs and ICOS+ Treg identifies the patients with deleterious prognosis who could benefit from adjuvant therapy with TME-modulating agents and checkpoint blockade. This work also provides PBL phenotypic markers that could allow their identification. Clin Cancer Res; 23(15); 4416-28. ©2017 AACR.
Collapse
Affiliation(s)
- Nicolas A Giraldo
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Etienne Becht
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Yann Vano
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Georges Pompidou European Hospital, Oncology Department, Paris 5 - Descartes University, Assistance Publique Hopitaux de Paris, Paris, France
| | - Florent Petitprez
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,Carte d'Identité des Tumeurs, Ligue contre le Cancer, Paris, France
| | - Laetitia Lacroix
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Pierre Validire
- Pathology Department, Institut Mutualiste Montsouris, Paris, France
| | | | - Alexandre Ingels
- Urology Department, Institut Mutualiste Montsouris, Paris, France
| | - Stephane Oudard
- Georges Pompidou European Hospital, Oncology Department, Paris 5 - Descartes University, Assistance Publique Hopitaux de Paris, Paris, France
| | - Audrey Moatti
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Benedicte Buttard
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Sarah Bourass
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Claire Germain
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France.,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Xavier Cathelineau
- Urology Department, Institut Mutualiste Montsouris, Paris, France.,University Paris Descartes Paris, Medical School, Paris, France
| | - Wolf H Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France. .,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Cordeliers Research Center, Team "Cancer, immune control and escape", Paris, France. .,University Paris Descartes Paris, Sorbonne Paris Cite, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France.,UPMC University Paris, Sorbonne University, UMR_S 1138, Centre de Recherche des Cordeliers, Paris, France
| |
Collapse
|
139
|
Eggink FA, Van Gool IC, Leary A, Pollock PM, Crosbie EJ, Mileshkin L, Jordanova ES, Adam J, Freeman-Mills L, Church DN, Creutzberg CL, De Bruyn M, Nijman HW, Bosse T. Immunological profiling of molecularly classified high-risk endometrial cancers identifies POLE-mutant and microsatellite unstable carcinomas as candidates for checkpoint inhibition. Oncoimmunology 2016; 6:e1264565. [PMID: 28344870 PMCID: PMC5353925 DOI: 10.1080/2162402x.2016.1264565] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/16/2016] [Accepted: 11/17/2016] [Indexed: 12/31/2022] Open
Abstract
High-risk endometrial cancer (EC) is an aggressive disease for which new therapeutic options are needed. Aims of this study were to validate the enhanced immune response in highly mutated ECs and to explore immune profiles in other EC subgroups. We evaluated immune infiltration in 116 high-risk ECs from the TransPORTEC consortium, previously classified into four molecular subtypes: (i) ultramutated POLE exonuclease domain-mutant ECs (POLE-mutant); (ii) hypermutated microsatellite unstable (MSI); (iii) p53-mutant; and (iv) no specific molecular profile (NSMP). Within The Cancer Genome Atlas (TCGA) EC cohort, significantly higher numbers of predicted neoantigens were demonstrated in POLE-mutant and MSI tumors compared with NSMP and p53-mutants. This was reflected by enhanced immune expression and infiltration in POLE-mutant and MSI tumors in both the TCGA cohort (mRNA expression) and the TransPORTEC cohort (immunohistochemistry) with high infiltration of CD8+ (90% and 69%), PD-1+ (73% and 69%) and PD-L1+ immune cells (100% and 71%). Notably, a subset of p53-mutant and NSMP cancers was characterized by signs of an antitumor immune response (43% and 31% of tumors with high infiltration of CD8+ cells, respectively), despite a low number of predicted neoantigens. In conclusion, the presence of enhanced immune infiltration, particularly high numbers of PD-1 and PD-L1 positive cells, in highly mutated, neoantigen-rich POLE-mutant and MSI endometrial tumors suggests sensitivity to immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Florine A Eggink
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen , Groningen, the Netherlands
| | - Inge C Van Gool
- Department of Pathology, Leiden University Medical Center , Leiden, the Netherlands
| | - Alexandra Leary
- Department of Medical Oncology, INSERM U981, Gustave Roussy Cancer Center , Villejuif, France
| | - Pamela M Pollock
- Queensland University of Technology (QUT), Translational Research Institute , Brisbane, QLD, Australia
| | - Emma J Crosbie
- Institute of Cancer Sciences, University of Manchester, St Marys Hospital , Manchester, UK
| | - Linda Mileshkin
- Division of Cancer Medicine, Peter MacCallum Cancer Centre , East Melbourne, VIC, Australia
| | - Ekaterina S Jordanova
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands; Center for Gynecological Oncology Amsterdam, VU Medical Center, Amsterdam, the Netherlands
| | - Julien Adam
- Department of Medical Oncology, INSERM U981, Gustave Roussy Cancer Center , Villejuif, France
| | - Luke Freeman-Mills
- Tumour Genomics and Immunology Group, Oxford Centre for Cancer Gene Research, The Wellcome Trust Centre for Human Genetics, University of Oxford , Oxford, UK
| | - David N Church
- Tumour Genomics and Immunology Group, Oxford Centre for Cancer Gene Research, The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK; Oxford Cancer Centre, Churchill Hospital, Oxford, UK
| | - Carien L Creutzberg
- Department of Clinical Oncology, Leiden University Medical Center , Leiden, the Netherlands
| | - Marco De Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen , Groningen, the Netherlands
| | - Hans W Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen , Groningen, the Netherlands
| | - Tjalling Bosse
- Department of Pathology, Leiden University Medical Center , Leiden, the Netherlands
| |
Collapse
|
140
|
Jin C, Yu D, Essand M. Prospects to improve chimeric antigen receptor T-cell therapy for solid tumors. Immunotherapy 2016; 8:1355-1361. [DOI: 10.2217/imt-2016-0125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Adoptive transfer of patient-derived T-cells engineered with a chimeric antigen receptor (CAR) targeting the pan-B-cell marker CD19 has led to complete remission in patients with B-cell leukemias while response rates are more modest for B-cell lymphomas. This can be attributed to the fact that the semi-solid structure of lymphomas impedes T-cell infiltration and that the immune suppressive microenvironment within these tumors dampens the effect of CAR T-cells. These obstacles are even more pronounced for solid tumors where dense and often highly immunosuppressive structures are found. This article focuses on different aspects of how to improve CAR T-cells for solid tumors, primarily by decreasing their sensitivity to the harsh tumor microenvironment, by altering the immunosuppressive microenvironment inside tumors and by inducing bystander immunity.
Collapse
Affiliation(s)
- Chuan Jin
- Department of Immunology, Genetics & Pathology, Science for Life Laboratory, Uppsala University, SE-75185 Uppsala, Sweden
| | - Di Yu
- Department of Immunology, Genetics & Pathology, Science for Life Laboratory, Uppsala University, SE-75185 Uppsala, Sweden
| | - Magnus Essand
- Department of Immunology, Genetics & Pathology, Science for Life Laboratory, Uppsala University, SE-75185 Uppsala, Sweden
| |
Collapse
|
141
|
|
142
|
Becht E, Giraldo NA, Lacroix L, Buttard B, Elarouci N, Petitprez F, Selves J, Laurent-Puig P, Sautès-Fridman C, Fridman WH, de Reyniès A. Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression. Genome Biol 2016; 17:218. [PMID: 27765066 PMCID: PMC5073889 DOI: 10.1186/s13059-016-1070-5] [Citation(s) in RCA: 1897] [Impact Index Per Article: 237.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/21/2016] [Indexed: 01/05/2023] Open
Abstract
We introduce the Microenvironment Cell Populations-counter (MCP-counter) method, which allows the robust quantification of the absolute abundance of eight immune and two stromal cell populations in heterogeneous tissues from transcriptomic data. We present in vitro mRNA mixture and ex vivo immunohistochemical data that quantitatively support the validity of our method’s estimates. Additionally, we demonstrate that MCP-counter overcomes several limitations or weaknesses of previously proposed computational approaches. MCP-counter is applied to draw a global picture of immune infiltrates across human healthy tissues and non-hematopoietic human tumors and recapitulates microenvironment-based patient stratifications associated with overall survival in lung adenocarcinoma and colorectal and breast cancer.
Collapse
Affiliation(s)
- Etienne Becht
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Nicolas A Giraldo
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Laetitia Lacroix
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Bénédicte Buttard
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Nabila Elarouci
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Florent Petitprez
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France.,Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Janick Selves
- Centre de Recherche en Cancérologie de Toulouse, Unité Mixte de Recherche, 1037 INSERM, Université Toulouse III, Toulouse, France.,Department of Pathology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Catherine Sautès-Fridman
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Wolf H Fridman
- INSERM UMR_S 1138, Cancer, Immune Control and Escape, Cordeliers Research Centre, Paris, France.,Université Paris Descartes, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France.
| |
Collapse
|
143
|
Sautès-Fridman C, Lawand M, Giraldo NA, Kaplon H, Germain C, Fridman WH, Dieu-Nosjean MC. Tertiary Lymphoid Structures in Cancers: Prognostic Value, Regulation, and Manipulation for Therapeutic Intervention. Front Immunol 2016; 7:407. [PMID: 27752258 PMCID: PMC5046074 DOI: 10.3389/fimmu.2016.00407] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 09/22/2016] [Indexed: 01/03/2023] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid aggregates that reflect lymphoid neogenesis occurring in tissues at sites of inflammation. They are detected in tumors where they orchestrate local and systemic anti-tumor responses. A correlation has been found between high densities of TLS and prolonged patient's survival in more than 10 different types of cancer. TLS can be regulated by the same set of chemokines and cytokines that orchestrate lymphoid organogenesis and by regulatory T cells. Thus, TLS offer a series of putative new targets that could be used to develop therapies aiming to increase the anti-tumor immune response.
Collapse
Affiliation(s)
- Catherine Sautès-Fridman
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Myriam Lawand
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Nicolas A Giraldo
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Hélène Kaplon
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Claire Germain
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Wolf Herman Fridman
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- INSERM, UMR_S 1138, Team "Cancer, Immune Control and Escape", Cordeliers Research Center, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, University Paris Descartes, Paris, France; UMR_S 1138, Centre de Recherche des Cordeliers, Sorbonne University, UPMC University Paris 06, Paris, France
| |
Collapse
|
144
|
Knee DA, Hewes B, Brogdon JL. Rationale for anti-GITR cancer immunotherapy. Eur J Cancer 2016; 67:1-10. [PMID: 27591414 DOI: 10.1016/j.ejca.2016.06.028] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 06/29/2016] [Indexed: 12/18/2022]
Abstract
Over the past decade, our understanding of cancer immunotherapy has evolved from assessing peripheral responses in the blood to monitoring changes in the tumour microenvironment. Both preclinical and clinical experience has taught us that modulation of the tumour microenvironment has significant implications to generating robust antitumour immunity. Clinical benefit has been well documented to correlate with a tumour microenvironment that contains a dense infiltration of CD8+CD45RO+ T effectors and a high ratio of CD8+ T cells to FoxP3+ regulatory T cells (Tregs). In preclinical tumour models, modulation of the Glucocorticoid induced TNF receptor (GITR)/GITR ligand (GITRL) axis suggests this pathway may provide the desired biological outcome of inhibiting Treg function while activating CD8+ T effector cells. This review will focus on the scientific rationale and considerations for the therapeutic targeting of GITR for cancer immunotherapy and will discuss possible combination strategies to enhance clinical benefit.
Collapse
Affiliation(s)
- Deborah A Knee
- Department of Cancer Immunotherapeutics, Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Drive, San Diego, CA 92121, USA.
| | - Becker Hewes
- Department of Translational & Clinical Oncology, Novartis Institute for Biomedical Research, 220 Massachusetts Ave, Cambridge, MA, USA.
| | - Jennifer L Brogdon
- Department of Exploratory Immuno-Oncology, Novartis Institute for Biomedical Research, 250 Massachusetts Ave, Cambridge, MA, USA.
| |
Collapse
|
145
|
Bobisse S, Foukas PG, Coukos G, Harari A. Neoantigen-based cancer immunotherapy. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:262. [PMID: 27563649 DOI: 10.21037/atm.2016.06.17] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Emerging clinical evidence on the role of the antitumor activity of the immune system has generated great interest in immunotherapy in all cancer types. Recent clinical data clearly demonstrated that human tumor cells express antigenic peptides (epitopes) that can be recognized by autologous tumor-specific T cells and that enhancement of such immune reactivity can potentially lead to cancer control and cancer regression in patients with advanced disease. However, in most cases, it is unclear which tumor antigens (Ags) mediated cancer regression. Mounting evidence indicates that numerous endogenous mutated cancer proteins, a hallmark of tumor cells, can be processed into peptides and presented on the surface of tumor cells, leading to their immune recognition in vivo as "non-self" or foreign. Massively parallel sequencing has now overcome the challenge of rapidly identifying the comprehensive mutational spectrum of individual tumors (i.e., the "mutanome") and current technologies, as well as computational tools, have emerged that allow the identification of private epitopes derived from their mutanome and called neoantigens (neoAgs). On this basis, both CD4(+) and CD8(+) neoantigen-specific T cells have been identified in multiple human cancers and shown to be associated with a favorable clinical outcome. Notably, emerging data also indicate that neoantigen recognition represents a major factor in the activity of clinical immunotherapies. In the post-genome era, the mutanome holds promise as a long-awaited 'gold mine' for the discovery of unique cancer cell targets, which are exclusively tumor-specific and unlikely to drive immune tolerance, hence offering the chance for highly promising clinical programs of cancer immunotherapy.
Collapse
Affiliation(s)
- Sara Bobisse
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland
| | - Periklis G Foukas
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland;; 2nd Department of Pathology, National and Kapodistrian University of Athens, School of Medicine, Attikon University Hospital, Athens, Greece
| | - George Coukos
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland
| | - Alexandre Harari
- Ludwig Cancer Center and Department of Oncology, University of Lausanne, Lausanne, Switzerland;; Center of Experimental Therapeutics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
146
|
Drakes ML, Stiff PJ. Understanding dendritic cell immunotherapy in ovarian cancer. Expert Rev Anticancer Ther 2016; 16:643-52. [DOI: 10.1080/14737140.2016.1178576] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
147
|
Becht E, Giraldo NA, Dieu-Nosjean MC, Sautès-Fridman C, Fridman WH. Cancer immune contexture and immunotherapy. Curr Opin Immunol 2016; 39:7-13. [DOI: 10.1016/j.coi.2015.11.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/27/2015] [Accepted: 11/30/2015] [Indexed: 01/01/2023]
|
148
|
Fridman WH. [History of immunotherapy. Paradigm change?]. Bull Cancer 2016:S0007-4551(16)00034-5. [PMID: 26976507 DOI: 10.1016/j.bulcan.2016.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 01/17/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Wolf Hervé Fridman
- Université Paris Descartes, centre de recherche des Cordeliers, UMRS 1138, 15, rue de l'École-de-Médecine, 75005 Paris, France.
| |
Collapse
|