351
|
Elia AR, Caputo S, Bellone M. Immune Checkpoint-Mediated Interactions Between Cancer and Immune Cells in Prostate Adenocarcinoma and Melanoma. Front Immunol 2018; 9:1786. [PMID: 30108594 PMCID: PMC6079266 DOI: 10.3389/fimmu.2018.01786] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/19/2018] [Indexed: 01/05/2023] Open
Abstract
Prostate adenocarcinoma (PCa) and melanoma are paradigmatic examples of tumors that are either poorly or highly sensitive to therapies based on monoclonal antibodies directed against regulatory pathways in T lymphocytes [i.e., immune checkpoint blockade (ICB)]. Yet, approximately 40% of melanoma patients are resistant or acquire resistance to ICB. What characterize the microenvironment of PCa and ICB-resistant melanoma are a scanty cytotoxic T cell infiltrate and a strong immune suppression, respectively. Here, we compare the tumor microenvironment in these two subgroups of cancer patients, focusing on some among the most represented immune checkpoint molecules: cytotoxic T lymphocyte-associated antigen-4, programmed death-1, lymphocyte activation gene-3, and T cell immunoglobulin and mucin-domain containing-3. We also report on several examples of crosstalk between cancer and immune cells that are mediated by inhibitory immune checkpoints and identify promising strategies aimed at overcoming ICB resistance both in PCa and melanoma.
Collapse
Affiliation(s)
- Angela Rita Elia
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Sara Caputo
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Bellone
- Cellular Immunology Unit, Department of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
352
|
Nguyen A, Ho L, Workenhe ST, Chen L, Samson J, Walsh SR, Pol J, Bramson JL, Wan Y. HDACi Delivery Reprograms Tumor-Infiltrating Myeloid Cells to Eliminate Antigen-Loss Variants. Cell Rep 2018; 24:642-654. [PMID: 30021162 DOI: 10.1016/j.celrep.2018.06.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/02/2018] [Accepted: 06/08/2018] [Indexed: 01/05/2023] Open
Abstract
Immune recognition of tumor-expressed antigens by cytotoxic CD8+ T cells is the foundation of adoptive T cell therapy (ACT) and has been shown to elicit significant tumor regression. However, therapy-induced selective pressure can sculpt the antigenicity of tumors, resulting in outgrowth of variants that lose the target antigen. We demonstrate that tumor relapse from ACT and subsequent oncolytic viral vaccination can be prevented using class I HDACi, MS-275. Drug delivery subverted the phenotype of tumor-infiltrating CD11b+ Ly6Chi Ly6G- myeloid cells, favoring NOS2/ROS secretion and pro-inflammatory genes characteristic of M1 polarization. Simultaneously, MS-275 abrogated the immunosuppressive function of tumor-infiltrating myeloid cells and reprogrammed them to eliminate antigen-negative tumor cells in a caspase-dependent manner. Elevated IFN-γ within the tumor microenvironment suggests that MS-275 modulates the local cytokine landscape to favor antitumor myeloid polarization through the IFN-γR/STAT1 signaling axis. Exploiting tumor-infiltrating myeloid cell plasticity thus complements T cell therapy in targeting tumor heterogeneity and immune escape.
Collapse
Affiliation(s)
- Andrew Nguyen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Louisa Ho
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Samuel T Workenhe
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lan Chen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | | | - Scott R Walsh
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jonathan Pol
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
353
|
Lu X, Pan X, Wu CJ, Zhao D, Feng S, Zang Y, Lee R, Khadka S, Amin SB, Jin EJ, Shang X, Deng P, Luo Y, Morgenlander WR, Weinrich J, Lu X, Jiang S, Chang Q, Navone NM, Troncoso P, DePinho RA, Wang YA. An In Vivo Screen Identifies PYGO2 as a Driver for Metastatic Prostate Cancer. Cancer Res 2018; 78:3823-3833. [PMID: 29769196 PMCID: PMC6381393 DOI: 10.1158/0008-5472.can-17-3564] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 03/27/2018] [Accepted: 05/10/2018] [Indexed: 01/08/2023]
Abstract
Advanced prostate cancer displays conspicuous chromosomal instability and rampant copy number aberrations, yet the identity of functional drivers resident in many amplicons remain elusive. Here, we implemented a functional genomics approach to identify new oncogenes involved in prostate cancer progression. Through integrated analyses of focal amplicons in large prostate cancer genomic and transcriptomic datasets as well as genes upregulated in metastasis, 276 putative oncogenes were enlisted into an in vivo gain-of-function tumorigenesis screen. Among the top positive hits, we conducted an in-depth functional analysis on Pygopus family PHD finger 2 (PYGO2), located in the amplicon at 1q21.3. PYGO2 overexpression enhances primary tumor growth and local invasion to draining lymph nodes. Conversely, PYGO2 depletion inhibits prostate cancer cell invasion in vitro and progression of primary tumor and metastasis in vivo In clinical samples, PYGO2 upregulation associated with higher Gleason score and metastasis to lymph nodes and bone. Silencing PYGO2 expression in patient-derived xenograft models impairs tumor progression. Finally, PYGO2 is necessary to enhance the transcriptional activation in response to ligand-induced Wnt/β-catenin signaling. Together, our results indicate that PYGO2 functions as a driver oncogene in the 1q21.3 amplicon and may serve as a potential prognostic biomarker and therapeutic target for metastatic prostate cancer.Significance: Amplification/overexpression of PYGO2 may serve as a biomarker for prostate cancer progression and metastasis. Cancer Res; 78(14); 3823-33. ©2018 AACR.
Collapse
Affiliation(s)
- Xin Lu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Biological Sciences, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana
- Tumor Microenvironment and Metastasis Program, Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana
| | - Xiaolu Pan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chang-Jiun Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Di Zhao
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shan Feng
- Department of Biological Sciences, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana
| | - Yong Zang
- Department of Biostatistics, Indiana University, Indianapolis, Indiana
| | - Rumi Lee
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunada Khadka
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samirkumar B Amin
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eun-Jung Jin
- Department of Biological Science, Wonkwang University, Cheonbuk, Iksan, South Korea
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pingna Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yanting Luo
- Department of Biological Sciences, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana
| | - William R Morgenlander
- Department of Biological Sciences, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana
| | - Jacqueline Weinrich
- Department of Biological Sciences, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana
| | - Xuemin Lu
- Department of Biological Sciences, Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, Indiana
| | - Shan Jiang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qing Chang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nora M Navone
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Patricia Troncoso
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
354
|
Palanissami G, Paul SFD. RAGE and Its Ligands: Molecular Interplay Between Glycation, Inflammation, and Hallmarks of Cancer—a Review. Discov Oncol 2018; 9:295-325. [DOI: 10.1007/s12672-018-0342-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/25/2018] [Indexed: 12/15/2022] Open
|
355
|
Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, Senju S, Ihn H, Nishimura Y, Oshiumi H. Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res 2018; 78:5011-5022. [PMID: 29967259 DOI: 10.1158/0008-5472.can-18-0118] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/24/2018] [Accepted: 06/27/2018] [Indexed: 11/16/2022]
Abstract
Recently emerging cancer immunotherapies combine the applications of therapeutics to disrupt the immunosuppressive conditions in tumor-bearing hosts. In this study, we found that targeting the proinflammatory cytokine IL6 enhances tumor-specific Th1 responses and subsequent antitumor effects in tumor-bearing mice. IL6 blockade upregulated expression of the immune checkpoint molecule programmed death-ligand 1 (PD-L1) on melanoma cells. This PD-L1 induction was canceled in IFNγ-deficient mice or CD4+ T cell-depleted mice, suggesting that CD4+ T cell-derived IFNγ is important for PD-L1 induction in tumor-bearing hosts. In some patients with melanoma, however, treatment with the anti-PD-1 antibody nivolumab increased systemic levels of IL6, which was associated with poor clinical responses. This PD-L1 blockade-evoked induction of IL6 was reproducible in melanoma-bearing mice. We found that PD-1/PD-L1 blockade prompted PD-1+ macrophages to produce IL6 in the tumor microenvironment. Depletion of macrophages in melanoma-bearing mice reduced the levels of IL6 during PD-L1 blockade, suggesting macrophages are responsible for the IL6-mediated defective CD4+ Th1 response. Combined blockade of the mutually regulated immunosuppressive activities of IL6 and PD-1/PD-L1 signals enhanced expression of T cell-attracting chemokines and promoted infiltration of IFNγ-producing CD4+ T cells in tumor tissues, exerting a synergistic antitumor effect, whereas PD-L1 blockade alone did not promote Th1 response. Collectively, these findings suggest that IL6 is a rational immunosuppressive target for overcoming the narrow therapeutic window of anti-PD-1/PD-L1 therapy.Significance: These findings advance our understanding of IL6-PD1/PD-L1 cross-talk in the tumor microenvironment and provide clues for targeted interventional therapy that may prove more effective against cancer. Cancer Res; 78(17); 5011-22. ©2018 AACR.
Collapse
Affiliation(s)
| | - Koji Fujieda
- Department of Immunogenetics, Kumamoto University, Kumamoto, Japan
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Kubo
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
356
|
Targeting Wnt/β-Catenin Signaling for Cancer Immunotherapy. Trends Pharmacol Sci 2018; 39:648-658. [DOI: 10.1016/j.tips.2018.03.008] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/19/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
|
357
|
Calcinotto A, Spataro C, Zagato E, Di Mitri D, Gil V, Crespo M, De Bernardis G, Losa M, Mirenda M, Pasquini E, Rinaldi A, Sumanasuriya S, Lambros MB, Neeb A, Lucianò R, Bravi CA, Nava-Rodrigues D, Dolling D, Prayer-Galetti T, Ferreira A, Briganti A, Esposito A, Barry S, Yuan W, Sharp A, de Bono J, Alimonti A. IL-23 secreted by myeloid cells drives castration-resistant prostate cancer. Nature 2018; 559:363-369. [PMID: 29950727 DOI: 10.1038/s41586-018-0266-0] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 05/29/2018] [Indexed: 01/25/2023]
Abstract
Patients with prostate cancer frequently show resistance to androgen-deprivation therapy, a condition known as castration-resistant prostate cancer (CRPC). Acquiring a better understanding of the mechanisms that control the development of CRPC remains an unmet clinical need. The well-established dependency of cancer cells on the tumour microenvironment indicates that the microenvironment might control the emergence of CRPC. Here we identify IL-23 produced by myeloid-derived suppressor cells (MDSCs) as a driver of CRPC in mice and patients with CRPC. Mechanistically, IL-23 secreted by MDSCs can activate the androgen receptor pathway in prostate tumour cells, promoting cell survival and proliferation in androgen-deprived conditions. Intra-tumour MDSC infiltration and IL-23 concentration are increased in blood and tumour samples from patients with CRPC. Antibody-mediated inactivation of IL-23 restored sensitivity to androgen-deprivation therapy in mice. Taken together, these results reveal that MDSCs promote CRPC by acting in a non-cell autonomous manner. Treatments that block IL-23 can oppose MDSC-mediated resistance to castration in prostate cancer and synergize with standard therapies.
Collapse
Affiliation(s)
- Arianna Calcinotto
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Clarissa Spataro
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Elena Zagato
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Diletta Di Mitri
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Veronica Gil
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Mateus Crespo
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Gaston De Bernardis
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Marco Losa
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Michela Mirenda
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Emiliano Pasquini
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland
| | - Semini Sumanasuriya
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Maryou B Lambros
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Antje Neeb
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Roberta Lucianò
- Division of Oncology, Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Carlo A Bravi
- Division of Oncology, Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Daniel Nava-Rodrigues
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - David Dolling
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | | | - Ana Ferreira
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Alberto Briganti
- Division of Oncology, Unit of Urology, URI, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Antonio Esposito
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy
| | - Simon Barry
- IMED Oncology AstraZeneca, Li Ka Shing Centre, Cambridge, UK
| | - Wei Yuan
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Adam Sharp
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Johann de Bono
- The Institute of Cancer Research and The Royal Marsden NHS Foundation Trust, London, UK
| | - Andrea Alimonti
- Institute of Oncology Research (IOR), Oncology Institute of Southern Switzerland, Bellinzona, Switzerland. .,Università della Svizzera italiana, Faculty of Biomedical Sciences, Lugano, Switzerland. .,Faculty of Biology and Medicine, University of Lausanne UNIL, Lausanne, Switzerland. .,Department of Medicine, Venetian Institute of Molecular Medicine, University of Padova, Padova, Italy.
| |
Collapse
|
358
|
Lerman I, Hammes SR. Neutrophil elastase in the tumor microenvironment. Steroids 2018; 133:96-101. [PMID: 29155217 PMCID: PMC5870895 DOI: 10.1016/j.steroids.2017.11.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/07/2017] [Accepted: 11/11/2017] [Indexed: 12/12/2022]
Abstract
Myeloid cell production within the bone marrow is accelerated in the setting of cancer, and the numbers of circulating and infiltrating neutrophils and granulocytic myeloid derived suppressor cells (MDSCs) correlate with tumor progression and patient survival. Cancer is therefore able to hijack the normally host-protective immune system and use it to further fuel growth and metastasis. Myeloid cells secrete neutrophil elastase and neutrophil extracellular traps (NETs) in response to cues within the tumor microenvironment, thereby leading to enhanced activity in a variety of cancer types. Neutrophil elastase may indeed be a driver of tumorigenesis, since genetic deletion and pharmacological inhibition markedly reduces tumor burden and metastatic potential in numerous preclinical studies. In this review, we examine the current evidence for neutrophil elastase as a stimulatory factor in cancer, focusing on precise mechanisms by which it facilitates primary tumor growth and secondary organ metastasis. We conclude with a brief overview of neutrophil elastase inhibitors and discuss their potential use in cancer therapy.
Collapse
Affiliation(s)
- Irina Lerman
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Rochester, NY 14642, United States.
| | - Stephen R Hammes
- Department of Medicine, Division of Endocrinology and Metabolism, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave., Rochester, NY 14642, United States
| |
Collapse
|
359
|
Gourdin T, Sonpavde G. Utility of cell-free nucleic acid and circulating tumor cell analyses in prostate cancer. Asian J Androl 2018; 20:230-237. [PMID: 29578115 PMCID: PMC5952476 DOI: 10.4103/aja.aja_1_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/28/2017] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer is characterized by bone metastases and difficulty of objectively measuring disease burden. In this context, cell-free circulating tumor DNA (ctDNA) and circulating tumor cell (CTC) quantitation and genomic profiling afford the ability to noninvasively and serially monitor the tumor. Recent data suggest that ctDNA and CTC quantitation are prognostic for survival. Indeed, CTC enumeration using the CellSearch® platform is validated as a prognostic factor and warrants consideration as a stratification factor in randomized trials. Changes in quantities of CTCs using CellSearch also are prognostic and may be employed to detect a signal of activity of new agents. Molecular profiling of both CTCs and ctDNA for androgen receptor (AR) variants has been associated with outcomes in the setting of novel androgen inhibitors. Serial profiling to detect the evolution of new alterations may inform drug development and help develop precision medicine. The costs of these assays and the small quantities in which they are detectable in blood are a limitation, and novel platforms are required to address this challenge. The presence of multiple platforms to assay CTCs and ctDNA also warrants the consideration of a mechanism to allow comparison of data across platforms. Further validation and the continued development and standardization of these promising modalities will facilitate their adoption in the clinic.
Collapse
Affiliation(s)
| | - Guru Sonpavde
- Dana Farber Cancer Institute, Genitourinary Oncology Section, Boston, MA 02215, USA
| |
Collapse
|
360
|
Zhang J, Wang G, Zhou Y, Chen Y, Ouyang L, Liu B. Mechanisms of autophagy and relevant small-molecule compounds for targeted cancer therapy. Cell Mol Life Sci 2018; 75:1803-1826. [PMID: 29417176 PMCID: PMC11105210 DOI: 10.1007/s00018-018-2759-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/15/2018] [Accepted: 01/23/2018] [Indexed: 02/05/2023]
Abstract
Autophagy is an evolutionarily conserved, multi-step lysosomal degradation process for the clearance of damaged or superfluous proteins and organelles. Accumulating studies have recently revealed that autophagy is closely related to a variety of types of cancer; however, elucidation of its Janus role of either tumor-suppressive or tumor-promoting still remains to be discovered. In this review, we focus on summarizing the context-dependent role of autophagy and its complicated molecular mechanisms in different types of cancer. Moreover, we discuss a series of small-molecule compounds targeting autophagy-related proteins or the autophagic process for potential cancer therapy. Taken together, these findings would shed new light on exploiting the intricate mechanisms of autophagy and relevant small-molecule compounds as potential anti-cancer drugs to improve targeted cancer therapy.
Collapse
Affiliation(s)
- Jin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Yuxin Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Yi Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
361
|
Dai W, Chen W, Debnath B, Wu Y, Neamati N. Synthesis, Structure–Activity Relationship Studies, and ADMET Properties of 3‐Aminocyclohex‐2‐en‐1‐ones as Chemokine Receptor 2 (CXCR2) Antagonists. ChemMedChem 2018; 13:916-930. [PMID: 29493096 DOI: 10.1002/cmdc.201800027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/23/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Weiyang Dai
- Department of Medicinal Chemistry, College of Pharmacy University of Michigan 1600 Huron Parkway Ann Arbor MI USA
- Key Laboratory of Drug Targeting and Drug Delivery, System of Ministry of Education, West China School of Pharmacy Sichuan University No. 17 People's South Road Chengdu 610041 P.R. China
| | - Wenmin Chen
- Department of Medicinal Chemistry, College of Pharmacy University of Michigan 1600 Huron Parkway Ann Arbor MI USA
| | - Bikash Debnath
- Department of Medicinal Chemistry, College of Pharmacy University of Michigan 1600 Huron Parkway Ann Arbor MI USA
| | - Yong Wu
- Key Laboratory of Drug Targeting and Drug Delivery, System of Ministry of Education, West China School of Pharmacy Sichuan University No. 17 People's South Road Chengdu 610041 P.R. China
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy University of Michigan 1600 Huron Parkway Ann Arbor MI USA
| |
Collapse
|
362
|
Toor SM, Elkord E. Therapeutic prospects of targeting myeloid-derived suppressor cells and immune checkpoints in cancer. Immunol Cell Biol 2018; 96:888-897. [PMID: 29635843 DOI: 10.1111/imcb.12054] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 12/28/2022]
Abstract
Immune evasion is a characteristic of most human malignancies and is induced via various mechanisms. Immunosuppressive cells, including myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg), are key mediators in assisting tumors to escape immune surveillance. Expansion of MDSC, Treg and elevated levels of immune checkpoints (IC) are frequently detected in the tumor microenvironment and periphery of cancer patients. Various therapeutic agents have been shown to target MDSC and to block IC for inducing anti-tumor immunity and reversal of tumor immune escape. Importantly, some recent studies have shown that MDSC targeting improves the efficacy of IC blockade in cancer therapy. However, there is a pressing need to improve our understanding of the distinct role of these cells to develop combination therapy that attacks tumor cells from all frontiers to improve cancer therapeutics. Herein, we discuss the role of MDSC in cancer progression, interactions with IC in the context of anti-cancer immunity and the current therapeutic strategies to target MDSC and block IC in cancer.
Collapse
Affiliation(s)
- Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute, College of Science and Engineering, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Institute of Cancer Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
363
|
Zhang Y, Wu L, Li Z, Zhang W, Luo F, Chu Y, Chen G. Glycocalyx-Mimicking Nanoparticles Improve Anti-PD-L1 Cancer Immunotherapy through Reversion of Tumor-Associated Macrophages. Biomacromolecules 2018; 19:2098-2108. [DOI: 10.1021/acs.biomac.8b00305] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Yufei Zhang
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Libin Wu
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Zhen Li
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Weiyi Zhang
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Guosong Chen
- Department of Macromolecular Science, The State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, China
| |
Collapse
|
364
|
Huck BR, Kötzner L, Urbahns K. Kleine Moleküle, ganz groß: niedermolekulare immunonkologische Kombinationstherapien. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201707816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Bayard R. Huck
- Healthcare R&D, Discovery Technologies, Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| | - Lisa Kötzner
- Healthcare R&D, Discovery Technologies, Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| | - Klaus Urbahns
- Healthcare R&D, Discovery Technologies, Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| |
Collapse
|
365
|
Huck BR, Kötzner L, Urbahns K. Small Molecules Drive Big Improvements in Immuno-Oncology Therapies. Angew Chem Int Ed Engl 2018; 57:4412-4428. [PMID: 28971564 PMCID: PMC5900885 DOI: 10.1002/anie.201707816] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Indexed: 12/16/2022]
Abstract
Immuno-oncology therapies have the potential to revolutionize the armamentarium of available cancer treatments. To further improve clinical response rates, researchers are looking for novel combination regimens, with checkpoint blockade being used as a backbone of the treatment. This Review highlights the significance of small molecules in this approach, which holds promise to provide increased benefit to cancer patients.
Collapse
Affiliation(s)
- Bayard R. Huck
- Healthcare R&D, Discovery Technologies, Merck KGaAFrankfurter Strasse 25064293DarmstadtGermany
| | - Lisa Kötzner
- Healthcare R&D, Discovery Technologies, Merck KGaAFrankfurter Strasse 25064293DarmstadtGermany
| | - Klaus Urbahns
- Healthcare R&D, Discovery Technologies, Merck KGaAFrankfurter Strasse 25064293DarmstadtGermany
| |
Collapse
|
366
|
Attili I, Karachaliou N, Bonanno L, Berenguer J, Bracht J, Codony-Servat J, Codony-Servat C, Ito M, Rosell R. STAT3 as a potential immunotherapy biomarker in oncogene-addicted non-small cell lung cancer. Ther Adv Med Oncol 2018; 10:1758835918763744. [PMID: 29636826 PMCID: PMC5888808 DOI: 10.1177/1758835918763744] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/15/2018] [Indexed: 12/27/2022] Open
Abstract
Immune checkpoint blockade has modified the treatment landscape for many types of tumors, including lung cancer. Still our knowledge on the biology of the interaction between tumor cells and the microenvironment is limited, preventing the optimal use of these new compounds and the maximum benefit that the patients can derive from them. We have actively worked on the role of STAT3, a transcriptional factor that causes innate resistance to targeted therapies in oncogene-addicted tumors. In this short review we take the opportunity to express our opinion and review existing knowledge on the immune role of STAT3 and the possible implications that this may have for the discovery of new biomarkers to predict response to immunotherapy, as well as new partners to combine with and increase the efficacy of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Ilaria Attili
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 53, Padova 35128, Italy
| | - Niki Karachaliou
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
- Instituto Oncológico Dr Rosell (IOR), University Hospital Sagrat Cor, Barcelona, Spain
| | | | - Jordi Berenguer
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Jillian Bracht
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Jordi Codony-Servat
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Carles Codony-Servat
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
| | - Masaoki Ito
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Rafael Rosell
- Coyote Research Group, Pangaea Oncology, Laboratory of Molecular Biology, Quirón-Dexeus University Institute, Barcelona, Spain
- Instituto Oncológico Dr Rosell (IOR), Quirón-Dexeus University Institute, Barcelona, Spain
- Institut d’Investigació en Ciències Germans Trias i Pujol, Badalona, Spain
- Institut Català d’Oncologia, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
367
|
Profiling Prostate Cancer Therapeutic Resistance. Int J Mol Sci 2018; 19:ijms19030904. [PMID: 29562686 PMCID: PMC5877765 DOI: 10.3390/ijms19030904] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 03/16/2018] [Accepted: 03/16/2018] [Indexed: 02/06/2023] Open
Abstract
The major challenge in the treatment of patients with advanced lethal prostate cancer is therapeutic resistance to androgen-deprivation therapy (ADT) and chemotherapy. Overriding this resistance requires understanding of the driving mechanisms of the tumor microenvironment, not just the androgen receptor (AR)-signaling cascade, that facilitate therapeutic resistance in order to identify new drug targets. The tumor microenvironment enables key signaling pathways promoting cancer cell survival and invasion via resistance to anoikis. In particular, the process of epithelial-mesenchymal-transition (EMT), directed by transforming growth factor-β (TGF-β), confers stem cell properties and acquisition of a migratory and invasive phenotype via resistance to anoikis. Our lead agent DZ-50 may have a potentially high efficacy in advanced metastatic castration resistant prostate cancer (mCRPC) by eliciting an anoikis-driven therapeutic response. The plasticity of differentiated prostate tumor gland epithelium allows cells to de-differentiate into mesenchymal cells via EMT and re-differentiate via reversal to mesenchymal epithelial transition (MET) during tumor progression. A characteristic feature of EMT landscape is loss of E-cadherin, causing adherens junction breakdown, which circumvents anoikis, promoting metastasis and chemoresistance. The targetable interactions between androgens/AR and TGF-β signaling are being pursued towards optimized therapeutic regimens for the treatment of mCRPC. In this review, we discuss the recent evidence on targeting the EMT-MET dynamic interconversions to overcome therapeutic resistance in patients with recurrent therapeutically resistant prostate cancer. Exploitation of the phenotypic landscape and metabolic changes that characterize the prostate tumor microenvironment in advanced prostate cancer and consequential impact in conferring treatment resistance are also considered in the context of biomarker discovery.
Collapse
|
368
|
Che JX, Wang ZL, Dong XW, Hu YH, Xie X, Hu YZ. Bicyclo[2.2.1]heptane containing N, N'-diarylsquaramide CXCR2 selective antagonists as anti-cancer metastasis agents. RSC Adv 2018; 8:11061-11069. [PMID: 35541503 PMCID: PMC9078949 DOI: 10.1039/c8ra01806e] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/09/2018] [Indexed: 11/21/2022] Open
Abstract
CXCR1 and CXCR2 are CXC chemokine receptors (CXCRs), corresponding to cytokines of the CXC chemokine family. CXCR2 was found to be 77% homologous to CXCR1. Antagonism of the chemokine receptor CXCR2 has been proposed as a new strategy for the treatment of metastatic cancer. In order to find a CXCR2 selective antagonist, a bicyclo[2.2.1]heptane containing N,N'-diarylsquaramide (compound 2e) was identified by introducing a bridge ring system into the N,N'-diarylsquaramide skeleton, and it exhibited good CXCR2 antagonistic activity (CXCR2IC50 = 48 nM) and good selectivity (CXCR1IC50/CXCR2IC50 = 60.4). Furthermore, an in vitro biological assay of compound 2e also demonstrated its good anti-cancer metastatic effect against the pancreatic cancer cell line CFPAC1. In addition, compound 2e showed an extremely high stability in simulated intestinal fluid (SIF) and simulated gastric fluid (SGF), as well as in rat and human plasma, but not in rat and human liver microsomes. In vivo pharmacokinetic studies in rats indicated that 2e has an excellent PK profile (10 mg kg-1 po, C max = 2863 ng mL-1, t 1/2 = 2.58 h). Moreover, molecular docking was further implemented to propose the preponderant configuration of compound 2e, providing important and useful guidelines for further development.
Collapse
Affiliation(s)
- Jin-Xin Che
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University Hangzhou PR China
| | - Zhi-Long Wang
- State Key Laborarory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai PR China
| | - Xiao-Wu Dong
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University Hangzhou PR China
| | - You-Hong Hu
- State Key Laborarory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai PR China
| | - Xin Xie
- State Key Laborarory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai PR China
- CAS Key Laboratory of Receptor Research, The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai PR China
| | - Yong-Zhou Hu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, College of Pharmaceutical Sciences, Zhejiang University Hangzhou PR China
| |
Collapse
|
369
|
Dosset M, Vargas TR, Lagrange A, Boidot R, Végran F, Roussey A, Chalmin F, Dondaine L, Paul C, Lauret Marie-Joseph E, Martin F, Ryffel B, Borg C, Adotévi O, Ghiringhelli F, Apetoh L. PD-1/PD-L1 pathway: an adaptive immune resistance mechanism to immunogenic chemotherapy in colorectal cancer. Oncoimmunology 2018; 7:e1433981. [PMID: 29872568 PMCID: PMC5980491 DOI: 10.1080/2162402x.2018.1433981] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 01/08/2023] Open
Abstract
Background: Chemotherapy is currently evaluated in order to enhance the efficacy of immune checkpoint blockade (ICB) therapy in colorectal cancer. However, the mechanisms by which these drugs could synergize with ICB remains unclear. The impact of chemotherapy on the PD-1/PD-L1 pathway and the resulting anticancer immune responses was assessed in two mouse models of colorectal cancer and validated in tumor samples from metastatic colorectal cancer patients that received neoadjuvant treatment. We demonstrated that 5-Fluorouracil plus Oxaliplatin (Folfox) drove complete tumor cure in mice when combined to anti-PD-1 treatment, while each monotherapy failed. This synergistic effect relies on the ability of Folfox to induce tumor infiltration by activated PD-1+ CD8 T cells in a T-bet dependent manner. This effect was concomitantly associated to the expression of PD-L1 on tumor cells driven by IFN-γ secreted by PD-1+ CD8 T cells, indicating that Folfox triggers tumor adaptive immune resistance. Finally, we observed an induction of PD-L1 expression and high CD8 T cell infiltration in the tumor microenvironment of colorectal cancer patients treated by Folfox regimen. Our study delineates a molecular pathway involved in Folfox-induced adaptive immune resistance in colorectal cancer. The results strongly support the use of immune checkpoint blockade therapy in combination with chemotherapies like Folfox.
Collapse
Affiliation(s)
- Magalie Dosset
- INSERM, U1231, Dijon, France.,INSERM, U1098, Besançon, France.,LabEx LipSTIC, Besançon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Thaiz Rivera Vargas
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Anaïs Lagrange
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Romain Boidot
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Frédérique Végran
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Aurélie Roussey
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Fanny Chalmin
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Lucile Dondaine
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Catherine Paul
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | | | - François Martin
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France
| | - Bernhard Ryffel
- University of Cape Town, RSA, CNRS, UMR7355, Orleans, France, IDM
| | - Christophe Borg
- INSERM, U1098, Besançon, France.,Université de Bourgogne Franche Comté, Dijon, France.,Department of Medical Oncology, University Hospital of Besançon, France
| | - Olivier Adotévi
- INSERM, U1098, Besançon, France.,Université de Bourgogne Franche Comté, Dijon, France.,Department of Medical Oncology, University Hospital of Besançon, France
| | - François Ghiringhelli
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| | - Lionel Apetoh
- INSERM, U1231, Dijon, France.,Université de Bourgogne Franche Comté, Dijon, France.,Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
370
|
Boibessot C, Toren P. Sex steroids in the tumor microenvironment and prostate cancer progression. Endocr Relat Cancer 2018; 25:R179-R196. [PMID: 29317479 DOI: 10.1530/erc-17-0493] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/30/2022]
Abstract
Prostate cancer is uniquely dependent on androgens. Despite years of research on the relationship between androgens and prostate cancer, many questions remain as to the biological effects of androgens and other sex steroids during prostate cancer progression. This article reviews the clinical and basic research on the influence of sex steroids such as androgens, estrogens and progesterone within the prostate tumor microenvironment on the progression of prostate cancer. We review clinical studies to date evaluating serum sex steroids as prognostic biomarkers and discuss their respective biological effects within the prostate tumor microenvironment. We also review the link between genomic alterations and sex steroid levels within prostate tumors. Finally, we highlight the links between sex steroid levels and the function of the immune system within the tumor microenvironment. As the context of treatment of lethal prostate cancer evolves over time, an understanding of this underlying biology remains central to developing optimal treatment approaches.
Collapse
Affiliation(s)
- Clovis Boibessot
- Department of SurgeryLaval University, Quebec City, Quebec, Canada
| | - Paul Toren
- Department of SurgeryLaval University, Quebec City, Quebec, Canada
| |
Collapse
|
371
|
Chauhan K, Hernandez-Meza JM, Rodríguez-Hernández AG, Juarez-Moreno K, Sengar P, Vazquez-Duhalt R. Multifunctionalized biocatalytic P22 nanoreactor for combinatory treatment of ER+ breast cancer. J Nanobiotechnology 2018; 16:17. [PMID: 29463260 PMCID: PMC5819296 DOI: 10.1186/s12951-018-0345-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/13/2018] [Indexed: 12/20/2022] Open
Abstract
Background Tamoxifen is the standard endocrine therapy for breast cancers, which require metabolic activation by cytochrome P450 enzymes (CYP). However, the lower and variable concentrations of CYP activity at the tumor remain major bottlenecks for the efficient treatment, causing severe side-effects. Combination nanotherapy has gained much recent attention for cancer treatment as it reduces the drug-associated toxicity without affecting the therapeutic response. Results Here we show the modular design of P22 bacteriophage virus-like particles for nanoscale integration of virus-driven enzyme prodrug therapy and photodynamic therapy. These virus capsids carrying CYP activity at the core are decorated with photosensitizer and targeting moiety at the surface for effective combinatory treatment. The estradiol-functionalized nanoparticles are recognized and internalized into ER+ breast tumor cells increasing the intracellular CYP activity and showing the ability to produce reactive oxygen species (ROS) upon UV365 nm irradiation. The generated ROS in synergy with enzymatic activity drastically enhanced the tamoxifen sensitivity in vitro, strongly inhibiting tumor cells. Conclusions This work clearly demonstrated that the targeted combinatory treatment using multifunctional biocatalytic P22 represents the effective nanotherapeutics for ER+ breast cancer. Electronic supplementary material The online version of this article (10.1186/s12951-018-0345-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kanchan Chauhan
- Department of Bionanotechnology, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km. 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico
| | - Juan M Hernandez-Meza
- Department of Bionanotechnology, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km. 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico
| | - Ana G Rodríguez-Hernández
- Department of Bionanotechnology, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km. 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico
| | - Karla Juarez-Moreno
- Department of Bionanotechnology, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km. 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico
| | - Prakhar Sengar
- Department of Bionanotechnology, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km. 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico
| | - Rafael Vazquez-Duhalt
- Department of Bionanotechnology, Centro de Nanociencias y Nanotecnología, Universidad Nacional Autónoma de México, Km. 107 carretera Tijuana-Ensenada, 22860, Ensenada, Baja California, Mexico.
| |
Collapse
|
372
|
Abstract
The clinical effectiveness of immunotherapies for prostate cancer remains subpar compared with that for other cancers. The goal of most immunotherapies is the activation of immune effectors, such as T cells and natural killer cells, as the presence of these activated mediators positively correlates with patient outcomes. Clinical evidence shows that prostate cancer is immunogenic, accessible to the immune system, and can be targeted by antitumour immune responses. However, owing to the detrimental effects of prostate-cancer-associated immunosuppression, even the newest immunotherapeutic approaches fail to initiate the clinically desired antitumour immune reaction. Oncolytic viruses, originally used for their preferential cancer-killing activity, are now being recognized for their ability to overturn cancer-associated immune evasion and promote otherwise absent antitumour immunity. This oncolytic-virus-induced subversion of tumour-associated immunosuppression can potentiate the effectiveness of current immunotherapeutics, including immune checkpoint inhibitors (for example, antibodies against programmed cell death protein 1 (PD1), programmed cell death 1 ligand 1 (PDL1), and cytotoxic T lymphocyte antigen 4 (CTLA4)) and chemotherapeutics that induce immunogenic cell death (for example, doxorubicin and oxaliplatin). Importantly, oncolytic-virus-induced antitumour immunity targets existing prostate cancer cells and also establishes long-term protection against future relapse. Hence, the strategic use of oncolytic viruses as monotherapies or in combination with current immunotherapies might result in the next breakthrough in prostate cancer immunotherapy.
Collapse
|
373
|
Fankhauser CD, Schüffler PJ, Gillessen S, Omlin A, Rupp NJ, Rueschoff JH, Hermanns T, Poyet C, Sulser T, Moch H, Wild PJ. Comprehensive immunohistochemical analysis of PD-L1 shows scarce expression in castration-resistant prostate cancer. Oncotarget 2018; 9:10284-10293. [PMID: 29535806 PMCID: PMC5828186 DOI: 10.18632/oncotarget.22888] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/15/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND We aimed to analyze the frequency and distribution of PD-L1 expression in specimens from prostate cancer (PC) patients using two different anti-PD-L1 antibodies (E1L3N, SP263). MATERIALS AND METHODS PD-L1 immunohistochemistry was performed in a tissue microarray consisting of 82 castration-resistant prostate cancer (CRPC) specimens, 70 benign prostate hyperplasia (BPH) specimens, 96 localized PC cases, and 3 PC cell lines, using two different antibodies (clones E1L3N, and SP263). Staining images for CD4, CD8, PD-L1, and PanCK of a single PD-L1 positive case were compared, using a newly developed dot-wise correlation method for digital images to objectively test for co-expression. RESULTS Depending on the antibody used, in tumor cells (TC) only five (E1L3N: 6%) and three (SP263: 3.7%) samples were positive. In infiltrating immune cells (IC) 12 (SP263: 14.6%) and 8 (E1L3N: 9.9%) specimens showed PD-L1 expression. Two PC cell lines (PC3, LnCaP) also displayed membranous immunoreactivity. All localized PCs or BPH samples tested were negative. Dot-wise digital correlation of expression patterns revealed a moderate positive correlation between PD-L1 and PanCK expression, whereas both PanCK and PD-L1 showed a weak negative Pearson correlation coefficient between CD4 and CD8. CONCLUSIONS PD-L1 was not expressed in localized PC or BPH, and was only found in a minority of CRPC tumors and infiltrating immune cells. Protein expression maps and systematic dot-wise comparison could be a useful objective way to describe the relationship between immuno- and tumor-related proteins in the future, without the need to develop multiplex staining methods.
Collapse
Affiliation(s)
- Christian D. Fankhauser
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter J. Schüffler
- Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Silke Gillessen
- Department of Medical Oncology and Hematology, Cantonal Hospital, St. Gallen, Switzerland
| | - Aurelius Omlin
- Department of Medical Oncology and Hematology, Cantonal Hospital, St. Gallen, Switzerland
| | - Niels J. Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jan H. Rueschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cedric Poyet
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter J. Wild
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
374
|
Lu H, Yang T, Xu Z, Lin X, Ding Q, Zhang Y, Cai X, Dong K, Gong S, Zhang W, Patel M, Copley RCB, Xiang J, Guan X, Wren P, Ren F. Discovery of Novel 1-Cyclopentenyl-3-phenylureas as Selective, Brain Penetrant, and Orally Bioavailable CXCR2 Antagonists. J Med Chem 2018; 61:2518-2532. [DOI: 10.1021/acs.jmedchem.7b01854] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Hongfu Lu
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Ting Yang
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Zhongmiao Xu
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Xichen Lin
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Qian Ding
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Yueting Zhang
- R&D Projects Clinical Platforms and Sciences, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Xin Cai
- Platform Technology Sciences, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Kelly Dong
- Platform Technology Sciences, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Sophie Gong
- Platform Technology Sciences, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Wei Zhang
- Platform Technology Sciences, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Metul Patel
- Platform Technology Sciences, GSK Pharmaceuticals R&D, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Royston C. B. Copley
- Platform Technology & Science, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Jianing Xiang
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Xiaoming Guan
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| | - Paul Wren
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 1250 South Collegeville Road, Collegeville, Pennsylvania 19426, United States
| | - Feng Ren
- Neurosciences Therapeutic Area Unit, GSK Pharmaceuticals R&D, 898 Halei Road, Zhangjiang Hi-Tech Park, Pudong, Shanghai 201203, P. R. China
| |
Collapse
|
375
|
Suleman S, Wei GH. Combined immunotherapy for advanced prostate cancer: Empowering the T cell army. Asian J Urol 2018; 4:199-200. [PMID: 29387551 PMCID: PMC5772898 DOI: 10.1016/j.ajur.2017.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Sufyan Suleman
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Gong-Hong Wei
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| |
Collapse
|
376
|
Bezzi M, Seitzer N, Ishikawa T, Reschke M, Chen M, Wang G, Mitchell C, Ng C, Katon J, Lunardi A, Signoretti S, Clohessy JG, Zhang J, Pandolfi PP. Diverse genetic-driven immune landscapes dictate tumor progression through distinct mechanisms. Nat Med 2018; 24:165-175. [PMID: 29309058 DOI: 10.1038/nm.4463] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 11/29/2017] [Indexed: 12/23/2022]
Abstract
Multiple immune-cell types can infiltrate tumors and promote progression and metastasis through different mechanisms, including immunosuppression. How distinct genetic alterations in tumors affect the composition of the immune landscape is currently unclear. Here, we characterized the immune-cell composition of prostate cancers driven by the loss of the critical tumor suppressor gene Pten, either alone or in combination with the loss of Trp53, Zbtb7a or Pml. We observed a striking quantitative and qualitative heterogeneity that was directly dependent on the specific genetic events in the tumor and ranged from 'cold', noninflamed tumors to massively infiltrated landscapes-results with important therapeutic implications. Further, we showed these qualitative differences in transcriptomic analysis of human prostate cancer samples. These data suggest that patient stratification on the basis of integrated genotypic-immunophenotypic analyses may be necessary for successful clinical trials and tailored precision immunological therapies.
Collapse
Affiliation(s)
- Marco Bezzi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nina Seitzer
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Tomoki Ishikawa
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Markus Reschke
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ming Chen
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Guocan Wang
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Mitchell
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher Ng
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jesse Katon
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea Lunardi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - John G Clohessy
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
- Preclinical Murine Pharmacogenetics Facility, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jiangwen Zhang
- School of Biological Sciences, University of Hong Kong, Hong Kong SAR, China
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
377
|
Myeloid-derived suppressor cells coming of age. Nat Immunol 2018; 19:108-119. [PMID: 29348500 DOI: 10.1038/s41590-017-0022-x] [Citation(s) in RCA: 1234] [Impact Index Per Article: 205.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/07/2017] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells generated during a large array of pathologic conditions ranging from cancer to obesity. These cells represent a pathologic state of activation of monocytes and relatively immature neutrophils. MDSCs are characterized by a distinct set of genomic and biochemical features, and can, on the basis of recent findings, be distinguished by specific surface molecules. The salient feature of these cells is their ability to inhibit T cell function and thus contribute to the pathogenesis of various diseases. In this Review, we discuss the origin and nature of these cells; their distinctive features; and their biological roles in cancer, infectious diseases, autoimmunity, obesity and pregnancy.
Collapse
|
378
|
Shi F, Zhang J, Liu H, Wu L, Jiang H, Wu Q, Liu T, Lou M, Wu H. The dual PI3K/mTOR inhibitor dactolisib elicits anti-tumor activity in vitro and in vivo. Oncotarget 2018; 9:706-717. [PMID: 29416647 PMCID: PMC5787502 DOI: 10.18632/oncotarget.23091] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/01/2017] [Indexed: 11/25/2022] Open
Abstract
Glioblastomas (GBMs) are among the most malignant of all human tumors and have poor prognosis. The current standard of care (SOC) includes maximal surgical tumor resection followed by adjuvant temozolomide (TMZ) and concomitant radiotherapy (RT). However, even with this treatment, the 5-year survival rate is less than 10%, and thus, follow-up treatment is required to improve efficacy. In GBMs as well as many other solid cancers, PI3K/mTOR signaling is overactivated. Therefore, multiple tumor-based PI3K inhibitors have been studied in various cancers. In the current study, we investigated the effect of the dual PI3K/mTOR inhibitor dactolisib on TMZ+RT treatment in three human GBM cell lines and a orthotopic xenograft model. Dactolisib alone induced cytotoxicity and pro-apoptotic effects, which act as antitumor factors. Combined with SOC treatment, dactolisib inhibited cell viability, induced enhanced pro-apoptotic effect, and attenuated migration/invasion in all three cell lines, thereby enhancing the SOC therapeutic effect. Protein microarray analysis showed that A172 cells treated with TMZ+RT+dactolisib had higher p27 and lower Bcl-2 expression than other groups. Moreover, in the xenograft model, oral dactolisib combined with TMZ+RT inhibited tumor growth and prolonged survival. Thus, SOC combined with dactolisib shows potent anti-tumor activity and has promising potential for solid tumor treatment.
Collapse
Affiliation(s)
- Fei Shi
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Jinying Zhang
- Institute of Basic Medicine Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Hongyu Liu
- Key Laboratory of Cancer Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Liangliang Wu
- Key Laboratory of Cancer Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Hongyu Jiang
- Department of Anesthesiology, Wuxi Third People’s Hospital, Wuxi, Jiangsu 214000, China
| | - Qiyan Wu
- Key Laboratory of Cancer Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Tianyi Liu
- Key Laboratory of Cancer Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| |
Collapse
|
379
|
Plourde G. Case Report # 8—Treatment Modalities in Development. Prostate Cancer 2018. [DOI: 10.1016/b978-0-12-815966-8.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
380
|
|
381
|
Synergistic Effects of Cabozantinib and EGFR-Specific CAR-NK-92 Cells in Renal Cell Carcinoma. J Immunol Res 2017; 2017:6915912. [PMID: 29423418 PMCID: PMC5750507 DOI: 10.1155/2017/6915912] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 09/17/2017] [Accepted: 10/08/2017] [Indexed: 12/16/2022] Open
Abstract
The chimeric antigen receptor-modified immune effector cell (CAR-T and CAR-NK) therapies are newly developed adoptive treatments of cancers. However, their therapeutic efficacy against solid tumors is limited. Combining CAR-T or CAR-NK cells with chemotherapeutic drugs to treat solid tumor may be a promising strategy. We developed an epidermal growth factor- (EGFR-) specific third-generation CAR. NK-92 cells were modified with the CAR by lentivirus infection. The specific killing ability of the CAR-modified NK-92 cells (CAR-NK-92) against renal cell carcinoma (RCC) cell lines was confirmed in vitro. The synergistic effects of cabozantinib and EGFR-specific CAR-NK-92 cells were investigated in vitro and in vivo. Our results showed that the CAR-NK-92 cells lyse RCC cells in an EGFR-specific manner. Treatment with cabozantinib could increase EGFR and decrease PD-L1 membrane surface expression in RCC cells and enhance the killing ability of CAR-NK-92 cells against the RCC cells in vitro. Furthermore, the CAR-NK-92 cells show synergistic therapeutic efficacy with cabozantinib against human RCC xenograft models. Our results provided the basis for combination with chemotherapy as a novel strategy for enhancing the therapeutic efficacy of CAR-modified immune effector cells for solid tumors.
Collapse
|
382
|
De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol 2017; 18:e731-e741. [PMID: 29208439 DOI: 10.1016/s1470-2045(17)30607-1] [Citation(s) in RCA: 491] [Impact Index Per Article: 70.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/19/2022]
Abstract
Use of immune checkpoint inhibitors targeting the programmed cell death protein-1/programmed cell death-ligand 1 and cytotoxic T lymphocyte-associated protein-4 axes has yielded impressive results in some clinical trials. However, only a subset of patients initially respond to these inhibitors, and increasing clinical evidence indicates that a substantial proportion of initial responders ultimately relapse with lethal, drug-resistant disease months or years later. Studies that have used massively parallel sequencing have shed light on the rich functional landscape of mutations that endow tumour cells with the ability to evade T-cell-mediated immunosurveillance. Cancer genomes bear signatures of clonal evolution and selection, particularly implicating acquired defects in interferon receptor signalling and antigen presentation. In this Review, we discuss the biological processes that operate in the formation of so-called immunoresistant niches, and describe the latest progress in the development of combination strategies to reinstate immunosurveillance in immune-refractory tumours.
Collapse
|
383
|
Wang C, Peng G, Huang H, Liu F, Kong DP, Dong KQ, Dai LH, Zhou Z, Wang KJ, Yang J, Cheng YQ, Gao X, Qu M, Wang HR, Zhu F, Tian QQ, Liu D, Cao L, Cui XG, Xu CL, Xu DF, Sun YH. Blocking the Feedback Loop between Neuroendocrine Differentiation and Macrophages Improves the Therapeutic Effects of Enzalutamide (MDV3100) on Prostate Cancer. Clin Cancer Res 2017; 24:708-723. [PMID: 29191973 DOI: 10.1158/1078-0432.ccr-17-2446] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/22/2017] [Accepted: 11/20/2017] [Indexed: 11/16/2022]
Affiliation(s)
- Chao Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
- Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Guang Peng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hai Huang
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Fei Liu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - De-Pei Kong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Ke-Qin Dong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li-He Dai
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhe Zhou
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kai-Jian Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jun Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yan-Qiong Cheng
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xu Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Min Qu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hong-Ru Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Feng Zhu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qin-Qin Tian
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dan Liu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Li Cao
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of the Ministry of Education, Second Military Medical University, Shanghai, China
| | - Xin-Gang Cui
- Department of Urinary Surgery, Third Affiliated Hospital, Second Military Medical University, Shanghai, China
| | - Chuan-Liang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Dan-Feng Xu
- Department of Urinary Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Ying-Hao Sun
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
384
|
Owen KL, Parker BS. Beyond the vicious cycle: The role of innate osteoimmunity, automimicry and tumor-inherent changes in dictating bone metastasis. Mol Immunol 2017; 110:57-68. [PMID: 29191489 DOI: 10.1016/j.molimm.2017.11.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/24/2017] [Indexed: 12/12/2022]
Abstract
Bone metastasis is a fatal consequence of a subset of solid malignancies that fail to respond to conventional therapies. While a myriad of factors contribute to osteotropism and disseminated cell survival and outgrowth in bone, efforts to inhibit tumor cell growth in the bone-metastatic niche have largely relied on measures that disrupt the bi-directional interactions between bone resident and tumor cells. However, the targeting of isolated stromal interactions has proven ineffective to date in inhibiting bone-metastatic progression and patient mortality. Osteoimmune regulation is now emerging as a critical determinant of metastatic growth in the bone microenvironment. While this has highlighted the importance of innate immune populations in dictating the temporal development of overt bone metastases, the osteoimmunological processes that underpin tumor cell progression in bone remain severely underexplored. Along with tumor-intrinsic alterations that occur specifically within the bone microenvironment, innate osteoimmunological crosstalk poses an exciting area of future discovery and therapeutic development. Here we review current knowledge of the unique exchange that occurs between bone resident cells, innate immune populations and tumor cells that leads to the establishment of a tumor-permissive milieu.
Collapse
Affiliation(s)
- Katie L Owen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
385
|
Tripathi M, Nandana S, Billet S, Cavassani KA, Mishra R, Chung LW, Posadas EM, Bhowmick NA. Modulation of cabozantinib efficacy by the prostate tumor microenvironment. Oncotarget 2017; 8:87891-87902. [PMID: 29152128 PMCID: PMC5675680 DOI: 10.18632/oncotarget.21248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is increasingly recognized as the arbiter of metastatic progression and drug resistance in advanced prostate cancer (PCa). Cabozantinib is a potent tyrosine kinase inhibitor (TKI) with reported biological activity in the PCa epithelia, but failed to provide an overall survival benefit in phase 3 clinical trials. However, the promising biologic efficacy of the drug in early trials warranted a better understanding of the mechanism of action, with the goal of improving patient selection for TKI-based therapy such as cabozantinib. We found a 100-fold lower cabozantinib IC50 in macrophages, PCa associated fibroblasts, and bone marrow fibroblasts compared to PCa epithelia. In PCa mouse models, pre-treatment with cabozantinib potentiated osseous and visceral tumor engraftment, suggesting a pro-tumorigenic host response to the drug. We further found that the host effects of cabozantinib impacted bone turnover, but not necessarily tumor expansion. Cabozantinib affected M1 macrophage polarization in mice. Analogously, circulating monocytes from PCa patients treated with cabozantinib, demonstrated a striking correlation of monocyte reprograming with therapeutic bone responsivity, to support patient selection at early stages of treatment. Thus, a re-evaluation of TKI-based therapeutic strategies in PCa can be considered for suitable patient populations based on TME responses.
Collapse
Affiliation(s)
- Manisha Tripathi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Srinivas Nandana
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Sandrine Billet
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Karen A. Cavassani
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Rajeev Mishra
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Leland W.K. Chung
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Edwin M. Posadas
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Neil A. Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
- Department of Research, Greater Los Angeles Veterans Administration, Los Angeles, California 90048, USA
| |
Collapse
|
386
|
Karachaliou N, Gonzalez-Cao M, Sosa A, Berenguer J, Bracht JWP, Ito M, Rosell R. The combination of checkpoint immunotherapy and targeted therapy in cancer. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:388. [PMID: 29114546 PMCID: PMC5653508 DOI: 10.21037/atm.2017.06.47] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 06/08/2017] [Indexed: 02/05/2023]
Abstract
The therapeutic possibilities for patients with metastatic melanoma have changed due to the development of targeted therapies that inhibit oncogenic signaling pathways as well as immune modulating therapies that unleash the patient antitumor immunity. These therapeutic changes have impressively increased the median overall survival of the patients. Considering the dramatic but transient responses that occur with targeted therapies for a subgroup of patients and the durable responses that can be achieved with immunotherapy in a subset of patients, a lot of effort is ongoing for the clinical development of combinations of these two therapeutic approaches. Herein we discuss the existing preclinical and clinical data for the combination of targeted therapies and immunotherapy focusing mainly on melanoma and non-small cell lung cancer (NSCLC).
Collapse
Affiliation(s)
- Niki Karachaliou
- Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor, Barcelona, Spain
| | - Maria Gonzalez-Cao
- Institute of Oncology Rosell (IOR), Quirón-Dexeus University Institute, Barcelona, Spain
| | - Aaron Sosa
- Institute of Oncology Rosell (IOR), University Hospital Sagrat Cor, Barcelona, Spain
| | - Jordi Berenguer
- Pangaea Oncology, Quirón-Dexeus University Institute, Barcelona, Spain
| | | | - Masaoki Ito
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Rafael Rosell
- Germans Trias i Pujol Research Institute, Badalona, Spain
- Catalan Institute of Oncology, Germans Trias i Pujol University Hospital, Badalona, Spain
| |
Collapse
|
387
|
Zhang B, Chen F, Xu Q, Han L, Xu J, Gao L, Sun X, Li Y, Li Y, Qian M, Sun Y. Revisiting ovarian cancer microenvironment: a friend or a foe? Protein Cell 2017; 9:674-692. [PMID: 28929459 PMCID: PMC6053350 DOI: 10.1007/s13238-017-0466-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/21/2017] [Indexed: 02/06/2023] Open
Abstract
Development of ovarian cancer involves the co-evolution of neoplastic cells together with the adjacent microenvironment. Steps of malignant progression including primary tumor outgrowth, therapeutic resistance, and distant metastasis are not determined solely by genetic alterations in ovarian cancer cells, but considerably shaped by the fitness advantage conferred by benign components in the ovarian stroma. As the dynamic cancer topography varies drastically during disease progression, heterologous cell types within the tumor microenvironment (TME) can actively determine the pathological track of ovarian cancer. Resembling many other solid tumor types, ovarian malignancy is nurtured by a TME whose dark side may have been overlooked, rather than overestimated. Further, harnessing breakthrough and targeting cures in human ovarian cancer requires insightful understanding of the merits and drawbacks of current treatment modalities, which mainly target transformed cells. Thus, designing novel and precise strategies that both eliminate cancer cells and manipulate the TME is increasingly recognized as a rational avenue to improve therapeutic outcome and prevent disease deterioration of ovarian cancer patients.
Collapse
Affiliation(s)
- Boyi Zhang
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Fei Chen
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qixia Xu
- Institute of Health Sciences, Shanghai Jiao Tong University, School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Liu Han
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiaqian Xu
- Institute of Health Sciences, Shanghai Jiao Tong University, School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Libin Gao
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiaochen Sun
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yiwen Li
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yan Li
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Min Qian
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Sun
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
388
|
Shinko D, Diakos CI, Clarke SJ, Charles KA. Cancer-Related Systemic Inflammation: The Challenges and Therapeutic Opportunities for Personalized Medicine. Clin Pharmacol Ther 2017; 102:599-610. [PMID: 28699186 DOI: 10.1002/cpt.789] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/23/2017] [Accepted: 07/06/2017] [Indexed: 12/15/2022]
Abstract
Over the last decade there has been significant progress towards the development of personalized or "precision" medicine for many patients with cancer. However, there still remain subpopulations of cancer patients that do not possess a tumor mutation profile that is successfully targeted by the newer molecular anticancer drugs and further personalized approaches are needed. The presence of cancer-related systemic inflammation represents an underappreciated subpopulation of cancer patients needing personalized therapy. For ∼25% of all advanced cancer patients, regardless of histological subtype, the patients with systemic inflammation have significantly poorer response to chemotherapy and also shorter overall survival compared to those cancer patients without inflammation. The development of cancer-related systemic inflammation involves interactions between host and tumor cells that are potential new drug targets in cancer chemotherapy. In this review we discuss the challenges and clinical opportunities to develop new therapeutic strategies for this underappreciated drug target.
Collapse
Affiliation(s)
- Diana Shinko
- Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Connie I Diakos
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Bill Walsh Translational Research Laboratories, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Stephen J Clarke
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia.,Bill Walsh Translational Research Laboratories, Kolling Institute of Medical Research, St Leonards, NSW, Australia
| | - Kellie A Charles
- Discipline of Pharmacology, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
389
|
Gibbons Johnson RM, Dong H. Functional Expression of Programmed Death-Ligand 1 (B7-H1) by Immune Cells and Tumor Cells. Front Immunol 2017; 8:961. [PMID: 28848559 PMCID: PMC5554355 DOI: 10.3389/fimmu.2017.00961] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/27/2017] [Indexed: 01/05/2023] Open
Abstract
The programmed death-1 (PD-1) and its ligand PD-L1 (B7-H1) signaling pathway has been the focus of much enthusiasm in the fields of tumor immunology and oncology with recent FDA approval of the anti-PD-1 antibodies pembrolizumab and nivolumab and the anti-PD-L1 antibodies durvalumab, atezolimuab, and avelumab. These therapies, referred to here as PD-L1/PD-1 checkpoint blockade therapies, are designed to block the interaction between PD-L1, expressed by tumor cells, and PD-1, expressed by tumor-infiltrating CD8+ T cells, leading to enhanced antitumor CD8+ T cell responses and tumor regression. The influence of PD-L1 expressed by tumor cells on antitumor CD8+ T cell responses is well characterized, but the impact of PD-L1 expressed by immune cells has not been well defined for antitumor CD8+ T cell responses. Although PD-L1 expression by tumor cells has been used as a biomarker in selection of patients for PD-L1/PD-1 checkpoint blockade therapies, patients whose tumor cells lack PD-L1 expression often respond positively to PD-L1/PD-1 checkpoint blockade therapies. This suggests that PD-L1 expressed by non-malignant cells may also contribute to antitumor immunity. Here, we review the functions of PD-L1 expressed by immune cells in the context of CD8+ T cell priming, contraction, and differentiation into memory populations, as well as the role of PD-L1 expressed by tumor cells in regulating antitumor CD8+ T cell responses.
Collapse
Affiliation(s)
| | - Haidong Dong
- Department of Urology, College of Medicine, Mayo Clinic, Rochester, MN, United States.,Department of Immunology, College of Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
390
|
Green M, Feng FY, Mehra R, Spratt DE. Convergence of immunotherapy, radiotherapy and prostate cancer: challenges and opportunities. Immunotherapy 2017; 9:695-699. [PMID: 28771102 DOI: 10.2217/imt-2017-0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Michael Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel E Spratt
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
391
|
Raddaoui N, Stazzoni S, Möckl L, Viverge B, Geiger F, Engelke H, Bräuchle C, Carell T. Dendrimer-Based Signal Amplification of Click-Labelled DNA in Situ. Chembiochem 2017. [DOI: 10.1002/cbic.201700209] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Nada Raddaoui
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Samuele Stazzoni
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Leonhard Möckl
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Bastien Viverge
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Florian Geiger
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Hanna Engelke
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Christoph Bräuchle
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| | - Thomas Carell
- Center for Integrated Protein Science (CiPSM); Department of Chemistry; LMU München; Butenandtstrasse 5-13 81377 München Germany
| |
Collapse
|
392
|
Overcoming Oncogenic Mediated Tumor Immunity in Prostate Cancer. Int J Mol Sci 2017; 18:ijms18071542. [PMID: 28714919 PMCID: PMC5536030 DOI: 10.3390/ijms18071542] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 06/26/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is being tested intensively in clinical trials for prostate cancer; it includes immune checkpoint inhibition, prostate specific antigen (PSA) vaccines and dendritic cell-based strategies. Despite increasing evidence for clinical responses, the consensus of multiple trials is that prostate cancers are poorly responsive to immunotherapy. Prostate cancer has a high degree of pathological and genetic heterogeneity compared to other cancer types, which may account for immunotherapeutic resistance. This hypothesis also implies that select types of prostate tumors may be differentially responsive to immune-based strategies and that the clinical stage, pathological grade and underlying genetic landscape may be important criteria in identifying tumors that respond to immune therapies. One strategy is to target oncogenic driver pathways in combination with immunotherapies with the goal of overcoming tumor immunity and broadening the number of patients achieving a clinical response. In this analysis, we address the hypothesis that driver oncogenic signaling pathways regulate cancer progression, tumor immunity and resistance to current immune therapeutics in prostate cancer. We propose that increased responsiveness may be achieved through the combined use of immunotherapies and inhibitors targeting tumor cell autonomous pathways that contribute towards anti-tumor immunity in patients with prostate cancer.
Collapse
|
393
|
Rationally combining immunotherapies to improve efficacy of immune checkpoint blockade in solid tumors. Cytokine Growth Factor Rev 2017; 36:5-15. [PMID: 28693973 DOI: 10.1016/j.cytogfr.2017.06.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/13/2022]
Abstract
With the widespread application of immune checkpoint blocking antibodies (ICBs) for the treatment of advanced cancer, immunotherapy has proven to be capable of yielding unparalleled clinical results. However, despite the initial success of ICB-treatment, still a minority of patients experience durable responses to ICB therapy. A plethora of mechanisms underlie ICB resistance ranging from low immunogenicity, inadequate generation or recruitment of tumor-specific T cells or local suppression by stromal cells to acquired genetic alterations leading to immune escape. Increasing the response rates to ICBs requires insight into the mechanisms underlying resistance and the subsequent design of rational therapeutic combinations on a per patient basis. In this review, we aim to establish order into the mechanisms governing primary and secondary ICB resistance, offer therapeutic options to circumvent different modes of resistance and plea for a personalized medicine approach to maximize immunotherapeutic benefit for all cancer patients.
Collapse
|
394
|
Liu Y, Hu Y, Gu F, Liang J, Zeng Y, Hong X, Zhang K, Liu L. Phenotypic and clinical characterization of low density neutrophils in patients with advanced lung adenocarcinoma. Oncotarget 2017; 8:90969-90978. [PMID: 29207617 PMCID: PMC5710898 DOI: 10.18632/oncotarget.18771] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 06/10/2017] [Indexed: 12/31/2022] Open
Abstract
Purpose An immunosuppressive subgroup of neutrophils, low density neutrophils (LDNs) was reported to be closely related to several diseases. This study was designed to explore the association between LDNs and advanced lung adenocarcinoma, as well as potential mechanisms. Results The expression levels of surface CD molecules on LDNs were different from high density neutrophils (HDNs), consistent with previous studies. The ratio of LDNs/HDNs, rather than the percentage of LDNs in peripheral blood mononuclear cells (PBMCs), was significantly higher in lung adenocarcinoma patients than healthy controls. It was also observed that the ratio decreased when patients received anti-cancer treatments, and increased when disease relapsed. Patients harboring positive epidermal growth factor receptor (EGFR) mutation had significantly higher ratios. Both the ratio and the percentage showed positive correlation with CD8+ T cells. Although significantly increased TGF-β was detected in lung adenocarcinoma patients, relationship between TGF-β and LDNs was not obvious. Materials and Methods LDNs and HDNs levels of peripheral blood from 52 lung adenocarcinoma patients and 13 healthy controls were determined by flow cytometry. Lymphocytes and cytokines were also detected. Conclusions Two kinds of neutrophils with different phenotypes were identified in lung adenocarcinoma patients. Besides, we found the existence of high ratio of LDNs/HDNs in these patients, which is related to disease prognosis, EGFR mutation and bad immune status.
Collapse
Affiliation(s)
- Yangyang Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yue Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feifei Gu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinyan Liang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yulan Zeng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaohua Hong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kai Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
395
|
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
|
396
|
Levingston CA, Young MRI. Local Immune Responsiveness of Mice Bearing Premalignant Oral Lesions to PD-1 Antibody Treatment. Cancers (Basel) 2017; 9:cancers9060062. [PMID: 28574425 PMCID: PMC5483881 DOI: 10.3390/cancers9060062] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 12/11/2022] Open
Abstract
A carcinogen-induced premalignant oral lesion model that progresses to oral cancer was used to examine the immunological impact of a 5-week treatment regimen to block programmed cell death protein 1 (PD-1). PD-1 antibody treatment resulted in concurrent, but transient, increases in interleukin (IL)-2, IFN-γ and IL-17, and delayed increases in IL-6 and IL-10 within the lesion-bearing tongue epithelium. In contrast, cytokine secretion by lymph node cells of PD-1 antibody-treated mice was lower than for mice treated with control antibodies, with the exception of interferon (IFN)-γ, whose secretion increased late in the treatment period. This delayed secretion of IFN-γ coincided with an increase in CD4+ lymph node cells expressing IFN-γ. Lymph node cells of PD-1 antibody-treated mice reacted to a challenge with lysates of lesions or cancer by early production of IFN-γ, but this rapidly subsided. There also was increased production IL-17 and tumor necrosis factor (TNF)-α in response to the challenge, but the response was greatest by cells of control lesion-bearing mice. Clinical assessment showed an early but transient, stabilization of disease in mice treated with PD-1 antibody. These results show an early beneficial, but time-limited, response to PD-1 antibody treatment, which then fails with continued lesion progression.
Collapse
Affiliation(s)
- Corinne A Levingston
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA.
| | - M Rita I Young
- Research Service, Ralph H. Johnson VA Medical Center, Charleston, SC 29401, USA.
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, Charleston, SC 29425, USA.
| |
Collapse
|
397
|
Thoma C. New model to test immunotherapy combinations. Nat Rev Urol 2017; 14:323. [DOI: 10.1038/nrurol.2017.44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
398
|
Erratum: Effective combinatorial immunotherapy for castration-resistant prostate cancer. Nature 2017; 545:116. [PMID: 28470201 DOI: 10.1038/nature22348] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|