51
|
Wang YN, Lou DF, Li DY, Jiang W, Dong JY, Gao W, Chen HC. Elevated levels of IL-17A and IL-35 in plasma and bronchoalveolar lavage fluid are associated with checkpoint inhibitor pneumonitis in patients with non-small cell lung cancer. Oncol Lett 2020; 20:611-622. [PMID: 32565986 PMCID: PMC7285943 DOI: 10.3892/ol.2020.11618] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/01/2020] [Indexed: 12/11/2022] Open
Abstract
Advances in the immunology have identified that interleukin (IL)-17 and IL-35 are cytokines with diverse functions, serving important roles in autoimmune diseases and chronic inflammation. Checkpoint inhibitor pneumonitis (CIP) is focal or diffuse lung inflammation induced by immune checkpoint inhibitors and the underlying pathogenesis has not been fully explored. The aim of the present study was to investigate the roles of IL-17A and IL-35, and the correlation between their levels and different T cell subsets in CIP. The levels of IL-17A and IL-35 in peripheral blood and bronchoalveolar lavage fluid (BALF) were measured in patients with non-small cell lung cancer (NSCLC) with CIP, and the corresponding controls. The percentages of helper T lymphocyte (Th)1, Th2 and Th17 cells, and regulatory T cells (Tregs) in the peripheral blood were synchronically detected. Serum levels of IL-17A and IL-35 were significantly increased at the time of CIP diagnosis compared with the baseline, and significantly decreased upon clinical recovery or improvement. IL-17A and IL-35 were also increased in the BALF during the development of CIP compared with the baseline. Serum levels of IL-17A were positively correlated with the percentages of Th1 and Th17 cells as well as the ratio of Th17 to Tregs, but negatively associated with the frequency of Tregs in CIP. Serum levels of IL-35 were positively correlated with the percentages of Th1 and Tregs, and with the ratio of Th1 to Th2 cells in CIP. A higher frequency of Th1 and Th17 cells, as well as higher ratios of Th17 to Tregs and Th1 to Th2 cells were detected upon development of CIP comparing with the baseline. These data suggested that the activation of Th1 and Th17 cells, as well as Treg inhibition contributed to the imbalanced ratios of Th1 to Th2 and Th17 to Tregs, which resulted in increased secretion of IL-17A and IL-35 in the plasma and BALF; this may present a valuable index to monitor the development and severity of CIP in patients with NSCLC receiving immunotherapy.
Collapse
Affiliation(s)
- Yi Na Wang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Dan Feng Lou
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang 310000, P.R. China
| | - Dan Yang Li
- Department of Intensive Care Unit, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wei Jiang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Jing Yin Dong
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, P.R. China
| | - Wei Gao
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, P.R. China
| | - Hong Chao Chen
- Department of Clinical Laboratory, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
52
|
Yi M, Jiao D, Qin S, Chu Q, Li A, Wu K. Manipulating Gut Microbiota Composition to Enhance the Therapeutic Effect of Cancer Immunotherapy. Integr Cancer Ther 2020; 18:1534735419876351. [PMID: 31517538 PMCID: PMC7242797 DOI: 10.1177/1534735419876351] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In the past decade, a growing set of immunotherapies including immune checkpoint
blockade, chimeric antigen receptor T cells, and bispecific antibodies propelled
the advancement of oncology therapeutics. Accumulating evidence demonstrates
that immunotherapy could eliminate tumors better than traditional chemotherapy
or radiotherapy with lower risk of adverse events in numerous cancer types.
Unfortunately, a substantial proportion of patients eventually acquire
resistance to immunotherapy. By analyzing the differences between
immunotherapy-sensitive and immunotherapy-resistant populations, it was noticed
that the composition of gut microbiota is closely related to treatment effect.
Moreover, in xenograft models, interventional regulation of gut microbiota could
effectively enhance efficacy and relieve resistance during immunotherapy. Thus,
we believe that gut microbiota composition might be helpful to explain the
heterogeneity of treatment effect, and manipulating gut microbiota could be a
promising adjuvant treatment for cancer immunotherapy. In this mini review, we
focus on the latest understanding of the cross-talk between gut microbiota and
host immunity. Moreover, we highlight the role of gut microbiota in cancer
immunotherapy including immune checkpoint inhibitor and adoptive cell
transfer.
Collapse
Affiliation(s)
- Ming Yi
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dechao Jiao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuang Qin
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Chu
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anping Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kongming Wu
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
53
|
Ciciola P, Cascetta P, Bianco C, Formisano L, Bianco R. Combining Immune Checkpoint Inhibitors with Anti-Angiogenic Agents. J Clin Med 2020; 9:E675. [PMID: 32138216 PMCID: PMC7141336 DOI: 10.3390/jcm9030675] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has recently emerged as a novel strategy for treating different types of solid tumors, with promising results. However, still a large fraction of patients do not primarily respond to such approaches, and even responders sooner or later develop resistance. Moreover, immunotherapy is a promising strategy for certain malignancies but not for others, with this discrepancy having been attributed to a more immunogenic microenvironment of some tumors. As abnormal and augmented tumor vessels often occur in cancerogenesis, anti-angiogenic drugs have already demonstrated their effectiveness both in preclinical and in clinical settings. By targeting abnormal formation of tumor vessels, anti-angiogenetic agents potentially result in an enhanced infiltration of immune effector cells. Moreover, crosstalks downstream of the immune checkpoint axis and vascular endothelial growth factor receptor (VEGFR) signaling may result in synergistic effects of combined treatment in tumor cells. In this review, we will describe and discuss the biological rationale of a combined therapy, underlying the modification in tumor microenvironment as well as in tumor cells after exposure to checkpoint inhibitors and anti-angiogenic drugs. Moreover, we will highlight this strategy as a possible way for overcoming drug resistance. By first discussing potential prognostic and predictive factors for combined treatment, we will then turn to clinical settings, focusing on clinical trials where this strategy is currently being investigated.
Collapse
Affiliation(s)
- Paola Ciciola
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| | - Priscilla Cascetta
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| | - Cataldo Bianco
- Department of Experimental and Clinical Medicine, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, 80131 Naples, Italy; (P.C.); (P.C.)
| |
Collapse
|
54
|
Aurilio G, Santoni M, Cimadamore A, Massari F, Scarpelli M, Lopez-Beltran A, Cheng L, Battelli N, Nolé F, Montironi R. Renal Cell Carcinoma: genomic landscape and clinical implications. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1733407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Gaetano Aurilio
- Medical Oncology Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, Ancona, Italy
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, Ancona, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Franco Nolé
- Medical Oncology Division of Urogenital and Head and Neck Tumours, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, Ancona, Italy
| |
Collapse
|
55
|
PDL1-targeted vaccine exhibits potent antitumor activity by simultaneously blocking PD1/PDL1 pathway and activating PDL1-specific immune responses. Cancer Lett 2020; 476:170-182. [PMID: 32092355 DOI: 10.1016/j.canlet.2020.02.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/10/2020] [Accepted: 02/17/2020] [Indexed: 12/30/2022]
Abstract
Despite the clinical success of immune checkpoint blockade, only a subset of people exhibits durable responses, suggesting that an alternative immunotherapeutic strategy is required. This paper reported a two-in-one cancer vaccine that targets programmed death ligand 1 (PDL1) that blocks the PD1/PDL1 pathway and also activates antitumor immune response. The PDL1- NitraTh vaccine, which consists of the extracellular domain of PDL1 and nitrated T cell epitope, effectively broke the immune tolerance of PDL1 and elicited PDL1-specific humoral and cellular immunity. The treatment of PDL1-NitraTh exhibited potent antitumor activity. Moreover, immunization of PDL1 vaccine increased the infiltration of tumor lymphocytes and decreased the proportion of Treg cells in tumor tissues, suggesting that the vaccine may remodel the tumor microenvironment. The upregulation of PDL1 in tumor tissues was induced by PDL1-NitraTh vaccine but not in spleen and lymphomas. This upregulation of PDL1 is beneficial to the antitumor activity of PDL1-specific humoral and cellular immunity induced by PDL1-NitraTh. In summary, PDL1-targeted vaccine exhibits potent antitumor activity and may provide an alternative immunotherapy strategy for patients who are not sensitive to PDL1 antibody drugs.
Collapse
|
56
|
Holmström MO, Cordua S, Skov V, Kjær L, Pallisgaard N, Ellervik C, Hasselbalch HC, Andersen MH. Evidence of immune elimination, immuno-editing and immune escape in patients with hematological cancer. Cancer Immunol Immunother 2020; 69:315-324. [PMID: 31915854 DOI: 10.1007/s00262-019-02473-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/31/2019] [Indexed: 10/25/2022]
Abstract
There is mounting evidence that the immune system can spontaneously clear malignant lesions before they manifest as overt cancer, albeit this activity has been difficult to demonstrate in humans. The calreticulin (CALR) exon 9 mutations are driver mutations in patients with chronic myeloproliferative neoplasms (MPN), which are chronic blood cancers. The CALR mutations generate a neo-antigen that is recognized by patient T cells, and T cells isolated from a patient with a CALR-mutation can recognize and kill autologous CALR-mutant cells. Surprisingly, healthy individuals display frequent and strong T cell responses to the CALR neo-antigens too. Furthermore, healthy individuals display immune responses to all parts of the mutant CALR epitope, and the CALR neo-epitope specific responses are memory T cell responses. These data suggest that although healthy individuals might acquire a CALR mutation, the mutant cells can be eliminated by the immune system. Additionally, a small fraction of healthy individuals harbor a CALR exon 9 mutation. Four healthy individuals carrying CALR mutations underwent a full medical examination including a bone marrow biopsy after a median follow up of 6.2 years. None of these patients displayed any signs of CALR-mutant MPN. Additionally, all healthy individuals displayed strong CALR neo-epitope specific T cell responses suggesting that these healthy individuals retained their CALR-mutant cells in the editing stage for several years. Thus, we suggest that CALR-mutant MPN could be a disease model of cancer immuno-editing, as we have demonstrated that CALR-mutant MPN displays all three stages described in the theory of cancer immuno-editing.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, Borgmester Ib Juuls Vej 25C, 5. Sal, 2730, Herlev, Denmark.
| | - Sabrina Cordua
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Niels Pallisgaard
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark.,Department of Laboratory Medicine, Harvard Medical School, Boston Children's Hospital, Boston, USA
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, Borgmester Ib Juuls Vej 25C, 5. Sal, 2730, Herlev, Denmark.,Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
57
|
Grépin R, Guyot M, Dumond A, Durivault J, Ambrosetti D, Roussel JF, Dupré F, Quintens H, Pagès G. The combination of bevacizumab/Avastin and erlotinib/Tarceva is relevant for the treatment of metastatic renal cell carcinoma: the role of a synonymous mutation of the EGFR receptor. Theranostics 2020; 10:1107-1121. [PMID: 31938054 PMCID: PMC6956821 DOI: 10.7150/thno.38346] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 08/14/2019] [Indexed: 01/25/2023] Open
Abstract
Metastatic clear cell renal cell carcinomas (mRCC) over-express the vascular endothelial growth factor (VEGF). Hence, the anti-VEGF antibody bevacizumab/Avastin (BVZ) combined with interferon alpha (IFN) was approved for the treatment of mRCC. However, approval was lost in July 2016 due to the absence of sustained efficacy. We previously showed that BVZ accelerates tumor growth in experimental models of mRCC in mice, results in part explained by down-regulation of the phospho tyrosine phosphatase receptor kappa (PTPRκ) in tumor cells. The epidermal growth factor receptor (EGFR) is a direct target of PTPRκ. Its down-regulation leads to constitutive activation of EGFR, an observation which prompted us to test the effect of the EGFR inhibitor erlotinib/Tarceva (ERLO) in addition to BVZ/IFN. The influence of the long non-coding RNA, EGFR-AS1, on ERLO efficacy was also addressed. Methods: The effect of BVZ/IFN/ERLO was tested on the growth of experimental tumors in nude mice. The presence of germline mutation in the EGFR was evaluated on cell lines and primary RCC cells. In vitro translation and transfections of expression vectors coding the wild-type or the EGFR mutated gene in HEK-293 cells were used to test the role of EGFR mutation of the ERLO efficacy. Correlation between EGFR/EGFR-AS1 expression and survival was analyzed with an online available data base (TCGA). Results: Tumor growth was strongly reduced by the triple combination BVZ/IFN/ERLO and linked to reduced levels of pro-angiogenic/pro-inflammatory cytokines of the ELR+CXCL family and to subsequent inhibition of vascularization, a decreased number of lymphatic vessels and polarization of macrophages towards the M1 phenotype. Cells isolated from surgical resection of human tumors presented a range of sensitivity to ERLO depending on the presence of a newly detected mutation in the EGFR and to the presence of EGFR-AS1. Conclusions: Our results point-out that the BVZ/IFN/ERLO combination deserves testing for the treatment of mRCC that have a specific mutation in the EGFR.
Collapse
|
58
|
Laha D, Nilubol N, Boufraqech M. New Therapies for Advanced Thyroid Cancer. Front Endocrinol (Lausanne) 2020; 11:82. [PMID: 32528402 PMCID: PMC7257776 DOI: 10.3389/fendo.2020.00082] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/07/2020] [Indexed: 12/14/2022] Open
Abstract
Thyroid cancer is the most common endocrine cancer. The discovery of new biomarkers for thyroid cancer has significantly improved the understanding of the molecular pathogenesis of thyroid cancer, thus allowing more personalized treatments for patients with thyroid cancer. Most of the recently discovered targeted therapies inhibit the known oncogenic mechanisms in thyroid cancer initiation and progression such as MAPK pathway, PI3K/Akt-mTOR pathways, or VEGF. Despite the significant advances in molecular testing and the discoveries of new and promising therapeutics, effective treatments for advanced and metastatic, iodine-refractory thyroid cancer are still lacking. Here, we aim to summarize the current understanding of the genetic alterations and the dysregulated pathways in thyroid cancer and to discuss the most recent targeted therapies and immunotherapy for advanced thyroid cancer with a promising anti-tumor activity and clinical benefit.
Collapse
|
59
|
Chung JS, Ramani V, Kobayashi M, Fattah F, Popat V, Zhang S, Cruz PD, Gerber DE, Ariizumi K. DC-HIL/Gpnmb Is a Negative Regulator of Tumor Response to Immune Checkpoint Inhibitors. Clin Cancer Res 2019; 26:1449-1459. [PMID: 31822499 DOI: 10.1158/1078-0432.ccr-19-2360] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 11/07/2019] [Accepted: 12/04/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Immune checkpoint inhibitors (ICI) benefit only a minority of treated patients with cancer. Identification of biomarkers distinguishing responders and nonresponders will improve management of patients with cancer. Because the DC-HIL checkpoint differs from the PD1 pathway in expression and inhibitory mechanisms, we examined whether DC-HIL expression regulates ICI responsiveness. EXPERIMENTAL DESIGN Plasma samples were collected from patients with advanced non-small cell lung carcinoma (NSCLC) (n = 76) at baseline and/or follow-up after ICI monotherapy. Blood-soluble DC-HIL (sDC-HIL) was determined and analyzed for correlation with the early tumor response. To study the mechanisms, we measured effect of anti-DC-HIL versus anti-PDL1 mAb on growth of mouse tumor cells in experimentally metastatic lung. Influence of DC-HIL to anti-PDL1 treatment was assessed by changes in tumor response after deletion of host-DC-HIL gene, injection of DC-HIL-expressing myeloid-derived suppressor cells (MDSC), or induction of sDC-HIL expression. RESULTS Nonresponders expressed significantly higher levels of baseline sDC-HIL levels than responders. Among patients (n = 28) for fluctuation with time, nonresponders (14/15 cases) showed increasing or persistently elevated levels. Responders (12/13) had decreasing or persistently low levels. Among various tumors, B16 melanoma exhibited resistance to anti-PDL1 but responded to anti-DC-HIL mAb. Using B16 melanoma and LL2 lung cancer, we showed that deletion of host-derived DC-HIL expression converted the resistant tumor to one responsive to anti-PDL1 mAb. The responsive state was reversed by infusion of DC-HIL+MDSC or induction of sDC-HIL expression. CONCLUSIONS sDC-HIL in the blood and probably DC-HIL receptor expressed by MDSC play an important role in regulating response to ICI in advanced NSCLC.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Animals
- Antibodies, Monoclonal/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- B7-H1 Antigen/metabolism
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Line, Tumor
- Disease Models, Animal
- Female
- Humans
- Immunologic Factors/metabolism
- Immunotherapy/methods
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Male
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Middle Aged
- Myeloid-Derived Suppressor Cells/immunology
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Skin Neoplasms/therapy
Collapse
Affiliation(s)
- Jin-Sung Chung
- Department of Dermatology, the University of Texas Southwestern Medical Center, and Dermatology Section (Medical Service), North Texas Veterans Affairs Medical Center, Dallas, Texas
| | - Vijay Ramani
- Department of Dermatology, the University of Texas Southwestern Medical Center, and Dermatology Section (Medical Service), North Texas Veterans Affairs Medical Center, Dallas, Texas
| | - Masato Kobayashi
- Department of Dermatology, the University of Texas Southwestern Medical Center, and Dermatology Section (Medical Service), North Texas Veterans Affairs Medical Center, Dallas, Texas
| | - Farjana Fattah
- Department of Hematology Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vinita Popat
- Department of Hematology Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Song Zhang
- Department of Population Data Sciences, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ponciano D Cruz
- Department of Dermatology, the University of Texas Southwestern Medical Center, and Dermatology Section (Medical Service), North Texas Veterans Affairs Medical Center, Dallas, Texas
| | - David E Gerber
- Department of Hematology Oncology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kiyoshi Ariizumi
- Department of Dermatology, the University of Texas Southwestern Medical Center, and Dermatology Section (Medical Service), North Texas Veterans Affairs Medical Center, Dallas, Texas.
| |
Collapse
|
60
|
Wang W, Ren S, Wang Z, Zhang C, Huang J. Increased expression of TTC21A in lung adenocarcinoma infers favorable prognosis and high immune infiltrating level. Int Immunopharmacol 2019; 78:106077. [PMID: 31812070 DOI: 10.1016/j.intimp.2019.106077] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 10/27/2019] [Accepted: 11/21/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a crucial pathological type of lung cancer. Immune-infiltration of the tumor microenvironment positively associated with overall survival in LUAD. TTC21A is a gene has not reported in cancer, and the mechanism behind it is still unclear. Our study assesses TTC21A role in LUAD, via TCGA data. METHODS GEPIA was utilized to analyze the expression of TTC21A in LUAD. We evaluated the influence of TTC21A on survival of LUAD patients by survival module. Then, data sets of LUAD were downloaded from TCGA. The correlations between clinical information and TTC21A expression were analyzed using logistic regression. Clinicopathologic characteristics associated with overall survival in TCGA patients using Cox regression. In addition, we explored the correlation between TTC21A and cancer immune infiltrates using CIBERSORT and "Correlation" module of GEPIA. RESULTS The univariate analysis using logistic regression, wherein TTC21A expression served as a categorical dependent variable (with a median expression value of 2.5), indicated that increased TTC21A expression is significantly correlated with pathological stage, tumor status and lymph nodes. Moreover, multivariate analysis revealed that the up-regulated TTC21A expression, negative results of pathological stage and distant metastasis are independent prognostic factors for good prognosis. Specifically, a positive correlation between increased TTC21A expression and immune infiltrating level of B cells, Neutrophils, Mast cells and T cells was established using CIBERSORT analysis. Furthermore, we confirmed it in "correlation" module of GEPIA. CONCLUSION Together with all these findings, increased TTC21A expression correlates with favorable prognosis and increased proportion of immune cells, such as B cells, Neutrophils, Mast cells and T cells in LUAD. These conclusions indicate that TTC21A could serve as a potential biomarker to assess prognosis and immune infiltration level in LUAD.
Collapse
Affiliation(s)
- Wei Wang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, PR China; Department of Medicine, Nantong University Xinling College, Nantong, PR China
| | - Shiqi Ren
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, PR China; Department of Medicine, Nantong University Xinling College, Nantong, PR China
| | - Ziheng Wang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, PR China; Department of Medicine, Nantong University Xinling College, Nantong, PR China
| | - Chenlin Zhang
- Department of Spine, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi 214000, PR China
| | - Jianfei Huang
- Department of Clinical Biobank, Nantong University Affiliated Hospital, Nantong, PR China; Institute of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
61
|
Abstract
Introduction: Metastatic renal cell carcinoma (mRCC) in recent years has been one of the diseases in the oncology that has experienced the greatest development of targeted therapies.Area covered: On one hand, more and more refined acquisitions of molecular biology are allowing a better sub-classification of this tumor, identifying new and peculiar biological profiles of patients with metastatic renal cancer that will soon be destined for increasingly tailored treatments. On the other hand, immunotherapy has, in a short time, redesigned the perimeter of our knowledge, entering clinical practice in an overpowering and convincing way. Monoclonal antibodies targeting immune checkpoint inhibitors have now entered clinical trials and have demonstrated promising antitumor effect for refractory metastatic renal cancer.Expert opinion: This review summarizes the results of published and reported studies of immune checkpoint inhibitors targeted therapies enrolling patients with metastatic renal cancer.
Collapse
Affiliation(s)
- Andrea Muto
- Division of Medical Oncology, "S. G. Moscati" Hospital, Avellino, Contrada Amoretta, Italy
| | - Cesare Gridelli
- Division of Medical Oncology, "S. G. Moscati" Hospital, Avellino, Contrada Amoretta, Italy
| |
Collapse
|
62
|
Peinemann F, Unverzagt S, Hadjinicolaou AV, Moldenhauer I. Immunotherapy for metastatic renal cell carcinoma: A systematic review. J Evid Based Med 2019; 12:253-262. [PMID: 31430052 DOI: 10.1111/jebm.12362] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 04/02/2019] [Accepted: 08/01/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Various approaches have been developed for the treatment of metastatic renal cell carcinoma (MRCC). The objective was to assess the efficacy of immunotherapeutic approaches. METHODS We included participants diagnosed with all types of histologically confirmed MRCC, the test intervention was immunotherapy alone or combined with other immunotherapy or targeted therapies, and the study design was restricted to randomized controlled trials (RCTs). Primary outcomes were overall survival, adverse events, and health-related quality of life. We conducted the last search in March 31, 2018. RESULTS We included nine RCTs, and we established seven different treatment comparisons according to the type of data. Two RCTs favored nivolumab as monotherapy or combined with ipilimumab on account of all primary outcomes. The efficacy data of all other comparisons were either indifferent or favored the control group. CONCLUSION The immune checkpoint inhibitors nivolumab as monotherapy or combined with ipilimumab appears to be the only new immunotherapy that may improve overall survival in participants with metastatic renal cell carcinoma. Interferon-α alone is unfavorable to targeted therapies with respect to overall survival and adverse events.
Collapse
Affiliation(s)
- Frank Peinemann
- Pediatric Oncology and Hematology, Children's Hospital, University Hospital of Cologne, Cologne, Germany
- FOM University of Applied Science for Economics & Management, Essen, Germany
| | - Susanne Unverzagt
- Institute of Medical Epidemiology, Biostatistics, and Informatics, University Hospital Halle, Halle (Saale), Germany
| | - Andreas V Hadjinicolaou
- Human Immunology Unit, Institute of Molecular Medicine, Radcliffe Department of Medicine, Oxford University, Oxford, UK
| | - Ines Moldenhauer
- Department of Internal Medicine I, University Hospital Halle, Halle (Saale), Germany
| |
Collapse
|
63
|
Mantia CM, McDermott DF. Vascular endothelial growth factor and programmed death-1 pathway inhibitors in renal cell carcinoma. Cancer 2019; 125:4148-4157. [PMID: 31532565 DOI: 10.1002/cncr.32361] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 01/05/2023]
Abstract
Advanced renal cell carcinoma has historically carried a poor prognosis with very limited treatment options. However, in recent years, the treatment landscape has changed drastically, with many new therapeutic options and improved survival for patients. Novel treatments consist of molecularly targeted agents against the vascular endothelial growth factor (VEGF) pathway as well as the immune checkpoint inhibitors, which stimulate an antitumor immune response. Recent strategy has focused on the development of combination therapy with the use of VEGF inhibitors and immune checkpoint inhibitors in the first-line setting. As more treatments are approved and the options for therapy expand further, there is a growing need for predictive biomarkers to personalize treatment choices for individual patients. Prospective clinical trials comparing the sequencing of treatments are needed to help determine the best therapeutic approach.
Collapse
Affiliation(s)
- Charlene M Mantia
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - David F McDermott
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
64
|
Synergism of PDL/PD1 and IL33/ST2 Axis in Tumor Immunology. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2018-0033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Abstract
When it comes to tumor immunology, understanding of molecular pathways is rather important. During oncogenesis, many molecules should be taken in consideration altogether in context of a single malignancy. It is of a great significance to determine whether these molecules act synergistically or contrary, whether to understand a malignant disease more thoroughly, or even more important, to reveal new approaches of therapy. In this review, we discuss whether and how IL-33/ST2 and PD-1/PDL axis involve in antitumor immunity.
Collapse
|
65
|
Lombardo SD, Presti M, Mangano K, Petralia MC, Basile MS, Libra M, Candido S, Fagone P, Mazzon E, Nicoletti F, Bramanti A. Prediction of PD-L1 Expression in Neuroblastoma via Computational Modeling. Brain Sci 2019; 9:E221. [PMID: 31480495 PMCID: PMC6770763 DOI: 10.3390/brainsci9090221] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/26/2019] [Accepted: 08/31/2019] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy is a promising new therapeutic approach for neuroblastoma (NBM): an anti-GD2 vaccine combined with orally administered soluble beta-glucan is undergoing a phase II clinical trial and nivolumab and ipilimumab are being tested in recurrent and refractory tumors. Unfortunately, predictive biomarkers of response to immunotherapy are currently not available for NBM patients. The aim of this study was to create a computational network model simulating the different intracellular pathways involved in NBM, in order to predict how the tumor phenotype may be influenced to increase the sensitivity to anti-programmed cell death-ligand-1 (PD-L1)/programmed cell death-1 (PD-1) immunotherapy. The model runs on COPASI software. In order to determine the influence of intracellular signaling pathways on the expression of PD-L1 in NBM, we first developed an integrated network of protein kinase cascades. Michaelis-Menten kinetics were associated to each reaction in order to tailor the different enzymes kinetics, creating a system of ordinary differential equations (ODEs). The data of this study offers a first tool to be considered in the therapeutic management of the NBM patient undergoing immunotherapeutic treatment.
Collapse
Affiliation(s)
- Salvo Danilo Lombardo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Mario Presti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Maria Cristina Petralia
- IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico) Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124- Messina, Italy
| | - Maria Sofia Basile
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Paolo Fagone
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy.
| | - Emanuela Mazzon
- IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico) Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124- Messina, Italy
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123- Catania, Italy
| | - Alessia Bramanti
- IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico) Centro Neurolesi Bonino Pulejo, C.da Casazza, 98124- Messina, Italy
| |
Collapse
|
66
|
BET protein targeting suppresses the PD-1/PD-L1 pathway in triple-negative breast cancer and elicits anti-tumor immune response. Cancer Lett 2019; 465:45-58. [PMID: 31473251 DOI: 10.1016/j.canlet.2019.08.013] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/08/2019] [Accepted: 08/24/2019] [Indexed: 12/19/2022]
Abstract
Therapeutic strategies aiming to leverage anti-tumor immunity are being intensively investigated as they show promising results in cancer therapy. The PD-1/PD-L1 pathway constitutes an important target to restore functional anti-tumor immune response. Here, we report that BET protein inhibition suppresses PD-1/PD-L1 in triple-negative breast cancer. BET proteins control PD-1 expression in T cells, and PD-L1 in breast cancer cell models. BET protein targeting reduces T cell-derived interferon-γ production and signaling, thereby suppressing PD-L1 induction in breast cancer cells. Moreover, BET protein inhibition improves tumor cell-specific T cell cytotoxic function. Overall, we demonstrate that BET protein targeting represents a promising strategy to overcome tumor-reactive T cell exhaustion and improve anti-tumor immune responses, by reducing the PD-1/PD-L1 axis in triple-negative breast cancer.
Collapse
|
67
|
Liu F, Zhang H, Xue L, Yang Q, Yan W. Molecular profiling of transcription factors pinpoints MYC-estrogen related receptor α-regulatory factor X5 panel for characterizing the immune microenvironment and predicting the efficacy of immune checkpoint inhibitors in renal cell carcinoma. Oncol Lett 2019; 18:1895-1903. [PMID: 31423259 PMCID: PMC6614680 DOI: 10.3892/ol.2019.10523] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
Transcription factors (TFs) play key roles in biological processes, and previous studies revealed that they can control oncogenic processes. However, the functional impact of TFs on the prognosis of patients with cancer has not been extensively elucidated. In the context of The Cancer Genome Atlas, few studies have focused on the roles of TFs in tumorigenesis. In the present study, a TF-based robust MYC-estrogen related receptor α-regulatory factor X5 (MYC-ESRRA-RFX5) signature was developed for predicting the survival of patients with renal cell carcinoma. Functional enrichment analysis of this signature revealed that it was associated with the immune system of these patients. Further analysis demonstrated that this panel could characterize the immune microenvironment and potentially predicts the effectiveness of immune checkpoint inhibitors. Therefore, the present study recommends future exploration on TF-based biomarkers for their potential as prognostic predictors. Overall, the highlights of this study are: i) This novel study pinpoints a TF panel for the robust prediction of renal cell carcinoma prognosis, and ii) the MYC-ESRRA-RFX5 panel is proposed as a signature for characterizing the immune microenvironment, and to potentially predict the effectiveness of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Fei Liu
- Department of Nephrology, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu 730000, P.R. China
| | - Hongxia Zhang
- Department of Emergency Medicine, The First Hospital of The Chinese People's Liberation Army, Lanzhou, Gansu 730000, P.R. China
| | - Lihua Xue
- Department of Obstetrics and Gynecology, The Family Planning Service Center for Maternal and Child Health in Zhouqu County, Lanzhou, Gansu 730000, P.R. China
| | - Qiankun Yang
- Department of Bone and Soft Tissue, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, P.R. China
| | - Wanchun Yan
- Department of Geriatrics, The First Hospital of The Chinese People's Liberation Army, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
68
|
Bex A, van Thienen JV, Schrier M, Graafland N, Kuusk T, Hendricksen K, Lagerveld B, Zondervan P, van Moorselaar JA, Blank C, Wilgenhof S, Haanen J. A Phase II, single-arm trial of neoadjuvant axitinib plus avelumab in patients with localized renal cell carcinoma who are at high risk of relapse after nephrectomy (NEOAVAX). Future Oncol 2019; 15:2203-2209. [DOI: 10.2217/fon-2019-0111] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Surgery is the standard treatment for nonmetastatic renal cell carcinoma. Despite curative intent, patients with a high risk of relapse have a 5-year metastasis-free survival rate of only 30% and prevention of recurrence is an unmet need. In a Phase III trial (JAVELIN Renal 101), progression-free survival of axitinib + avelumab was superior to sunitinib with a favorable objective response rate and no added toxicity profiles as known for axitinib or avelumab single agent. NEOAVAX is designed as open label, single arm, Phase II trial with a Simon’s two-stage design evaluating neoadjuvant axitinib + avelumab followed by complete surgical resection in 40 patients with high-risk nonmetastatic clear-cell renal cell carcinoma. Primary end point is remission of the primary tumor (RECIST 1.1; Response Evaluation Criteria In Solid Tumors) following neoadjuvant therapy. Secondary end points include disease-free survival, overall survival, rate of metastasis and local recurrence, safety, and tolerability. Exploratory end points include investigation of effects on neoangiogenesis, immune infiltrates and myeloid-derived suppressor cell components to support a rationale for the combined use of axitinib and avelumab (NCT03341845).
Collapse
Affiliation(s)
- Axel Bex
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Urology, Specialist Centre for Kidney Cancer, Royal Free London NHS Foundation Trust, London, UK
- Department of Urology, UCL Division of Surgery & Interventional Science, London, UK
| | - Johan V van Thienen
- Department of Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Mariette Schrier
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Niels Graafland
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Teele Kuusk
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kees Hendricksen
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Brunolf Lagerveld
- Department of Urology, Onze Lieve Vrouwe Gasthuis, Amsterdam, The Netherlands
| | - Patricia Zondervan
- Department of Urology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | | | - Christian Blank
- Department of Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sofie Wilgenhof
- Department of Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John Haanen
- Department of Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
69
|
Jiang P, Gao W, Ma T, Wang R, Piao Y, Dong X, Wang P, Zhang X, Liu Y, Su W, Xiang R, Zhang J, Li N. CD137 promotes bone metastasis of breast cancer by enhancing the migration and osteoclast differentiation of monocytes/macrophages. Theranostics 2019; 9:2950-2966. [PMID: 31244935 PMCID: PMC6568184 DOI: 10.7150/thno.29617] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 03/25/2019] [Indexed: 12/13/2022] Open
Abstract
Rationale: Bone is one of the most common metastatic sites of breast cancer. CD137 (4-1BB), a member of the tumor necrosis factor (TNF) receptor superfamily, is mainly expressed in activated leukocytes. Previous study demonstrates the effect of CD137-CD137L bidirectional signaling pathway on RANKL-mediated osteoclastogenesis. However, the role of CD137 in bone metastasis of breast cancer needs further study. Methods: Stable monocyte/macrophage cell lines with Cd137 overexpression and silencing were established. Western blot, real-time PCR, transwell and tartrate-resistant acid phosphatase staining were used to detect the regulatory effect of CD137 on migration and osteoclastogenesis of monocytes/macrophages in vitro. Spontaneous bone metastasis mouse model was established, bioluminescent images, immunohistochemistry and histology assay were performed to detect the function of CD137 in bone metastasis in vivo. Results: We found that CD137 promotes the migration of monocytes/macrophages to tumor microenvironment by upregulating the expression of Fra1. It also promoted the differentiation of monocytes/macrophages into osteoclasts at the same time, thus providing a favorable microenvironment for the colonization and growth of breast cancer cells in bone. Based on these findings, a novel F4/80-targeted liposomal nanoparticle encapsulating the anti-CD137 blocking antibody (NP-αCD137 Ab-F4/80) was synthesized. This nanoparticle could inhibit both bone and lung metastases of 4T1 breast cancer cells with high efficacy in vivo. In addition, it increased the therapeutic efficacy of Fra1 inhibitor on tumor metastasis. Conclusions: Taken together, these findings reveal the promotion effect of macrophage/monocyte CD137 on bone metastases and provide a promising therapeutic strategy for metastasis of breast cancer.
Collapse
|
70
|
Barrington DA, Dilley SE, Smith HJ, Straughn JM. Pembrolizumab in advanced recurrent endometrial cancer: A cost-effectiveness analysis. Gynecol Oncol 2019; 153:381-384. [DOI: 10.1016/j.ygyno.2019.02.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/29/2019] [Accepted: 02/17/2019] [Indexed: 02/02/2023]
|
71
|
Kushlinskii NE, Gershtein ES, Goryatcheva IO, Morozov AA, Alferov AA, Bezhanova SD, Kazantseva IA, Bazaev VV, Matveev VB. Soluble forms of the immune check-point receptor PD-1 and its ligand PD-L1 in blood serum of patients with renal cell carcinoma: clinical and pathologic correlations. ACTA ACUST UNITED AC 2019. [DOI: 10.17650/1726-9776-2019-15-1-15-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- N. E. Kushlinskii
- National Medical Research Center of Oncology, Ministry of Health of Russia
| | - E. S. Gershtein
- National Medical Research Center of Oncology, Ministry of Health of Russia
| | - I. O. Goryatcheva
- National Medical Research Center of Oncology, Ministry of Health of Russiaatcheva1
| | - A. A. Morozov
- M.F. Vladimirsky Moscow Regional Research and Clinical Institute
| | - A. A. Alferov
- National Medical Research Center of Oncology, Ministry of Health of Russia
| | - S. D. Bezhanova
- National Medical Research Center of Oncology, Ministry of Health of Russia
| | - I. A. Kazantseva
- M.F. Vladimirsky Moscow Regional Research and Clinical Institute
| | - V. V. Bazaev
- M.F. Vladimirsky Moscow Regional Research and Clinical Institute
| | - V. B. Matveev
- National Medical Research Center of Oncology, Ministry of Health of Russia
| |
Collapse
|
72
|
Zhao W, Zhao F, Yang K, Lu Y, Zhang Y, Wang W, Xie H, Deng K, Yang C, Rong Z, Hou Y, Li K. An immunophenotyping of renal clear cell carcinoma with characteristics and a potential therapeutic target for patients insensitive to immune checkpoint blockade. J Cell Biochem 2019; 120:13330-13341. [PMID: 30916827 DOI: 10.1002/jcb.28607] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/07/2019] [Accepted: 01/14/2019] [Indexed: 12/11/2022]
Abstract
Renal clear cell carcinoma (RCC) patients who do not achieve optimal control of progression with immune checkpoint blockade (ICB) should be further studied. Unsupervised consensus clustering was used to group 525 RCC patients based on two typical ICB pathways, CTLA-4 and pogrammed death 1 (PD-1)/programmed death-ligand 1 (PD-L1), as well as two new discovered regulators, CMTM6 and CMTM4. Three immune molecular subtypes (IMMSs) with different clinical and immunological characteristics were identified (type I, II, and III), among which there were more stage I and low-grade tumors in type I RCC than in type II and III. The proportion of males was highest in type II RCC. Overall survival of type II and III was similar (5.2 and 6 years) and statistically shorter than that of type I (7.6 years) before and after adjusting for age and gender. When conducting stratified analysis, our IMMSs were able to identify high-risk patients among middle-aged patients, males, and stage IV patients. Among the differentially expressed genes, approximately 84% were highly expressed in type II and III RCC. Genes related to ICB (CTLA-4, CD274, and PDCD1LG2) and cytotoxic lymphocytes (CD8A, GZMA, and PRF1) were all highly expressed in type II and III RCC. These results documented that patients with type II and III cancer may be more sensitive to anti-CTLA-4 therapy, anti-PD-1/PD-L1 therapy, and a combination of immunotherapies. High expression of CMTM4 in type I RCC (69%) and a statistically significant interaction of CD274 and CMTM6 indicated that CMTM4/6 might be new therapy targets for type I, who are resistant to ICB.
Collapse
Affiliation(s)
- Weiwei Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Falin Zhao
- Department of Health Management, School of Medicine, Hangzhou Normal University, Hangzhou, People's Republic of China
| | - Kai Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yaxin Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yuanyuan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Wenjie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Hongyu Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Kui Deng
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Chunyan Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Zhiwei Rong
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Yan Hou
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| | - Kang Li
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, People's Republic of China
| |
Collapse
|
73
|
Wu J, Jin S, Gu W, Wan F, Zhang H, Shi G, Qu Y, Ye D. Construction and Validation of a 9-Gene Signature for Predicting Prognosis in Stage III Clear Cell Renal Cell Carcinoma. Front Oncol 2019; 9:152. [PMID: 30941304 PMCID: PMC6433707 DOI: 10.3389/fonc.2019.00152] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose: Aim of this study was to develop a multi-gene signature to help better predict prognosis for stage III renal cell carcinoma (RCC) patients. Methods: Fourteen pairs of stage III tumor and normal tissues mRNA expression data from GSE53757 and 16 pairs mRNA expression data from TCGA clear cell RCC database were used to analyze differentially expressed genes between tumor and normal tissues. Common different expressed genes in both datasets were used for further modeling. Lasso Cox regression analysis was performed to select and build prognostic multi-gene signature in TCGA stage III kidney cancer patients (N = 122). Then, the multi-gene signature was validated in stage III renal cancer cases in Fudan University Shanghai Cancer Center (N = 77). C-index and time-dependent ROC were used to test the efficiency of this signature in predicting overall survival. Results: In total, 1,370 common different expressed genes were found between tumor and normal tissues in both datasets. After Lasso Cox modeling, nine mRNAs were finally identified to build a classifier. Using this classifier, we could classify stage III clear cell RCC patients into high-risk group and low-risk group. Prognosis was significantly different between these groups in discovery TCGA cohort, validation FUSCC cohort and entire set (All P < 0.001). Multivariate cox regression in entire set (N = 199) revealed that risk group classified by 9-gene signature, age of diagnosis, pN stage and ISUP grade were independent prognostic factor of overall survival in stage III kidney cancer patients. Conclusion: We developed a robust multi-gene classifier that can effectively classify stage III RCC patients into groups with low and high risk of poor prognosis. This signature may help select high-risk patients who require more aggressive adjuvant target therapy or immune therapy.
Collapse
Affiliation(s)
- Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengming Jin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weijie Gu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fangning Wan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guohai Shi
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuanyuan Qu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
74
|
Study of Cathepsin B inhibition in VEGFR TKI treated human renal cell carcinoma xenografts. Oncogenesis 2019; 8:15. [PMID: 30796200 PMCID: PMC6386754 DOI: 10.1038/s41389-019-0121-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 12/19/2022] Open
Abstract
Several therapeutic options are available for metastatic RCC, but responses are almost never complete, and resistance to therapy develops in the vast majority of patients. Consequently, novel treatments are needed to combat resistance to current therapies and to improve patient outcomes. We have applied integrated transcriptome and proteome analyses to identify cathepsin B (CTSB), a cysteine proteinase of the papain family, as one of the most highly upregulated gene products in established human RCC xenograft models of resistance to vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKI). We used established RCC models to test the significance of CTSB in the progression of renal cancer. Our evaluation of CTSB showed that stable CTSB knockdown suppressed RCC growth in vitro and in vivo. Stable over-overexpression of wild-type CTSB (CTSBwt/hi), but not of an CTSB active site mutant (CTSBN298A), rescued cell growth in CTSB knockdown cells and abolished the efficacy of VEGFR TKI treatment. Genome-wide transcriptome profiling of CTSB knockdown cells demonstrated significant effects on multiple metabolic and stem cell-related pathways, with ALDHA1A (ALDH1) as one of the most significantly downregulated genes. Importantly, survival analysis across 16 major TCGA cancers revealed that CTSB overexpression is associated with low rates of three and five year patient survival rates (P = 2.5e-08, HR = 1.4). These data strongly support a contribution of CTSB activity to RCC cell growth and tumorigenicity. They further highlight the promise of CTSB inhibition in development of novel combination therapies designed to improve efficacy of current TKI treatments of metastatic RCC.
Collapse
|
75
|
MHC Class 1 and PDL-1 Status of Primary Tumor and Lymph Node Metastatic Tumor Tissue in Gastric Cancers. Gastroenterol Res Pract 2019; 2019:4785098. [PMID: 30881447 PMCID: PMC6381579 DOI: 10.1155/2019/4785098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/08/2018] [Accepted: 12/19/2018] [Indexed: 12/24/2022] Open
Abstract
The prognosis of metastatic gastric cancer is poor. Despite the use of VEGF-, EGFR-, and HER2-targeting agents, prognosis is still poor in advanced gastric cancer. Although cancer immunotherapy responds well in some patients, clinical use is limited due to unanswered patients. For this reason, it is necessary to know the characteristics of primary and metastatic cancer cells for patient selection for immunotherapy and additional criteria are required. MHC-1 downregulation is most frequently observed in the tumor escape mechanism of cancer cells from the immune system. MHC-1 downregulation with increased PDL-1 expression of cancer cells has an important role in immune escape. MHC-1 downregulation and PDL-1 expression have been shown in many types of cancers. However, there is no study on the status of MHC-1 and PDL-1 in primary and metastatic tumor tissue. In this study, MHC-1 and PDL-1 score in primary and metastatic tumor cells was evaluated in 43 gastric cancer patients with lymph node metastasis. According to our results, the primary tumor PDL-1 score was correlated with the number of metastatic lymph nodes (r = 0.258; p = 0.024) and primary tumor size (r = 0.341; p = 0.045). A similar correlation was found between the primary tumor PDL-1 score and the metastatic tumor PDL-1 score (r = 0.213; p = 0.015). In our study, MHC-1 was found to be higher in primary tumors than metastatic tumors, although not statistically significant (p = 0.054). The results of our study showed high MHC-1 and low PDL-1 expression in primary tumors and low MHC-1 and high PDL-1 expression in metastatic tumors. These results reveal different biological characteristics of primary and metastatic tumor cells.
Collapse
|
76
|
Xia B, Wu DW, Wang TT, Guo SQ, Wang Y, Yang HL, Xu W, Tian C, Zhang LY, Sun BC, Sotomayor EM, Zhang YZ. [Expressions and prognostic significance of PTEN and PD-1 protein in patients with classical Hodgkin's lymphoma]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2019; 39:839-844. [PMID: 30373357 PMCID: PMC7348287 DOI: 10.3760/cma.j.issn.0253-2727.2018.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
目的 分析肿瘤免疫耐受信号通路的重要因子PD-1和PTEN在经典型霍奇金淋巴瘤(CHL)患者中的表达及其与患者临床特征和预后的相关性。 方法 回顾性分析2003年2月至2013年8月诊治的56例CHL患者的临床资料。采用免疫组织化学染色法检测CHL患者PD-1和PTEN蛋白的表达,采用原位杂交法检测EBV及EBV编码的小mRNA(EBER),并结合患者的临床特征与生存状态进行相关性分析。 结果 ①56例患者中,男34例,女22例,中位年龄25(7~71)岁,PTEN阳性者11例(19.64%),PD-1阳性者14例(25.00%)。②PTEN和PD-1表达呈正相关(rs=0.320,P=0.016);PTEN表达与Ann Arbor分期、IPS评分和有无大包块(≥5 cm)明显相关,PD-1仅与有无大包块相关(P值均<0.05)。③中位随访43(5~86)个月,多因素分析结果显示:年龄≥45岁(P<0.001)、IPS评分>2分(P=0.026)、EBER阳性(P=0.004)、PTEN蛋白高表达(P=0.035)是影响患者5年总生存的不良预后因素,也是影响5年无进展生存的不良预后因素(P值分别为0.007、0.014、0.002、0.024)。 结论 肿瘤免疫逃逸信号通路因子PTEN与CHL患者的预后相关,对CHL患者的预后判断有一定作用,同时也为CHL的免疫治疗提供了新思路和理论依据。
Collapse
Affiliation(s)
- B Xia
- Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | | | | | | | | | | | | | | | | | | | | | - Y Z Zhang
- Department of hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
77
|
Rong L, Liu Y, Hui Z, Zhao Z, Zhang Y, Wang B, Yuan Y, Li W, Guo L, Ying J, Song Y, Wang L, Zhou Z, Xue L, Lu N. PD-L1 expression and its clinicopathological correlation in advanced esophageal squamous cell carcinoma in a Chinese population. Diagn Pathol 2019; 14:6. [PMID: 30684971 PMCID: PMC6347821 DOI: 10.1186/s13000-019-0778-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 01/02/2019] [Indexed: 11/16/2022] Open
Abstract
Background Programmed death ligand 1 (PD-L1) is a ligand for the inhibitory programmed cell death protein 1 (PD-1), which are targeted by several anti-PD-1 and PD-L1 drugs for a variety of human cancers. However, only a few studies have evaluated PD-L1 expression in esophageal squamous cell carcinoma (ESCC) with a large Chinese cohort. Our present study is to evaluate the association of PD-L1 expression with clinicopathological features on ESCC. Methods Using tissue microarray and immunohistochemistry, PD-L1 expression on tumor cells and tumor-infiltrating immune cells was studied in 378 advanced ESCC patients without neoadjuvant chemoradiotherapy. Its correlation with clinicopathological parameters was analyzed. Results PD-L1 was expressed on 29.9% (113/378) ESCC tumor cells and 40.2% (152/378) tumor-infiltrating immune cells. PD-L1 expression in tumor cells was significantly correlated with age, degree of differentiation, T stage, N stage and metachronous hematogenous metastasis, and PD-L1 expression in tumor-infiltrating immune cells was significantly associated with N stage (P < 0.05). Patients with PD-L1 expression in tumor cells had poor disease-free survival (Hazard ratio [HR] = 1.436, P = 0.009). There was a positive association between tumor cells and tumor-infiltrating immune cells for PD-L1 expression (r = 0.16, P = 0.002). However, PD-L1 expression in tumor-infiltrating immune cells was not significantly correlated with disease-free survival and overall survival. Conclusions PD-L1 expression in tumor cells and tumor infiltrating immune cells is not only an indicator for immunotherapy, but also significantly related with age, differentiation, stage, metastasis and disease free survival.
Collapse
Affiliation(s)
- Lulu Rong
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yong Liu
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhouguang Hui
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yueming Zhang
- Department of Endoscopy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bingzhi Wang
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yanling Yuan
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenbin Li
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Luhua Wang
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhongren Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Liyan Xue
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China. .,Center for Cancer Precision Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Ning Lu
- Department of Pathology and Resident Training Base, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
78
|
Soluble Ligand of the Immune Checkpoint Receptor (sPD-L1) in Blood Serum of Patients with Renal Cell Carcinoma. Bull Exp Biol Med 2019; 166:353-357. [PMID: 30627905 DOI: 10.1007/s10517-019-04349-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Indexed: 10/27/2022]
Abstract
The content of the soluble ligand of the immune checkpoint receptor (sPD-L1) was determined in the blood serum of 106 patients with renal cell carcinoma and 11 patients with benign kidney tumors by direct ELISA (Human sPD-L1 Platinum ELISA; Affimetrix, eBioscience). The control group included 19 healthy men and 18 women. Serum level of sPD-L1 significantly surpassed the control values in both patients with primary renal cancer (p<0.0001) and in patients examined during disease progression (p<0.05). In patients with benign kidney tumors, the level of this marker was significantly higher than in the control (p<0.05), but lower than in patients with renal cell carcinoma. The sPD-L1 level significantly increased with disease stage (p<0.001); it was higher in the presence of metastases in regional lymph nodes irrespective of their number (N1 or N2) than in the absence of metastases (N0); it was also increased in patients with distant metastases (M1) and patients with grade III-IV tumors in comparison with grade III-IV tumors (p<0.05). The highest sPD-L1 levels were recorded in patients with tumor size corresponding to T2 and T3 and decreased in patients with T4 tumors. Thus, sPD-L1 level in patients with renal cell carcinoma correlated with tumor grade and metastasizing and can be considered as a promising marker in monitoring of the effect of anti-PD1/PD-L1 therapy.
Collapse
|
79
|
Bersanelli M, Gnetti L, Varotti E, Ampollini L, Carbognani P, Leonardi F, Rusca M, Campanini N, Ziglioli F, Dadomo CI, Pilato FP, Cortellini A, Rapacchi E, Caruso G, Silini EM, Maestroni U, Buti S. Immune context characterization and heterogeneity in primary tumors and pulmonary metastases from renal cell carcinoma. Immunotherapy 2019; 11:21-35. [PMID: 30702014 DOI: 10.2217/imt-2018-0097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 10/11/2018] [Indexed: 12/25/2022] Open
Abstract
AIM The knowledge of the immune context of renal cell carcinoma (RCC) is useful to predict benefit from immunotherapy. We retrospectively characterized the immune context of RCC patients underwent primary nephrectomy and pulmonary metastasectomy. MATERIALS & METHODS Intratumoral infiltrating lymphocytes and peritumoral renal infiltrating lymphocytes, lymphocyte subpopulations (CD4+, CD8+), PD-1, PD-L1 were explored in paired samples of primary RCC (T) and respective pulmonary metastases (M). RESULTS The immune variables demonstrated intralesional and intratumoral heterogeneity. Intralesional lymphocyte heterogeneity reached 76% of cases in T, 28% in M. The heterogeneity rate for PD-L1 expression was from 44% (T) to 56% (M); it correlated with better survival. CONCLUSION The immune context of RCC is highly variable both within a given tumor and among primary and metastases.
Collapse
Affiliation(s)
- Melissa Bersanelli
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Letizia Gnetti
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Elena Varotti
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Luca Ampollini
- Department of Thoracic Surgery, University Hospital of Parma, Parma, 43126, Italy
| | - Paolo Carbognani
- Department of Thoracic Surgery, University Hospital of Parma, Parma, 43126, Italy
| | - Francesco Leonardi
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Michele Rusca
- Department of Thoracic Surgery, University Hospital of Parma, Parma, 43126, Italy
| | - Nicoletta Campanini
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Francesco Ziglioli
- Department of Urology, University Hospital of Parma, Parma, 43126, Italy
| | - Clara I Dadomo
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Francesco P Pilato
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Alessio Cortellini
- Department of Biotechnological & Applied Clinical Sciences, Medical Oncology, St Salvatore Hospital, University of L'Aquila, L'Aquila, 67100, Italy
| | - Elena Rapacchi
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Giuseppe Caruso
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| | - Enrico M Silini
- Department of Pathological Anatomy & Histology, University Hospital of Parma, Parma, 43126, Italy
| | - Umberto Maestroni
- Department of Urology, University Hospital of Parma, Parma, 43126, Italy
| | - Sebastiano Buti
- Department of Medical Oncology, University Hospital of Parma, Parma, 43126, Italy
| |
Collapse
|
80
|
Abstract
Inflammatory skin diseases encompass a vast array of conditions. The field continues to expand and evolve with resurgence of conditions, through newly recognized medication adverse effects, and via more detailed descriptions of known dermatoses. The importance of clinicopathologic correlation and an up to date knowledge of dermatologic conditions cannot be overstated. This review focuses on an array of recent important developments in the histologic diagnosis of inflammatory conditions that affect the skin.
Collapse
|
81
|
Yan C, Chang J, Song X, Yan F, Yu W, An Y, Wei F, Yang L, Ren X. Memory stem T cells generated by Wnt signaling from blood of human renal clear cell carcinoma patients. Cancer Biol Med 2019; 16:109-124. [PMID: 31119051 PMCID: PMC6528452 DOI: 10.20892/j.issn.2095-3941.2018.0118] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective Memory stem T cells (Tscm) have attracted attention because of their enhanced self-renewal, multipotent capacity, and anti-tumor capacities. However, little is known about Tscm in patients with renal clear cell carcinoma (RCC) and the role of Wnt signaling in these cells. We evaluated Tscm from RCC patients concerning their activation of Wnt signaling in vitro and explored the mechanism of preferential survival.
Methods Flow cytometry identified surface markers and cytokines produced from accumulated Tscm in the presence of the glycogen synthase kinase beta inhibitor TWS119. Apoptosis was evaluated after induction using tumor necrosis factor-alpha. Immunofluorescence and Western blot analyses were used to investigate the activation of the nuclear factor-kappa B (NF-КB) pathway. Results RCC patients had a similar percentage of CD4+ and CD8+ Tscm as healthy donors. Activation of Wnt signaling by TWS119 resulted in the accumulation of Tscm in activated T cells, but reversal of differentiated T cells to Tscm was not achieved. Preferential survival of Tscm was associated with increased anti-apoptotic ability mediated downstream of the NF-КB activation pathway.
Conclusions The finding that Tscm can accumulate by Wnt signaling in vitro in blood from RCC patients will help in devising new cancer therapy strategies of Tscm-based adoptive immunotherapy, such as dendritic cell-stimulated Tscm, and T cell receptor or chimeric antigen receptor-engineered Tscm.
Collapse
Affiliation(s)
- Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jingjing Chang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xinmiao Song
- Department of Electromyogram, 3rd Affiliated Hospital of Hebei Medical University, Shijiazhuang 050051, China
| | - Fan Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Wenwen Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yang An
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lili Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
82
|
Gershtein ES, Utkin DO, Goryacheva IO, Khulamkhanova MM, Petrikova NA, Vinogradov II, Alferov AA, Stilidi IS, Kushlinskii NE. Soluble forms of immune checkpoint receptor PD-1 and its ligand PD-L1 in plasma of patients with ovarian neoplasms. ACTA ACUST UNITED AC 2018. [DOI: 10.18786/2072-0505-2018-46-7-690-698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background:Ovarian cancer is one of the most common oncologic diseases holding the frst place in mortality related to neoplasms of female genitalia. Along with active surgical intervention, contemporary ovarian cancer treatment includes various chemotherapeutic regimens which in many cases are quite effective, but relapse and death rates still remain high. In the recent years, major attention has been paid to the possibility of ovarian cancer immunotherapy associated with the discovery of the so-called “immune checkpoint” signaling, i.e. programmed cell death-1 / programmed death-ligand 1 (PD-1/PD-L) pathway, controlling intensity and duration of autoimmune response at physiologic conditions. Tumor PD-1 and/or PD-L1 expression is being actively studied as a predictor of anti-PD-1/PD-L treatment efficacy; however, this approach has certain limitations and problems that might be probably bypassed by determination of soluble PD-1 (sPD-1) and its ligand (sPD-L1) in serum or plasma.Aim:Comparative evaluation of sPD-1 and sPD-L1 content in plasma of healthy women and of patients with benign or borderline ovarian tumors and ovarian cancer, as well as the analysis of associations between these markers and main clinical and pathologic characteristics of ovarian cancer.Materials and methods:Sixty two (62) patients with ovarian neoplasms aged 32 to 77 (median, 56.5) years were enrolled into the study. Fifteen (15) patients had benign tumors, 9 had borderline, and 38, ovarian cancer. The control group included 17 healthy women aged 24 to 67 (median, 49) years. Plasma sPD-L1 and sPD-1 concentrations were measured with standard enzyme immunoassay kits (Afmetrix, eBioscience, USA).Results:Plasma sPD-L1 and sPD-1 levels in ovarian cancer patients (median, 41.3 and 48.0 pg/ml, respectively) did not differ significantly from those in the control group (49.5 and 43.8 pg/ml). sPD-L1 level in the patients with benign tumors (median, 22.2 pg/ml) was signifcantly lower than in the control (p < 0.01). The lowest sPD-1 level in plasma was found in the patients with borderline ovarian neoplasms, the difference with the ovarian cancer group being statistically signifcant (p < 0.05). No correlations between sPD-L1 and sPD-1 plasma levels were found in any of the study groups. sPD-L1 level signifcantly increased with disease stage (R = 0.44; p < 0.01), the most signifcant increase being observed at the most advanced IIIC stage (p < 0.05 as compared to all other stages). sPD-L1 was also signifcantly higher in the patients with ascites than in those without ascites. Plasma sPD-1 concentration was not associated with the indices of ovarian cancer progression, though its median was 1.3–1.44 times lower in the stage I than in the stage II–III patients, and decreased in those with the tumor size above 10 cm (assessed by ultrasound examination) and in the patients with ascites. No statistically signifcant associations of the markers' levels with tumor histological type and differentiation grade of ovarian cancer were found.Conclusion:sPD-L1 level in ovarian cancer patients correlates with disease progression and can be considered as a promising marker for monitoring of anti-PD-1/PD-L1 treatment efficacy. Potential clinical implications of sPD-1 require further studies.
Collapse
Affiliation(s)
| | - D. O. Utkin
- Ryazan Regional Clinical Oncology Dispensary
| | | | | | | | | | - A. A. Alferov
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | - I. S. Stilidi
- N.N. Blokhin National Medical Research Centre of Oncology
| | | |
Collapse
|
83
|
Wang Z, Song Q, Yang Z, Chen J, Shang J, Ju W. Construction of immune-related risk signature for renal papillary cell carcinoma. Cancer Med 2018; 8:289-304. [PMID: 30516029 PMCID: PMC6346237 DOI: 10.1002/cam4.1905] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/24/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022] Open
Abstract
The kidney renal papillary cell carcinoma (KIRP) is a relatively rare type of kidney cancer. There has been no investigation to find a robust signature to predict the survival outcome of KIRP patients in the aspect of tumor immunology. In this study, 285 KIRP samples from The Cancer Genome Atlas (TCGA) were randomly divided into training and testing set. A total of 1534 immune‐related genes from The Immunology Database and Analysis Portal (ImmPort) were used as candidates to construct the signature. Using univariate Cox analysis, we evaluated the relationship between overall survival and immune‐related genes expression and found 272 immune‐related genes with predicting prognostic ability. In order to construct an efficient predictive model, the Cox proportional hazards model with an elastic‐net penalty was used and identified 23 groups after 1000 iterations. As a result, 15‐genes model showing more stable than other gene groups was chosen to construct our immune‐related risk signature. In line with our expectations, the signature can independently predict the survival outcome of KIRP patients. Patients with high‐immune risk were found correlated with advanced stage. We also found that the high‐immune risk patients with higher PBRM1 and SETD2 mutations, increasing chromosomal instability, together with the gene set enrichment analysis (GSEA) results showing intensive connection of our signature with immune pathways. In conclusion, our study constructs a robust 15‐gene signature for predicting KIRP patients’ survival outcome on the basis of tumor immune environment and may provide possible relationship between prognosis and immune‐related biological function.
Collapse
Affiliation(s)
- Zhongyu Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Qian Song
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zuyi Yang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianing Chen
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Shang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wen Ju
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
84
|
Zhang X, Shi X, Li J, Hu Z, Gao J, Wu S, Long Z. Combination immunotherapy with interleukin-2 surface-modified tumor cell vaccine and programmed death receptor-1 blockade against renal cell carcinoma. Cancer Sci 2018; 110:31-39. [PMID: 30343514 PMCID: PMC6317916 DOI: 10.1111/cas.13842] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 01/23/2023] Open
Abstract
Immunotherapy may be an effective way to prevent postoperative recurrence of renal cell carcinoma. Streptavidin‐interleukin‐2 (SA‐IL‐2) surface‐modified tumor cell vaccine developed through our protein‐anchor technology could induce specific antitumor T‐cell responses, but this immunotherapy cannot completely eradicate the tumor. These effector T cells highly expressed programmed death receptor‐1 (PD‐1), and the expression of programmed death ligand‐1 (PD‐L1) in the tumor environment also was upregulated after SA‐IL‐2‐modified vaccine therapy. PD‐1/PD‐L1 interaction promotes tumor immune evasion. Adding PD‐1 blockade to SA‐IL‐2‐modified vaccine therapy increased the number of CD4+, CD8+ and CD8+interferon‐γ+ but not CD4+Foxp3+ T cells. PD‐1 blockade could rescue the activity of tumor‐specific T lymphocytes induced by the SA‐IL‐2‐modified vaccine. Combination therapy delayed tumor growth and protected mice against a second Renca cells but not melanoma cells challenge. Taken together, PD‐1 blockade could reverse immune evasion in the treatment with SA‐IL‐2‐modified vaccine, and eventually induce a stronger specific antitumor immune response against renal cell carcinoma.
Collapse
Affiliation(s)
- Xinji Zhang
- Department of Urology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Xiaojun Shi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinlong Li
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhiming Hu
- Institute of Biotherapy, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shihao Wu
- Department of Urology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Zhaolin Long
- Department of Urology, Shunde Hospital, Southern Medical University, Foshan, China
| |
Collapse
|
85
|
Lopez-Beltran A, Henriques V, Cimadamore A, Santoni M, Cheng L, Gevaert T, Blanca A, Massari F, Scarpelli M, Montironi R. The Identification of Immunological Biomarkers in Kidney Cancers. Front Oncol 2018; 8:456. [PMID: 30450335 PMCID: PMC6225533 DOI: 10.3389/fonc.2018.00456] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/01/2018] [Indexed: 12/18/2022] Open
Abstract
The recent approval of several agents have revolutionized the scenario of therapeutic management of metastatic renal cell carcinoma (RCC) allowing us to reach important clinical end points with extended patients' survival. Actually, every new drug approved has represented an important step forward to the improvement of patient's survival. On the other hand, we now understand that RCC includes a large group of tumor entities, each of them with different genetic and mutational alterations, but also showing different clinical behavior; a reason behind the needs of subtype specific personalized approach to therapy of RCC. Immunotherapy is gradually becoming a key factor in the therapeutic algorithm for patients with locally advanced or metastatic RCC. Due to the combination of potent treatment success and potentially deadly adverse effects from immune checkpoint inhibitors (ICI), gathering prognostic and predictive information about FDA-indicated tumors seems to be prudent. Robust and reliable biomarkers are crucial for patient's selection of treatments with immunomodulatory drugs. PD-L1 expression is a poor prognostic factor and predictive of better responses from both PD-1 and PD-L1 inhibitors in a variety of tumor types including RCC. Each FDA approved PD-1/PD-L1 drug is paired with a PD-L1 Immunohistochemistry (IHC) assay. Thus, there is need for improved knowledge and application of PD-1/PD-L1 IHC biomarkers in daily practice. IHC staining appears in membranous fashion. The atezolizumab approved IHC assay is unique in that only immune cell staining is quantified for the use of this assay in RCC. A single biomarker for patient selection may not be feasible, given that immune responses are dynamic and evolve over time. Biomarker development for ICI drugs will likely require integration of multiple biologic components like PD-L1 expression, TILs and mutational load. New methodological approaches based on digital pathology may be relevant since they will allow recognition of the biomarker and to objectively quantitate its expression, and therefore might produce objective and reproducible cut-off assessment. Multidisciplinary approach is very much needed to fully develop the current and future value of ICI in clinical practice.
Collapse
Affiliation(s)
- Antonio Lopez-Beltran
- Department of Pathology and Surgery, Faculty of Medicine, Cordoba University, Cordoba, Spain
| | | | - Alessia Cimadamore
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Thomas Gevaert
- Laboratory of Experimental Urology, Organ Systems, KU Leuven, Leuven, Belgium.,Department of Pathology, AZ Klina, Brasschaat, Belgium
| | - Ana Blanca
- Instituto Maimonides de Investigación Biomédica de Córdoba, Córdoba, Spain
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, United Hospital, School of Medicine, Polytechnic University of the Marche Region, Ancona, Italy
| |
Collapse
|
86
|
Carrozza F, Santoni M, Piva F, Cheng L, Lopez-Beltran A, Scarpelli M, Montironi R, Battelli N, Tamberi S. Emerging immunotherapeutic strategies targeting telomerases in genitourinary tumors. Crit Rev Oncol Hematol 2018; 131:1-6. [PMID: 30293699 DOI: 10.1016/j.critrevonc.2018.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 06/12/2018] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
Telomerase activity and telomere length are essential for the pathogenesis of several human diseases, including genitourinary tumors. Telomerase constitutes a complex system that includes human telomerase reverse transcriptase (hTERT), human telomerase RNA component (hTR) and telomerase associated protein 1 (TEP1), which are overexpressed in tumor cells compared to normal cells and are involved in the carcinogenesis and progression of renal cell carcinoma (RCC), bladder (BC) and prostate cancer (PCa). In addition, telomerase degraded peptide fragments expressed on the surface of tumor cells lead to their recognition by immune cells. On this scenario, in vitro and in vivo studies have shown effective anti-tumor activity of hTERT-tailored strategies in genitourinary tumors, including active immunotherapy with hTERT-peptide vaccines and passive immunotherapy with hTERT-transduced T cell infusion. This review emphasizes the role of telomerase in the carcinogenesis and progression of genitourinary tumors, thus underlying the potential of emerging telomerase-tailored immunotherapies in these patients.
Collapse
Affiliation(s)
| | | | - Francesco Piva
- Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | | |
Collapse
|
87
|
Santoni M, Massari F, Di Nunno V, Conti A, Cimadamore A, Scarpelli M, Montironi R, Cheng L, Battelli N, Lopez-Beltran A. Immunotherapy in renal cell carcinoma: latest evidence and clinical implications. Drugs Context 2018; 7:212528. [PMID: 29899754 PMCID: PMC5992965 DOI: 10.7573/dic.212528] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 04/11/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Advances in understanding the mechanisms of tumour-induced immunosuppression have led to the development of immune-checkpoint inhibitors in cancer patients, including those with renal cell carcinoma (RCC). The optimal combination between immunotherapy and targeted agents (as well as the possible favourable sequential therapy of these two classes of drugs) remains an open question at this moment. Several trials are currently underway to assess the combination of anti-programmed-death 1 (PD-1) or anti-PD-ligand(L)1 agents with other immunotherapies or with anti-vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitors (TKIs). In this editorial, we described the results of the most recent clinical trials on the use of immunotherapies in RCC and the emerging data on the research for reliable biomarkers of tumour response in this setting. In addition, we have focused on the role of the gut microbiome and tumour microenvironment in the development of future therapeutic strategies for RCC patients.
Collapse
Affiliation(s)
| | | | | | - Alessandro Conti
- Department of Urology, Bressanone/Brixen Hospital, Bressanone, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Antonio Lopez-Beltran
- Department of Pathology and Surgery, University of Cordoba, Faculty of Medicine, Cordoba, Spain
| |
Collapse
|
88
|
Zizzari IG, Napoletano C, Botticelli A, Caponnetto S, Calabrò F, Gelibter A, Rughetti A, Ruscito I, Rahimi H, Rossi E, Schinzari G, Marchetti P, Nuti M. TK Inhibitor Pazopanib Primes DCs by Downregulation of the β-Catenin Pathway. Cancer Immunol Res 2018; 6:711-722. [PMID: 29700053 DOI: 10.1158/2326-6066.cir-17-0594] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 02/15/2018] [Accepted: 04/06/2018] [Indexed: 11/16/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) target angiogenesis by affecting, for example, the VEGF receptors in tumors and have improved outcomes for patients with metastatic renal cell carcinoma (mRCC). Immune checkpoint inhibitors (ICIs) have also been proposed for treatment of mRCC with encouraging results. A better understanding of the activity of immune cells in mRCC, the immunomodulatory effects of TKIs, and the characteristics defining patients most likely to benefit from various therapies will help optimize immunotherapeutic approaches. In this study, we investigated the influence of the TKI pazopanib on dendritic cell (DC) performance and immune priming. Pazopanib improved DC differentiation and performance by promoting upregulation of the maturation markers HLA-DR, CD40, and CCR7; decreasing IL10 production and endocytosis; and increasing T-cell proliferation. PD-L1 expression was also downregulated. Our results demonstrate that pazopanib inhibits the Erk/β-catenin pathway, suggesting this pathway might be involved in increased DC activation. Similar results were confirmed in DCs differentiated from mRCC patients during pazopanib treatment. In treated patients pazopanib appeared to enhance a circulating CD4+ T-cell population that expresses CD137 (4-1BB). These results suggest that a potentially exploitable immunomodulatory effect induced by pazopanib could improve responses of patients with mRCC in customized protocols combining TKIs with ICI immunotherapy. Cancer Immunol Res; 6(6); 711-22. ©2018 AACR.
Collapse
Affiliation(s)
- Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy.
| | - Chiara Napoletano
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Andrea Botticelli
- Division of Oncology, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Salvatore Caponnetto
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Fabio Calabrò
- Division of Medical Oncology B, San Camillo Forlanini Hospital Rome, Rome, Italy
| | - Alain Gelibter
- Division of Oncology, Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Aurelia Rughetti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Ilary Ruscito
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Hassan Rahimi
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| | - Ernesto Rossi
- Department of Medical Oncology, Fondazione Policlinico A. Gemelli Rome, Italy
| | - Giovanni Schinzari
- Department of Medical Oncology, Fondazione Policlinico A. Gemelli Rome, Italy
| | - Paolo Marchetti
- Division of Oncology, Department of Clinical and Molecular Medicine, Ospedale Sant'Andrea, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
89
|
In papillary thyroid carcinoma, expression by immunohistochemistry of BRAF V600E, PD-L1, and PD-1 is closely related. Virchows Arch 2018; 472:779-787. [PMID: 29651624 DOI: 10.1007/s00428-018-2357-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 11/14/2017] [Accepted: 04/03/2018] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitor therapies targeting PD-L1/PD-1 have been shown to be effective in treating several types of human cancer. In papillary thyroid carcinoma (PTC), little is known about the expression of PD-L1/PD-1 in the tumor microenvironment or its potential correlation with BRAF V600E mutation status. In this study, we examined the expression of PD-L1, PD-1, and BRAF V600E in PTC by immunohistochemistry and investigated the clinical significance of expression status. We studied the expression of PD-L1, PD-1, and BRAF V600E by immunohistochemical staining in 110 cases of PTC with a diameter > 1 cm. Cases with a background of chronic lymphocytic thyroiditis (CLT) were excluded, as differentiating lymphocytes in the context of CLT from tumor-infiltrating lymphocytes (TILs) is difficult. We classified PD-L1+/PD-1+ expression as type 1 (41%), PD-L1-/PD-1- as type 2 (17%), PD-L1+/PD-1- as type 3 (5%), and PD-L1-/PD-1+ as type 4 (37%). Significant correlations were found between expression of BRAF V600E and that of PD-L1 and PD-1. The positive correlation observed between expression of BRAF V600E and PD-L1/PD-1 suggests that immunotherapies targeting PD-L1/PD-1 might be effective for PTC patients with the BRAF V600E mutation, which are refractory to radioiodine therapy.
Collapse
|
90
|
PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2018; 8:2171-2186. [PMID: 27974689 PMCID: PMC5356790 DOI: 10.18632/oncotarget.13895] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/21/2016] [Indexed: 12/31/2022] Open
Abstract
Checkpoint programmed death-1 (PD-1)/programmed cell death ligands (PD-Ls) have been identified as negative immunoregulatory molecules that promote immune evasion of tumor cells. The interaction of PD-1 and PD-Ls inhibits the function of T cells and tumor-infiltrating lymphocytes (TIL) while increasing the function of immunosuppressive regulatory T cells (Tregs). This condition causes the tumor cells to evade immune response. Thus, the blockade of PD-1/PD-L1 enhances anti-tumor immunity by reducing the number and/or the suppressive activity of Tregs and by restoring the activity of effector T cells. Furthermore, some monoclonal antibodies blockading PD-1/PD-Ls axis have achieved good effect and received Food and Drug Administration approval. The role of PD-1/PD-Ls in tumors has been well studied, but little is known on the mechanism by which PD-1 blocks T-cell activation. In this study, we provide a brief overview on the discovery and regulatory mechanism of PD-1 and PD-L1 dysregulation in tumors, as well as the function and signaling pathway of PD-1 and its ligands; their roles in tumor evasion and clinical treatment were also studied.
Collapse
|
91
|
Zakharia Y, Bhattacharya A, Rustum YM. Selenium targets resistance biomarkers enhancing efficacy while reducing toxicity of anti-cancer drugs: preclinical and clinical development. Oncotarget 2018; 9:10765-10783. [PMID: 29535842 PMCID: PMC5828194 DOI: 10.18632/oncotarget.24297] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 01/13/2018] [Indexed: 12/31/2022] Open
Abstract
Selenium (Se)-containing molecules exert antioxidant properties and modulate targets associated with tumor growth, metastasis, angiogenesis, and drug resistance. Prevention clinical trials with low-dose supplementation of different types of Se molecules have yielded conflicting results. Utilizing several xenograft models, we earlier reported that the enhanced antitumor activity of various chemotherapeutic agents by selenomethione and Se-methylselenocysteine in several human tumor xenografts is highly dose- and schedule-dependent. Further, Se pretreament offered selective protection of normal tissues from drug-induced toxicity, thereby allowing higher dosing than maximum tolerated doses. These enhanced therapeutic effects were associated with inhibition of hypoxia-inducible factor 1- and 2-alpha (HIF1α, HIF2α) protein, nuclear factor (erythyroid-derived 2)-like 2 (Nrf2) and pair-related homeobox-1 (Prx1) transcription factors, downregulation of oncogenic- and upregulation of tumor suppressor miRNAs. This review provides: 1) a brief update of clinical prevention trials with Se; 2) advances in the use of specific types, doses, and schedules of Se that selectively modulate antitumor activity and toxicity of anti-cancer drugs; 3) identification of targets selectively modulated by Se; 4) plasma and tumor tissue Se levels achieved after oral administration of Se in xenograft models and cancer patients; 5) development of a phase 1 clinical trial with escalating doses of orally administered selenomethionine in sequential combination with axitinib to patients with advanced clear cell renal cell carcinoma; and 6) clinical prospects for future therapeutic use of Se in combination with anticancer drugs.
Collapse
Affiliation(s)
- Yousef Zakharia
- University of Iowa Division of Medical Oncology and Hematology, Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Arup Bhattacharya
- Roswell Park Cancer Institute, Department of Pharmacology and Therapeutics, Buffalo, NY, USA
| | - Youcef M. Rustum
- University of Iowa Division of Medical Oncology and Hematology, Holden Comprehensive Cancer Center, Iowa City, IA, USA
- Roswell Park Cancer Institute, Department of Pharmacology and Therapeutics, Buffalo, NY, USA
| |
Collapse
|
92
|
Juengel E, Krueger G, Rutz J, Nelson K, Werner I, Relja B, Seliger B, Fisslthaler B, Fleming I, Tsaur I, Haferkamp A, Blaheta RA. Renal cell carcinoma alters endothelial receptor expression responsible for leukocyte adhesion. Oncotarget 2018; 7:20410-24. [PMID: 26943029 PMCID: PMC4991464 DOI: 10.18632/oncotarget.7804] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 02/16/2016] [Indexed: 12/17/2022] Open
Abstract
Renal cell carcinoma (RCC) escapes immune recognition. To elaborate the escape strategy the influence of RCC cells on endothelial receptor expression and endothelial leukocyte adhesion was evaluated. Human umbilical vein endothelial cells (HUVEC) were co-cultured with the RCC cell line, Caki-1, with and without tumor necrosis factor (TNF)-alpha. Intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), endothelial (E)-selectin, standard and variants (V) of CD44 were then analysed in HUVEC, using flow cytometry and Western blot analysis. To determine which components are responsible for HUVEC-Caki-1 interaction causing receptor alteration, Caki-1 membrane fragments versus cell culture supernatant were applied to HUVECS. Adhesion of peripheral blood lymphocytes (PBL) and polymorphonuclear neutrophils (PMN) to endothelium was evaluated by co-culture adhesion assays. Relevance of endothelial receptor expression for adhesion to endothelium was determined by receptor blockage. Co-culture of RCC and HUVECs resulted in a significant increase in endothelial ICAM-1, VCAM-1, E-selectin, CD44 V3 and V7 expression. Previous stimulation of HUVECs with TNF-alpha and co-cultivation with Caki-1 resulted in further elevation of endothelial CD44 V3 and V7 expression, whereas ICAM-1, VCAM-1 and E-selectin expression were significantly diminished. Since Caki-1 membrane fragments also caused these alterations, but cell culture supernatant did not, cell-cell contact may be responsible for this process. Blocking ICAM-1, VCAM-1, E-selectin or CD44 with respective antibodies led to a significant decrease in PBL and PMN adhesion to endothelium. Thus, exposing HUVEC to Caki-1 results in significant alteration of endothelial receptor expression and subsequent endothelial attachment of PBL and PMN.
Collapse
Affiliation(s)
- Eva Juengel
- Department of Urology, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Geraldine Krueger
- Department of Urology, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Jochen Rutz
- Department of Urology, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Karen Nelson
- Department of Vascular and Endovascular Surgery, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Isabella Werner
- Department of Thoracic, Cardiac and Vascular Surgery, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Barbara Seliger
- Institute for Medical Immunology, Medical Faculty, Martin Luther University, Halle, Germany
| | - Beate Fisslthaler
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Igor Tsaur
- Department of Urology, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Axel Haferkamp
- Department of Urology, Goethe-University Hospital, Frankfurt am Main, Germany
| | - Roman A Blaheta
- Department of Urology, Goethe-University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
93
|
Hahn SA, Neuhoff A, Landsberg J, Schupp J, Eberts D, Leukel P, Bros M, Weilbaecher M, Schuppan D, Grabbe S, Tueting T, Lennerz V, Sommer C, Jonuleit H, Tuettenberg A. A key role of GARP in the immune suppressive tumor microenvironment. Oncotarget 2018; 7:42996-43009. [PMID: 27248166 PMCID: PMC5190003 DOI: 10.18632/oncotarget.9598] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/14/2016] [Indexed: 12/26/2022] Open
Abstract
In melanoma patients, one of the main reasons for tumor immune escape and therapy failure is the immunosuppressive tumor microenvironment. Herein, suppressive immune cells and inhibitory factors secreted by the tumor itself play a central role. In the present study we show that the Treg activation marker GARP (glycoprotein A repetitions predominant), known to induce peripheral tolerance in a TGF-β dependent way, is also expressed on human primary melanoma. Interestingly, membrane bound GARP is shed from the surface of both, activated Treg and melanoma cells, and, in its soluble form (sGARP), not only induces peripheral Treg but also a tumor associated (M2) macrophage phenotype. Notably, proliferation of cytotoxic T cells and their effector function is inhibited in the presence of sGARP. GARP expression on Treg and melanoma cells is significantly decreased in the presence of agents such as IFN-α, thus explaining at least in part a novel mechanism of action of this adjuvant therapy. In conclusion, GARP in its soluble and membrane bound form contributes to peripheral tolerance in a multipronged way, potentiates the immunosuppressive tumor microenvironment and thus acts as a negative regulator in melanoma patients. Therefore, it may qualify as a promising target and a new checkpoint for cancer immunotherapy.
Collapse
Affiliation(s)
- Susanne A Hahn
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Annemarie Neuhoff
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Jenny Landsberg
- Department of Dermatology, University Medical Center, Bonn, Germany
| | - Jonathan Schupp
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Daniela Eberts
- Department of Medicine II, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Petra Leukel
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Martin Weilbaecher
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immunotherapy (FZI), University Medical Center, Johannes Gutenberg-University, Mainz, Germany.,Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Thomas Tueting
- Department of Dermatology, University Medical Center, Bonn, Germany
| | - Volker Lennerz
- Department of Medicine II, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Clemens Sommer
- Institute of Neuropathology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
94
|
Joung JY, Kwon WA, Lim J, Oh CM, Jung KW, Kim SH, Seo HK, Park WS, Chung J, Lee KH, Won YJ. Second Primary Cancer Risk among Kidney Cancer Patients in Korea: A Population-Based Cohort Study. Cancer Res Treat 2018; 50:293-301. [PMID: 28421722 PMCID: PMC5784635 DOI: 10.4143/crt.2016.543] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/14/2017] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Secondary primary cancers (SPCs) commonly arise in patients with renal cell carcinoma (RCC). We designed the present study to estimate the SPC incidence in Korean patients with RCC. MATERIALS AND METHODS The study cohort was population-based and consisted of 40,347 individuals from the Korean Central Cancer Registry who were diagnosed with primary renal cancer between 1993 and 2013. Standardized incidence ratios (SIRs) for SPCs were estimated for different ages at diagnosis, latencies, diagnostic periods, and treatments. RESULTS For patients with primary RCC, the risk of developing a SPC was higher than the risk of developing cancer in the general population (SIR, 1.13; 95% confidence interval, 1.08 to 1.18). Most cancer types showed higher incidences in patients with RCC than in the general population. However, the relative incidence of gastric cancer as an SPC varied by age. Gastric cancer incidence was elevated in young patients (< 30 years) with RCC, but reduced in older (≥ 30) patients with RCC. Patients with advanced RCC died prematurely, regardless of SPC development. In contrast, those with early-stage RCC survived for longer periods, although SPC development affected their post-RCC survival. After SPC development, women had better survival than men. CONCLUSION In Korean patients with primary RCC, the incidence of SPC was 13% higher than the incidence of cancer in the general population. These findings may play important roles in the conduct of follow-up evaluations and education for patients with RCC.
Collapse
Affiliation(s)
- Jae Young Joung
- Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Whi-An Kwon
- Department of Urology, Institute of Wonkwang Medical Science, Wonkwang University Sanbon Hospital, Wonkwang University School of Medicine, Gunpo, Korea
| | - Jiwon Lim
- Cancer Registration and Statistics Branch, National Cancer Center, Goyang, Korea
| | - Chang-Mo Oh
- Cancer Registration and Statistics Branch, National Cancer Center, Goyang, Korea
| | - Kyu-Won Jung
- Cancer Registration and Statistics Branch, National Cancer Center, Goyang, Korea
| | - Sung Han Kim
- Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Ho Kyung Seo
- Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Weon Seo Park
- Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Jinsoo Chung
- Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Kang Hyun Lee
- Center for Prostate Cancer, National Cancer Center, Goyang, Korea
| | - Young-Joo Won
- Cancer Registration and Statistics Branch, National Cancer Center, Goyang, Korea
| |
Collapse
|
95
|
Lin Z, Xu Y, Zhang Y, He Q, Zhang J, He J, Liang W. The prevalence and clinicopathological features of programmed death-ligand 1 (PD-L1) expression: a pooled analysis of literatures. Oncotarget 2017; 7:15033-46. [PMID: 26930715 PMCID: PMC4924769 DOI: 10.18632/oncotarget.7590] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/20/2016] [Indexed: 11/25/2022] Open
Abstract
Background & Aims Programmed death-ligand 1 (PD-L1) has been recognized as a critical and promising target in therapies that direct immune escape of cancers. However, its association with aggressive clinicopathological features in solid tumors remains unclear. We investigated this question by synthesizing published articles. Methods Electronic databases were searched for relevant studies. Outcomes of interest included age, gender, tumor size, tumor size, lymph node metastasis and tumor cell differentiation. Results A total of 61 studies involving 17 types of malignancies were included. The overall expression rate of PD-L1 was 44.5% (95% CI, 37.5% to 51.6 %). Patients with regional lymph node metastases (OR 1.38; P < 0.01), large size tumor (OR 1.89; P < 0.01) or poor differentiated tumors (OR 1.71; P < 0.01) were associated with higher PD-L1 expression rate. However, no significant association was observed between young and elder patients (OR 1.04; P = 0.58), or male and female patients (OR 1.13; P = 0.06). A numerically higher PD-L1 expression rate was detected in polyclonal antibodies (57.2%) than monoclonal antibodies (39.6%). In addition, the PD-L1 expression rate reported by studies from Asian areas (52.3%) was numerically higher than those from non-Asian areas, namely Caucasians (32.7%). Conclusions This meta-analysis indicated that patients with larger tumors, regional lymph node metastases, or poor-differentiated tumors were associated with a higher PD-L1 expression rate; in addition the expression rate of PD-L1 in Asians might be higher than that of Caucasians. This information might be useful in screening candidates for relevant tests and treatments.
Collapse
Affiliation(s)
- Ziying Lin
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yutong Xu
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaxiong Zhang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China.,Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Medical Oncology of Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qihua He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Jianrong Zhang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Wenhua Liang
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Guangzhou Institute of Respiratory Disease & China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| |
Collapse
|
96
|
Rasti A, Madjd Z, Abolhasani M, Mehrazma M, Janani L, Saeednejad Zanjani L, Asgari M. Cytoplasmic expression of Twist1, an EMT-related transcription factor, is associated with higher grades renal cell carcinomas and worse progression-free survival in clear cell renal cell carcinoma. Clin Exp Med 2017; 18:177-190. [PMID: 29204790 DOI: 10.1007/s10238-017-0481-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 11/19/2017] [Indexed: 12/17/2022]
Abstract
Twist1 is a key transcription factor, which confers tumor cells with cancer stem cell (CSC)-like characteristics and enhances epithelial-mesenchymal transition in pathological conditions including tumor malignancy and metastasis. This study aimed to evaluate the expression patterns and clinical significance of Twist1 in renal cell carcinoma (RCC). The cytoplasmic and nuclear expression of Twist1 were examined in 252 well-defined renal tumor tissues, including 173 (68.7%) clear cell renal cell carcinomas (ccRCC), 45 (17.9%) papillary renal cell carcinomas (pRCC) and 34 (13.5%) chromophobe renal cell carcinoma, by immunohistochemistry on a tissue microarray. The association between expression of this marker and clinicopathologic parameters and survival outcomes were then analyzed. Twist1 was mainly localized to the cytoplasm of tumor cells (98.8%). Increased cytoplasmic expression of Twist1 was associated with higher grade tumors (P = 0.045), renal vein invasion (P = 0.031) and microvascular invasion (P = 0.044) in RCC. It was positively correlated with higher grade tumors (P = 0.026), shorter progression-free survival time (P = 0.027) in patients with ccRCC, and also with higher stage in pRCC patients (P = 0.036). Significantly higher cytoplasmic expression levels of Twist1 were found in ccRCC and pRCC subtypes, due to their more aggressive tumor behavior. Increased cytoplasmic expression of Twist1 had a critical role in worse prognosis in ccRCC. These findings suggest that cytoplasmic, rather than nuclear expression of Twist1 can be considered as a prognostic and therapeutic marker for targeted therapy of RCC, especially for ccRCC patients.
Collapse
Affiliation(s)
- Arezoo Rasti
- Oncopathology Research Centre, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next TO Milad Tower, Tehran, 14496-14530, Iran
| | - Zahra Madjd
- Oncopathology Research Centre, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next TO Milad Tower, Tehran, 14496-14530, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maryam Abolhasani
- Oncopathology Research Centre, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next TO Milad Tower, Tehran, 14496-14530, Iran. .,Hasheminejad Kidney Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran.
| | - Mitra Mehrazma
- Oncopathology Research Centre, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next TO Milad Tower, Tehran, 14496-14530, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran
| | - Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Leili Saeednejad Zanjani
- Oncopathology Research Centre, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next TO Milad Tower, Tehran, 14496-14530, Iran
| | - Mojgan Asgari
- Oncopathology Research Centre, Iran University of Medical Sciences (IUMS), Hemmat Street (Highway), Next TO Milad Tower, Tehran, 14496-14530, Iran.,Hasheminejad Kidney Center, Iran University of Medical Sciences, (IUMS), Tehran, Iran
| |
Collapse
|
97
|
Rotz SJ, Leino D, Szabo S, Mangino JL, Turpin BK, Pressey JG. Severe cytokine release syndrome in a patient receiving PD-1-directed therapy. Pediatr Blood Cancer 2017; 64. [PMID: 28544595 DOI: 10.1002/pbc.26642] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/24/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
Cytokine release syndrome (CRS) is a phenomenon of immune hyperactivation described in the setting of cellular and bispecific T-cell engaging immunotherapy. Checkpoint blockade using anti-programmed cell death 1 (anti-PD-1) inhibitors is an approach to antitumor immune system stimulation. A 29-year-old female with alveolar soft part sarcoma developed severe CRS after treatment with anti-PD-1 therapy. CRS was characterized by high fevers, encephalopathy, hypotension, hypoxia, hepatic dysfunction, and evidence of coagulopathy, and resolved after infusion of the interleukin-6 inhibitor tocilizumab and corticosteroids.
Collapse
Affiliation(s)
- Seth J Rotz
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Daniel Leino
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Sara Szabo
- Department of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jennifer L Mangino
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Brian K Turpin
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Joseph G Pressey
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
98
|
Xu J, Zhang Q, Tian K, Wang H, Yin H, Zheng J. Current status and future prospects of the strategy of combining CAR‑T with PD‑1 blockade for antitumor therapy (Review). Mol Med Rep 2017; 17:2083-2088. [PMID: 29207115 DOI: 10.3892/mmr.2017.8129] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 08/04/2017] [Indexed: 11/06/2022] Open
Abstract
The immune system serves an important role in controlling and eradicating malignant cells. Immunotherapy for treating tumors has received much attention in recent years due to its marked effect. There are two approaches which currently lead this field: Chimeric antigen receptor‑modified T‑cell immunotherapy (CAR‑T) and programmed cell death protein-1 blockade (PD‑1 blockade). CAR‑T has emerged as a promising regimen for the treatment of a range of types of cancer, including chronic lymphoid leukemia and neuroblastoma, with studies of long term remission in certain patients. PD‑1 blockade has been reported to exert marked clinical responses in patients against a range of types of solid cancer, including advanced melanoma, non‑small‑cell lung cancer and renal cell carcinoma, in addition to hematological malignancies. While increasing the power of the immune system to fight cancer has been a long‑standing goal in oncology, a number of studies have demonstrated the synergistic antitumor effects of combination therapies under the umbrella of immunotherapy. The present review focused on a novel combination approach involving CAR‑T and PD‑1 blockade. The present reviews aimed to discuss the following four aspects of such an approach: i) Current monotherapy status; ii) rationale for the combination of CAR‑T and PD‑1 blockade; iii) current status of the combination of CAR‑T and PD‑1 blockade; and iv) conclusions and future perspectives.
Collapse
Affiliation(s)
- Jinjing Xu
- Cancer Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Qing Zhang
- Cancer Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Kang Tian
- Cancer Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Haiyu Wang
- Cancer Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Hong Yin
- Cancer Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Junnian Zheng
- Cancer Institute of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
99
|
Mauzo SH, Tetzlaff MT, Nelson K, Amaria R, Patel S, Aung PP, Nagarajan P, Torres‐Cabala CA, Diab A, Prieto VG, Curry JL. Regressed melanocytic nevi secondary to pembrolizumab therapy: an emerging melanocytic dermatologic effect from immune checkpoint antibody blockade. Int J Dermatol 2017; 58:1045-1052. [DOI: 10.1111/ijd.13833] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 09/03/2017] [Accepted: 10/06/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Shakuntala H. Mauzo
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Michael T. Tetzlaff
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Kelly Nelson
- Department of Dermatology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Rodabe Amaria
- Department of Melanoma Medical Oncology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Sapna Patel
- Department of Melanoma Medical Oncology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Phyu P. Aung
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Priyadharsini Nagarajan
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Carlos A. Torres‐Cabala
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
- Department of Dermatology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Adi Diab
- Department of Melanoma Medical Oncology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Victor G. Prieto
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
- Department of Dermatology The University of Texas MD Anderson Cancer Center Houston TX USA
| | - Jonathan L. Curry
- Department of Pathology Section of Dermatopathology The University of Texas MD Anderson Cancer Center Houston TX USA
- Department of Dermatology The University of Texas MD Anderson Cancer Center Houston TX USA
| |
Collapse
|
100
|
Chen Y, Li Q, Li X, Ma D, Fang J, Luo L, Liu X, Wang X, Lui VWY, Xia J, Cheng B, Wang Z. Blockade of PD-1 effectively inhibits in vivo malignant transformation of oral mucosa. Oncoimmunology 2017; 7:e1388484. [PMID: 29308315 DOI: 10.1080/2162402x.2017.1388484] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/25/2017] [Accepted: 09/30/2017] [Indexed: 12/11/2022] Open
Abstract
Curbing PD-1 immunosuppressive signaling represents an effective immune awakening or immune-reactivation approach for tumor eradication for many cancers. Yet, the potential involvement of this critical PD-1 immunosuppressive signaling in de novo malignant transformation of epithelial cells to pre-cancerous or cancerous lesions is largely unknown. In this study, we demonstrate that PD-1 signaling is critically involved in de novo malignant transformation of oral mucosa upon carcinogen exposure in vivo. Our findings revealed that 4NQO-treated mice had almost double the numbers of PD-1-positive CD4+ cells and PD-1-positive CD8+ cells in peripheral blood lymphocytes as well as elevated PD-1 expression in tumor infiltrating lymphocytes (when compared to that of control-treated mice), strongly supportive of a general immune-suppression induced by carcinogen challenges in vivo. Importantly, inhibition of PD-1 signaling during the carcinogenesis process (immediately after 4NQO challenge) significantly reduced and delayed de novo formation of both pre-cancerous and cancerous lesions in vivo, in conjunction with effective PD-1 down-modulation in the tumor infiltrating leukocyte and peripheral lymph organs. Lastly, reduction of carcinogen-induced lesions upon PD-1 mAb treatment in vivo was accompanied by reduction of potent immunosuppressive myeloid-derived suppressor cells (MDSCs), and increase in "activated" T cell accumulations in the lesion-microenvironment (127% increase) and peripheral lymph nodes (25% increase). These data support PD-1 blockade as a new approach to enhance the efficacy of T-cell immunotherapy and reduce canceration rate in premalignant lesions.
Collapse
Affiliation(s)
- Yichen Chen
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Qiusheng Li
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Xinye Li
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Da Ma
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Juan Fang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Liqun Luo
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiangqi Liu
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Xi Wang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Vivian Wai Yan Lui
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong SAR
| | - Juan Xia
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Bin Cheng
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| | - Zhi Wang
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Stomatological Hospital, Sun Yat-Sen University, No. 56, Lingyuan West Road, Guangzhou, Guangdong, China
| |
Collapse
|