1
|
Sabbatino F, Scognamiglio G, Liguori L, Marra A, Anniciello AM, Polcaro G, Dal Col J, Caputo A, Peluso AL, Botti G, Zeppa P, Ferrone S, Pepe S. Peritumoral Immune Infiltrate as a Prognostic Biomarker in Thin Melanoma. Front Immunol 2020; 11:561390. [PMID: 33117345 PMCID: PMC7550791 DOI: 10.3389/fimmu.2020.561390] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Thin melanomas are tumors less than 1 mm thick according to Breslow classification. Their prognosis is in most cases excellent. However, a small subset of these tumors relapses. These clinical findings emphasize the need of novel prognostic biomarkers to identify this subset of tumors. Characterization of tumor immune microenvironment (TIME) is currently investigated as a prognostic and predictive biomarker for cancer immunotherapy in several solid tumors including melanoma. Here, taking into account the limited availability of tumor tissues, by characterizing some of the characteristics of TIME such as number of infiltrating lymphocytes, HLA class I antigen and PD-L1 expression, we show that number of infiltrating CD8+ and FOXP3+ T cells as well as CD8+/FOXP3+ T cell ratio can represent a useful prognostic biomarker in thin melanoma. Although further investigations in a larger patient cohort are needed, these findings have potential clinical significance since they can be used to define subgroups of thin melanoma patients who have a worse prognosis and might need different treatment modalities.
Collapse
Affiliation(s)
- Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Oncology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, Salerno, Italy
| | - Giosuè Scognamiglio
- Pathology Unit, Istituto Nazionale Tumori, IRCSS, “Fondazione G. Pascale”, Naples, Italy
| | - Luigi Liguori
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Naples, Italy
| | - Antonio Marra
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology, Milan, Italy
| | - Anna Maria Anniciello
- Pathology Unit, Istituto Nazionale Tumori, IRCSS, “Fondazione G. Pascale”, Naples, Italy
| | - Giovanna Polcaro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Jessica Dal Col
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
| | - Alessandro Caputo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Pathology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, Salerno, Italy
| | - Anna Lucia Peluso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Pathology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, Salerno, Italy
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Pio Zeppa
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Pathology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, Salerno, Italy
| | - Soldano Ferrone
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Stefano Pepe
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, Italy
- Oncology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, Salerno, Italy
| |
Collapse
|
2
|
Cell death-based treatments of melanoma:conventional treatments and new therapeutic strategies. Cell Death Dis 2018; 9:112. [PMID: 29371600 PMCID: PMC5833861 DOI: 10.1038/s41419-017-0059-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/17/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022]
Abstract
The incidence of malignant melanoma has continued to rise during the past decades. However, in the last few years, treatment protocols have significantly been improved thanks to a better understanding of the key oncogenes and signaling pathways involved in its pathogenesis and progression. Anticancer therapy would either kill tumor cells by triggering apoptosis or permanently arrest them in the G1 phase of the cell cycle. Unfortunately, melanoma is often refractory to commonly used anticancer drugs. More recently, however, some new anticancer strategies have been developed that are “external” to cancer cells, for example stimulating the immune system’s response or inhibiting angiogenesis. In fact, the increasing knowledge of melanoma pathogenetic mechanisms, in particular the discovery of genetic mutations activating specific oncogenes, stimulated the development of molecularly targeted therapies, a form of treatment in which a drug (chemical or biological) is developed with the goal of exclusively destroying cancer cells by interfering with specific molecules that drive growth and spreading of the tumor. Again, after the initial exciting results associated with targeted therapy, tumor resistance and/or relapse of the melanoma lesion have been observed. Hence, very recently, new therapeutic strategies based on the modulation of the immune system function have been developed. Since cancer cells are known to be capable of evading immune-mediated surveillance, i.e., to block the immune system cell activity, a series of molecular strategies, including monoclonal antibodies, have been developed in order to “release the brakes” on the immune system igniting immune reactivation and hindering metastatic melanoma cell growth. In this review we analyze the various biological strategies underlying conventional chemotherapy as well as the most recently developed targeted therapies and immunotherapies, pointing at the molecular mechanisms of cell injury and death engaged by the different classes of therapeutic agents.
Collapse
|
3
|
Li Z, Zhao K, Tian H. Integrated analysis of differential expression and alternative splicing of non-small cell lung cancer based on RNA sequencing. Oncol Lett 2017; 14:1519-1525. [PMID: 28789374 PMCID: PMC5529932 DOI: 10.3892/ol.2017.6300] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 03/03/2017] [Indexed: 12/11/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, with high morbidity and mortality rates. Numerous diagnosis and treatment methods have been proposed, and the prognosis of NSCLC has improved to a certain extent. However, the mechanisms of NSCLC remain largely unknown, and additional studies are required. In the present study, the RNA sequencing dataset of NSCLC was downloaded from the Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo/). The clean reads obtained from the raw data were mapped to the University of California Santa Cruz human genome (hg19), based on TopHat, and were assembled into transcripts via Cufflink. The differential expression (DE) and differential alternative splicing (DAS) genes were screened out through Cuffdiff and rMATS, respectively. The significantly enriched gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes pathways were obtained through the Database of Annotation, Visualization and Integrated Discovery (DAVID). Different numbers of DE and DAS genes were identified in different types of NSCLC samples, but a number of common functions and pathways were obtained, including biological processes associated with abnormal immune and cell activity. GO terms and pathways associated with substance metabolism, including the insulin signaling pathway and oxidative phosphorylation, were enriched in DAS genes rather than DE genes. Integrated analysis of differential expression and alternative splicing may be helpful in understanding the mechanisms of NSCLC, in addition to its early diagnosis and treatment.
Collapse
Affiliation(s)
- Zulei Li
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Thoracic Surgery, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Kai Zhao
- Department of Thoracic Surgery, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
4
|
Wang X, Bao Z, Zhang X, Li F, Lai T, Cao C, Chen Z, Li W, Shen H, Ying S. Effectiveness and safety of PD-1/PD-L1 inhibitors in the treatment of solid tumors: a systematic review and meta-analysis. Oncotarget 2017; 8:59901-59914. [PMID: 28938692 PMCID: PMC5601788 DOI: 10.18632/oncotarget.18316] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 03/01/2017] [Indexed: 01/01/2023] Open
Abstract
Background PD-1/PD-L1 inhibitors have been implicated as potentially effective anti-cancer therapies. Some clinical randomized controlled trials (RCTs) have been completed for a variety of PD-1/PD-L1 inhibitors to treat various malignancies, and more RCTs are still under way. We carried out this systematic meta-analysis to evaluate the efficacy and safety of PD-1/PD-L1 inhibitors in the treatment of solid tumors. Methods We searched PubMed, EMBASE, clinical trial registers, conference reports, and related reviews. Eligible RCTs that compared PD-1/PD-L1 inhibitors with other chemotherapy agents or placebo in solid tumor patients were included. For each RCT, progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), stable disease rate (SDR), progressive disease rate (PDR), and adverse events (AEs) were pooled for meta-analysis. Findings Based on an analysis of 10 eligible RCTs, PD-1/PD-L1 inhibitors were found to significantly improve PFS (Hazard ratio (HR), 0.65; 95% confidence interval (CI) 0.53 to 0.79, P<0.001), OS (HR, 0.69; 95%CI 0.62 to 0.76, P<0.001), and ORR (Risk Ratio (RR) 292; 95% confidence interval (CI) 2.06 to 4.15, P<0.00001) in all populations, including melanoma and NSCLC subgroups. However, they failed to increase the DCR of cancer patients (RR 1.15; 95%CI 0.91 to 1.45, P=0.25). Furthermore, less AEs were observed in the PD-1/PD-L1 inhibitor groups than the control groups. Interpretation PD-1 inhibitors are more effective for improving the PFS, OS, and ORR of cancer patients with little toxicity, despite having little effect on increasing of the DCR.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengqiang Bao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoju Zhang
- Department of Respiratory Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Fei Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianwen Lai
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Chen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China
| | - Huahao Shen
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,State Key Laboratory of Respiratory Diseases, Guangzhou, China
| | - Songmin Ying
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Institute of Respiratory Diseases, Zhejiang University School of Medicine, Hangzhou, China.,Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
Wang H, Zhang L, Yang L, Liu C, Zhang Q, Zhang L. Targeting macrophage anti-tumor activity to suppress melanoma progression. Oncotarget 2017; 8:18486-18496. [PMID: 28060744 PMCID: PMC5392344 DOI: 10.18632/oncotarget.14474] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 12/27/2016] [Indexed: 01/09/2023] Open
Abstract
By phagocytosing cancer cells and their cellular debris, macrophages play a critical role in nonspecific defense (innate immunity) and, as antigen presenters, they help initiate specific defense mechanisms (adaptive immunity). Malignant melanoma is a lethal disease due to its aggressive capacity for metastasis and resistance to therapy. For decades, considerable effort has gone into development of an effective immunotherapy for treatment of metastatic melanoma. In this review, we focus on the anti-tumor activities of macrophages in melanoma and their potential as therapeutic targets in melanoma. Although macrophages can be re-educated through intercellular signaling to promote tumor survival owing to their plasticity, we expect that targeting the anti-tumor activity of macrophages remains a promising strategy for melanoma inhibition. The combination of tumoricidal macrophage activation and other treatments such as surgery, chemotherapy, and radiotherapy, may provide an effective and comprehensive anti-melanoma strategy.
Collapse
Affiliation(s)
- Huafeng Wang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Lijuan Zhang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Luhong Yang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| | - Chengfang Liu
- Department of Human Anatomy, Shanxi Medical University, Shanxi Sheng, China
| | | | - Linjing Zhang
- Modern College of Arts and Science, or School of Life Science, Shanxi Normal University, Linfen, China
| |
Collapse
|
6
|
Iwata T, Kondo Y, Kimura O, Morosawa T, Fujisaka Y, Umetsu T, Kogure T, Inoue J, Nakagome Y, Shimosegawa T. PD-L1 +MDSCs are increased in HCC patients and induced by soluble factor in the tumor microenvironment. Sci Rep 2016; 6:39296. [PMID: 27966626 PMCID: PMC5155242 DOI: 10.1038/srep39296] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/21/2016] [Indexed: 12/11/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) could have important roles in immune regulation, and MDSCs can be induced in patients with various malignant tumors. The immune-suppressive functions of MDSCs in hepatocellular carcinoma (HCC) patients have not been clarified. Therefore, we tried to analyze the biological significance of MDSCs in HCC patients. We quantified PD-L1+MDSCs of HCC patients in various conditions by using multi-color flow cytometry analysis. PBMCs from HCC patients contained significantly higher percentages of PD-L1+MDSCs in comparison to those from healthy subjects (p < 0.001). The percentages of PD-L1+MDSCs were reduced by curative treatment for HCC (p < 0.05), and the percentages of PD-L1+MDSCs before treatment were inversely correlated with disease-free survival time. After we cocultivated PBMCs and several liver cancer cell lines in a transwell coculture system, the percentages of PD-L1+MDSCs were significantly increased compared with control (p < 0.05). The expression of M-CSF and VEGFA was higher in the cell lines that strongly induced PD-L1+MDSCs. Peripheral blood from HCC patients had significantly higher percentages of PD-L1+MDSCs in comparison to those of healthy subjects, and the percentages of PD-L1+MDSCs were reduced by HCC treatment, suggesting that we might use PD-L1+MDSCs as a new biomarker of HCC.
Collapse
Affiliation(s)
- Tomoaki Iwata
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Yasuteru Kondo
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
- Department of Hepatology, Sendai Kousei Hospital, 4-15 Hirose, Aoba, Sendai City, 980-0873, Miyagi, Japan
| | - Osamu Kimura
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Tatsuki Morosawa
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Yasuyuki Fujisaka
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Teruyuki Umetsu
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Takayuki Kogure
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Jun Inoue
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Yu Nakagome
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Hospital, 1-1 Seiryo, Aoba, Sendai City, 980-8574, Miyagi, Japan
| |
Collapse
|
7
|
Agur Z, Halevi-Tobias K, Kogan Y, Shlagman O. Employing dynamical computational models for personalizing cancer immunotherapy. Expert Opin Biol Ther 2016; 16:1373-1385. [PMID: 27564141 DOI: 10.1080/14712598.2016.1223622] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Recently, cancer immunotherapy has shown considerable success, but due to the complexity of the immune-cancer interactions, clinical outcomes vary largely between patients. A possible approach to overcome this difficulty may be to develop new methodologies for personal predictions of therapy outcomes, by the integration of patient data with dynamical mathematical models of the drug-affected pathophysiological processes. AREAS COVERED This review unfolds the story of mathematical modeling in cancer immunotherapy, and examines the feasibility of using these models for immunotherapy personalization. The reviewed studies suggest that response to immunotherapy can be improved by patient-specific regimens, which can be worked out by personalized mathematical models. The studies further indicate that personalized models can be constructed and validated relatively early in treatment. EXPERT OPINION The suggested methodology has the potential to raise the overall efficacy of the developed immunotherapy. If implemented already during drug development it may increase the prospects of the technology being approved for clinical use. However, schedule personalization, per se, does not comply with the current, 'one size fits all,' paradigm of clinical trials. It is worthwhile considering adjustment of the current paradigm to involve personally tailored immunotherapy regimens.
Collapse
Affiliation(s)
- Zvia Agur
- a Institute for Medical BioMathematics (IMBM) , Bene Ataroth , Israel
| | | | - Yuri Kogan
- a Institute for Medical BioMathematics (IMBM) , Bene Ataroth , Israel
| | - Ofer Shlagman
- a Institute for Medical BioMathematics (IMBM) , Bene Ataroth , Israel
| |
Collapse
|