1
|
Trojan A, Lone YC, Briceno I, Trojan J. Anti-Gene IGF-I Vaccines in Cancer Gene Therapy: A Review of a Case of Glioblastoma. Curr Med Chem 2024; 31:1983-2002. [PMID: 38031775 DOI: 10.2174/0109298673237968231106095141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 06/27/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023]
Abstract
OBJECTIVE Vaccines for the deadliest brain tumor - glioblastoma (GBM) - are generally based on targeting growth factors or their receptors, often using antibodies. The vaccines described in the review were prepared to suppress the principal cancer growth factor - IGF-I, using anti-gene approaches either of antisense (AS) or of triple helix (TH) type. Our objective was to increase the median survival of patients treated with AS and TH cell vaccines. METHODOLOGY The cells were transfected in vitro by both constructed IGF-I AS and IGF-I TH expression episomal vectors; part of these cells was co-cultured with plant phytochemicals, modulating IGF-I expression. Both AS and TH approaches completely suppressed IGF-I expression and induced MHC-1 / B7 immunogenicity related to the IGF-I receptor signal. RESULTS This immunogenicity proved to be stronger in IGF-I TH than in IGF-I AS-prepared cell vaccines, especially in TH / phytochemical cells. The AS and TH vaccines generated an important TCD8+ and TCD8+CD11b- immune response in treated GBM patients and increased the median survival of patients up to 17-18 months, particularly using TH vaccines; in some cases, 2- and 3-year survival was reported. These clinical results were compared with those obtained in therapies targeting other growth factors. CONCLUSION The anti-gene IGF-I vaccines continue to be applied in current GBM personalized medicine. Technical improvements in the preparation of AS and TH vaccines to increase MHC-1 and B7 immunogenicity have, in parallel, allowed to increase in the median survival of patients.
Collapse
Affiliation(s)
- Annabelle Trojan
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- Faculty of Medicine, University of Cartagena, PO Box: 130014 Cartagena de Indias, Colombia
| | - Yu-Chun Lone
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- CEDEA / ICGT - Center of Oncological Diseases Diagnosis, PO Box: 110231 Bogota, Colombia
| | - Ignacio Briceno
- Faculty of Medicine, University of La Sabana, PO Box: 250008 Chia, Colombia
| | - Jerzy Trojan
- INSERM UMR 1197, Cancer Center & University of Paris / Saclay, PO Box: 94802 Villejuif, France
- CEDEA / ICGT - Center of Oncological Diseases Diagnosis, PO Box: 110231 Bogota, Colombia
- National Academy of Medicine - ANM, PO Box: 75272 Paris, France
| |
Collapse
|
2
|
Jyotsna F, Ikram J, Nageeta F, Komal F, Anjlee F, Patel H, Nassri T, Kumari M, Kumar R, Shah SU, Kashif M, Varrassi G, Kumar S, Patel T. Unlocking the Potential of Immunotherapy in Cardiovascular Disease: A Comprehensive Review of Applications and Future Directions. Cureus 2023; 15:e42790. [PMID: 37664375 PMCID: PMC10469982 DOI: 10.7759/cureus.42790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023] Open
Abstract
Immunotherapy has emerged as a pioneering therapeutic approach that harnesses the immune system's abilities to combat diseases, particularly in the field of oncology where it has led to significant advancements. However, despite its significant impact in the field of oncology, the potential of immunotherapy in the context of cardiovascular disease (CVD) has not been thoroughly investigated. The purpose of this narrative review is to address the existing knowledge and potential uses of immunotherapy in the field of cardiovascular disease (CVD), with the intention of filling the existing gap in understanding. Furthermore, the review thoroughly examines the future prospects of this swiftly advancing field, providing insights into the aspects that necessitate further investigation and addressing the forthcoming challenges. The review is organized into four distinct sections to enhance comprehension. The first section introduces immunotherapy, presenting the fundamental concepts and principles. The second section explores the immunomodulatory mechanisms in cardiovascular disease (CVD), with a specific focus on the intricate interplay between the immune system and the development of cardiovascular pathogenesis. The utilization of immunotherapy in specific cardiovascular conditions will be examined, investigating the application of immunotherapy in the context of different cardiovascular diseases. The future prospects and challenges in immunotherapy for cardiovascular diseases will be discussed, highlighting the potential areas for future research and addressing the barriers that must be overcome to effectively implement immunotherapeutic interventions in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Fnu Jyotsna
- Medicine, Dr. B.R. Ambedkar Medical College & Hospital, Mohali, IND
| | - Jibran Ikram
- Orthopaedics and Trauma, Rehman Medical Institute, Peshawar, PAK
| | - Fnu Nageeta
- Medicine, Ghulam Muhammad Mahar Medical College, Sukkur, PAK
| | - Fnu Komal
- Medicine, Chandka Medical College, Larkana, PAK
| | - Fnu Anjlee
- Medicine, Ghulam Muhammad Mahar Medical College, Sukkur, PAK
| | - Harshkumar Patel
- Internal Medicine, PDU (Pandit Dindayal Upadhyay) Medical College, Rajkot, IND
| | - Taleb Nassri
- Medicine, Heart and Vascular Institute, Dearborn, USA
| | - Meena Kumari
- Internal Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Rajesh Kumar
- Business Intelligence and Data Analytics, Westcliff University, Irvine, USA
| | | | - Maham Kashif
- Medicine, Khawaja Muhammad Safdar Medical College, Wazirabad, PAK
| | | | - Satesh Kumar
- Medicine and Surgery, Shaheed Mohtarma Benazir Bhutto Medical College, Karachi, PAK
| | - Tirath Patel
- Medicine, American University of Antigua, St. John, ATG
| |
Collapse
|
3
|
Li C, Liu W, Liu C, Luo Q, Luo K, Wei C, Li X, Qin J, Zheng C, Lan C, Wei S, Tan R, Chen J, Chen Y, Huang H, Zhang G, Huang H, Wang X. Integrating machine learning and bioinformatics analysis to m6A regulator-mediated methylation modification models for predicting glioblastoma patients' prognosis and immunotherapy response. Aging (Albany NY) 2023; 15:204495. [PMID: 37244287 DOI: 10.18632/aging.204495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/30/2022] [Indexed: 05/29/2023]
Abstract
BACKGROUND Epigenetic regulations of immune responses are essential for cancer development and growth. As a critical step, comprehensive and rigorous explorations of m6A methylation are important to determine its prognostic significance, tumor microenvironment (TME) infiltration characteristics and underlying relationship with glioblastoma (GBM). METHODS To evaluate m6A modification patterns in GBM, we conducted unsupervised clustering to determine the expression levels of GBM-related m6A regulatory factors and performed differential analysis to obtain m6A-related genes. Consistent clustering was used to generate m6A regulators cluster A and B. Machine learning algorithms were implemented for identifying TME features and predicting the response of GBM patients receiving immunotherapy. RESULTS It is found that the m6A regulatory factor significantly regulates the mutation of GBM and TME. Based on Europe, America, and China data, we established m6Ascore through the m6A model. The model accurately predicted the results of 1206 GBM patients from the discovery cohort. Additionally, a high m6A score was associated with poor prognoses. Significant TME features were found among the different m6A score groups, which demonstrated positive correlations with biological functions (i.e., EMT2) and immune checkpoints. CONCLUSIONS m6A modification was important to characterize the tumorigenesis and TME infiltration in GBM. The m6Ascore provided GBM patients with valuable and accurate prognosis and prediction of clinical response to various treatment modalities, which could be useful to guide patient treatments.
Collapse
Affiliation(s)
- Chuanyu Li
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Wangrui Liu
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Chengming Liu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu, China
| | - Qisheng Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Kunxiang Luo
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Cuicui Wei
- Department of Outpatient, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Xueyu Li
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Jiancheng Qin
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Chuanhua Zheng
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Chuanliu Lan
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Shiyin Wei
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Rong Tan
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Jiaxing Chen
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yuanbiao Chen
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Huadong Huang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Gaolian Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning 530023, Guangxi, China
| | - Haineng Huang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou 510632, Guangdong Province, China
| |
Collapse
|
4
|
Möhn N, Mahjoub S, Duzzi L, Narten E, Grote-Levi L, Körner G, Seeliger T, Beutel G, Bollmann BA, Wirth T, Huss A, Tumani H, Grimmelmann I, Gutzmer R, Ivanyi P, Skripuletz T. Monocyte chemoattractant protein 1 as a potential biomarker for immune checkpoint inhibitor-associated neurotoxicity. Cancer Med 2023; 12:9373-9383. [PMID: 36794673 PMCID: PMC10166892 DOI: 10.1002/cam4.5695] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/25/2023] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Oncological patients can benefit substantially from treatment with immune checkpoint inhibitors (ICI). However, there is a growing awareness of immune-related adverse events (irAE). Especially ICI-mediated neurological adverse events (nAE(+)), are tough to diagnose and biomarkers to identify patients at risk are missing. METHODS A prospective register with prespecified examinations was established for ICI treated patients in December 2019. At the time of data cut-off, 110 patients were enrolled and completed the clinical protocol. Herein, cytokines and serum neurofilament light chain (sNFL) from 21 patients were analyzed. RESULTS nAE of any grade were observed in 31% of the patients (n = 34/110). In nAE(+) patients a significant increase in sNFL concentrations over time was observed. Patients with higher-grade nAE had significantly elevated serum-concentrations of monocyte chemoattractant protein 1 (MCP-1) and brain-derived neurotrophic factor (BDNF) at baseline compared to individuals without any nAE (p < 0.01 and p < 0.05). CONCLUSION Here, we identified nAE to occur more frequently than previously reported. Increase of sNFL during nAE confirms the clinical diagnosis of neurotoxicity and might be a suitable marker for neuronal damage associated with ICI therapy. Furthermore, MCP-1 and BDNF are potentially the first clinical-class nAE predictors for patients under ICI therapy.
Collapse
Affiliation(s)
- Nora Möhn
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Susann Mahjoub
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Laura Duzzi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Emily Narten
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Lea Grote-Levi
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Gudrun Körner
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Tabea Seeliger
- Department of Neurology, Hannover Medical School, Hannover, Germany
| | - Gernot Beutel
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | | - Thomas Wirth
- Department of Gastroenterology, Hannover Medical School, Hannover, Germany
| | - André Huss
- Department of Neurology, University Hospital Ulm, Ulm, Germany
| | | | | | - Ralf Gutzmer
- Skin-Cancer-Center, Hannover Medical School, Hannover, Germany
- Department of Dermatology Venerology, Allergy and Phlebology, Hannover Medical School, Minden, Germany
| | - Philipp Ivanyi
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
5
|
Aldrich J, Ekpo P, Rupji M, Switchenko JM, Torres MA, Kalinsky K, Bhave MA. Racial Disparities in Clinical Outcomes on Investigator-Initiated Breast Cancer Clinical Trials at an Urban Medical Center. Clin Breast Cancer 2023; 23:38-44. [PMID: 36333193 DOI: 10.1016/j.clbc.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/22/2022] [Accepted: 10/09/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND Black women are 40% more likely to die of breast cancer compared to White women. Inadequate representation of Black patients in clinical trials may contribute to health care inequity. We aimed to assess breast cancer clinical outcomes in Non-Hispanic Black (Black) versus Non-Hispanic White (White) women with metastatic breast cancer (MBC) enrolled on investigator-initiated clinical trials at Winship Cancer Institute at Emory University, given the significant number of patients from underrepresented minority groups seen at Winship. MATERIALS AND METHODS Black and White women with MBC on investigator-initiated trials at Emory between 2009 and 2019 were retrospectively evaluated. Univariate analyses and multiple logistic regression models were used to assess clinical response and treatment toxicities. Differences in overall survival between groups was assessed using quantile analysis. RESULTS Sixty-two women with MBC were included (66% White vs. 34% Black). Black patients had less clinical benefit from the trial therapy as only 57% had partial response or stable disease as best response compared to 78% of White women (P = .09). Quantile analysis showed significant difference in mean survival between Whites and Blacks by the end of follow up (64 vs. 38 months). There were no significant differences in toxicities between groups. CONCLUSION Participation rates of Black women with MBC on investigator-initiated clinical trials at an urban cancer center were higher compared to key national trials. Black women had worse treatment response and survival. These results reinforce the need for assessment of tumor differences by ancestry and continued improvement in minority representation on clinical trials.
Collapse
Affiliation(s)
- Jeffrey Aldrich
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Princess Ekpo
- Department of Biology, Emory University, Atlanta, GA
| | - Manali Rupji
- Biostatistics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Jeffrey M Switchenko
- Biostatistics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Mylin A Torres
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Kevin Kalinsky
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Manali A Bhave
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA.
| |
Collapse
|
6
|
Sun X, Sun P, Sui Y, Tan C, Chen Y. Prognostic model based on six feature genes of intestinal flora subtypes predicts survival in colon cancer. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2126898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Xiao Sun
- Department of Gastrointestinal Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Peng Sun
- Department of Gastrointestinal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, People’s Republic of China
| | - Yi Sui
- Department of IVD Medical Marketing, 3D Medicine Inc., Shanghai, People’s Republic of China
| | - Canliang Tan
- Departmemt of General Surgery, the Third Affilliated Hospital of Southern Medical University, Guangzhou, People’s Republic of China
| | - Yinggang Chen
- Department of Gastrointestinal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, People’s Republic of China
| |
Collapse
|
7
|
SINGH G. Resveratrol Delivery <i>via</i> Gene Therapy: Entering the Modern Era. Turk J Pharm Sci 2022; 19:104-109. [DOI: 10.4274/tjps.galenos.2020.89577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
8
|
Shen J, Shen H, Ke L, Chen J, Dang X, Liu B, Hua Y. Knowledge Mapping of Immunotherapy for Hepatocellular Carcinoma: A Bibliometric Study. Front Immunol 2022; 13:815575. [PMID: 35173728 PMCID: PMC8841606 DOI: 10.3389/fimmu.2022.815575] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is one of the most common malignant tumors, and many patients are diagnosed with advanced disease. The treatment of advanced liver cancer has made significant strides in recent years, owing to the practice of immunotherapy drugs. Numerous studies have been published on immunotherapy for HCC; however, no relevant bibliometric study has been published. This study aims to gain a better understanding of the current situation and to identify potential new research directions by conducting a bibliometric analysis on immunotherapy for HCC. Methods We searched the Web of Science Core Collection (WoSCC) for articles related to immunotherapy for HCC. Three software (VOSviewer, CiteSpace, and python) were primarily used to assess the contribution and co-occurrence relationships of various countries/regions, institutes, journals, and, authors as well as to identify research hotspots and promising future trends in this research field. Results A total of 1,641 English articles published between 2011 and 2020 were collected, with the number of articles increasing nearly every year. The majority of publications originated from China (n = 893, 54.42%), followed by the United States and Japan. The Sun Yat-sen University contributed the most publications (n = 97, 5.91%). Nakatsura Tetsuya (n = 26) and Llovet JM (n = 366) were ranked first in the top ten authors and co-cited authors. Cancer Immunology Immunotherapy was the most productive academic journal on immunotherapy for HCC [n = 46, 2.80%; impact factor (IF) 2020 = 6.9679]. Aggregation and identification of critical nodes in the co-cited network demonstrated a shift in the field of HCC immunotherapy. Initially, the hotspots were predominantly “glypican-3”, “cytokine-induced killer cells”, and “ny-eso-1”, while the emphasis has shifted in recent years to “landscape”, “camrelizumab”, “combination therapy”, and “immune score”. Conclusion Increased attention has been paid to HCC with the advancement of immunotherapy. At the moment, the most active frontiers are focused on better understanding the immunological landscape of liver cancer, screening the population that can benefit from immunotherapy, and the clinical application of immune checkpoint inhibitors, particularly in combination with other therapeutic options (such as local therapy and targeted therapy).
Collapse
Affiliation(s)
- Jianming Shen
- Hepatobiliary and Pancreatic Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Shen
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lixin Ke
- Hepatobiliary and Pancreatic Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jialin Chen
- Hepatobiliary and Pancreatic Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi Dang
- Hepatobiliary and Pancreatic Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Baoxian Liu
- Department of Medical Ultrasonics, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Baoxian Liu, ; Yunpeng Hua,
| | - Yunpeng Hua
- Hepatobiliary and Pancreatic Center, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Baoxian Liu, ; Yunpeng Hua,
| |
Collapse
|
9
|
Motofei IG. Nobel Prize for immune checkpoint inhibitors, understanding the immunological switching between immunosuppression and autoimmunity. Expert Opin Drug Saf 2021; 21:599-612. [PMID: 34937484 DOI: 10.1080/14740338.2022.2020243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a revolutionary form of immunotherapy in cancer. However, the percentage of patients responding to therapy is relatively low, while adverse effects occur in a large number of patients. In addition, the therapeutic mechanisms of ICIs are not yet completely described. AREAS COVERED The initial view (articles published in PubMed, Scopus, Web of Science, etc.) was that ICIs increase tumor-specific immunity. Recent data (collected from the same databases) suggest that the ICIs pharmacotherapy actually extends beyond the topic of immune reactivity, including additional immune pathways, such as disrupting immunosuppression and increasing tumor-specific autoimmunity. Unfortunately, there is no clear delimitation between these specific autoimmune reactions that are therapeutically beneficial, and nonspecific autoimmune reactions/toxicity that can be extremely severe side effects. EXPERT OPINION Immune checkpoint mechanisms perform a non-selective immune regulation, maintaining a dynamic balance between immunosuppression and autoimmunity. By blocking these mechanisms, ICIs actually perform an immunological reset, decreasing immunosuppression and increasing tumor-specific immunity and predisposition to autoimmunity. The predisposition to autoimmunity induces both side effects and beneficial autoimmunity. Consequently, further studies are necessary to maximize the beneficial tumor-specific autoimmunity, while reducing the counterproductive effect of associated autoimmune toxicity.
Collapse
Affiliation(s)
- Ion G Motofei
- Department of Surgery/ Oncology, Carol Davila University, Bucharest, Romania.,Department of Surgery/ Oncology, St. Pantelimon Hospital, Bucharest, Romania
| |
Collapse
|
10
|
Chen H, Sun Q, Zhang C, She J, Cao S, Cao M, Zhang N, Adiila AV, Zhong J, Yao C, Wang Y, Xia H, Lan L. Identification and Validation of CYBB, CD86, and C3AR1 as the Key Genes Related to Macrophage Infiltration of Gastric Cancer. Front Mol Biosci 2021; 8:756085. [PMID: 34950700 PMCID: PMC8688826 DOI: 10.3389/fmolb.2021.756085] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is rampant around the world. Most of the GC cases are detected in advanced stages with poor prognosis. The identification of marker genes for early diagnosis is of great significance. Studying the tumor environment is helpful to acknowledge the process of tumorigenesis, development, and metastasis. Twenty-two kinds of immune cells were calculated by CIBERSORT from Gene Expression Omnibus (GEO) database. Subsequently, higher infiltration of macrophages M0 was discovered in GC compared with normal tissues. WGCNA was utilized to construct the network and then identify key modules and genes related to macrophages in TCGA. Finally, 18 hub genes were verified. In the PPI bar chart, the top 3 genes were chosen as hub genes involved in most pathways. On the TIMER and THPA websites, it is verified that the expression levels of CYBB, CD86, and C3AR1 genes in tumor tissues were higher than those in normal tissues. These genes may work as biomarkers or targets for accurate diagnosis and treatment of GC in the future. Our findings may be a new strategy for the treatment of GC.
Collapse
Affiliation(s)
- Haiyan Chen
- Institute for Cancer Research, School of Basic Medical Science of Xi'an Jiaotong University, Xi'an, China
| | - Qi Sun
- Department of Pathology, School of Basic Medical Sciences and Sir Run Run Hospital and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Junjun She
- Department of High Talent and General Surgery and Center for Gut Microbiome Research and Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Shuai Cao
- Department of Orthopedics, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Cao
- Institute for Cancer Research, School of Basic Medical Science of Xi'an Jiaotong University, Xi'an, China
| | - Nana Zhang
- Institute for Cancer Research, School of Basic Medical Science of Xi'an Jiaotong University, Xi'an, China
| | - Ayarick Vivian Adiila
- Institute for Cancer Research, School of Basic Medical Science of Xi'an Jiaotong University, Xi'an, China
| | - Jinjin Zhong
- Institute for Cancer Research, School of Basic Medical Science of Xi'an Jiaotong University, Xi'an, China
| | - Chengyun Yao
- Jiangsu Cancer Hospital and the Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Yili Wang
- Institute for Cancer Research, School of Basic Medical Science of Xi'an Jiaotong University, Xi'an, China
| | - Hongping Xia
- Department of Pathology, School of Basic Medical Sciences and Sir Run Run Hospital and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China.,Department of High Talent and General Surgery and Center for Gut Microbiome Research and Med-X Institute, the First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Liu W, Li C, Wu Y, Xu W, Chen S, Zhang H, Huang H, Zhao S, Wang J. Integrating m6A Regulators-Mediated Methylation Modification Models and Tumor Immune Microenvironment Characterization in Caucasian and Chinese Low-Grade Gliomas. Front Cell Dev Biol 2021; 9:725764. [PMID: 34900988 PMCID: PMC8661096 DOI: 10.3389/fcell.2021.725764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 10/11/2021] [Indexed: 01/21/2023] Open
Abstract
Background: As an important epigenetic modification, m6A methylation plays an essential role in post-transcriptional regulation and tumor development. It is urgently needed to comprehensively and rigorously explore the prognostic value of m6A regulators and its association with tumor microenvironment (TME) infiltration characterization of low-grade glioma (LGG). Methods: Based on the expression of 20 m6A regulatory factors, we comprehensively evaluated the m6A modification patterns of LGG after unsupervised clustering. Subsequent analysis of the differences between these groups was performed to obtain m6A-related genes, then consistent clustering was conducted to generate m6AgeneclusterA and m6AgeneclusterB. A Random Forest and machining learning algorithms were used to reduce dimensionality, identify TME characteristics and predict responses for LGG patients receiving immunotherapies. Results: Evident differential m6A regulators were found in mutation, CNV and TME characteristics of LGG. Based on TCGA and CGGA databases, we identified that m6A regulators clusterA could significantly predict better prognosis (p = 0.00016) which enriched in mTOR signaling pathway, basal transcription factors, accompanied by elevated immune cells infiltration, and decreased IDH and TP53 mutations. We also investigated the distribution of differential genes in m6A regulators clusters which was closely associated with tumor immune microenvironment through three independent cohort comparisons. Next, we established m6Ascore based on previous m6A model, which accurately predicts outcomes in 1089 LGG patients (p < 0.0001) from discovering cohort and 497 LGG patients from testing cohort. Significant TME characteristics, including genome heterogeneity, abidance of immune cells, and clinicopathologic parameters have been found between m6Ascore groups. Importantly, LGG patients with high m6Ascore are confronted with significantly decreased responses to chemotherapies, but benefit more from immunotherapies. Conclusion: In conclusion, this study first demonstrates that m6A modification is crucial participant in tumorigenesis and TME infiltration characterization of LGG based on large-scale cohorts. The m6Ascore provides useful and accurately predict of prognosis and clinical responses to chemotherapy, immunotherapy and therapeutic strategy development for LGG patients.
Collapse
Affiliation(s)
- Wangrui Liu
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, China
| | - Chuanyu Li
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, China
| | - Yuhao Wu
- School of Medicine, Tongji University, Shanghai, China
| | - Wenhao Xu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuxian Chen
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hailiang Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haineng Huang
- Department of Neurosurgery, Affiliated Hospital of Youjiang Medical University for Nationalities, Guangxi, China
| | - Shuai Zhao
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Dual Targeting of Cancer Cells and MMPs with Self-Assembly Hybrid Nanoparticles for Combination Therapy in Combating Cancer. Pharmaceutics 2021; 13:pharmaceutics13121990. [PMID: 34959271 PMCID: PMC8707712 DOI: 10.3390/pharmaceutics13121990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/05/2021] [Accepted: 11/19/2021] [Indexed: 01/21/2023] Open
Abstract
The co-delivery of chemotherapeutic agents and immune modulators to their targets remains to be a great challenge for nanocarriers. Here, we developed a hybrid thermosensitive nanoparticle (TMNP) which could co-deliver paclitaxel-loaded transferrin (PTX@TF) and marimastat-loaded thermosensitive liposomes (MMST/LTSLs) for the dual targeting of cancer cells and the microenvironment. TMNPs could rapidly release the two payloads triggered by the hyperthermia treatment at the site of tumor. The released PTX@TF entered cancer cells via transferrin-receptor-mediated endocytosis and inhibited the survival of tumor cells. MMST was intelligently employed as an immunomodulator to improve immunotherapy by inhibiting matrix metalloproteinases to reduce chemokine degradation and recruit T cells. The TMNPs promoted the tumor infiltration of CD3+ T cells by 2-fold, including memory/effector CD8+ T cells (4.2-fold) and CD4+ (1.7-fold), but not regulatory T cells. Our in vivo anti-tumor experiment suggested that TMNPs possessed the highest tumor growth inhibitory rate (80.86%) compared with the control group. We demonstrated that the nanoplatform could effectively inhibit the growth of tumors and enhance T cell recruitment through the co-delivery of paclitaxel and marimastat, which could be a promising strategy for the combination of chemotherapy and immunotherapy for cancer treatment.
Collapse
|
13
|
Della Sala F, Fabozzi A, di Gennaro M, Nuzzo S, Makvandi P, Solimando N, Pagliuca M, Borzacchiello A. Advances in Hyaluronic-Acid-Based (Nano)Devices for Cancer Therapy. Macromol Biosci 2021; 22:e2100304. [PMID: 34657388 DOI: 10.1002/mabi.202100304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Cancer is the main cause of fatality all over the world with a considerable growth rate. Many biologically active nanoplatforms are exploited for tumor treatment. Of nanodevices, hyaluronic acid (HA)-based systems have shown to be promising candidates for cancer therapy due to their high biocompatibility and cell internalization. Herein, surface functionalization of different nanoparticles (NPs), e.g., organic- and inorganic-based NPs, is highlighted. Subsequently, HA-based nanostructures and their applications in cancer therapy are presented.
Collapse
Affiliation(s)
- Francesca Della Sala
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Antonio Fabozzi
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Mario di Gennaro
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Stefano Nuzzo
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Pooyan Makvandi
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| | - Nicola Solimando
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Maurizio Pagliuca
- Altergon Italia s.r.l, Zona Industriale ASI, Morra De Sanctis (AV), 83040, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council, IPCB-CNR, Viale J.F. Kennedy 54, Naples, 80125, Italy
| |
Collapse
|
14
|
Pham MM, Ngoi NYL, Peng G, Tan DSP, Yap TA. Development of poly(ADP-ribose) polymerase inhibitor and immunotherapy combinations: progress, pitfalls, and promises. Trends Cancer 2021; 7:958-970. [PMID: 34158277 PMCID: PMC8458234 DOI: 10.1016/j.trecan.2021.05.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 12/21/2022]
Abstract
The efficacy of poly(ADP-ribose) polymerase inhibitors (PARPi) is restricted by inevitable drug resistance, while their use in combination with chemotherapy and targeted agents is commonly associated with dose-limiting toxicities. Immune checkpoint blockade (ICB) has demonstrated durable responses in different solid tumors and is well-established across multiple cancers. Despite this, single agent activity is limited to a minority of patients and drug resistance remains an issue. Building on the monotherapy success of both drug classes, combining PARPi with ICB may be a safe and well-tolerated strategy with the potential to improve survival outcomes. In this review, we present the preclinical, translational, and clinical data supporting the combination of DNA damage response (DDR) and ICB as well as consider important questions to be addressed with future research.
Collapse
Affiliation(s)
- Melissa M Pham
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie Y L Ngoi
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Guang Peng
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S P Tan
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute, National University of Singapore, Singapore
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
15
|
Stege H, Haist M, Nikfarjam U, Schultheis M, Heinz J, Pemler S, Loquai C, Grabbe S. The Status of Adjuvant and Neoadjuvant Melanoma Therapy, New Developments and Upcoming Challenges. Target Oncol 2021; 16:537-552. [PMID: 34554353 PMCID: PMC8484171 DOI: 10.1007/s11523-021-00840-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 12/14/2022]
Abstract
The global incidence of malignant melanoma, the leading cause of skin cancer death, has steadily increased in recent years. Surgical excision is the treatment of choice for early-stage melanoma. However, 40-60% of patients with high-risk melanoma or with nodal involvement eventually experience loco-regional relapse or tumor progression. Adjuvant therapy aims to reduce the rate of recurrence in radically operated high-risk patients with melanoma and thus improves survival. Interferon-α has long been the only approved drug for adjuvant melanoma therapy, despite an unclear survival benefit. The landmark success of immune-checkpoint inhibitors and BRAF/MEK-directed targeted therapies in the treatment of patients with stage IV melanoma led to the initiation of clinical trials in the adjuvant setting. These trials demonstrated the efficacy of immune-checkpoint inhibitors and targeted therapies for the adjuvant treatment of high-risk patients with melanoma, as shown both by an increase in recurrence-free survival and the emergence of long-term survivors, finally resulting in the approval of the cytotoxic T-lymphocyte antigen 4 inhibitor ipilimumab, PD1 inhibitors (nivolumab, pembrolizumab), and BRAF/MEK inhibitors for adjuvant melanoma therapy. This review aims to delineate the advances in adjuvant melanoma therapy, issuing particularly recent results from clinical trials. Moreover, we also discuss pending issues and future challenges, which comprise the adequate selection of adjuvant regimens for patient subgroups and the identification of markers likely to predict the individual response to adjuvant treatments. Last, we outline the role of emerging neoadjuvant approaches, which may complement adjuvant strategies and are currently investigated in clinical trials.
Collapse
Affiliation(s)
- Henner Stege
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany.
| | - Maximilian Haist
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Ulrike Nikfarjam
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Michael Schultheis
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Jaqueline Heinz
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Saskia Pemler
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
16
|
Ye Z, Zheng M, Zeng Y, Wei S, Huang H, Wang Y, Liu Q, Lin Z, Chen S, Zheng Q, Chen L. A 13-Gene Metabolic Prognostic Signature Is Associated With Clinical and Immune Features in Stomach Adenocarcinoma. Front Oncol 2021; 11:612952. [PMID: 34235071 PMCID: PMC8256842 DOI: 10.3389/fonc.2021.612952] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 05/19/2021] [Indexed: 12/11/2022] Open
Abstract
Patients with advanced stomach adenocarcinoma (STAD) commonly show high mortality and poor prognosis. Increasing evidence has suggested that basic metabolic changes may promote the growth and aggressiveness of STAD; therefore, identification of metabolic prognostic signatures in STAD would be meaningful. An integrative analysis was performed with 407 samples from The Cancer Genome Atlas (TCGA) and 433 samples from Gene Expression Omnibus (GEO) to develop a metabolic prognostic signature associated with clinical and immune features in STAD using Cox regression analysis and least absolute shrinkage and selection operator (LASSO). The different proportions of immune cells and differentially expressed immune-related genes (DEIRGs) between high- and low-risk score groups based on the metabolic prognostic signature were evaluated to describe the association of cancer metabolism and immune response in STAD. A total of 883 metabolism-related genes in both TCGA and GEO databases were analyzed to obtain 184 differentially expressed metabolism-related genes (DEMRGs) between tumor and normal tissues. A 13-gene metabolic signature (GSTA2, POLD3, GLA, GGT5, DCK, CKMT2, ASAH1, OPLAH, ME1, ACYP1, NNMT, POLR1A, and RDH12) was constructed for prognostic prediction of STAD. Sixteen survival-related DEMRGs were significantly related to the overall survival of STAD and the immune landscape in the tumor microenvironment. Univariate and multiple Cox regression analyses and the nomogram proved that a metabolism-based prognostic risk score (MPRS) could be an independent risk factor. More importantly, the results were mutually verified using TCGA and GEO data. This study provided a metabolism-related gene signature for prognostic prediction of STAD and explored the association between metabolism and the immune microenvironment for future research, thereby furthering the understanding of the crosstalk between different molecular mechanisms in human STAD. Some prognosis-related metabolic pathways have been revealed, and the survival of STAD patients could be predicted by a risk model based on these pathways, which could serve as prognostic markers in clinical practice.
Collapse
Affiliation(s)
- Zaisheng Ye
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Miao Zheng
- Department of Clinical Laboratory, Fujian Provincial Maternity and Children Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yi Zeng
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Shenghong Wei
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - He Huang
- Department of Digestive Endoscopy, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Yi Wang
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Qinying Liu
- Department of Clinical Laboratory, Fujian Provincial Maternity and Children Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Zhitao Lin
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Shu Chen
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Qiuhong Zheng
- Department of Clinical Laboratory, Fujian Provincial Maternity and Children Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Luchuan Chen
- Department of Gastrointestinal Surgical Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
17
|
Thomas PL, Groves SM, Zhang YK, Li J, Gonzalez-Ericsson P, Sivagnanam S, Betts CB, Chen HC, Liu Q, Lowe C, Chen H, Boyd KL, Kopparapu PR, Yan Y, Coussens LM, Quaranta V, Tyson DR, Iams W, Lovly CM. Beyond Programmed Death-Ligand 1: B7-H6 Emerges as a Potential Immunotherapy Target in SCLC. J Thorac Oncol 2021; 16:1211-1223. [PMID: 33839362 DOI: 10.1016/j.jtho.2021.03.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The programmed death-ligand 1 (PD-L1) immune checkpoint inhibitors, atezolizumab and durvalumab, have received regulatory approval for the first-line treatment of patients with extensive-stage SCLC. Nevertheless, when used in combination with platinum-based chemotherapy, these PD-L1 inhibitors only improve overall survival by 2 to 3 months. This may be due to the observation that less than 20% of SCLC tumors express PD-L1 at greater than 1%. Evaluating the composition and abundance of checkpoint molecules in SCLC may identify molecules beyond PD-L1 that are amenable to therapeutic targeting. METHODS We analyzed RNA-sequencing data from SCLC cell lines (n = 108) and primary tumor specimens (n = 81) for expression of 39 functionally validated inhibitory checkpoint ligands. Furthermore, we generated tissue microarrays containing SCLC cell lines and patient with SCLC specimens to confirm expression of these molecules by immunohistochemistry. We annotated patient outcomes data, including treatment response and overall survival. RESULTS The checkpoint protein B7-H6 (NCR3LG1) exhibited increased protein expression relative to PD-L1 in cell lines and tumors (p < 0.05). Higher B7-H6 protein expression correlated with longer progression-free survival (p = 0.0368) and increased total immune infiltrates (CD45+) in patients. Furthermore, increased B7-H6 gene expression in SCLC tumors correlated with a decreased activated natural killer cell gene signature, suggesting a complex interplay between B7-H6 expression and immune signature in SCLC. CONCLUSIONS We investigated 39 inhibitory checkpoint molecules in SCLC and found that B7-H6 is highly expressed and associated with progression-free survival. In addition, 26 of 39 immune checkpoint proteins in SCLC tumors were more abundantly expressed than PD-L1, indicating an urgent need to investigate additional checkpoint targets for therapy in addition to PD-L1.
Collapse
Affiliation(s)
- Portia L Thomas
- Department of Microbiology, Immunology & Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee; School of Graduate Studies & Research, Meharry Medical College, Nashville, Tennessee
| | - Sarah M Groves
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Yun-Kai Zhang
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jia Li
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Paula Gonzalez-Ericsson
- Breast Cancer Research Program, Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee
| | - Shamilene Sivagnanam
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Courtney B Betts
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Hua-Chang Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Qi Liu
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cindy Lowe
- Department of Pathology, Immunology and Microbiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Kelli L Boyd
- Department of Pathology, Immunology and Microbiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Prasad R Kopparapu
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yingjun Yan
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lisa M Coussens
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon; Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Darren R Tyson
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee
| | - Wade Iams
- Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Christine M Lovly
- School of Graduate Studies & Research, Meharry Medical College, Nashville, Tennessee; Division of Hematology-Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
18
|
Li X, Zhang W. Expression of PD-L1 in EBV-associated malignancies. Int Immunopharmacol 2021; 95:107553. [PMID: 33765613 DOI: 10.1016/j.intimp.2021.107553] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/22/2021] [Accepted: 02/28/2021] [Indexed: 02/06/2023]
Abstract
Epstein-Barr virus infection is closely related to the occurrence and development of a variety of malignant tumors. Tumor immunotherapy has been combined with modern biological high-tech technology, and has become the fourth cancer treatment mode after surgery, chemotherapy and radiotherapy. In 2013, immunotherapy was named the first of ten scientific breakthroughs by science. It aims to control and destroy tumor cells by stimulating and enhancing autoimmune function. In recent years, immune checkpoint inhibitors (ICIs) targeting PD-L1 have become a research hotspot in the field of cancer. Recent studies have shown that EBV infection can upregulate PD-L1 through complex mechanisms. Further understanding of these mechanisms and prevention of hyperprogressive disease (HPD) can make PD-L1 immune checkpoint inhibitors an effective way of immunotherapy for EBV related malignant tumors.
Collapse
Affiliation(s)
- Xiaoxu Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China; Clinical Laboratory, The Second People's Hospital of Wuhu City, Wuhu 241001, Anhui, People's Republic of China
| | - Wenling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
19
|
Zhang H, Yang Z, Du G, Cao L, Tan B. CD155-Prognostic and Immunotherapeutic Implications Based on Multiple Analyses of Databases Across 33 Human Cancers. Technol Cancer Res Treat 2021; 20:1533033820980088. [PMID: 33576304 PMCID: PMC7887689 DOI: 10.1177/1533033820980088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Growing evidence has suggested that CD155 participates in the regulation of many biological processes ranging cell growth, invasion, and migration from regulation of immune responses in most malignances. However, the impact of prognostic value and CD115-related immune response on the survival in multiple cancers remains incompletely clear. In our study, we assessed the prognostic significance and immune-associated mechanism of CD155 based on data from multiple databases and methods, including UCSC Xena, Oncomine, PrognoScan. We identified that CD155 was commonly upregulated in most human cancers, and High expression of CD155 was closely correlated with unfavorable clinical outcomes in 10/33 of human cancers, while CD155 at low level was responsible for better survival in KICH and PAAD. More intriguingly, CD155 expression had a significant interaction with immune function in several tumors by analyzing Tumor mutational burden and microsatellite in stability, immune score and stromal score. The correlation between immune infiltration and CD155 expression also indicated that CD155 expression positively correlated with CD4+ T cells in Head and Neck squamous cell carcinoma, Lung adenocarcinoma and Colon adenocarcinoma, while had inversely interaction with CD8+ T in Kidney renal clear cell carcinoma and Head and Neck squamous cell carcinoma as well as Tregs in Skin Cutaneous Melanoma, Head and Neck squamous cell carcinoma and Bladder Urothelial Carcinoma. These findings indicate CD155 correlates with cancer immunotherapy function. In conclusions, our observations revealed CD155 might function as immune-associated system in the development of human cancers, and acted as a promising prognostic and therapeutic target against human cancers.
Collapse
Affiliation(s)
- Hongpan Zhang
- Department of Oncology, 117913Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Zhihao Yang
- BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, People's Republic of China
| | - Guobo Du
- Department of Oncology, 117913Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - Lu Cao
- Department of Oncology, 117913Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| | - BangXian Tan
- Department of Oncology, 117913Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, People's Republic of China
| |
Collapse
|
20
|
Chang YW, Zhu WJ, Gu W, Sun J, Li ZQ, Wei XE. Anti-osteosarcoma Biological Activity Evaluation and Complete Chloroplast Genome Sequencing of Populus yunnanensis. J Oleo Sci 2021; 70:1429-1435. [PMID: 34615829 DOI: 10.5650/jos.ess21171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recently, the Populus yunnanensis extract has drawn the attention of most researchers, because of their anti-cancer activity. In this present research, the anti-cancer activity of the Populus yunnanensis extract was measured with Cell Counting Kit-8 (CCK-8) detection kit on the cancer cells. Then, the inhibitory activity of the Populus yunnanensis extract on the migration and invasion ability of the cancer cells was also determined in this present research with trans-well assay. Subsequently, to reveal the evolutionary genome evolution evaluation of the Populus yunnanensis and other Populus species, the high-throughput Illumina pair-end sequencing was performed and the chloroplast (cp) genome of Populus yunnanensis was determined, and the phylogenetic analysis was finished as wells. The results of the CCK-8 assay indicated that the Populus yunnanensis extract showed inhibitory effect on the cancer cell viability. Besides, the migration and invasion ability of the cancer cell was also reduced by the Populus yunnanensis extract. The complete chloroplast genome sequence results revealed that the Populus yunnanensis has a 156,505 bp circular cp genome. The phylogenetic analysis further revealed that the Populus yunnanensis has closely relationship with Populus simonii.
Collapse
Affiliation(s)
- Yue-Wen Chang
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Wen-Jun Zhu
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Wei Gu
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Jun Sun
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Zhi-Qiang Li
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| | - Xiao-En Wei
- Department of Orthopedics, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine
| |
Collapse
|
21
|
Jiang M, Qi L, Li L, Li Y. The caspase-3/GSDME signal pathway as a switch between apoptosis and pyroptosis in cancer. Cell Death Discov 2020; 6:112. [PMID: 33133646 PMCID: PMC7595122 DOI: 10.1038/s41420-020-00349-0] [Citation(s) in RCA: 309] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/09/2020] [Accepted: 10/14/2020] [Indexed: 12/18/2022] Open
Abstract
Apoptosis has long been recognized as a mechanism that kills the cancer cells by cytotoxic drugs. In recent years, studies have proved that pyroptosis can also shrink tumors and inhibit cells proliferation. Both apoptosis and pyroptosis are caspase-dependent programmed cell death pathways. Cysteinyl aspartate specific proteinase-3 (Caspase-3) is a common key protein in the apoptosis and pyroptosis pathways, and when activated, the expression level of tumor suppressor gene Gasdermin E (GSDME) determines the mechanism of tumor cell death. When GSDME is highly expressed, the active caspase-3 cuts it and releases the N-terminal domain to punch holes in the cell membrane, resulting in cell swelling, rupture, and death. When the expression of GSDME is low, it will lead to the classical mechanism of tumor cell death, which is apoptosis. More interestingly, researchers have found that GSDME can also be located upstream of caspase-3, connecting extrinsic, and intrinsic apoptotic pathways. Then, promoting caspase-3 activation, and forming a self-amplifying feed-forward loop. GSDME-mediated pyroptosis is correlated with the side effects of chemotherapy and anti-tumor immunity. This article mainly reviews the caspase-3/GSDME signal pathway as a switch between apoptosis and pyroptosis in cancer, to provide new strategies and targets for cancer treatment.
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang 150081 P. R. China
| | - Ling Qi
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang 150081 P. R. China
| | - Lisha Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang 150081 P. R. China
| | - Yanjing Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, 150 Haping St, Nangang District, Harbin, Heilongjiang 150081 P. R. China
| |
Collapse
|
22
|
Combination of Detoxified Pneumolysin Derivative ΔA146Ply and Berbamine as a Treatment Approach for Breast Cancer. MOLECULAR THERAPY-ONCOLYTICS 2020; 18:247-261. [PMID: 32728613 PMCID: PMC7369532 DOI: 10.1016/j.omto.2020.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/19/2020] [Indexed: 12/13/2022]
Abstract
Increasing evidence demonstrates that microorganisms and their products can modulate host responses to cancer therapies and contribute to tumor shrinkage via various mechanisms, including intracellular signaling pathways modulation and immunomodulation. Detoxified pneumolysin derivative ΔA146Ply is a pneumolysin mutant lacking hemolytic activity. To determine the antitumor activity of ΔA146Ply, the combination of ΔA146Ply and berbamine, a well-established antitumor agent, was used for breast cancer therapy, especially for triple-negative breast cancer. The efficacy of the combination therapy was evaluated in vitro using four breast cancer cell lines and in vivo using a synergistic mouse tumor model. We demonstrated that in vitro, the combination therapy significantly inhibited cancer cell proliferation, promoted cancer cell apoptosis, caused cancer cell-cycle arrest, and suppressed cancer cell migration and invasion. In vivo, the combination therapy significantly suppressed tumor growth and prolonged the median survival time of tumor-bearing mice partially through inhibiting tumor cell proliferation, promoting tumor cell apoptosis, and activating systemic antitumor immune responses. The safety analysis demonstrated that the combination therapy showed no obvious liver and kidney toxicity to tumor-bearing mice. Our study provides a new treatment option for breast cancer and lays the experimental basis for the development of ΔA146Ply as an antitumor agent.
Collapse
|
23
|
Cong X, Tian H, Liu S, Mao K, Chen H, Xin Y, Liu F, Wang X, Meng X, Zhu G, Wang J, Gao X, Tan H, Yang YG, Sun T. Cationic Liposome/DNA Complexes Mediate Antitumor Immunotherapy by Promoting Immunogenic Tumor Cell Death and Dendritic Cell Activation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:28047-28056. [PMID: 32478501 DOI: 10.1021/acsami.0c08112] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Immunotherapy has been successfully used in the treatment of multiple malignancies, but clinical studies revealed low response rates. Thus, the development of new effective immunotherapeutic modalities is urgently needed. Successfully inducing tumor cell death with enhanced antigenicity is important for the expansion and differentiation of tumor-specific CD8+ cytotoxic T lymphocytes. Cationic liposome/DNA complexes (CLN/DNA), which usually have obvious cytotoxic effects, may improve the antitumor immunity through enhancing the immunogenicity of dying tumor cells. Herein, we report that a plasmid DNA-encapsulated cationic lipid nanoparticle formulated with cholesterol, DOTAP, and DSPE-mPEG2000 significantly increases the tumor cell death with high antigenicity in vitro. Furthermore, the cationic liposome/DNA complex (CLN/DNA) treatment promotes the activation of dendritic cells (DCs). We also find that the intratumorally injected CLN/DNA successfully promoted the activation of DCs in the tumor-draining lymph node. Importantly, both local tumor growth and distant tumor formation were significantly inhibited by T cell-dependent antitumor immune responses after intratumoral injection of CLN/DNA. This study presents a simple and effective strategy for improving the cancer immunotherapy.
Collapse
Affiliation(s)
- Xiuxiu Cong
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Huimin Tian
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Shuhan Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Kuirong Mao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
| | - Hongmei Chen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
| | - Yanbao Xin
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Feiqi Liu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
| | - Xin Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Xiandi Meng
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Ge Zhu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Jialiang Wang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Xue Gao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Huizhu Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
| | - Tianmeng Sun
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, The First Hospital, Jilin University, Changchun, Jilin 130061, China
- National-local Joint Engineering Laboratory of Animal Models for Human Diseases, Changchun, Jilin 130062, China
- International Center of Future Science at Jilin University, Changchun, Jilin 130015, China
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
24
|
O'Connell L, Winter DC. Organoids: Past Learning and Future Directions. Stem Cells Dev 2020; 29:281-289. [DOI: 10.1089/scd.2019.0227] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Lauren O'Connell
- Department of Surgery, St. Vincent's University Hospital, Elm Park, Dublin, Ireland
| | - Des C. Winter
- Department of Surgery, St. Vincent's University Hospital, Elm Park, Dublin, Ireland
| |
Collapse
|
25
|
Granier C, Vinatier E, Colin E, Mandavit M, Dariane C, Verkarre V, Biard L, El Zein R, Lesaffre C, Galy-Fauroux I, Roussel H, De Guillebon E, Blanc C, Saldmann A, Badoual C, Gey A, Tartour É. Multiplexed Immunofluorescence Analysis and Quantification of Intratumoral PD-1+ Tim-3+ CD8+ T Cells. J Vis Exp 2018. [PMID: 29553498 DOI: 10.3791/56606] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Immune cells are important components of the tumor microenvironment and influence tumor growth and evolution at all stages of carcinogenesis. Notably, it is now well established that the immune infiltrate in human tumors can correlate with prognosis and response to therapy. The analysis of the immune infiltrate in the tumor microenvironment has become a major challenge for the classification of patients and the response to treatment. The co-expression of inhibitory receptors such as Program Cell Death Protein 1 (PD1; also known as CD279), Cytotoxic T Lymphocyte Associated Protein 4 (CTLA-4), T-Cell Immunoglobulin and Mucin Containing Protein-3 (Tim-3; also known as CD366), and Lymphocyte Activation Gene 3 (Lag-3; also known as CD223), is a hallmark of T cell exhaustion. We developed a multiparametric in situ immunofluorescence staining to identify and quantify at the cellular level the co-expression of these inhibitory receptors. On a retrospective series of frozen tissue of renal cell carcinomas (RCC), using a fluorescence multispectral imaging technology coupled with an image analysis software, it was found that co-expression of PD-1 and Tim-3 on tumor infiltrating CD8+ T cells is correlated with a poor prognosis in RCC. To our knowledge, this represents the first study demonstrating that this automated multiplex in situ technology may have some clinical relevance.
Collapse
Affiliation(s)
| | - Emeline Vinatier
- INSERM U970, Université Paris Descartes; Equipe Labellisée Ligue Contre le Cancer; Department of Immunology, Hopital Européen Georges Pompidou
| | - Elia Colin
- INSERM U970, Université Paris Descartes; Equipe Labellisée Ligue Contre le Cancer
| | - Marion Mandavit
- INSERM U970, Université Paris Descartes; Equipe Labellisée Ligue Contre le Cancer
| | - Charles Dariane
- INSERM U970, Université Paris Descartes; Equipe Labellisée Ligue Contre le Cancer; Department of Medical Urology, Hopital Européen Georges Pompidou
| | | | | | | | | | | | - Hélène Roussel
- INSERM U970, Université Paris Descartes; Equipe Labellisée Ligue Contre le Cancer; Department of Pathology, Hopital Européen Georges Pompidou
| | | | - Charlotte Blanc
- INSERM U970, Université Paris Descartes; Equipe Labellisée Ligue Contre le Cancer
| | | | - Cécile Badoual
- INSERM U970, Université Paris Descartes; Department of Pathology, Hopital Européen Georges Pompidou
| | - Alain Gey
- Department of Immunology, Hopital Européen Georges Pompidou
| | - Éric Tartour
- INSERM U970, Université Paris Descartes; Department of Immunology, Hopital Européen Georges Pompidou;
| |
Collapse
|
26
|
Benzaquen J, Marquette CH, Glaichenhaus N, Leroy S, Hofman P, Ilié M. [The biological rationale for immunotherapy in cancer]. Rev Mal Respir 2018; 35:206-222. [PMID: 29428191 DOI: 10.1016/j.rmr.2017.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Immunotherapy aims to promote the immune system's activity against malignant cells by stimulating the response to several tumor antigens. STATE OF THE ART Immunosurveillance may adjust the immunogenicity of tumors. To be effective, immunity must induce the specific activation of CD4+ and CD8+ T lymphocytes, as well as activation of innate immunity. Activator and inhibitory costimulatory molecules regulate T lymphocyte activation at immunity checkpoints such as PD-1/PD-L1 and CTLA-4. Adaptive immune resistance confers tumour resistance to immunosurveillance through these immune checkpoints. PERSPECTIVES Approaches involving the combination of several immunotherapies with each other or with chemotherapy and radiotherapy and antibodies against other molecules of costimulation are under development. The development of biomarkers, which can select a targeted population and predict therapeutic response, represents a major challenge. Tumour high-throughput sequencing could refine "immunoscore". Intratumoral T cell receptor seems to represent a promising biomarker. CONCLUSIONS Numerous challenges still remain in developing research approaches for the development of immunotherapies.
Collapse
Affiliation(s)
- J Benzaquen
- Department of Pulmonary Medicine and Oncology, CHU de Nice, FHU OncoAge, 06100 Nice, France
| | - C-H Marquette
- Department of Pulmonary Medicine and Oncology, CHU de Nice, FHU OncoAge, 06100 Nice, France.
| | - N Glaichenhaus
- Université Côte-d'Azur, CNRS, Inserm, institut de pharmacologie moleculaire et cellulaire, FHU-OncoAge, 06560 Valbonne, France
| | - S Leroy
- Department of Pulmonary Medicine and Oncology, CHU de Nice, FHU OncoAge, 06100 Nice, France
| | - P Hofman
- Laboratory of Clinical and Experimental Pathology and Hospital-related Biobank (BB-0033-00025), IRCAN, FHU OncoAge, 06100 Nice, France
| | - M Ilié
- Laboratory of Clinical and Experimental Pathology and Hospital-related Biobank (BB-0033-00025), IRCAN, FHU OncoAge, 06100 Nice, France
| |
Collapse
|
27
|
Szor DJ, Dias AR, Pereira MA, Ramos MFKP, Zilberstein B, Cecconello I, Ribeiro U. Prognostic Role of Neutrophil/Lymphocyte Ratio in Resected Gastric Cancer: A Systematic Review and Meta-analysis. Clinics (Sao Paulo) 2018; 73:e360. [PMID: 29924187 PMCID: PMC5996440 DOI: 10.6061/clinics/2018/e360] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 03/05/2018] [Indexed: 02/05/2023] Open
Abstract
High levels of inflammatory markers and the neutrophil-lymphocyte ratio appear to be associated with worse overall survival in solid tumors. However, few studies have analyzed the role of the neutrophil-lymphocyte ratio in gastric cancer patients scheduled to undergo curative resection. In the present study, a systematic review and meta-analysis was performed to analyze the relationship between the neutrophil-lymphocyte ratio and overall survival in patients with gastric cancer submitted to curative resection and to identify the clinicopathological features (age, gender, tumor depth, nodal involvement and tumor differentiation) that are correlated with high neutrophil-lymphocyte ratios. A literature search of PubMed, Scopus, Cochrane and EMBASE through November 2017 was conducted. Articles that included gastric cancer patients submitted to curative resection and preoperatory neutrophil-lymphocyte ratio values were included. A total of 7 studies comprising 3264 patients from 5 different countries were included. The meta-analysis revealed an association of high neutrophil-lymphocyte ratios with older age, male gender, lower 5-year overall survival, increased depth of tumor invasion, positive nodal involvement but not with histological differentiation. Evaluation of the neutrophil-lymphocyte ratio is a cost-effective method that is widely available in preoperatory settings. Furthermore, it can effectively predict prognosis, as high values of this biomarker are related to more aggressive tumor characteristics. This ratio can also be used to stratify risk in patients within the same disease stage and may be used to assist in individualized follow-up and treatment.
Collapse
Affiliation(s)
- Daniel Jose Szor
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- *Corresponding author. E-mail:
| | - Andre Roncon Dias
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marina Alessandra Pereira
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Marcus Fernando Kodama Pertille Ramos
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Bruno Zilberstein
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ivan Cecconello
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Ulysses Ribeiro
- Instituto do Cancer do Estado de Sao Paulo (ICESP), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|
28
|
Outh-Gauer S, Le Tourneau C, Broudin C, Scotte F, Roussel H, Hans S, Mandavit M, Tartour E, Badoual C. Actualités sur l’immunothérapie en pathologie des voies aérodigestives supérieures. Ann Pathol 2017; 37:79-89. [DOI: 10.1016/j.annpat.2016.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/07/2016] [Indexed: 02/06/2023]
|
29
|
Zhang X, Zhu S, Li T, Liu YJ, Chen W, Chen J. Targeting immune checkpoints in malignant glioma. Oncotarget 2017; 8:7157-7174. [PMID: 27756892 PMCID: PMC5351697 DOI: 10.18632/oncotarget.12702] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 10/12/2016] [Indexed: 12/31/2022] Open
Abstract
Malignant glioma is the most common and a highly aggressive cancer in the central nervous system (CNS). Cancer immunotherapy, strategies to boost the body's anti-cancer immune responses instead of directly targeting tumor cells, recently achieved great success in treating several human solid tumors. Although once considered "immune privileged" and devoid of normal immunological functions, CNS is now considered a promising target for cancer immunotherapy, featuring the recent progresses in neurobiology and neuroimmunology and a highly immunosuppressive state in malignant glioma. In this review, we focus on immune checkpoint inhibitors, specifically, antagonizing monoclonal antibodies for programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and indoleamine 2,3-dioxygenase (IDO). We discuss advances in the working mechanisms of these immune checkpoint molecules, their status in malignant glioma, and current preclinical and clinical trials targeting these molecules in malignant glioma.
Collapse
Affiliation(s)
- Xuhao Zhang
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Tete Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
- Sanofi Research and Development, Cambridge, MA, USA
| | - Wei Chen
- ADC Biomedical Research Institute, Saint Paul, MN, USA
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|