1
|
Zhu X, Ding CKC, Aggarwal RR. Emerging Therapeutic Targets of Neuroendocrine Prostate Cancer. Curr Oncol Rep 2025; 27:362-374. [PMID: 40011325 DOI: 10.1007/s11912-025-01643-9] [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] [Accepted: 01/27/2025] [Indexed: 02/28/2025]
Abstract
PURPOSE OF REVIEW Treatment-emergent neuroendocrine prostate cancer (NEPC) is aggressive and lethal. As androgen receptor signaling inhibitors (ARSIs) are increasingly used in earlier disease settings, treatment-emergent NEPC becomes more prevalent, and effective therapies are urgently needed. The purpose of this review was to summarize recent progress on emerging therapeutic targets of NEPC. RECENT FINDINGS A multitude of therapeutic targets have emerged in NEPC over recent years. These targets may represent drivers of treatment-emergent lineage plasticity or simply be overexpressed on the surface of NEPC cells. Multiple modalities have been employed to drug these targets, with promising preclinical and clinical results. Treatment-emergent NEPC represents a distinct and clinically significant subset of castration-resistant prostate cancer (CRPC). Emerging therapeutic approaches have demonstrated encouraging efficacy and safety profiles, offering the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Xiaolin Zhu
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Chien-Kuang C Ding
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomic Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Rahul R Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.
- Division of Hematology and Oncology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Xu Y, Yan Y, Liu D, Tang J, Zhang H, Liu X, Wu Y, Cui X. Risk of transplant rejection associated with ICIs prior to liver transplantation in HCC: A multicenter retrospective study. Int Immunopharmacol 2024; 143:113400. [PMID: 39467348 DOI: 10.1016/j.intimp.2024.113400] [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: 05/17/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Studies on the safety of immune checkpoint inhibitors (ICIs) prior to liver transplantation (LT) are still limited to case reports or case series. It is vital to be aware of possible risk of transplant rejection with regards to ICIs after LT. OBJECTIVE To explore the possible risk of transplant rejection induced by ICIs in hepatocellular carcinoma (HCC) patients and investigate the safe washout interval between ICIs administration and LT. METHODS HCC patients from 3 tertiary hospitals in China who received ICIs prior to LT over the past 5 years were analyzed retrospectively. Patients who had experienced transplant rejection were reported in detail. Additionally, a comprehensive search of databases was conducted to identify case reports of HCC patients who underwent LT after receiving ICIs until October 1, 2024. RESULTS In our study, a total of 25 patients were analyzed. Programmed cell death protein 1 (PD-1) inhibitors were most commonly used (68 %, 17/25). The median interval between the last dose of ICIs and LT was 64 (40-150.75) days. Three patients (12 %) experienced T-cell mediated rejection (TCMR), 1 of which was induced by ICIs, and the other 2 of which could not be excluded from the influence of immunosuppressant concentrations. In literature review, a total of 96 cases of HCC patients who had received ICIs prior to LT were included. PD-1 inhibitor monotherapy resulted in significantly higher rejection than PD-L1 inhibitor monotherapy and other ICIs combination regimens (P = 0.021). In patients with pembrolizumab, the interval from ICIs to LT was shorter in the rejection group than in the non-rejection group (P = 0.045). Twenty-one cases (21.88 %) experienced transplant rejection, and 3 patients passed away following transplant rejection. CONCLUSION ICIs prior to LT was associated with the risk of transplant rejection, especially with factors such as the type of ICIs and the interval between ICIs and LT. Multicenter prospective studies are needed to further explore the safety of ICIs.
Collapse
Affiliation(s)
- Ye Xu
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Yan Yan
- Department of Clinical Pharmacy, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Donghua Liu
- Department of Clinical Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Jing Tang
- Department of Clinical Pharmacy, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| | - Haiming Zhang
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Xiangduan Liu
- Department of Pharmacy, Zhengzhou People's Hospital, Zhengzhou, China.
| | - Yi Wu
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| | - Xiangli Cui
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
4
|
Mekala JR, Nalluri HP, Reddy PN, S B S, N S SK, G V S D SK, Dhiman R, Chamarthy S, Komaragiri RR, Manyam RR, Dirisala VR. Emerging trends and therapeutic applications of monoclonal antibodies. Gene 2024; 925:148607. [PMID: 38797505 DOI: 10.1016/j.gene.2024.148607] [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: 08/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Monoclonal antibodies (mAbs) are being used to prevent, detect, and treat a broad spectrum of malignancies and infectious and autoimmune diseases. Over the past few years, the market for mAbs has grown exponentially. They have become a significant part of many pharmaceutical product lines, and more than 250 therapeutic mAbs are undergoing clinical trials. Ever since the advent of hybridoma technology, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some of the benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies, which are affordable versions of therapeutic antibodies. Along with biosimilars, innovations in antibody engineering have helped to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. In the future, mAbs generated by applying next-generation sequencing (NGS) are expected to become a powerful tool in clinical therapeutics. This article describes the methods of mAb production, pre-clinical and clinical development of mAbs, approved indications targeted by mAbs, and novel developments in the field of mAb research.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA.
| | - Hari P Nalluri
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Prakash Narayana Reddy
- Department of Microbiology, Dr. V.S. Krishna Government College, Visakhapatnam 530013, India
| | - Sainath S B
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524320, AP, India
| | - Sampath Kumar N S
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Sai Kiran G V S D
- Santhiram Medical College and General Hospital, Nandyal, Kurnool 518501, AP, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Sciences, National Institute of Technology Rourkela-769008, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA
| | - Raghava Rao Komaragiri
- Department of CSE, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522302, Andhra Pradesh, INDIA
| | - Rajasekhar Reddy Manyam
- Amrita School of Computing, Amrita Vishwa Vidyapeetham, Amaravati Campus, Amaravati, Andhra Pradesh, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India.
| |
Collapse
|
5
|
Perovic D, Dusanovic Pjevic M, Perovic V, Grk M, Rasic M, Milickovic M, Mijovic T, Rasic P. B7 homolog 3 in pancreatic cancer. World J Gastroenterol 2024; 30:3654-3667. [PMID: 39193002 PMCID: PMC11346158 DOI: 10.3748/wjg.v30.i31.3654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024] Open
Abstract
Despite advances in cancer treatment, pancreatic cancer (PC) remains a disease with high mortality rates and poor survival outcomes. The B7 homolog 3 (B7-H3) checkpoint molecule is overexpressed among many malignant tumors, including PC, with low or absent expression in healthy tissues. By modulating various immunological and nonimmunological molecular mechanisms, B7-H3 may influence the progression of PC. However, the impact of B7-H3 on the survival of patients with PC remains a subject of debate. Still, most available scientific data recognize this molecule as a suppressive factor to antitumor immunity in PC. Furthermore, it has been demonstrated that B7-H3 stimulates the migration, invasion, and metastasis of PC cells, and enhances resistance to chemotherapy. In preclinical models of PC, B7-H3-targeting monoclonal antibodies have exerted profound antitumor effects by increasing natural killer cell-mediated antibody-dependent cellular cytotoxicity and delivering radioisotopes and cytotoxic drugs to the tumor site. Finally, PC treatment with B7-H3-targeting antibody-drug conjugates and chimeric antigen receptor T cells is being tested in clinical studies. This review provides a comprehensive analysis of all PC-related studies in the context of B7-H3 and points to deficiencies in the current data that should be overcome by future research.
Collapse
Affiliation(s)
- Dijana Perovic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Marija Dusanovic Pjevic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Vladimir Perovic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Milka Grk
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Milica Rasic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Maja Milickovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic”, Belgrade 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade 11000, Serbia
| | - Tanja Mijovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic”, Belgrade 11000, Serbia
| | - Petar Rasic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia “Dr. Vukan Cupic”, Belgrade 11000, Serbia
| |
Collapse
|
6
|
Zhang R, Jiang Q, Zhuang Z, Zeng H, Li Y. A bibliometric analysis of drug resistance in immunotherapy for breast cancer: trends, themes, and research focus. Front Immunol 2024; 15:1452303. [PMID: 39188717 PMCID: PMC11345160 DOI: 10.3389/fimmu.2024.1452303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
While breast cancer treatments have advanced significantly nowadays, yet metastatic, especially triple-negative breast cancer (TNBC), remains challenging with low survival. Cancer immunotherapy, a promising approach for HER2-positive and TNBC, still faces resistance hurdles. Recently, numerous studies have set their sights on the resistance of immunotherapy for breast cancer. Our study provides a thorough comprehension of the current research landscape, hotspots, and emerging breakthroughs in this critical area through a meticulous bibliometric analysis. As of March 26, 2024, a total of 1341 articles on immunology resistance in breast cancer have been gathered from Web of Science Core Collection, including 765 articles and 576 reviews. Bibliometrix, CiteSpace and VOSviewer software were utilized to examine publications and citations per year, prolific countries, contributive institutions, high-level journals and scholars, as well as highly cited articles, references and keywords. The research of immunotherapy resistance in breast cancer has witnessed a remarkable surge over the past seven years. The United States and China have made significant contributions, with Harvard Medical School being the most prolific institution and actively engaging in collaborations. The most contributive author is Curigliano, G from the European Institute of Oncology in Italy, while Wucherpfennig, K. W. from the Dana-Farber Cancer Institute in the USA, had the highest citations. Journals highly productive primarily focus on clinical, immunology and oncology research. Common keywords include "resistance", "expression", "tumor microenvironment", "cancer", "T cell", "therapy", "chemotherapy" and "cell". Current research endeavors to unravel the mechanisms of immune resistance in breast cancer through the integration of bioinformatics, basic experiments, and clinical trials. Efforts are underway to develop strategies that improve the effectiveness of immunotherapy, including the exploration of combination therapies and advancements in drug delivery systems. Additionally, there is a strong focus on identifying novel biomarkers that can predict patient response to immunology. This study will provide researchers with an up-to-date overview of the present knowledge in drug resistance of immunology for breast cancer, serving as a valuable resource for informed decision-making and further research on innovative approaches to address immunotherapy resistance.
Collapse
Affiliation(s)
- Rendong Zhang
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Qiongzhi Jiang
- Department of Radiation Oncology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Zhemin Zhuang
- Engineering College, Shantou University, Shantou, Guangdong, China
| | - Huancheng Zeng
- Department of Breast Surgery, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Yaochen Li
- The Central Laboratory, Cancer Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
7
|
Duan Z, Shi R, Gao B, Cai J. N-linked glycosylation of PD-L1/PD-1: an emerging target for cancer diagnosis and treatment. J Transl Med 2024; 22:705. [PMID: 39080767 PMCID: PMC11290144 DOI: 10.1186/s12967-024-05502-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
During tumorigenesis and progression, the immune checkpoint programmed death-1 (PD-1) and its ligand programmed death ligand-1 (PD-L1) play critical roles in suppressing T cell-mediated anticancer immune responses, leading to T-cell exhaustion and subsequent tumor evasion. Therefore, anti-PD-L1/PD-1 therapy has been an attractive strategy for treating cancer over the past decade. However, the overall efficacy of this approach remains suboptimal, revealing an urgent need for novel insights. Interestingly, increasing evidence indicates that both PD-L1 on tumor cells and PD-1 on tumor-specific T cells undergo extensive N-linked glycosylation, which is essential for the stability and interaction of these proteins, and this modification promotes tumor evasion. In various preclinical models, targeting the N-linked glycosylation of PD-L1/PD-1 was shown to significantly increase the efficacy of PD-L1/PD-1 blockade therapy. Furthermore, deglycosylation of PD-L1 strengthens the signal intensity in PD-L1 immunohistochemistry (IHC) assays, improving the diagnostic and therapeutic relevance of this protein. In this review, we provide an overview of the regulatory mechanisms underlying the N-linked glycosylation of PD-L1/PD-1 as well as the crucial role of N-linked glycosylation in PD-L1/PD-1-mediated immune evasion. In addition, we highlight the promising implications of targeting the N-linked glycosylation of PD-L1/PD-1 in the clinical diagnosis and treatment of cancer. Our review identifies knowledge gaps and sheds new light on the cancer research field.
Collapse
Affiliation(s)
- Zhiyun Duan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, P.R. China
| | - Runhan Shi
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, P.R. China
- Department of Ophthalmology and Vision Science, Shanghai Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, 200031, P.R. China
| | - Bo Gao
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, P.R. China
| | - Jiabin Cai
- Department of Liver Surgery and Transplantation, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Liver Cancer Institute, Fudan University, Shanghai, 200032, P.R. China.
- Department of Liver Surgery, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, 361015, P.R. China.
| |
Collapse
|
8
|
Schlegel LS, Werbrouck C, Boettcher M, Schlegel P. Universal CAR 2.0 to overcome current limitations in CAR therapy. Front Immunol 2024; 15:1383894. [PMID: 38962014 PMCID: PMC11219820 DOI: 10.3389/fimmu.2024.1383894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has effectively complemented the treatment of advanced relapsed and refractory hematological cancers. The remarkable achievements of CD19- and BCMA-CAR T therapies have raised high expectations within the fields of hematology and oncology. These groundbreaking successes are propelling a collective aspiration to extend the reach of CAR therapies beyond B-lineage malignancies. Advanced CAR technologies have created a momentum to surmount the limitations of conventional CAR concepts. Most importantly, innovations that enable combinatorial targeting to address target antigen heterogeneity, using versatile adapter CAR concepts in conjunction with recent transformative next-generation CAR design, offer the promise to overcome both the bottleneck associated with CAR manufacturing and patient-individualized treatment regimens. In this comprehensive review, we delineate the fundamental prerequisites, navigate through pivotal challenges, and elucidate strategic approaches, all aimed at paving the way for the future establishment of multitargeted immunotherapies using universal CAR technologies.
Collapse
Affiliation(s)
- Lara Sophie Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Coralie Werbrouck
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Pediatric Hematology and Oncology, Westmead Children’s Hospital, Sydney, NSW, Australia
| |
Collapse
|
9
|
Mitra A, Kumar A, Amdare NP, Pathak R. Current Landscape of Cancer Immunotherapy: Harnessing the Immune Arsenal to Overcome Immune Evasion. BIOLOGY 2024; 13:307. [PMID: 38785789 PMCID: PMC11118874 DOI: 10.3390/biology13050307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Cancer immune evasion represents a leading hallmark of cancer, posing a significant obstacle to the development of successful anticancer therapies. However, the landscape of cancer treatment has significantly evolved, transitioning into the era of immunotherapy from conventional methods such as surgical resection, radiotherapy, chemotherapy, and targeted drug therapy. Immunotherapy has emerged as a pivotal component in cancer treatment, harnessing the body's immune system to combat cancer and offering improved prognostic outcomes for numerous patients. The remarkable success of immunotherapy has spurred significant efforts to enhance the clinical efficacy of existing agents and strategies. Several immunotherapeutic approaches have received approval for targeted cancer treatments, while others are currently in preclinical and clinical trials. This review explores recent progress in unraveling the mechanisms of cancer immune evasion and evaluates the clinical effectiveness of diverse immunotherapy strategies, including cancer vaccines, adoptive cell therapy, and antibody-based treatments. It encompasses both established treatments and those currently under investigation, providing a comprehensive overview of efforts to combat cancer through immunological approaches. Additionally, the article emphasizes the current developments, limitations, and challenges in cancer immunotherapy. Furthermore, by integrating analyses of cancer immunotherapy resistance mechanisms and exploring combination strategies and personalized approaches, it offers valuable insights crucial for the development of novel anticancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ankita Mitra
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, Uttar Pradesh, India
| | - Nitin P. Amdare
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
10
|
Jing ZL, Liu GL, Zhou N, Xu DY, Feng N, Lei Y, Ma LL, Tang MS, Tong GH, Tang N, Deng YJ. Interferon-γ in the tumor microenvironment promotes the expression of B7H4 in colorectal cancer cells, thereby inhibiting cytotoxic T cells. Sci Rep 2024; 14:6053. [PMID: 38480774 PMCID: PMC10937991 DOI: 10.1038/s41598-024-56681-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/09/2024] [Indexed: 03/17/2024] Open
Abstract
The bioactivity of interferon-γ (IFN-γ) in cancer cells in the tumor microenvironment (TME) is not well understood in the current immunotherapy era. We found that IFN-γ has an immunosuppressive effect on colorectal cancer (CRC) cells. The tumor volume in immunocompetent mice was significantly increased after subcutaneous implantation of murine CRC cells followed by IFN-γ stimulation, and RNA sequencing showed high expression of B7 homologous protein 4 (B7H4) in these tumors. B7H4 promotes CRC cell growth by inhibiting the release of granzyme B (GzmB) from CD8+ T cells and accelerating apoptosis in CD8+ T cells. Furthermore, interferon regulatory factor 1 (IRF1), which binds to the B7H4 promoter, is positively associated with IFN-γ stimulation-induced expression of B7H4. The clinical outcome of patients with CRC was negatively related to the high expression of B7H4 in cancer cells or low expression of CD8 in the microenvironment. Therefore, B7H4 is a biomarker of poor prognosis in CRC patients, and interference with the IFN-γ/IRF1/B7H4 axis might be a novel immunotherapeutic method to restore the cytotoxic killing of CRC cells.
Collapse
Affiliation(s)
- Zhi-Liang Jing
- Department of Pathology, School of Basic Medical Sciences and Nan Fang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China
- Department of Pathology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Guang-Long Liu
- Department of Pathology, School of Basic Medical Sciences and Nan Fang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China
| | - Na Zhou
- Department of Pathology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, 510120, China
| | - Dong-Yan Xu
- Department of Pathology, School of Basic Medical Sciences and Nan Fang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China
| | - Na Feng
- Department of Pathology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, 523413, China
| | - Yan Lei
- Department of Pathology, School of Basic Medical Sciences and Nan Fang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China
| | - Li-Li Ma
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Min-Shan Tang
- Department of Pathology, School of Basic Medical Sciences and Nan Fang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China
| | - Gui-Hui Tong
- Department of Pathology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510415, China
| | - Na Tang
- Department of Pathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, China.
| | - Yong-Jian Deng
- Department of Pathology, School of Basic Medical Sciences and Nan Fang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Shakiba S, Haddadi NS, Afshari K, Lubov JE, Raef HS, Li R, Yildiz-Altay Ü, Daga M, Refat MA, Kim E, de Laflin JG, Akabane A, Sherman S, MacDonald E, Strassner JP, Zhang L, Leon M, Baer CE, Dresser K, Liang Y, Whitley JB, Skopelja-Gardner S, Harris JE, Deng A, Vesely MD, Rashighi M, Richmond J. Spatial characterization of interface dermatitis in cutaneous lupus reveals novel chemokine ligand-receptor pairs that drive disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.05.574422. [PMID: 38260617 PMCID: PMC10802382 DOI: 10.1101/2024.01.05.574422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Chemokines play critical roles in the recruitment and activation of immune cells in both homeostatic and pathologic conditions. Here, we examined chemokine ligand-receptor pairs to better understand the immunopathogenesis of cutaneous lupus erythematosus (CLE), a complex autoimmune connective tissue disorder. We used suction blister biopsies to measure cellular infiltrates with spectral flow cytometry in the interface dermatitis reaction, as well as 184 protein analytes in interstitial skin fluid using Olink targeted proteomics. Flow and Olink data concordantly demonstrated significant increases in T cells and antigen presenting cells (APCs). We also performed spatial transcriptomics and spatial proteomics of punch biopsies using digital spatial profiling (DSP) technology on CLE skin and healthy margin controls to examine discreet locations within the tissue. Spatial and Olink data confirmed elevation of interferon (IFN) and IFN-inducible CXCR3 chemokine ligands. Comparing involved versus uninvolved keratinocytes in CLE samples revealed upregulation of essential inflammatory response genes in areas near interface dermatitis, including AIM2. Our Olink data confirmed upregulation of Caspase 8, IL-18 which is the final product of AIM2 activation, and induced chemokines including CCL8 and CXCL6 in CLE lesional samples. Chemotaxis assays using PBMCs from healthy and CLE donors revealed that T cells are equally poised to respond to CXCR3 ligands, whereas CD14+CD16+ APC populations are more sensitive to CXCL6 via CXCR1 and CD14+ are more sensitive to CCL8 via CCR2. Taken together, our data map a pathway from keratinocyte injury to lymphocyte recruitment in CLE via AIM2-Casp8-IL-18-CXCL6/CXCR1 and CCL8/CCR2, and IFNG/IFNL1-CXCL9/CXCL11-CXCR3.
Collapse
Affiliation(s)
- Saeed Shakiba
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | | | | | - Janet E. Lubov
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | - Haya S. Raef
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | - Robert Li
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | | | - Mridushi Daga
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | | | - Evangeline Kim
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | | | - Andressa Akabane
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | - Shany Sherman
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | | | | | | | | | - Christina E. Baer
- UMass Chan Medical School, Sanderson Center for Optical Experimentation, Dept of Microbiology and Physiological Systems, Worcester, MA, USA
| | - Karen Dresser
- UMass Chan Medical School, Dept of Pathology, Worcester, MA, USA
| | - Yan Liang
- NanoString Technologies, Seattle, WA, USA
| | - James B Whitley
- Dartmouth Hitchcock Medical Center, Dept of Medicine, Lebanon, NH, USA
| | | | - John E Harris
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | - April Deng
- UMass Chan Medical School, Dept of Pathology, Worcester, MA, USA
| | - Matthew D. Vesely
- Yale University School of Medicine, Dept of Dermatology, New Haven, CT, USA
| | - Mehdi Rashighi
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| | - Jillian Richmond
- UMass Chan Medical School, Dept of Dermatology, Worcester, MA, USA
| |
Collapse
|
12
|
Siqueira JM, Mitani Y, Hoff CO, Bonini F, Guimaraes de Sousa L, Marques-Piubelli ML, Purushothaman A, Mitani M, Dai H, Lin SY, Spiotto MT, Hanna EY, McGrail DJ, El-Naggar AK, Ferrarotto R. Analysis of B7-H4 Expression Across Salivary Gland Carcinomas Reveals Adenoid Cystic Carcinoma-Specific Prognostic Relevance. Mod Pathol 2024; 37:100371. [PMID: 38015043 DOI: 10.1016/j.modpat.2023.100371] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/11/2023] [Accepted: 10/22/2023] [Indexed: 11/29/2023]
Abstract
B7-H4 (VTCN1), a member of the B7 family, is overexpressed in several types of cancer. Here we investigated the pattern of expression of B7-H4 in salivary gland carcinomas (SGC) and assessed its potential as a prognostic marker and therapeutic target. Immunohistochemistry (IHC) analyses were performed in a cohort of 340 patient tumors, composed of 124 adenoid cystic carcinomas (ACC), 107 salivary duct carcinomas (SDC), 64 acinic cell carcinomas, 36 mucoepidermoid carcinomas (MEC), 9 secretory carcinomas (SC), as well as 20 normal salivary glands (controls). B7-H4 expression was scored and categorized into negative (<5% expression of any intensity), low (5%-70% expression of any intensity or >70% with weak intensity), or high (>70% moderate or strong diffuse intensity). The associations between B7-H4 expression and clinicopathologic characteristics, as well as overall survival, were assessed. Among all tumors, B7-H4 expression was more prevalent in ACC (94%) compared with those of SC (67%), MEC (44%), SDC (32%), and acinic cell carcinomas (0%). Normal salivary gland tissue did not express B7-H4. High expression of B7-H4 was found exclusively in ACC (27%), SDC (11%), and MEC (8%). In SDC, B7-H4 expression was associated with female gender (P = .002) and lack of androgen receptor expression (P = .012). In ACC, B7-H4 expression was significantly associated with solid histology (P < .0001) and minor salivary gland primary (P = .02). High B7-H4 expression was associated with a poorer prognosis in ACC, regardless of clinical stage and histologic subtype. B7-H4 expression was not prognostic in the non-ACC SGC evaluated. Our comparative study revealed distinct patterns of B7-H4 expression according to SGC histology, which has potential therapeutic implications. B7-H4 expression was particularly high in solid ACC and was an independent prognostic marker in this disease but not in the other SGC assessed.
Collapse
Affiliation(s)
- Juliana Mota Siqueira
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Stomatology, Discipline of Oral and Maxillofacial Pathology, School of Dentistry, University of São Paulo, São Paulo, SP, Brazil
| | - Yoshitsugu Mitani
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Camilla Oliveira Hoff
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Flavia Bonini
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Luana Guimaraes de Sousa
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mario L Marques-Piubelli
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anurag Purushothaman
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mutsumi Mitani
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hui Dai
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael T Spiotto
- Department of Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ehab Y Hanna
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Adel K El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
13
|
Mortezaee K, Majidpoor J. Alternative immune checkpoints in immunoregulatory profile of cancer stem cells. Heliyon 2023; 9:e23171. [PMID: 38144305 PMCID: PMC10746460 DOI: 10.1016/j.heliyon.2023.e23171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Tumor-mediated bypass of immune checkpoint inhibitor (ICI) therapy with anti-programmed death-1 (PD-1), anti-programmed death-ligand 1 (PD-L1, also called B7-H1 or CD274) or anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4) is a challenge of current years in the area of cancer immunotherapy. Alternative immune checkpoints (AICs) are molecules beyond the common PD-1, PD-L1 or CTLA-4, and are upregulated in patients who show low/no ICI responses. These are members of B7 family including B7-H2 (ICOS-L), B7-H3 (CD276), B7-H4 (B7x), V-domain immunoglobulin suppressor of T cell activation (VISTA), B7-H6, HHLA2 (B7-H5/B7-H7) and catabolic enzymes like indoleamine 2,3-dioxygenase 1 (IDO1), and others that are also contributed to the regulation of tumor immune microenvironment (TIME). There is also strong evidence supporting the implication of AICs in regulation of cancer stemness and expanding the population of cancer stem cells (CSCs). CSCs display immunoregulatory capacity and represent multiple immune checkpoints either on their surface or inside. Besides, they are active promoters of resistance to the common ICIs. The aim of this review is to investigate interrelations between AICs with stemness and differentiation profile of cancer. The key message of this paper is that targeted checkpoints can be selected based on their impact on CSCs along with their effect on immune cells. Studies published so far mainly focused on immune cells as a target for anti-checkpoints. Ex vivo engineering of extracellular vesicles (EVs) equipped with CSC-targeted anti-checkpoint antibodies is without a doubt a key therapeutic target that can be under consideration in future research.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
14
|
Liang Y, Wang L, Ma P, Ju D, Zhao M, Shi Y. Enhancing anti-tumor immune responses through combination therapies: epigenetic drugs and immune checkpoint inhibitors. Front Immunol 2023; 14:1308264. [PMID: 38077327 PMCID: PMC10704038 DOI: 10.3389/fimmu.2023.1308264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
Epigenetic mechanisms are processes that affect gene expression and cellular functions without involving changes in the DNA sequence. This abnormal or unstable expression of genes regulated by epigenetics can trigger cancer and other various diseases. The immune cells involved in anti-tumor responses and the immunogenicity of tumors may also be affected by epigenomic changes. This holds significant implications for the development and application of cancer immunotherapy, epigenetic therapy, and their combined treatments in the fight against cancer. We provide an overview of recent research literature focusing on how epigenomic changes in immune cells influence immune cell behavior and function, as well as the immunogenicity of cancer cells. And the combined utilization of epigenetic medications with immune checkpoint inhibitors that focus on immune checkpoint molecules [e.g., Programmed Death 1 (PD-1), Cytotoxic T-Lymphocyte-Associated Protein 4 (CTLA-4), T cell Immunoglobulin and Mucin Domain (TIM-3), Lymphocyte Activation Gene-3 (LAG-3)] present in immune cells and stromal cells associated with tumors. We highlight the potential of small-molecule inhibitors targeting epigenetic regulators to amplify anti-tumor immune responses. Moreover, we discuss how to leverage the intricate relationship between cancer epigenetics and cancer immunology to create treatment regimens that integrate epigenetic therapies with immunotherapies.
Collapse
Affiliation(s)
- Ying Liang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Lingling Wang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan Wuchang Hospital, Wuhan, China
| | - Peijun Ma
- Clinical Laboratory, Shanghai Mental Health Center, Shanghai, China
| | - Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Minggao Zhao
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yun Shi
- Department of Immunology and Theranostics, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| |
Collapse
|
15
|
Wang T, Zhang K, You F, Ma R, Yang N, Tian S, An G, Yang L. Preconditioning of radiotherapy enhances efficacy of B7-H3-CAR-T in treating solid tumor models. Life Sci 2023; 331:122024. [PMID: 37574043 DOI: 10.1016/j.lfs.2023.122024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/30/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
AIMS Limited efficacy of chimeric antigen receptor T (CAR-T) cells in treating solid tumors is largely due to the antigen heterogeneity and immunosuppressive tumor microenvironment (TME). B7-H3 is over-expressed in most kind of solid tumors, making it a promising target for cancer treatment. This study aims to explore the effect of B7-H3-CAR-T therapy combined with radiotherapy in treating solid tumor models. METHODS Irradiated tumor cell lines were prepared and tested. A humanized B7-H3-CAR-T was constructed, and it was evaluated that B7-H3-CAR-T cytotoxicity against solid tumor models with preconditioning of radiotherapy in vitro and vivo. RESULTS Irradiation was found to increase expression level of B7-H3 in pancreatic cancer (PANC-1), colorectal cancer (HCT-15, SW620), acute myelocytic leukemia (AML-5), epidermoid carcinoma (KB) and glioma (U87-MG) human cell lines significantly. 6Gy irradiation was also found to up-regulate tumor-infiltration molecule like intracellular adhesion molecule-1 ICAM-1 or FAS in HCT-15 cells, supporting a possible synergistic enhancement effect of radiotherapy. In vitro and in vivo experiments demonstrated that irradiation indeed significantly enhanced the ability of B7-H3-CAR-T to infiltrate and kill tumors. Interestingly in dual-tumor mouse model study, not only tumor cells on irradiation side were eradicated completely, irradiation also enhanced CAR-T tumor-killing ability on non-irradiated side, confirming the abscopal effect of irradiation existed with CAR-T therapy. CONCLUSIONS Our results suggest that B7-H3-CAR-T therapy combined with radiotherapy may be a promising modality in treating solid tumors.
Collapse
Affiliation(s)
- Tian Wang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; PersonGen BioTherapeutics Co., Ltd., Suzhou, PR China
| | - Kailu Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fengtao You
- PersonGen BioTherapeutics Co., Ltd., Suzhou, PR China
| | - Renyuxue Ma
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Nan Yang
- PersonGen BioTherapeutics Co., Ltd., Suzhou, PR China
| | - Shuaiyu Tian
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Gangli An
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Lin Yang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; PersonGen BioTherapeutics Co., Ltd., Suzhou, PR China.
| |
Collapse
|
16
|
Xiao L, Wang Q, Peng H. Tumor-associated macrophages: new insights on their metabolic regulation and their influence in cancer immunotherapy. Front Immunol 2023; 14:1157291. [PMID: 37426676 PMCID: PMC10325569 DOI: 10.3389/fimmu.2023.1157291] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/12/2023] [Indexed: 07/11/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are a dynamic and heterogeneous cell population of the tumor microenvironment (TME) that plays an essential role in tumor formation and progression. Cancer cells have a high metabolic demand for their rapid proliferation, survival, and progression. A comprehensive interpretation of pro-tumoral and antitumoral metabolic changes in TAMs is crucial for comprehending immune evasion mechanisms in cancer. The metabolic reprogramming of TAMs is a novel method for enhancing their antitumor effects. In this review, we provide an overview of the recent research on metabolic alterations of TAMs caused by TME, focusing primarily on glucose, amino acid, and fatty acid metabolism. In addition, this review discusses antitumor immunotherapies that influence the activity of TAMs by limiting their recruitment, triggering their depletion, and re-educate them, as well as metabolic profiles leading to an antitumoral phenotype. We highlighted the metabolic modulational roles of TAMs and their potential to enhance immunotherapy for cancer.
Collapse
Affiliation(s)
- Li Xiao
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qiao Wang
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongling Peng
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Mortezaee K, Majidpoor J, Najafi S. VISTA immune regulatory effects in bypassing cancer immunotherapy: Updated. Life Sci 2022; 310:121083. [DOI: 10.1016/j.lfs.2022.121083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
|
18
|
Mining of transcriptome identifies CD109 and LRP12 as possible biomarkers and deregulation mechanism of T cell receptor pathway in Acute Myeloid Leukemia. Heliyon 2022; 8:e11123. [PMID: 36299526 PMCID: PMC9589179 DOI: 10.1016/j.heliyon.2022.e11123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/16/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is a heterogeneous disease with highest mortality compared to other types of leukemia. There is a need to find the gene abnormalities and mechanisms behind them due to their heterogenic nature. The present study is aimed to understand genes, pathways and biomarker proteins influenced by transcriptomic deregulation due to AML. Differentially expressed gene (DEG), protein-protein interaction network, gene ontology, KEGG pathway, variant analysis and secretome analyses were performed using different AML RNAseq datasets. A total of 655 DEGs including 291 up-regulated and 364 down-regulated genes, which were satisfied with a fold change of 1.5 were identified. Top hub genes for AML were identified as TP53, PTPRC and AKT1. This integrative bioinformatics approach revealed the deregulation of T Cell Receptor (TCR) pathway and altered immune response related genes. The survival analysis revealed the associated deregulation of multiple TCR pathway related genes. Variant analysis identified the benign and likely benign nature of many important target genes and markers screened, which were found to have an important role in the progression of AML. DEGs and secretome analysis found out a set of seven molecules represents potential biomarkers for AML. In vitro analytical validation showed overexpression pattern of CD109 and LRP12 in AML cell line and HL-60 cells than the normal human bone marrow-derived stromal cell line HS-5. Here we report first time for CD109 and LRP12 as a possible biomarkers for the diagnostic significance. Amino acid substitutions detected by variant analysis and deregulation of immune checkpoint molecules revealed their role in reducing immune response and inability to fight cancer cells. In conclusion, this study highlights the possibility of new biomarkers for AML and the mechanism of decrease in immune response due to the downregulation of co-stimulatory immune molecules, which needs further clinical validation investigations. Using RNA-seq data of AML patients, two biomarkers including CD109 and LRP12 for the diagnostic significance were identified based on DEGs, GO/KEGG, and PPI network analysis. The transcriptome mining unmasked the complexity of gene alterations in AML by identifying immune response related genes deregulation and significance of TCR signalling. Several genes were identified as AML hub genes by network analysis, variant analysis identified non-synonymous variants in co-stimulatory checkpoint targets and the co-inhibitory targets.
Collapse
|
19
|
Yan X, Hong B, Feng J, Jin Y, Chen M, Li F, Qian Y. B7-H4 is a potential diagnostic and prognostic biomarker in colorectal cancer and correlates with the epithelial-mesenchymal transition. BMC Cancer 2022; 22:1053. [PMID: 36217128 PMCID: PMC9549643 DOI: 10.1186/s12885-022-10159-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/06/2022] [Indexed: 12/01/2022] Open
Abstract
Background As a negative co-stimulatory molecule of the B7 family, B7-H4 has recently attracted increased attention. However, the clinical value of B7-H4 in colorectal cancer (CRC) remains controversial and requires further investigation. This study aimed to investigate the role of B7-H4 in the clinical diagnosis and survival prognosis of CRC. Methods The relationships between B7-H4 expression, immune cell infiltration, epithelial-mesenchymal transition (EMT), clinicopathological features, and survival prognosis were determined through the TCGA database and verified in a large CRC cohort (n = 1118). Results The results showed the level of B7-H4 mRNA expression was significantly increased in the CRC tumor tissues compared with normal tissues (P < 0.001). Immunohistochemistry showed that B7-H4 protein expression was also up-regulated in CRC. The positive rate of B7-H4 in CRC tumor tissues was 76.38%, which was significantly higher than that in non-tumor tissues (P < 0.001). Overexpression of B7-H4 was positively correlated with lymph node metastasis, advanced TNM stage, and poor tumor differentiation (P = 0.012; 0.009; 0.014). Prognostic analysis showed high B7-H4 expression was associated with significantly shorter OS. Multivariate analysis demonstrated the risk of death in CRC patients with high B7-H4 expression is 1.487 times that of low B7-H4 expression. In addition, B7-H4 expression was negatively correlated with the epithelial marker E-cadherin (P < 0.001) and positively correlated with the mesenchymal marker vimentin (P < 0.001) in CRC tissues. However, B7-H4 expression was not associated with the immunosuppressive microenvironment in CRC. Conclusion B7-H4 may represent a potential biomarker for the diagnosis and prognosis of CRC and enhance CRC invasion by promoting EMT. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-10159-5.
Collapse
Affiliation(s)
- Xiaotian Yan
- Department of Clinical Laboratory, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 166 North Qiutao Road, Hangzhou, Zhejiang Province, 310006, China
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Jie Feng
- Department of Blood Transfusion, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Yuanqing Jin
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang Province, China
| | - Mengting Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang Province, China
| | - Fugang Li
- Shanghai Upper Bio Tech Pharma Company Limited, Shanghai, 201201, China
| | - Yun Qian
- Department of Clinical Laboratory, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, 166 North Qiutao Road, Hangzhou, Zhejiang Province, 310006, China.
| |
Collapse
|
20
|
Wang Y, Zhang H, Liu C, Wang Z, Wu W, Zhang N, Zhang L, Hu J, Luo P, Zhang J, Liu Z, Peng Y, Liu Z, Tang L, Cheng Q. Immune checkpoint modulators in cancer immunotherapy: recent advances and emerging concepts. J Hematol Oncol 2022; 15:111. [PMID: 35978433 PMCID: PMC9386972 DOI: 10.1186/s13045-022-01325-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 08/01/2022] [Indexed: 12/13/2022] Open
Abstract
The discovery of immune checkpoint inhibitors (ICIs) has now been universally acknowledged as a significant breakthrough in tumor therapy after the targeted treatment of checkpoint molecules: anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) on several cancer types achieved satisfying results. However, there are still quite a lot of patients suffering from severe side effects and ineffective treatment outcomes. Although the current ICI therapy is far from satisfying, a series of novel immune checkpoint molecules with remarkable preclinical and clinical benefits are being widely investigated, like the V-domain Ig suppressor of T cell activation (VISTA), which can also be called PD-1 homolog (PD-1H), and ectonucleotidases: CD39, CD73, and CD38, which belong to the ribosyl cyclase family, etc. In this review, we systematically summarized and discussed these molecules' biological structures, molecular features, and the corresponding targeted drugs, aiming to help the in-depth understanding of immune checkpoint molecules and promote the clinical practice of ICI therapy.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, People's Republic of China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, People's Republic of China
| | - Longbo Zhang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Department of Neurosurgery, and Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, USA
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jason Hu
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Department of Neonatology, Yale University School of Medicine, New Haven, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, People's Republic of China
| | - Yun Peng
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| | - Lanhua Tang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| |
Collapse
|
21
|
Wang Y, Zhang H, Liu C, Wang Z, Wu W, Zhang N, Zhang L, Hu J, Luo P, Zhang J, Liu Z, Peng Y, Liu Z, Tang L, Cheng Q. Immune checkpoint modulators in cancer immunotherapy: recent advances and emerging concepts. J Hematol Oncol 2022. [PMID: 35978433 DOI: 10.1186/s13045-022-01325-0.pmid:35978433;pmcid:pmc9386972.[125]robertc.adecadeofimmune-checkpointinhibitorsincancertherapy.natcommun.2020jul30;11(1):3801.doi:10.1038/s41467-020-17670-y.pmid:32732879;pmcid:pmc7393098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
The discovery of immune checkpoint inhibitors (ICIs) has now been universally acknowledged as a significant breakthrough in tumor therapy after the targeted treatment of checkpoint molecules: anti-programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) on several cancer types achieved satisfying results. However, there are still quite a lot of patients suffering from severe side effects and ineffective treatment outcomes. Although the current ICI therapy is far from satisfying, a series of novel immune checkpoint molecules with remarkable preclinical and clinical benefits are being widely investigated, like the V-domain Ig suppressor of T cell activation (VISTA), which can also be called PD-1 homolog (PD-1H), and ectonucleotidases: CD39, CD73, and CD38, which belong to the ribosyl cyclase family, etc. In this review, we systematically summarized and discussed these molecules' biological structures, molecular features, and the corresponding targeted drugs, aiming to help the in-depth understanding of immune checkpoint molecules and promote the clinical practice of ICI therapy.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Xiangya School of Medicine, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, People's Republic of China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- One-Third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, People's Republic of China
| | - Longbo Zhang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Department of Neurosurgery, and Department of Cellular & Molecular Physiology, Yale University School of Medicine, New Haven, USA
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jason Hu
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China
- Department of Neonatology, Yale University School of Medicine, New Haven, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, People's Republic of China
| | - Yun Peng
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| | - Lanhua Tang
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Center South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China.
| |
Collapse
|
22
|
Bączkowska M, Dutsch-Wicherek MM, Przytuła E, Faryna J, Wojtyła C, Ali M, Knafel A, Ciebiera M. Expression of the Costimulatory Molecule B7-H4 in the Decidua and Placental Tissues in Patients with Placental Abruption. Biomedicines 2022; 10:biomedicines10040918. [PMID: 35453668 PMCID: PMC9033103 DOI: 10.3390/biomedicines10040918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 12/10/2022] Open
Abstract
B7 homolog 4 protein (B7-H4), a member of the B7 family, is a immunomodulatory membrane protein. The aim of the study was to evaluate the expression of this protein in the decidua and placental tissues in case of placental abruption (PA) compared to cases of retained placental tissue (RPT) and controls. Tissue samples were obtained from 47 patients with PA, 60 patients with RPT, and 41 healthy controls. The samples were stained for B7-H4 expression, analyzed by an expert pathologist, and a semi-quantitative scale was applied. A statistical analysis revealed that the expression of B7-H4 was significantly higher in the decidua in PA samples compared to samples from patients with RPT (p-value < 0.001) and healthy controls (p-value < 0.001). The expression of B7-H4 in the placental chorionic villus was significantly higher in PA samples in relation to samples from healthy controls (p-value < 0.001) but not in relation to RPT samples (p-value = 0.0853). This finding suggests that B7-H4 might play an important role in mechanisms restoring reproductive tract homeostasis. Further research is necessary in regard to the role of B7-H4 in PA.
Collapse
Affiliation(s)
- Monika Bączkowska
- Centre of Postgraduate Medical Education, Second Department of Obstetrics and Gynecology, 01-809 Warsaw, Poland; (M.B.); (A.K.)
| | | | - Ewa Przytuła
- Department of Pathology, Bielański Hospital, 01-809 Warsaw, Poland; (E.P.); (J.F.)
| | - Jan Faryna
- Department of Pathology, Bielański Hospital, 01-809 Warsaw, Poland; (E.P.); (J.F.)
| | - Cezary Wojtyła
- International Prevention Research Institute-Collaborating Centre, Calisia University, 62-800 Kalisz, Poland;
| | - Mohamed Ali
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt;
| | - Anna Knafel
- Centre of Postgraduate Medical Education, Second Department of Obstetrics and Gynecology, 01-809 Warsaw, Poland; (M.B.); (A.K.)
| | - Michał Ciebiera
- Centre of Postgraduate Medical Education, Second Department of Obstetrics and Gynecology, 01-809 Warsaw, Poland; (M.B.); (A.K.)
- Correspondence: ; Tel.: +48-607-155-177
| |
Collapse
|
23
|
Wang P, Tang L, Zhou B, Cheng L, Zhao RC, Zhang J. Analytical methods for the detection of PD-1/PD-L1 and other molecules related to immune checkpoints. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2021.116505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
24
|
Chen Y, Feng R, He B, Wang J, Xian N, Huang G, Zhang Q. PD-1H Expression Associated With CD68 Macrophage Marker Confers an Immune-Activated Microenvironment and Favorable Overall Survival in Human Esophageal Squamous Cell Carcinoma. Front Mol Biosci 2021; 8:777370. [PMID: 34950702 PMCID: PMC8688962 DOI: 10.3389/fmolb.2021.777370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal carcinoma (EC) in China. Although the PD-1 inhibitor pembrolizumab has been approved to treat patients with EC, its therapeutic efficacy is limited. Thus, additional immunotherapeutic targets for EC treatment are needed. Programmed Death-1 Homolog (PD-1H) is a negative checkpoint regulator that inhibits antitumor immune responses. Here, PD-1H expression in 114 patients with ESCC was evaluated by immunohistochemistry. Next, 12 randomly selected tumor tissue sections were used to assess the colocalization of PD-1H protein and multiple immune markers by multiplex immunohistochemistry. Our results demonstrated that PD-1H was expressed at high frequency in ESCC tumor tissues (85.1%). PD-1H protein was predominantly expressed in CD68+ tumor-associated macrophages and expressed at low levels in CD4+ T cells and CD8+ T cells in ESCC tumor tissues. Furthermore, based on ESCC data in The Cancer Genome Atlas (TCGA), the gene expression levels of PD-1H were positively associated with the infiltration levels of immune-activated cells especially CD8+ cytotoxic T cells. In contrast, the gene expression levels of PD-1H were negatively correlated with myeloid-derived suppressor cells (MDSCs). Importantly, PD-1H expression in tumor sites was significantly correlated with favorable overall survival in patients with ESCC. Collectively, our findings first provided direct information on the PD-1H expression pattern and distribution in ESCC, and positive correlation of PD-1H expression with overall survival suggested PD-1H expression levels could be a significant prognostic indicator for patients with ESCC. Future studies need to explore the immunoregulatory of PD-1H in the tumor microenvironment of ESCC.
Collapse
Affiliation(s)
- Yuangui Chen
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui Feng
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.,Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bailin He
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jun Wang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Na Xian
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Gangxiong Huang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Qiuyu Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
25
|
Gao Y, Xu Y, Gao M, Huang A, Chi P. A three-phase trans-ethnic study reveals B7-H3 expression is a significant and independent biomarker associated with colon cancer overall survival. J Gastrointest Oncol 2021; 12:2891-2905. [PMID: 35070416 PMCID: PMC8748050 DOI: 10.21037/jgo-21-821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/20/2021] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND There have been inconsistent results and conflicting conclusions among the existing prognostic studies of B7-H3 expression in colon cancer patients. Therefore, the association between B7-H3 expression and colon cancer survival has remained largely unclear. METHODS We performed a three-phase and trans-ethnic prognostic study of B7-H3 expression in colon cancer patients involving perhaps the largest population to date. In the discovery phase, we utilized a Cox proportional hazards model adjusted for covariates to test the association between B7-H3 expression and colon cancer overall survival (OS) time in a European population from The Cancer Genome Atlas (TCGA) cohort (n=433). In the validation phase I, the association was replicated in a European population from Gene Expression Omnibus (GEO) cohort (n=811). In the validation phase II, we again confirmed the significant association in an Asian population from Fujian Medical University Union Hospital (UNION) cohort (n=179). Furthermore, a series of Kaplan-Meier analysis, bioinformatics analysis of tumor immune microenvironment (TIME), and immune checkpoint prognostic prediction analysis, as well as sensitivity analysis, were also conducted. RESULTS Highly expressed B7-H3 was a significant and robust biomarker to colon cancer survival, with a large hazard ratio (HR) [HRTCGA =4.60, 95% confidence interval (CI): 2.15 to 9.83, P=8.37×10-05; HRGEO =1.47, 95% CI: 1.12 to 1.94, P=0.0056; HRUNION =1.63, 95% CI: 1.36 to 1.95, P=7.91×10-08]. We detected an involvement of B7-H3 in the tumor immune microenvironment (TIME). Meanwhile, B7-H3 was significantly and weakly correlated with 6 out of 27 well-recognized immune checkpoint genes. Even after adjusting for effects of other immune checkpoint genes, B7-H3 still exhibited a harmful effect on colon cancer survival using samples from TCGA and GEO cohorts (HR =1.47, 95% CI: 1.07 to 2.02, P=0.0184), indicating that it was an independent prognostic factor of colon cancer. We also proposed an immune checkpoint prognostic risk score which possessed the capability to identify colon cancers with high risk of mortality. CONCLUSIONS The expression of B7-H3 is a significant, robust, and independent prognostic factor to colon cancer OS.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu Xu
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Institute of Oncology of Fujian Medical University, Fuzhou, China
| | - Meiqin Gao
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Institute of Oncology of Fujian Medical University, Fuzhou, China
| | - Aimin Huang
- Department of Pathology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- Institute of Oncology of Fujian Medical University, Fuzhou, China
| | - Pan Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
26
|
Feng R, Chen Y, Liu Y, Zhou Q, Zhang W. The role of B7-H3 in tumors and its potential in clinical application. Int Immunopharmacol 2021; 101:108153. [PMID: 34678689 DOI: 10.1016/j.intimp.2021.108153] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
B7-H3 (CD276 molecule) is an immune checkpoint from the B7 family of molecules that acts more as a co-inhibitory molecule to promote tumor progression. It is abnormally expressed on tumor cells and can be induced to express on antigen-presenting cells (APCs) including dendritic cells (DCs) and macrophages. In the tumor microenvironment (TME), B7-H3 promotes tumor progression by impairing T cell response, promoting the polarization of tumor-associated macrophages (TAMs) to M2, inhibiting the function of DCs, and promoting the migration and invasion of cancer-associated fibroblasts (CAFs). In addition, through non-immunological functions, B7-H3 promotes tumor cell proliferation, invasion, metastasis, resistance, angiogenesis, and metabolism, or in the form of exosomes to promote tumor progression. In this process, microRNAs can regulate the expression of B7-H3. B7-H3 may serve as a potential biomarker for tumor diagnosis and a marker of poor prognosis. Immunotherapy targeting B7-H3 and the combination of B7-H3 and other immune checkpoints have shown certain efficacy. In this review, we summarized the basic characteristics of B7-H3 and its mechanism to promote tumor progression by inducing immunosuppression and non-immunological functions, as well as the potential clinical applications of B7-H3 and immunotherapy based on B7-H3.
Collapse
Affiliation(s)
- Ranran Feng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Andrology, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yong Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qing Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Masoumi E, Tahaghoghi-Hajghorbani S, Jafarzadeh L, Sanaei MJ, Pourbagheri-Sigaroodi A, Bashash D. The application of immune checkpoint blockade in breast cancer and the emerging role of nanoparticle. J Control Release 2021; 340:168-187. [PMID: 34743998 DOI: 10.1016/j.jconrel.2021.10.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022]
Abstract
Breast cancer is the most common malignancy in the female population with a high mortality rate. Despite the satisfying depth of studies evaluating the contributory role of immune checkpoints in this malignancy, few articles have reviewed the pros and cons of immune checkpoint blockades (ICBs). In the current review, we provide an overview of immune-related inhibitory molecules and also discuss the original data obtained from international research laboratories on the aberrant expression of T and non-T cell-associated immune checkpoints in breast cancer. Then, we especially focus on recent studies that utilized ICBs as the treatment strategy in breast cancer and provide their efficiency reports. As there are always costs and benefits, we discuss the limitations and challenges toward ICB therapy such as adverse events and drug resistance. In the last section, we allocate an overview of the recent data concerning the application of nanoparticle systems for cancer immunotherapy and propose that nano-based ICB approaches may overcome the challenges related to ICB therapy in breast cancer. In conclusion, it seems it is time for nanoscience to more rapidly move forward into clinical trials and illuminates the breast cancer treatment area with its potent features for the target delivery of ICBs.
Collapse
Affiliation(s)
- Elham Masoumi
- Department of Immunology, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran; Student Research Committee, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Sahar Tahaghoghi-Hajghorbani
- Microbiology and Virology Research Center, Qaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Leila Jafarzadeh
- Department of Laboratory Science, Sirjan Faculty of Medical Science, Sirjan, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Ahmad A. Epigenetic regulation of immunosuppressive tumor-associated macrophages through dysregulated microRNAs. Semin Cell Dev Biol 2021; 124:26-33. [PMID: 34556420 DOI: 10.1016/j.semcdb.2021.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/02/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023]
Abstract
Macrophages are immune cells that play different roles under different physiological conditions. They are present in all tissues where they primarily protect from bacteria and pathogens in addition to assisting in tissue repair. During tumor progression, macrophages can exert contrasting effects based on the M1 vs. M2 polarization. The M2 macrophages support tumor growth through mechanisms that help suppress immune responses and/or circumvent immune-surveillance. A number of such mechanisms such as production of IL-10 and arginase, and expression of PD-L1, V-domain Ig suppressor of T cell activation and B7 family molecule B7-H4 are now believed central to the immunosuppressive effects of tumor-associated macrophages (TAMs). Emerging data has identified epigenetic regulation of these immunosuppressive mechanisms by small non-coding RNAs, the microRNAs (miRNAs). This review discusses the available literature on the subject, including the exosomes mediated transfer of miRNAs between cancer cells and the macrophages within the tumor microenvironment. A number of miRNAs are now believed to be involved in TAMs' production of IL-10 and expression of PD-L1 while the information on such regulation of other immunosuppressive mechanisms is slowly emerging. A better understanding of epigenetic regulation of macrophages-mediated immunosuppressive effect can help identify novel targets for therapy and aid the design of future studies aimed at sensitizing tumors to immune responses.
Collapse
Affiliation(s)
- Aamir Ahmad
- Interim Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
29
|
New insights into exosome mediated tumor-immune escape: Clinical perspectives and therapeutic strategies. Biochim Biophys Acta Rev Cancer 2021; 1876:188624. [PMID: 34487817 DOI: 10.1016/j.bbcan.2021.188624] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in extracellular vesicle biology have uncovered a substantial role in maintaining cell homeostasis in health and disease conditions by mediating intercellular communication, thus catching the scientific community's attention worldwide. Extracellular microvesicles, some called exosomes, functionally transfer biomolecules such as proteins and non-coding RNAs from one cell to another, influencing the local environment's biology. Although numerous advancements have been made in treating cancer patients with immune therapy, controlling the disease remains a challenge in the clinic due to tumor-driven interference with the immune response and inability of immune cells to clear cancer cells from the body. The present review article discusses the recent findings and knowledge gaps related to the role of exosomes derived from tumors and the tumor microenvironment cells in tumor escape from immunosurveillance. Further, we highlight examples where exosomal non-coding RNAs influence immune cells' response within the tumor microenvironment and favor tumor growth and progression. Therefore, exosomes can be used as a therapeutic target for the treatment of human cancers.
Collapse
|
30
|
Li TT, Jiang JW, Qie CX, Xuan CX, Hu XL, Liu WM, Chen WT, Liu J. Identification of active small-molecule modulators targeting the novel immune checkpoint VISTA. BMC Immunol 2021; 22:55. [PMID: 34380434 PMCID: PMC8359099 DOI: 10.1186/s12865-021-00446-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/30/2021] [Indexed: 12/18/2022] Open
Abstract
Background Cancer immunotherapy has gained increasing popularity as a novel approach to treat cancer. A member of the B7 family, V-domain immunoglobulin suppressor of T-cell activation (VISTA) is a novel immune checkpoint that regulates a broad spectrum of immune responses. VISTA is an acidic pH-selective ligand for P-selectin glycoprotein ligand-1(PSGL-1). CA-170, a first-in-class small-molecule dual antagonist of VISTA/PD-L1, was collaboratively developed by Aurigene Discovery Technologies Limited and Curis, Inc. It is currently in Phase I clinical trial. Results In this study, we develop homology modeling for the VISTA 3D structure and subsequent virtual screening for VISTA small-molecule hit ligands. Visualization of the binding postures of docked ligands with the VISTA protein indicates that some small molecular compounds target VISTA. The ability of antagonist to disrupt immune checkpoint VISTA pathways was investigated though functional studies in vitro. Conclusions Affinity active molecule for VISTA was obtained through virtual screening, and the antagonist compound activity to VISTA was assayed in cellular level. We reported a small molecule with high VISTA affinity as antagonist, providing ideas for development VISTA-targeted small molecule compound in cancer immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-021-00446-4.
Collapse
Affiliation(s)
- Ting-Ting Li
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Jing-Wei Jiang
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Chen-Xin Qie
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Chun-Xiao Xuan
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin-Lei Hu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Wan-Mei Liu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Wen-Ting Chen
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China
| | - Jun Liu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
31
|
Liu S, Liang J, Liu Z, Zhang C, Wang Y, Watson AH, Zhou C, Zhang F, Wu K, Zhang F, Lu Y, Wang X. The Role of CD276 in Cancers. Front Oncol 2021; 11:654684. [PMID: 33842369 PMCID: PMC8032984 DOI: 10.3389/fonc.2021.654684] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 03/02/2021] [Indexed: 02/05/2023] Open
Abstract
Objective Aberrant expression of the immune checkpoint molecule, CD276, also known as B7-H3, is associated with tumorigenesis. In this review, we aim to comprehensively describe the role of CD276 in malignancies and its potential therapeutic effect. Data Sources Database including PubMed, EMbase, Cochrane Library, CNKI, and Clinical Trails.gov were searched for eligible studies and reviews. Study selection: Original studies and review articles on the topic of CD276 in tumors were retrieved. Results CD276 is an immune checkpoint molecule in the epithelial mesenchymal transition (EMT) pathway. In this review, we evaluated the available evidence on the expression and regulation of CD276. We also assessed the role of CD276 within the immune micro-environment, effect on tumor progression, and the potential therapeutic effect of CD276 targeted therapy for malignancies. Conclusion CD276 plays an essential role in cell proliferation, invasion, and migration in malignancies. Results from most recent studies indicate CD276 could be a promising therapeutic target for malignant tumors.
Collapse
Affiliation(s)
- Shengzhuo Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.,William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Jiayu Liang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihong Liu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Chi Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Alice Helen Watson
- Clinical Science and Services, Royal Veterinary College, University of London, London, United Kingdom
| | - Chuan Zhou
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Fan Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Kan Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Fuxun Zhang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Yiping Lu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Xianding Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Javadrashid D, Baghbanzadeh A, Hemmat N, Hajiasgharzadeh K, Nourbakhsh NS, Lotfi Z, Baradaran B. Envisioning the immune system to determine its role in pancreatic ductal adenocarcinoma: Culprit or victim? Immunol Lett 2021; 232:48-59. [PMID: 33647329 DOI: 10.1016/j.imlet.2021.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma has a poor 5-year survival rate that makes it one of the most fatal human malignancies. Unfortunately, despite the serious improvement in the survival of most cancers, there has been a minor advance in pancreatic cancer (PC). Major advances in PC treatment have been assessed over the bygone twenty-year time span, yet some complications make the survival of the patients shorter. Getting to know the PC tumor microenvironment (TME) and the immunosuppression that happens during the pathogenesis of this malignancy could be a great help to understand the nature of the immune system and find better treatment modalities based on it. Although many immune cells are present in PC, immunosuppression of the TME leads to severe immune dysfunction in the patients, therefore immune effectors fail to do their functions. Lately, immunotherapy has been presented as one of the promising treatment strategies for different malignancies including hepatocellular carcinoma, melanoma, non-small cell lung cancer, and kidney cancer. In PC, there has been shown promising results centered around the TME, immune checkpoint inhibitors, cancer vaccines, and other approaches especially when used as combinational therapy. Here we dig a little deeper into the role of the immune system and possible therapeutic options in the treatment of PC.
Collapse
Affiliation(s)
- Darya Javadrashid
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Ziba Lotfi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Hu X, Qie C, Jiang J, Xie X, Chen W, Liu W, Liu J. M351-0056 is a novel low MW compound modulating the actions of the immune-checkpoint protein VISTA. Br J Pharmacol 2021; 178:1445-1458. [PMID: 33450048 PMCID: PMC9328666 DOI: 10.1111/bph.15357] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 10/13/2020] [Accepted: 12/20/2020] [Indexed: 01/29/2023] Open
Abstract
Background and Purpose The protein V‐domain immunoglobulin suppressor of T‐cell activation (VISTA) is a novel immune‐checkpoint molecule that belongs to the B7 family and regulates a broad spectrum of immune responses. So far, low MW compounds targeting VISTA for the treatment of autoimmune diseases or inflammation, have not been identified. Experimental Approach We developed a homology modelling for VISTA 3D structure and subsequent virtual screening for low MW ligands binding to VISTA. Visualization of the binding postures of docked ligands with protein VISTA indicated that compound M351‐0056 targeted VISTA. The biological activities of compound M351‐0056 targeting VISTA were investigated in vitro using monocytes and T cells and in vivo, using mice with imiquimod‐induced dermatitis. Key Results The KD value of M351‐0056 for human VISTA‐extracellular domain was 12.60 ± 3.84 μM as assessed by microscale thermophoresis. M351‐0056 decreased cytokine secretion from PBMCs or human CD4+ T cells, suppressed proliferation of PBMCs and enhanced expression of Foxp3+ T cells. These effects of M351‐0056 modulating VISTA involved the JAK2–STAT2 pathway. Daily administration of M351‐0056 ameliorated imiquimod‐induced psoriasis‐like dermatitis. Expression of mRNA and protein of inflammatory cytokines in psoriatic lesions was decreased after M351‐0056 treatment. Conclusion and Implications The compound M351‐0056 showed high affinity for VISTA and may modulate its immune function in vitro and in vivo. Our finding provides a lead compound for therapeutically enhancing VISTA‐mediated pathways to benefit the treatment of autoimmune diseases or inflammation.
Collapse
Affiliation(s)
- Xinlei Hu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Chenxin Qie
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Xiaoxue Xie
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Wenting Chen
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Wanmei Liu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- Jiangsu Key Lab of Drug Screening, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
34
|
Candiloro F, Borioli V, Borsellino G, Picozza M, Pellini R, Cereda E, Gargano F, Caraccia M, Nardi MT, Bellu L, Tondulli L, Imarisio I, Pozzi E, Pedrazzoli P, Caccialanza R, Battistini L. Influence of different lipid emulsions on specific immune cell functions in head and neck cancer patients receiving supplemental parenteral nutrition: An exploratory analysis. Nutrition 2021; 86:111178. [PMID: 33631618 DOI: 10.1016/j.nut.2021.111178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/05/2021] [Accepted: 01/21/2021] [Indexed: 12/28/2022]
Abstract
OBJECTIVES The effect of diet on immune responses is an area of intense investigation. Dietary lipids have been shown to differently influence and fine-tune the reactivity of immune cell subsets, thus potentially affecting clinical outcomes. Patients with head and neck squamous cell carcinoma face malnutrition, due to swallowing impairment related to the tumor site or to treatment sequalae, and may need supplemental parenteral nutrition (SPN) in addition to oral feeding when enteral nutrition is not feasible. Additionally, immune depression is a well-known complication in these patients. Parenteral nutrition (PN) bags contain amino acids, minerals, electrolytes and mostly lipids that provide calories in a concentrated form and are enriched with essential fatty acids. The aim of this study was to investigate multiple parameters of the immune responses in a cohort of patients with head and neck squamous cell carcinoma undergoing supplemental PN with bags enriched in ω-3 or ω-9 and ω-6 fatty acids. METHODS To our knowledge, this was the first exploratory study to investigate the effects of two different PN lipid emulsions on specific immune cells function of patients with advanced head and neck squamous carcinoma. ω-3-enriched fish-oil-based- and ω-6- and ω-9-enriched olive-oil-basedSPN was administered to two groups of patients for 1 wk in the context of an observational multicentric study. Polychromatic flow cytometry was used to investigate multiple subsets of leukocytes, with a special focus on cellular populations endowed with antitumor activity. RESULTS Patients treated with olive-oil-based PN showed an increase in the function of the innate (natural killer cells and monocytes) and adaptive (both CD4 and CD8 cells) arms of the immune response. CONCLUSION An increase in the function of the innate and adaptive arms of the immune response may favor antitumoral responses.
Collapse
Affiliation(s)
| | - Valeria Borioli
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Italy
| | | | - Mario Picozza
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Raul Pellini
- Department of Otolaryngology Head and Neck Surgery, IRCCS Regina Elena National Cancer Institute, Italy
| | - Emanuele Cereda
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Italy
| | | | - Marilisa Caraccia
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Italy
| | - Maria Teresa Nardi
- Nutritional Support Unit and Department of Clinical & Experimental Oncology, Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Italy
| | - Luisa Bellu
- Nutritional Support Unit and Department of Clinical & Experimental Oncology, Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Italy
| | - Luca Tondulli
- Medical Oncology, Azienda Ospedaliera Universitaria Integrata (AOUI), Italy
| | - Ilaria Imarisio
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Italy
| | - Emma Pozzi
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Italy
| | - Paolo Pedrazzoli
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Italy; Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Italy
| | - Luca Battistini
- Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
35
|
Wang YN, Lee HH, Hsu JL, Yu D, Hung MC. The impact of PD-L1 N-linked glycosylation on cancer therapy and clinical diagnosis. J Biomed Sci 2020; 27:77. [PMID: 32620165 PMCID: PMC7333976 DOI: 10.1186/s12929-020-00670-x] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/30/2020] [Indexed: 12/15/2022] Open
Abstract
N-linked glycosylation is one of the most abundant posttranslational modifications of membrane-bound proteins in eukaryotes and affects a number of biological activities, including protein biosynthesis, protein stability, intracellular trafficking, subcellular localization, and ligand-receptor interaction. Accumulating evidence indicates that cell membrane immune checkpoint proteins, such as programmed death-ligand 1 (PD-L1), are glycosylated with heavy N-linked glycan moieties in human cancers. N-linked glycosylation of PD-L1 maintains its protein stability and interaction with its cognate receptor, programmed cell death protein 1 (PD-1), and this in turn promotes evasion of T-cell immunity. Studies have suggested targeting PD-L1 glycosylation as a therapeutic option by rational combination of cancer immunotherapies. Interestingly, structural hindrance by N-glycan on PD-L1 in fixed samples impedes its recognition by PD-L1 diagnostic antibodies. Notably, the removal of N-linked glycosylation enhances PD-L1 detection in a variety of bioassays and more accurately predicts the therapeutic efficacy of PD-1/PD-L1 inhibitors, suggesting an important clinical implication of PD-L1 N-linked glycosylation. A detailed understanding of the regulatory mechanisms, cellular functions, and diagnostic limits underlying PD-L1 N-linked glycosylation could shed new light on the clinical development of immune checkpoint inhibitors for cancer treatment and deepen our knowledge of biomarkers to identify patients who would benefit the most from immunotherapy. In this review, we highlight the effects of protein glycosylation on cancer immunotherapy using N-linked glycosylation of PD-L1 as an example. In addition, we consider the potential impacts of PD-L1 N-linked glycosylation on clinical diagnosis. The notion of utilizing the deglycosylated form of PD-L1 as a predictive biomarker to guide anti-PD-1/PD-L1 immunotherapy is also discussed.
Collapse
Affiliation(s)
- Ying-Nai Wang
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Heng-Huan Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jennifer L Hsu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA. .,Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, 91 Hsueh-Shih Rd, North District, Taichung, 404, Taiwan. .,Department of Biotechnology, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
36
|
Cao D, Chen MK, Zhang QF, Zhou YF, Zhang MY, Mai SJ, Zhang YJ, Chen MS, Li XX, Wang HY. Identification of immunological subtypes of hepatocellular carcinoma with expression profiling of immune-modulating genes. Aging (Albany NY) 2020; 12:12187-12205. [PMID: 32544882 PMCID: PMC7343492 DOI: 10.18632/aging.103395] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022]
Abstract
Recent studies demonstrate that immune checkpoint inhibitor (ICI) therapy has achieved success in many types of advanced cancers including advanced hepatocellular carcinoma (HCC). However, ICI therapy is beneficial in only some HCC patients, suggesting that immune-responses are highly variable in HCCs. Therefore, understanding the immune status in HCC microenvironment will facilitate ICI immunotherapy and guide patient selection for the therapy. In this study, we first analyzed the expression profile of immune-modulating genes and their relationship with survival of HCC patients using the data downloaded from The Cancer Genome Atlas - Liver Hepatocellular Carcinoma (TCGA-LIHC) database, and found that the higher expressions of CD276 (B7-H3) and CD47 were significantly associated with poor survival. Then we identified 4 immune subtypes of HCCs with different survivals by using the combination expression of B7-H3 (or CD47) and CD8. Patients with B7-H3low/CD8high or CD47low/CD8high have the best survival while ones with B7-H3high/CD8low or CD47high/CD8low have the worst survival. The 4 immune subtypes were validated in another 72 HCC patients obtained from South China. In conclusion, our findings suggest that HCC patient prognosis is associated with immunophenotypes by T cell infiltration (CD8 expression) and the expression of the adaptive immune resistance gene (B7-H3 or CD47), and this immune classification system will facilitate HCC patient selection for ICI immunotherapy.
Collapse
Affiliation(s)
- Di Cao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Meng-Ke Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qing-Feng Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yu-Feng Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shi-Juan Mai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yao-Jun Zhang
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Min-Shan Chen
- Department of Hepatobiliary Oncology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xiao-Xing Li
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| |
Collapse
|
37
|
Lanuza PM, Pesini C, Arias MA, Calvo C, Ramirez-Labrada A, Pardo J. Recalling the Biological Significance of Immune Checkpoints on NK Cells: A Chance to Overcome LAG3, PD1, and CTLA4 Inhibitory Pathways by Adoptive NK Cell Transfer? Front Immunol 2020; 10:3010. [PMID: 31998304 PMCID: PMC6962251 DOI: 10.3389/fimmu.2019.03010] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022] Open
Abstract
Immune checkpoint receptors (IC) positively or negatively regulate the activation of the host immune response, preventing unwanted reactions against self-healthy tissues. In recent years the term IC has been mainly used for the inhibitory ICs, which are critical to control Natural Killer (NK) and Cytotoxic CD8+ T cells due to its high cytotoxic potential. Due to the different nature of the signals that regulate T and NK cell activation, specific ICs have been described that mainly regulate either NK cell or T cell activity. Thus, strategies to modulate NK cell activity are raising as promising tools to treat tumors that do not respond to T cell-based immunotherapies. NK cell activation is mainly regulated by ICs and receptors from the KIR, NKG2 and NCRs families and the contribution of T cell-related ICs is less clear. Recently, NK cells have emerged as contributors to the effect of inhibitors of T cell-related ICs like CTLA4, LAG3 or the PD1/PD-L1 axes in cancer patients, suggesting that these ICs also regulate the activity of NK cells under pathological conditions. Strikingly, in contrast to NK cells from cancer patients, the level of expression of these ICs is low on most subsets of freshly isolated and in vitro activated NK cells from healthy patients, suggesting that they do not control NK cell tolerance and thus, do not act as conventional ICs under non-pathological conditions. The low level of expression of T cell-related ICs in “healthy” NK cells suggest that they should not be restricted to the detrimental effects of these inhibitory mechanisms in the cancer microenvironment. After a brief introduction of the regulatory mechanisms that control NK cell anti-tumoral activity and the conventional ICs controlling NK cell tolerance, we will critically discuss the potential role of T cell-related ICs in the control of NK cell activity under both physiological and pathological (cancer) conditions. This discussion will allow to comprehensively describe the chances and potential limitations of using allogeneic NK cells isolated from a healthy environment to overcome immune subversion by T cell-related ICs and to improve the efficacy of IC inhibitors (ICIs) in a safer way.
Collapse
Affiliation(s)
- Pilar M Lanuza
- Immunotherapy, Inflammation and Cancer, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | - Cecilia Pesini
- Immunotherapy, Inflammation and Cancer, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain
| | | | - Carlota Calvo
- Immunotherapy, Inflammation and Cancer, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Medical Oncopediatry Department, Aragón Health Research Institute (IIS Aragón), Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Ariel Ramirez-Labrada
- Immunotherapy, Inflammation and Cancer, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Unidad de Nanotoxicología e Inmunotoxicología (UNATI), Centro de Investigación Biomédica de Aragón (CIBA), Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Julian Pardo
- Immunotherapy, Inflammation and Cancer, Aragón Health Research Institute (IIS Aragón), Biomedical Research Centre of Aragón (CIBA), Zaragoza, Spain.,Aragón i + D Foundation (ARAID), Government of Aragon, Zaragoza, Spain.,Department of Microbiology, Preventive Medicine and Public Health, University of Zaragoza, Zaragoza, Spain.,Nanoscience Institute of Aragon (INA), University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
38
|
Pourmaleki M, Young JH, Socci ND, Chiang S, Edelweiss M, Li Y, Zhang M, Roshal L, Chi DS, Busam KJ, Mellinghoff IK, Hollmann TJ. Extramammary Paget disease shows differential expression of B7 family members B7-H3, B7-H4, PD-L1, PD-L2 and cancer/testis antigens NY-ESO-1 and MAGE-A. Oncotarget 2019; 10:6152-6167. [PMID: 31692889 PMCID: PMC6817453 DOI: 10.18632/oncotarget.27247] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Extramammary Paget disease (EMPD) is a rare cutaneous adenocarcinoma of the anogenital region most commonly treated with surgical excision. Surgical margin clearance is often problematic and recurrence rates remain high indicating the need for additional therapeutic options. Topical immunomodulators have been used with reported success suggesting EMPD may respond to other immunotherapies. This study investigates EMPD protein expression of targetable B7 family members and cancer/testis antigens (CTAs) B7-H3, B7-H4, PD-L1, PD-L2, MAGE-A, and NY-ESO-1 and components of antigen presenting machinery B2M and MHC-I. Fifty-seven specimens from 48 patients (31 female and 17 male), representing in situ, invasive, and metastatic disease of primary and secondary origin were stained and scored (627 total slides). The percentage of cases expressing each immune regulatory molecule in the in situ followed by invasive tumor components was: B7-H3 (94, 90), B7-H4 (82, 78), PD-L1 (6, 10), MAGE-A (39, 50), NY-ESO-1 (16, 20), B2M (100, 89), and MHC-I (78, 79). PD-L2 was negative in all cases. There was high correlation between marker expression within the in situ and invasive tumor components of the same case. B7-H4 was preferentially expressed in primary cutaneous EMPD. Co-expression of B7 family members B7-H3 and B7-H4 was found within the in situ and invasive tumor components of 74% and 48% of cases, respectively. These findings provide an initial characterization of EMPD tumor cell expression of B7-H3, B7-H4, PD-L1, PD-L2, MAGE-A, and NY-ESO-1 and indicate the potential for new immunotherapeutic options for patients with EMPD.
Collapse
Affiliation(s)
- Maryam Pourmaleki
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jonathan H Young
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Present address: School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Nicholas D Socci
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sarah Chiang
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Marcia Edelweiss
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yanyun Li
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mianlei Zhang
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lev Roshal
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Dennis S Chi
- Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Klaus J Busam
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ingo K Mellinghoff
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Travis J Hollmann
- Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
39
|
Nakashima T, Liu T, Hu B, Wu Z, Ullenbruch M, Omori K, Ding L, Hattori N, Phan SH. Role of B7H3/IL-33 Signaling in Pulmonary Fibrosis-induced Profibrogenic Alterations in Bone Marrow. Am J Respir Crit Care Med 2019; 200:1032-1044. [PMID: 31106564 PMCID: PMC6794107 DOI: 10.1164/rccm.201808-1560oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/14/2019] [Indexed: 01/12/2023] Open
Abstract
Rationale: The impact of lung insult on the bone marrow (BM) and subsequent disease is unknown.Objectives: To study alterations in the BM in response to lung injury/fibrosis and examine their impact on subsequent lung insult.Methods: BM cells from control or bleomycin-treated donor mice were transplanted into naive mice, which were subsequently evaluated for bleomycin-induced pulmonary fibrosis. In addition, the effect of prior bleomycin treatment on subsequent fibrosis was examined in wild-type and B7H3-knockout mice. Samples from patients with idiopathic pulmonary fibrosis were analyzed for potential clinical relevance of the findings.Measurements and Main Results: Recipient mice transplanted with BM from bleomycin-pretreated donors showed significant exacerbation of subsequent fibrosis with increased B7H3+ cell numbers and a T-helper cell type 2-skewed phenotype. Pretreatment with a minimally fibrogenic/nonfibrogenic dose of bleomycin also caused exacerbation, but not in B7H3-deficient mice. Exacerbation was not observed if the mice received naive BM cell transplant after the initial bleomycin pretreatment. Soluble B7H3 stimulated BM Ly6Chi monocytic cell expansion in vitro and caused similar expansion in the lung in vivo. Notably, soluble B7H3 was elevated in plasma of patients with idiopathic pulmonary fibrosis and in BAL fluid in those with acute exacerbation. Finally, ST2 deficiency diminished the bleomycin-induced B7H3 and IL-13 upregulation, suggesting a role for type 2 innate lymphoid cells.Conclusions: Pulmonary fibrosis caused significant alterations in BM with expansion and activation of monocytic cells, which enhanced fibrosis when transplanted to naive recipients with potential mediation by a novel role for B7H3 in the pathophysiology of pulmonary fibrosis in both mice and humans.
Collapse
Affiliation(s)
- Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Keitaro Omori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Lin Ding
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Sem H. Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
40
|
Hu B, Yang XB, Sang XT. Liver graft rejection following immune checkpoint inhibitors treatment: a review. Med Oncol 2019; 36:94. [DOI: 10.1007/s12032-019-1316-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a variety of solid tumors; nonetheless, they have not been well investigated and are still recognized as a relative contraindication for patients with a liver transplantation (LT) history, since ICIs treatment might potentially lead to graft rejection. The program death-1 (PD-1) and the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathways are implicated in the tolerance of transplanted organ, as well as blockade of the pathways, which contribute to eliminating tumors and may inadvertently lead to peripheral transplant rejection. Currently, no guidelines are available regarding the treatment for ICIs patients with a prior LT history. Therefore, this study was carried out to review the recent studies, attempting to introduce the ICIs-related graft rejection after LT from various aspects. We believed that ICIs could be given for the well-informed patients receiving LT and developed recurrence in a controlled setting. Typically, these patients should be treated according to a clinical care path or a prospective clinical trial, so as obtain a persistent anti-tumor immune response in the meantime of avoiding graft rejection, adjust the immunosuppression, reduce the possibility of graft loss following rejection, and have the opportunity to develop biomarkers for tumor response and transplant rejection.
Collapse
|
41
|
Abstract
Immunomodulatory antibodies that directly trigger and reawaken suppressed T-cell effector function are termed 'checkpoint inhibitors'. CTLA-4 and PD-1/PD-L1 molecules are the most studied inhibitory immune check points against cancer and because of this therapeutic property have entered the clinic for treating a variety of tumor types. The results so far demonstrate a positive impact on cancer remission. Preclinical studies have demonstrated that targeting a number of other T-cell surface molecules including both positive and negative immune regulators, also possesses strong antitumor activity. Some of these molecules have already entered clinical trials. In this report, we briefly highlight the status of these immune checkpoint inhibitors and discuss their side effects and future directions for their use.
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy & Rheumatology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Byoung S Kwon
- Section of Clinical Immunology, Allergy & Rheumatology, School of Medicine, Tulane University, New Orleans, LA 70112, USA.,Eutilex Institute for Biomedical Research, Suite #1401 Daeryung Technotown 17, Gasan digital 1-ro 25, Geumcheon-gu, Seoul Korea
| |
Collapse
|
42
|
Dendritic cell-associated B7-H3 suppresses the production of autoantibodies and renal inflammation in a mouse model of systemic lupus erythematosus. Cell Death Dis 2019; 10:393. [PMID: 31113935 PMCID: PMC6529467 DOI: 10.1038/s41419-019-1623-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022]
Abstract
B7-H3 immune modulatory molecule has been implicated in the generation and pathogenesis of autoimmune diseases, the mechanism of action is less known. We explored the role of B7-H3 in the induction of autoantibodies and organ-directed inflammation in a murine systemic lupus erythematosus (SLE) model in which the immunization with DNA extracted from activated T cells induced the production of anti-DNA autoantibodies and subsequent glomerulonephritis, two hallmarks of human SLE. Mice deficient of B7-H3 or treated with a B7-H3 specific antibody produced significantly higher levels of anti-DNA autoantibodies and more severe glomerulonephritis than wild-type mice, indicating an inhibitory function of B7-H3 in this model. Interestingly, immunization of mice with DNA-pulsed dendritic cells induced severe SLE symptoms while B7-H3 on dendritic cells is required in this process. Importantly, treatment of mice with recombinant B7-H3Ig fusion protein effectively ameliorated progression of murine SLE, accompanied with decreased level of anti-DNA autoantibodies and alleviated glomerulonephritis, decreased autoantibody deposition and complement deposition in kidney. Our findings implicate a potential role of B7-H3 on dendritic cells in the induction of SLE and as a potential target for the treatment of autoimmune diseases.
Collapse
|
43
|
Tumor mechanisms of resistance to immune attack. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:61-100. [PMID: 31383409 DOI: 10.1016/bs.pmbts.2019.03.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immune system plays a key role in the interactions between host and tumor. Immune selection pressure is a driving force behind the sculpting and evolution of malignant cancer cells to escape this immune attack. Several common tumor cell-based mechanisms of resistance to immune attack have been identified and can be broadly categorized into three main classes: loss of antigenicity, loss of immunogenicity, and creation of an immunosuppressive microenvironment. In this review, we will discuss in detail the relevant literature associated with each class of resistance and will describe the relevance of these mechanisms to human cancer patients. To conclude, we will outline the implications these mechanisms have for the treatment of cancer using currently available therapeutic approaches. Immunotherapy has been a successful addition to current treatment approaches, but many patients either do not respond or quickly become resistant. This reflects the ability of tumors to continue to adapt to immune selection pressure at all stages of development. Additional study of immune escape mechanisms and immunotherapy resistance mechanisms will be needed to inform future treatment approaches.
Collapse
|
44
|
Co-signal Molecules in T-Cell Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:3-23. [DOI: 10.1007/978-981-32-9717-3_1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
45
|
Altan M, Kidwell KM, Pelekanou V, Carvajal-Hausdorf DE, Schalper KA, Toki MI, Thomas DG, Sabel MS, Hayes DF, Rimm DL. Association of B7-H4, PD-L1, and tumor infiltrating lymphocytes with outcomes in breast cancer. NPJ Breast Cancer 2018; 4:40. [PMID: 30564631 PMCID: PMC6288133 DOI: 10.1038/s41523-018-0095-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 11/13/2018] [Indexed: 02/07/2023] Open
Abstract
B7-H4 (VTCN1) is a member of the CD28/B7 family of immune co-inhibitory molecules. The relationship of tumor and stromal B7-H4 protein expression with PD-L1, tumor infiltrating lymphocytes (TILs) and its association with clinico-pathological variables are not well defined. Herein, we explore the expression level of B7-H4 protein in breast cancer and evaluate its association with TILs, levels of PD-L1 expression, and clinico-pathological characteristics in two independent populations. In this study, we used multiplexed automated quantitative immunofluorescence (QIF) to measure the levels of B7-H4 and PD-L1 protein and determined TILs through pathologist assessment of H&E-stained preparations in over a thousand breast cancer cases from two institutions represented in tissue microarray format. Associations between the marker levels, major clinico-pathological variables, and survival were analyzed. We detected B7-H4 protein was highly expressed in both breast cancer and stromal cells. Its expression was independent of breast cancer intrinsic subtypes. PD-L1 expression was higher in triple negative breast cancers. Neither B7-H4 nor PD-L1 were associated with survival in breast cancer. Our study shows there is a mutually exclusive pattern of B7-H4 with both tumor PD-L1 expression and TILs in all breast cancers, independent of breast cancer intrinsic subtype. This exclusive pattern suggests that some breast tumors may preferentially use one B7-related immune evasion mechanism/pathway. This could explain the clinical benefit that is seen only in a fraction of patients with immune checkpoint inhibitors directed exclusively towards PD-L1 in breast cancer.
Collapse
Affiliation(s)
- Mehmet Altan
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Thoracic/Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | - Kelley M. Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI USA
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | | | - Daniel E. Carvajal-Hausdorf
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
- Anatomic Pathology, Clinica Alemana-Facultad de Medicina Universidad de Desarrollo, Vitacura, Santiago Chile
| | - Kurt A. Schalper
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Maria I. Toki
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| | - Dafydd G. Thomas
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Michael S. Sabel
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - Daniel F. Hayes
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI USA
| | - David L. Rimm
- Section of Medical Oncology, Yale School of Medicine, New Haven, CT USA
- Department of Pathology, Yale School of Medicine, New Haven, CT USA
| |
Collapse
|
46
|
Genomic Characteristics of Invasive Mucinous Adenocarcinomas of the Lung and Potential Therapeutic Targets of B7-H3. Cancers (Basel) 2018; 10:cancers10120478. [PMID: 30513627 PMCID: PMC6316015 DOI: 10.3390/cancers10120478] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/27/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022] Open
Abstract
Pulmonary invasive mucinous adenocarcinoma (IMA) is considered a variant of lung adenocarcinomas based on the current World Health Organization classification of lung tumors. However, the molecular mechanism driving IMA development and progression is not well understood. Thus, we surveyed the genomic characteristics of IMA in association with immune-checkpoint expression to investigate new potential therapeutic strategies. Tumor cells were collected from surgical specimens of primary IMA, and sequenced to survey 53 genes associated with lung cancer. The mutational profiles thus obtained were compared in silico to conventional adenocarcinomas and other histologic carcinomas, thereby establishing the genomic clustering of lung cancers. Immunostaining was also performed to compare expression of programmed death ligand 1 (PD-L1) and B7-H3 in IMA and conventional adenocarcinomas. Mutations in Kirsten rat sarcoma viral oncogene homolog (KRAS) were detected in 75% of IMAs, but in only 11.6% of conventional adenocarcinomas. On the other hand, the frequency of mutations in epidermal growth factor receptor (EGFR) and tumor protein p53 (TP53) genes was 5% and 10%, respectively, in the former, but 48.8% and 34.9%, respectively, in the latter. Clustering of all 78 lung cancers indicated that IMA is distinct from conventional adenocarcinoma or squamous cell carcinoma. Strikingly, expression of PD-L1 in ≥1% of cells was observed in only 6.1% of IMAs, but in 59.7% of conventional adenocarcinomas. Finally, 42.4% and 19.4% of IMAs and conventional adenocarcinomas, respectively, tested positive for B7-H3. Although currently classified as a variant of lung adenocarcinoma, it is also reasonable to consider IMA as fundamentally distinct, based on mutation profiles and genetic clustering as well as immune-checkpoint status. The immunohistochemistry data suggest that B7-H3 may be a new and promising therapeutic target for immune checkpoint therapy.
Collapse
|
47
|
Ji H, Guo J, Yang Y, Xu C, Mao W. Construction, expression and functional analysis of anti-B7-H4- scFv-CH3 recombinant antibody. J Biosci 2018; 43:661-671. [PMID: 30207312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The B7-H4 molecule, a unique negative regulator of T lymphocytes which is overexpressed on the surface of various tumor cells, is a particularly important target candidate for tumor therapy because it can be blocked with anti-B7-H4 antibodies to inhibit the B7-H4 signaling pathway. Our previous work established an anti-B7-H4 single-chain variable fragment (scFv) library, so we have now amplified the genes encoding anti-B7-H4-scFv and human IgG1 CH3 and ligated them by overlap extension PCR to obtain a recombinant gene. After sequencing, the gene was cloned into the expression vector pET43.1a and expression was induced in E. coli BL21 (DE3) by isopropyl-β-D-1-thiogalactopyranoside (IPTG). The protein was purified on a nickel-nitrilotriacetic acid (Ni-NTA) resin column and its antigen specificity and affinity were examined by ELISA and western blotting. We also established a Lewis lung cancer model in C57BL/6 mice to further identify the biological function of the scFv protein in vivo. The results showed that tumor volume, body weight and necrotic tissues in the control group were significantly greater than in the experimental group, indicating that selected scFvs had good biological activity and could inhibit tumor growth in tumor-bearing mice. Our work thus offers a new approach for the development of cancer-targeted therapy.
Collapse
Affiliation(s)
- Hongshuai Ji
- Jiangsu Province Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210046, People's Republic of China
| | | | | | | | | |
Collapse
|
48
|
Azuma T, Sato Y, Ohno T, Azuma M, Kume H. Serum soluble B7-H4 is a prognostic marker for patients with non-metastatic clear cell renal cell carcinoma. PLoS One 2018; 13:e0199719. [PMID: 30044793 PMCID: PMC6059393 DOI: 10.1371/journal.pone.0199719] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 06/12/2018] [Indexed: 01/24/2023] Open
Abstract
Background B7-H4 is a member of the B7 family of immune-regulatory ligands and is considered to be a negative regulator of the immune response. We investigated the clinical significance of serum soluble B7-H4 in patients with non-metastatic clear cell renal cell carcinoma. Methods We analyzed 108 patients in whom non-metastatic clear cell renal cancer was diagnosed at Tokyo Metropolitan Tama Medical Center between 2008 and 2013. We measured the serum soluble B7-H4 level using the Enzyme-Linked ImmunoSorbent Assay (ELISA) and evaluated the association between the peripheral blood neutrophil count and sB7-H4 as well as the utility of soluble B7-H4 as a prognostic biomarker for clear cell renal cancer. The Cox proportional hazards regression model was used to assess the PFS and OS with the soluble B7-H4 level. Results We detected high levels of soluble B7-H4 in the sera of 56% of patients with non-metastatic clear cell renal cell carcinoma versus only 10% of healthy donors. Elevated soluble B7-H4 levels were associated with changes in an elevated peripheral blood neutrophil count. The increase of soluble B7-H4 also was significantly associated with poor PFS and OS. Multivariate analysis showed that the elevation of the soluble B7-H4 level was an independent prognostic factor for PFS and OS. Conclusions Our data suggest that the association between serum soluble B7-H4 and peripheral blood neutrophil count, as well as the evaluation of serum soluble B7-H4 expression is a useful tool for predicting the prognosis of patients with non-metastatic clear cell renal cell carcinoma.
Collapse
Affiliation(s)
- Takeshi Azuma
- Department of Urology, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Urology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- * E-mail:
| | - Yujiro Sato
- Department of Urology, Tokyo Metropolitan Tama Medical Center, Tokyo, Japan
| | - Tetsukuni Ohno
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Haruki Kume
- Department of Urology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Wang L, Yang C, Liu XB, Wang L, Kang FB. B7-H4 overexpression contributes to poor prognosis and drug-resistance in triple-negative breast cancer. Cancer Cell Int 2018; 18:100. [PMID: 30008617 PMCID: PMC6044050 DOI: 10.1186/s12935-018-0597-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 07/06/2018] [Indexed: 11/20/2022] Open
Abstract
Background The expression of the immunoregulatory protein B7-H4 has been reported in many types of cancer, including breast cancer. However, its role in triple-negative breast cancer (TNBC), especially its correlation with patients’ prognosis and chemoresistance remains unclear. Methods The expression of B7-H4 in TNBC tissues and cell lines were measured with Real-Time PCR and western blotting. 65 cases of TNBC tissue samples and adjacent non-tumor tissue samples were analyzed by immunochemistry to demonstrate the correlation between the B7-H4 expression and clinicopathological characteristics. In vitro studies assessed mAb MIH43 alone and in combination with transfecting B7-H4 siRNA on the growth of chemosensitive and chemoresistant TNBC cell lines by CCK-8 and apoptotic enzyme-linked immunosorbent assay (ELISA). Results B7-H4 expression was detected positive in 59 of 65 (90.8%) different stage TNBC patients, especially in the samples of recurrence TNBC patients after receiving neoadjuvant chemotherapy treatment. Survival curves showed that patients with B7-H4 overexpression had significantly shorter survival and recurrence time than those with low B7-H4 expression (p < 0.005). Univariate and multivariate COX regression analysis demonstrated that B7-H4 was an independent predictor for advanced tumor stage. The monoclonal antibody of B7-H4 has the potential anti-proliferative effects on inhibiting the chemoresistant TNBC cell lines and increasing the sensitivity of TNBC cell lines to doxorubicin, paclitaxel or carboplatin. RNAi-mediated silencing of B7-H4 in TNBC cells enhanced drug-induced apoptosis via inhibiting PTEN/PI3K/AKT pathway, whereas reexpression of B7-H4 in B7-H4 knockdown and low B7-H4 expressing cells increased the phosphorylation of PI3K and AKT along with restoration of PETN expression. Conclusions Our data show that B7-H4 is a biomarker indicative of a poor prognosis in TNBC patients and at least partially downregulated in chemoresistance via PTEN/PI3K/AKT pathway. Targeting B7-H4 might provide an attractive therapeutic approach specifically for TNBC patients. Electronic supplementary material The online version of this article (10.1186/s12935-018-0597-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ling Wang
- 1Department of Orthopedic Oncology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei People's Republic of China
| | - Chao Yang
- 2Department of General Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei People's Republic of China
| | - Xin-Bo Liu
- 3Department of Thoracic Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei People's Republic of China
| | - Li Wang
- Department of Pathology, the Fourth Hospital of Shijiazhuang, Shijiazhuang, China
| | - Fu-Biao Kang
- 5Department of Liver Diseases, Bethune International Peace Hospital, Shijiazhuang, Hebei People's Republic of China
| |
Collapse
|
50
|
Zhao Q, Hu F, Xiao Z, Li M, Wu X, Zhao Y, Wu Y, Yin J, Lin L, Zhang H, Zhang L, Cho CH, Shen J. Comprehensive molecular profiling of the B7 family in gastrointestinal cancer. Cell Prolif 2018; 51:e12468. [PMID: 29999557 DOI: 10.1111/cpr.12468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 04/13/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES B7 family has been identified as co-stimulatory or co-inhibitory molecules on T-cell response and plays an important role in tumour mortality and malignancy. In this study, the expression pattern of B7 family in gastrointestinal (GI) cancer was examined. Its upstream regulating mechanism, downstream targets and association with clinical parameters were also studied. MATERIALS AND METHODS The expression level of B7 members was analysed by FIREHOUSE. The gene mutation, DNA methylation, association with clinical parameters and downstream network of B7 members were analysed in cBioportal. The mutation frequency was analysed by Catalogue of Somatic Mutations in Cancer (COSMIC) analysis. The phylogenetic tree was constructed in MEGA7. The interaction protein domain analysis was performed by Pfam 31.0. RESULTS Differential expression of B7 family molecules was detected in different kinds of GI cancer. High-frequency gene alteration was found in tumour samples. There was negative correlation of promoter methylation and mRNA expression of B7 family members in tumour samples, suggesting the epigenetic basis of B7 family gene deregulation in GI cancer. The overexpression of B7-H1 in pancreatic cancer, B7-H5 in oesophageal cancer and B7-H6 in liver cancer were significantly associated with worse overall survival. Finally, by network analysis, we identified some potential interacting proteins for B7-1/2 and B7-H1/DC. CONCLUSIONS Overall, our study suggested that B7 member deregulation was strongly involved in GI cancer tumorigenesis.
Collapse
Affiliation(s)
- Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Fuyan Hu
- Department of Statistics, Faculty of Science, Wuhan University of Technology, Wuhan, Hubei, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuanlin Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianhua Yin
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Lin
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Hanyu Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Lingling Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|