1
|
He J, Wang C, Fang X, Li J, Shen X, Zhang J, Peng C, Li H, Li S, Karp JM, Kuai R. Tuning the fluidity and protein corona of ultrasound-responsive liposomal nanovaccines to program T cell immunity in mice. Nat Commun 2024; 15:8121. [PMID: 39284814 PMCID: PMC11405680 DOI: 10.1038/s41467-024-52104-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 08/27/2024] [Indexed: 09/22/2024] Open
Abstract
Inducing high levels of antigen-specific CD8α+ T cells in the tumor is beneficial for cancer immunotherapy, but achieving this in a safe and effective manner remains challenging. Here, we have developed a designer liposomal nanovaccine containing a sonosensitizer (LNVS) to efficiently program T cell immunity in mice. Following intravenous injection, LNVS accumulates in the spleen in a protein corona and fluidity-dependent manner, leading to greater frequencies of antigen-specific CD8α+ T cells than soluble vaccines (the mixture of antigens and adjuvants). Meanwhile, some LNVS passively accumulates in the tumor, where it responds to ultrasound (US) to increase the levels of chemokines and adhesion molecules that are beneficial for recruiting CD8α+ T cells to the tumor. LNVS + US induces higher levels of intratumoral antitumor T cells than traditional sonodynamic therapy, regresses established mouse MC38 tumors and orthotopic cervical cancer, and protects cured mice from relapse. Our platform sheds light on the importance of tuning the fluidity and protein corona of naovaccines to program T cell immunity in mice and may inspire new strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xiao Fang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Junyao Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Xueying Shen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Junxia Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China
| | - Cheng Peng
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China
| | - Hongjian Li
- School of Medicine, Tsinghua University, Beijing, China
| | - Sai Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China
| | - Jeffrey M Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
2
|
Meyiah A, Elkord E. What is the relevance of FoxP3 in the tumor microenvironment and cancer outcomes? Expert Rev Clin Immunol 2024; 20:803-809. [PMID: 38512803 DOI: 10.1080/1744666x.2024.2334258] [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: 01/13/2024] [Accepted: 03/20/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Forkhead box P3 (FoxP3) transcription factor plays critical roles in controlling immune responses and cancer progression in different cancers. FoxP3 expression within the tumor microenvironment (TME) may influence clinical outcomes negatively or positively, and it could play dual roles in cancer, either by promoting or inhibiting tumor development and progression. Some studies reported that high levels of FoxP3 could be associated with tumor progression and worse prognosis, while others reported contradictory results. AREAS COVERED In this special report, we present a brief account on the role and function of FoxP3 in the TME, and its contribution to the clinical outcomes of cancer patients. Importantly, we give insights on the potential factors that could contribute to different clinical outcomes in cancer patients. EXPERT OPINION Different studies showed that FoxP3 expression can be associated with bad prognoses in cancer patients. However, FoxP3 could have opposing roles by enhancing cancer progression or regression. Location and expression of FoxP3 in T cells or tumor cells can have different impacts on cancer prognoses. Different factors should be considered to establish FoxP3 as a more robust prognostic biomarker and a potential therapeutic target for enhancing anti-tumor immunity and improving clinical outcomes of cancer patients.
Collapse
Affiliation(s)
- Abdo Meyiah
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Eyad Elkord
- Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University, Suzhou, China
- Biomedical Research Center, School of Science, Engineering and Environment, University of Salford, Manchester, UK
| |
Collapse
|
3
|
Luan J, Liu Y, Cao M, Guo X, Guo N. The pathogenic response of cytotoxic T‑lymphocytes, a common therapeutic target for cancer, has a direct impact on treatment outcomes (Review). Oncol Rep 2024; 52:98. [PMID: 38904200 PMCID: PMC11200153 DOI: 10.3892/or.2024.8757] [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: 02/23/2024] [Accepted: 05/15/2024] [Indexed: 06/22/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs), also known as CD8+ T cells, participate in immune function by secreting various cytokines after recognizing specific antigens and class I major histocompatibility complex molecules associated with tumor cells, and thus have a key role in antitumor immunity. However, certain CD8+ T cells show low reactivity and thus cannot effectively remove tumor cells or viral antigens. Due to this heterogeneity, effective biomarkers representing these differences in CD8+ cells are needed. The identification of suitable biomarkers will also enhance the management of cancer treatment. Recent research has improved the understanding of CD8+ T lymphocytes in the tumor microenvironment and circulatory system. Treatment efficacy is impacted directly by the pathogenic response of CTLs, and thus, the use of adjuvant therapies to address these pathological changes, e.g., stimulating the increase in the proportion of reactive T cells or suppressing the proportion of terminally exhausted T cells, would be advantageous.
Collapse
Affiliation(s)
- Jing Luan
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Yuxin Liu
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Meng Cao
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Xianing Guo
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| | - Na Guo
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, P.R. China
| |
Collapse
|
4
|
Febriani V, Indrawati I, Sumpono ASB, Ferronika P, Bawono RG, Dwianingsih EK. Tumor Infiltrating Lymphocytes as an Independent Prognostic Factor in Undifferentiated Nasopharyngeal Carcinoma. Asian Pac J Cancer Prev 2024; 25:1997-2002. [PMID: 38918661 PMCID: PMC11382859 DOI: 10.31557/apjcp.2024.25.6.1997] [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: 11/17/2023] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a common type of cancer in Southeast Asia. This cancer usually spreads locally and to nearby lymph nodes. One unique feature of NPC is its many immune cells called tumor-infiltrating lymphocytes (TILs). Recent studies have suggested that TILs in many types of cancer can indicate a better prognosis. However, the role of TILs in NPC is still a matter of debate. Further research is necessary to determine whether TILs can be used as a prognostic factor of NPC's outcome. METHOD A retrospective cohort study was conducted at Sardjito Hospital to examine the records and pathological sections of patients treated for the undifferentiated subtype of NPC. Two pathologists analyzed the presence of TILs using HE-stained slides. TILs were evaluated in stromal compartments, and their association with clinicopathological variables was analyzed using the Chi-square and Fisher exact tests. The study compared overall survival in tumor patients with varying TIL levels using Kaplan-Meier survival curves and the log-rank test. A Cox regression model was used for univariate and multivariate analyses to test the significance of different factors. RESULT Out of the total 61 subjects, 16 (26.2%) had high stromal TILs (≥ 70%), and 45 (73.8%) had low stromal TILs (<70%). The subjects' sex, age, and tumor stage did not affect the OS. However, high stromal TILs (≥ 70%) showed a significant association with a longer OS (log-rank test p = 0.006, HR 0.37, 95% CI 0.17-0.79, log-rank p = 0.006). Moreover, multivariate analysis confirmed that TILs were an independent prognostic indicator for OS (aHR 0.015). CONCLUSION TILs correlate positively with overall survival in the undifferentiated NPC subtype and are an independent prognostic indicator.
Collapse
Affiliation(s)
- Vivin Febriani
- Departement of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Indrawati Indrawati
- Departement of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Auliya Suluk Brilliant Sumpono
- Departement of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Paranita Ferronika
- Departement of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Rheza Gandi Bawono
- Departement of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Ery Kus Dwianingsih
- Departement of Anatomical Pathology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| |
Collapse
|
5
|
Ma Y, Zhou H, Luo F, Zhang Y, Zhu C, Li W, Huang Z, Zhao J, Xue J, Zhao Y, Fang W, Yang Y, Huang Y, Zhang L, Zhao H. Remodeling the tumor-immune microenvironment by anti-CTLA4 blockade enhanced subsequent anti-PD-1 efficacy in advanced nasopharyngeal carcinoma. NPJ Precis Oncol 2024; 8:65. [PMID: 38448521 PMCID: PMC10917783 DOI: 10.1038/s41698-024-00558-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Sequential immunotherapy has shown certain advantages in malignancy. Here, we aim to evaluate the efficacy of sequential anti-CTLA-4 and anti-PD-1 treatment for recurrent or metastatic nasopharyngeal carcinoma patients (R/M NPC). We retrospectively analysis 2 phase I trial of ipilimumab and camrelizumab in Chinese R/M NPC patients. These patients were initially treated with ipilimumab, a CTLA4 blockade, followed by anti-PD-1 treatment. We observed a durable tumor remission in these patients (mPFS: 12.3 months; mDoR: 20.9 months). Multimodal investigations of biopsy samples disclosed remodeling of tumor-immune microenvironment triggered by ipilimumab. In responders, we found increased tumoral PD-L1/PD-L2 expression and T-cell infiltration after ipilimumab treatment, accompanied by reduced stroma and malignant cell components. In contrast, non-responders exhibited increased B-cell infiltration and increased peripheral CD19 + B cells, suggesting a defective transition from memory B cells to plasma cells. This study proposes that sequential therapy can potentially enhance treatment efficacy in chemotherapy-resistant NPC patients and provides insights into how preexisting anti-CTLA4 blockade can influence subsequent anti-PD-1 efficacy by remodeling the TME. Additionally, our results highlight the need for therapeutic strategies targeting naïve/memory B cells.
Collapse
Affiliation(s)
- Yuxiang Ma
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Huaqiang Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Fan Luo
- Intensive Care Unit Department, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yang Zhang
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Changbin Zhu
- Department of Translational Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Weiwei Li
- Department of Translational Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Zhan Huang
- Department of Translational Medicine, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Jingbo Zhao
- Department of Research and Development, Amoy Diagnostics Co., Ltd., Xiamen, China
| | - Jinhui Xue
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yuanyuan Zhao
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yunpeng Yang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Yan Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China
| | - Li Zhang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
| | - Hongyun Zhao
- Department of Clinical Research, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, China.
| |
Collapse
|
6
|
Suryani L, Lee HPY, Teo WK, Chin ZK, Loh KS, Tay JK. Precision Medicine for Nasopharyngeal Cancer-A Review of Current Prognostic Strategies. Cancers (Basel) 2024; 16:918. [PMID: 38473280 DOI: 10.3390/cancers16050918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/02/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV) driven malignancy arising from the nasopharyngeal epithelium. Current treatment strategies depend on the clinical stage of the disease, including the extent of the primary tumour, the extent of nodal disease, and the presence of distant metastasis. With the close association of EBV infection with NPC development, EBV biomarkers have shown promise in predicting treatment outcomes. Among the omic technologies, RNA and miRNA signatures have been widely studied, showing promising results in the research setting to predict treatment response. The transformation of radiology images into measurable features has facilitated the use of radiomics to generate predictive models for better prognostication and treatment selection. Nonetheless, much of this work remains in the research realm, and challenges remain in clinical implementation.
Collapse
Affiliation(s)
- Luvita Suryani
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Hazel P Y Lee
- Department of Otolaryngology-Head & Neck Surgery, National University Hospital, Singapore 119228, Singapore
| | - Wei Keat Teo
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Zhi Kang Chin
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Kwok Seng Loh
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | - Joshua K Tay
- Department of Otolaryngology-Head & Neck Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| |
Collapse
|
7
|
Qayoom H, Sofi S, Mir MA. Targeting tumor microenvironment using tumor-infiltrating lymphocytes as therapeutics against tumorigenesis. Immunol Res 2023; 71:588-599. [PMID: 37004645 DOI: 10.1007/s12026-023-09376-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 03/25/2023] [Indexed: 04/04/2023]
Abstract
The immune system plays a vital role in suppressing tumor cell progression. The tumor microenvironment augmented with significant levels of tumor-infiltrating lymphocytes has been widely investigated and it is suggested that tumor-infiltrating lymphocytes have shown a significant role in the prognosis of cancer patients. Compared to ordinary non-infiltrating lymphocytes, tumor-infiltrating lymphocytes (TILs) are a significant population of lymphocytes that infiltrate tumor tissue and have a higher level of specific immunological reactivity against tumor cells. They serve as an effective immunological defense against various malignancies. TILs are a diverse group of immune cells that are divided into immune subsets based on the pathological and physiological impact they have on the immune system. TILs mainly consist of B-cells, T-cells, or natural killer cells with diverse phenotypic and functional properties. TILs are known to be superior to other immune cells in that they can recognize a wide range of heterogeneous tumor antigens by producing many clones of T cell receptors (TCRs), outperforming treatments like TCR-T cell and CAR-T therapy. With the introduction of genetic engineering technologies, tumor-infiltrating lymphocytes (TILs) have become a ground-breaking therapeutic option for malignancies, but because of the hindrances opposed by the immune microenvironment and the mutation of antigens, the development of TILs as therapeutic has been hindered. By giving some insight into the many variables, such as the various barriers inhibiting its usage as a potential therapeutic agent, we have examined various aspects of TILs in this work.
Collapse
Affiliation(s)
- Hina Qayoom
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India
| | - Shazia Sofi
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India
| | - Manzoor A Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, 190006, Jammu and Kashmir, India.
| |
Collapse
|
8
|
Yi C, Wei W, Wan M, Chen Y, Zhang B, Wu W. Expression Patterns of HOX Gene Family Defines Tumor Microenvironment and Immunotherapy in Hepatocellular Carcinoma. Appl Biochem Biotechnol 2023; 195:5072-5093. [PMID: 36976502 DOI: 10.1007/s12010-023-04443-8] [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: 03/15/2023] [Indexed: 03/29/2023]
Abstract
Hepatocellular carcinoma (HCC) pathophysiology is prevalently related with HOX genes. However, the study on associations of extensive HOX genes with tumor microenvironment and drug sensitivity of HCC remains scarce. The data sets of HCC were downloaded from TCGA, ICGC, and GEO by bioinformatics method and analyzed. Based on a computational frame, HCC samples were divided into a high and a low HOXscore group, and significantly shorter survival time in the high HOXscore was observed relative to low HOXscore group using survival analysis. Gene set enrichment analysis (GSEA) revealed that the high HOXscore group was more likely to be enriched in cancer-specific pathways. Furthermore, the high HOXscore group was involved in the infiltration of inhibitory immune cells. In response to anti-cancer drugs, the high HOXscore group was more sensitive to mitomycin and cisplatin. Importantly, the HOXscore was associated with the therapeutic efficacy of PD-L1 blockade, suggesting that the development of potential drugs targeting these HOX genes to aid the clinical benefits of immunotherapy is needed. In addition, RT-qPCR and immunohistochemistry showed 10 HOX genes mRNA expression was higher in HCC compared to the normal tissues. This study provides a comprehensive analysis of HOX genes family in HCC and revealed the potential function of these HOX genes family in tumor microenvironment (TME) and identified their therapeutic liability in targeted therapy and immunotherapy. Eventually, this work highlights the cross-talk and potential clinical utility of HOX genes family in HCC therapy.
Collapse
Affiliation(s)
- Changhong Yi
- Department of Interventional Radiology, Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Wei Wei
- Department of Interventional, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Maolin Wan
- Department of Interventional, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Ya Chen
- Department of Interventional, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Bo Zhang
- Department of Interventional, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Wenze Wu
- Department of Interventional, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China.
| |
Collapse
|
9
|
Alilou M, Khorrami M, Prasanna P, Bera K, Gupta A, Viswanathan VS, Patil P, Velu PD, Fu P, Velcheti V, Madabhushi A. A tumor vasculature-based imaging biomarker for predicting response and survival in patients with lung cancer treated with checkpoint inhibitors. SCIENCE ADVANCES 2022; 8:eabq4609. [PMID: 36427313 PMCID: PMC9699671 DOI: 10.1126/sciadv.abq4609] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 10/06/2022] [Indexed: 05/30/2023]
Abstract
Tumor vasculature is a key component of the tumor microenvironment that can influence tumor behavior and therapeutic resistance. We present a new imaging biomarker, quantitative vessel tortuosity (QVT), and evaluate its association with response and survival in patients with non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitor (ICI) therapies. A total of 507 cases were used to evaluate different aspects of the QVT biomarkers. QVT features were extracted from computed tomography imaging of patients before and after ICI therapy to capture the tortuosity, curvature, density, and branching statistics of the nodule vasculature. Our results showed that QVT features were prognostic of OS (HR = 3.14, 0.95% CI = 1.2 to 9.68, P = 0.0006, C-index = 0.61) and could predict ICI response with AUCs of 0.66, 0.61, and 0.67 on three validation sets. Our study shows that QVT imaging biomarker could potentially aid in predicting and monitoring response to ICI in patients with NSCLC.
Collapse
Affiliation(s)
- Mehdi Alilou
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY 11790, USA
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Amit Gupta
- University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Pradnya Patil
- Department of Solid Tumor Oncology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Priya Darsini Velu
- Pathology and Laboratory Medicine, Weill Cornell Medicine Physicians, New York, NY 10021, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, CWRU, Cleveland, OH 44106, USA
| | - Vamsidhar Velcheti
- Department of Hematology and Oncology, NYU Langone Health, New York, NY 10016, USA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, USA
- Atlanta Veterans Administration Medical Center, Atlanta, GA 30322, USA
| |
Collapse
|
10
|
Liu Z, He J, Hu X. Ferroptosis regulators related scoring system by Gaussian finite mixture model to predict prognosis and immunotherapy efficacy in nasopharyngeal carcinoma. Front Genet 2022; 13:975190. [PMID: 36118882 PMCID: PMC9479336 DOI: 10.3389/fgene.2022.975190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
The role of ferroptosis in tumor progression and metastasis has been demonstrated. Nonetheless, potential biological function of ferroptosis regulatory pattern in nasopharyngeal carcinoma (NPC) remains unknown. Ferroptosis regulatory patterns of nasopharyngeal carcinoma samples were evaluated based on 113 ferroptosis regulators and three distinct ferroptosis subtypes were determined by unsupervised clustering. The ferroptosis score (FEP score) was identified to quantify ferroptosis patterns within individual tumors by Gaussian finite mixture model and systematically correlated with representative tumor characteristics. Subtype 1 and subtype 3 were consistent with immune activated phenotype, while subtype 2 was consistent with immune suppressed phenotype. High ferroptosis score, characterized by immune activation and suppression of mRNA based stemness index (mRNAsi) and Epstein-Barr virus (EBV) genes, indicated an immune activated tumor microenvironment (TME) phenotype, with better progression free survival (PFS) and lower risk of recurrence and metastasis. Low ferroptosis score, characterized by activation of Wnt and NF-κB signaling pathways and lack of effective immune infiltration, indicated an immune suppressed tumor microenvironment phenotype and poorer survival. High ferroptosis score was also correlated to enhanced response to immunotherapy, and was confirmed to correlate with therapeutic advantages and clinical benefits in an anti-programmed cell death 1 ligand 1 (PD-L1) immunotherapy cohort. As ferroptosis played a crucial role in the tumor microenvironment’s diversity, assessing the ferroptosis pattern within individual tumor with ferroptosis score could enhance our understanding of tumor microenvironment infiltration characterization and help develop more effective immunotherapy.
Collapse
Affiliation(s)
- Zijian Liu
- Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jinlan He
- Department of Head and Neck Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaolin Hu
- West China School of Nursing, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xiaolin Hu,
| |
Collapse
|
11
|
Rachmadi L, Susanto YDB, Manatar AF, Sitinjak D. Factors Associated with Dysplastic Changes in Sinonasal Inverted Papilloma: Study of Tumor Infiltrating Lymphocytes (TILs) FOXP3, CD4, CD8, and expression of p53. Asian Pac J Cancer Prev 2022; 23:3223-3227. [PMID: 36172688 PMCID: PMC9810298 DOI: 10.31557/apjcp.2022.23.9.3223] [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: 04/16/2022] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE This study examine FOXP3, CD4, CD8 and p53 expression in the transformation of the Sinonasal Inverted Papilloma (SIP) malignancy into sinonasal carcinoma. MATERIALS AND METHODS This study used a cross-sectional approach. The research sample from thirty-six paraffin block preparations with the diagnosis of SIP. Then, immunohistochemical staining was performed using FOXP3 mouse monoclonal antibody (236A/E7), CD8 rabbit monoclonal antibody (CD8/1179R), CD4 mouse monoclonal antibody (4B12) and p53 rabbit monoclonal antibody. Results: There was a significant difference between Foxp3 expression in SIP without dysplasia and SIP with dysplasia (p= 0.013). There was no significant difference between the expression of CD4 and CD8 in the two groups with p-values 0.1 and 0.062, respectively. The mean percentage of positive p53 expression in SIP without dysplasia was 0.45+0.63 and in the SIP with dysplasia 29.31+38.96. There was a significant difference between the two groups (p<0.001). CONCLUSION FOXP3 and p53 were overexpressed in SIP with malignant transformation. FOXP3 together with p53 status is associated with dysplastic changed in the SIP. FOXP3 and p53 status could be potential biomarker of malignant transformation in sinonasal inverted papilloma.
Collapse
|
12
|
Gondhowiardjo SA, Adham M, Rachmadi L, Atmakusuma TD, Tobing DL, Auzan M, Hariyanto AD, Sulaeman D, Permata TBM, Handoko. Immune cells markers within local tumor microenvironment are associated with EBV oncoprotein in nasopharyngeal cancer. BMC Cancer 2022; 22:887. [PMID: 35963999 PMCID: PMC9375267 DOI: 10.1186/s12885-022-09948-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/29/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction EBV infection in nasopharyngeal cancer ensued in latent infection mode. In this latent infection various EBV oncoproteins such as EBNA1 and LMP1 was expressed. EBV oncoproteins could theoretically recruit immune cells, which might help to control cancer. Therefore, this study was aimed to elucidate the association with EBV oncoproteins (EBNA1 and LMP1), immune markers (CD4, CD8, and FOXP3) from nasopharyngeal cancer microenvironment with tumor progression. Method Nasopharyngeal biopsy was obtained from patients suspected to have nasopharyngeal cancer. Those samples with microscopically confirmed nasopharyngeal cancer were tested for EBNA1, LMP1, CD4, CD8, and FOXP3 concentration with ELISA, then verified with IHC. Each patient tumor volume was assessed for primary nasopharyngeal tumor volume (GTVp) and neck nodal metastases tumor volume (GTVn). Correlation test with Spearman correlation and scatterplot were carried out. Result Total 23 samples with nasopharyngeal cancer were analyzed. There was moderate correlation (ρ = 0.45; p value = 0.032) between LMP1 and GTVp. There was strong correlation (ρ = 0.81; p value < 0.001) between CD8 and GTVp. There was also moderate correlation (ρ = 0.6; p value = 0.002) between FOXP3 and GTVp. The CD8 concentration has moderate correlation with both EBNA1 (ρ = 0.46; p value = 0.026) and LMP1 (ρ = 0.47; p value = 0.023). While FOXP3 has moderate correlation with only LMP1 (ρ = 0.58; p value = 0.004). No correlation was found between all the markers tested here with GTVn. Discussion We found larger primary nasopharyngeal tumor was associated with higher CD8 marker. This was thought due to the presence of abundance CD8 T cells in the nasopharynx, but those abundance CD8 T cells were suspected to be dysfunctional. The nasopharyngeal cancer was also known to upregulate chemokines that could recruit T regulatory FOXP3 cells. Furthermore, T regulatory FOXP3 cells differentiation was induced through several pathways which was triggered by EBNA1. The correlation found in this study could guide further study to understand nasopharyngeal carcinogenesis and the relationship with our immune system.
Collapse
Affiliation(s)
- Soehartati A Gondhowiardjo
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Marlinda Adham
- Department of ENT, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Lisnawati Rachmadi
- Department of Anatomical Pathology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Tubagus Djumhana Atmakusuma
- Department of Medical Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Demak Lumban Tobing
- Department of Clinical Pathology, Dharmais National Cancer Hospital, Jakarta - Indonesia, Jl. Letjen S. Parman No. 84-86, Jakarta, Indonesia, 11420
| | - Mahesa Auzan
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Agustinus Darmadi Hariyanto
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Dede Sulaeman
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Tiara Bunga Mayang Permata
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430
| | - Handoko
- Department of Radiation Oncology, Faculty of Medicine, Universitas Indonesia / Cipto Mangunkusumo National General Hospital, Jakarta - Indonesia, Jl. Salemba Raya No. 6, Jakarta, Indonesia, 10430.
| |
Collapse
|
13
|
Deng K, Yang D, Zhou Y. Nanotechnology-Based siRNA Delivery Systems to Overcome Tumor Immune Evasion in Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14071344. [PMID: 35890239 PMCID: PMC9315482 DOI: 10.3390/pharmaceutics14071344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 12/31/2022] Open
Abstract
Immune evasion is a common reason causing the failure of anticancer immune therapy. Small interfering RNA (siRNA), which can activate the innate and adaptive immune system responses by silencing immune-relevant genes, have been demonstrated to be a powerful tool for preventing or reversing immune evasion. However, siRNAs show poor stability in biological fluids and cannot efficiently cross cell membranes. Nanotechnology has shown great potential for intracellular siRNA delivery in recent years. Nano-immunotherapy can efficiently penetrate the tumor microenvironment (TME) and deliver multiple immunomodulatory agents simultaneously, which appears to be a promising method for combination therapy. Therefore, it provides a new perspective for siRNA delivery in immunomodulation and cancer immunotherapy. The current advances and challenges in nanotechnology-based siRNA delivery strategies for overcoming immune evasion will be discussed in this review. In addition, we also offer insights into therapeutic options, which may expand its applications in clinical cancer treatment.
Collapse
Affiliation(s)
- Kaili Deng
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; (K.D.); (D.Y.)
- School of Medicine, Ningbo University, Ningbo 315021, China
| | - Dongxue Yang
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; (K.D.); (D.Y.)
- Institute of Digestive Disease of Ningbo University, Ningbo 315020, China
| | - Yuping Zhou
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; (K.D.); (D.Y.)
- Institute of Digestive Disease of Ningbo University, Ningbo 315020, China
- Correspondence:
| |
Collapse
|
14
|
Yin W, Jin J, Bao H, Chen H, Wang C, Cheng G, Wu C, Wu M, Yan J, Wu X, Shao Y, Ni X, Su D. Tumor infiltrating lymphocytes-based subtypes and genomic characteristics of EBV- associated lymphoepithelioma-like carcinoma. J Pathol 2022; 257:650-662. [PMID: 35451500 DOI: 10.1002/path.5916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 03/19/2022] [Accepted: 04/19/2022] [Indexed: 11/08/2022]
Abstract
Tumor infiltrating lymphocytes (TILs) offer a key for morphological diagnosis of lymphoepithelioma-like carcinoma (LELC) and are the foundation of onco-immunology. To date, no reports have found a specific risk stratification value of TILs and related it to genomic variation in LELC. Based on the stromal TILs (str-TILs) ratio, we classified 105 EBV-associated LELC cases into two subtypes: patients with ≥ 60% str-TILs area ratio in tumor were classified as subtype I, and otherwise as subtype II. Subtype I patients had significantly better progression-free survival (PFS) and overall survival (OS). We also explored genomic characteristics of EBV-associated LELC within different involved organs. We performed whole-exome sequencing for 51 patients with enough tissue and analyzed genomic characteristics of EBV-associated LELC. Overall, EBV-associated LELCs were characterized by low somatic mutation rate and copy number variations; the enriched genetic lesions affected RTK-RAS, PI3K and cell cycle pathways. Moreover, EBV-associated LELCs from different organs were more similar to each other genetically as compared with other traditional carcinomas of the same sites -as evidenced by unsupervised clustering based on the quantitative data from both mutation signature and chromosomal aneuploidies. Notably, EBV-associated LELC patients with oncogenic driver alterations showed a worse prognosis compared with patients without such alterations. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- WenJuan Yin
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - JiaoYue Jin
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - Hua Bao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, PR China
| | - HanLin Chen
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, PR China
| | - CanMing Wang
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - GuoPing Cheng
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - ChaoQi Wu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - Meijuan Wu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - Junrong Yan
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, PR China
| | - Xue Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, PR China
| | - Yang Shao
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, PR China.,School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Xinghao Ni
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| | - Dan Su
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences(Zhejiang Cancer Hospital, Hangzhou, Zhejiang, PR China.,Institute of Basic Medicine and Cancer (IBMC)s, Chinese Academy of Sciences, Hangzhou, Zhejiang, PR China
| |
Collapse
|
15
|
Su ZY, Siak PY, Leong CO, Cheah SC. Nasopharyngeal Carcinoma and Its Microenvironment: Past, Current, and Future Perspectives. Front Oncol 2022; 12:840467. [PMID: 35311066 PMCID: PMC8924466 DOI: 10.3389/fonc.2022.840467] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/11/2022] [Indexed: 12/31/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy that raises public health concerns in endemic countries. Despite breakthroughs in therapeutic strategies, late diagnosis and drug resistance often lead to unsatisfactory clinical outcomes in NPC patients. The tumor microenvironment (TME) is a complex niche consisting of tumor-associated cells, such as fibroblasts, endothelial cells, leukocytes, that influences tumor initiation, progression, invasion, and metastasis. Cells in the TME communicate through various mechanisms, of note, exosomes, ligand-receptor interactions, cytokines and chemokines are active players in the construction of TME, characterized by an abundance of immune infiltrates with suppressed immune activities. The NPC microenvironment serves as a target-rich niche for the discovery of potential promising predictive or diagnostic biomarkers and the development of therapeutic strategies. Thus, huge efforts have been made to exploit the role of the NPC microenvironment. The whole picture of the NPC microenvironment remains to be portrayed to understand the mechanisms underlying tumor biology and implement research into clinical practice. The current review discusses the recent insights into the role of TME in the development and progression of NPC which results in different clinical outcomes of patients. Clinical interventions with the use of TME components as potential biomarkers or therapeutic targets, their challenges, and future perspectives will be introduced. This review anticipates to provide insights to the researchers for future preclinical, translational and clinical research on the NPC microenvironment.
Collapse
Affiliation(s)
- Zhi Yi Su
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Pui Yan Siak
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| | - Chee-Onn Leong
- Centre of Cancer and Stem Cells Research, International Medical University, Kuala Lumpur, Malaysia
- Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia
| | - Shiau-Chuen Cheah
- Faculty of Medicine and Health Sciences, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Liu Z, He J, Han J, Yang J, Liao W, Chen N. m6A Regulators Mediated Methylation Modification Patterns and Tumor Microenvironment Infiltration Characterization In Nasopharyngeal Carcinoma. Front Immunol 2022; 12:762243. [PMID: 35069534 PMCID: PMC8776994 DOI: 10.3389/fimmu.2021.762243] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 12/03/2021] [Indexed: 02/05/2023] Open
Abstract
Background The role of RNA N6-methyladenosine (m6A) modification in tumor progression and metastasis has been demonstrated. Nonetheless, potential biological function of m6A modification patterns in nasopharyngeal carcinoma (NPC) remains unknown. Methods The m6A modification patterns were comprehensively evaluated based on 26 m6A regulators in NPC, and m6A subtype and also m6A score were identified and systematically correlated with representative tumor characteristics. Results Two distinct m6A subtypes were determined and were highly consistent with immune activated and immune suppressed phenotypes, respectively. More representative m6A scores of individual tumors could predict tumor microenvironment (TME) infiltration, mRNA based stemness index (mRNAsi), EBV gene expression, genetic variation, and prognosis of NPC patients. Low m6A score, characterized by activation of immunity and suppression of mRNAsi and EBV gene, indicated an activated TME phenotype and better PFS and also lower risk of recurrence and metastasis. High m6A score, characterized by activation of Wnt and NF-κB signaling pathway and lack of effective immune infiltration, indicated an immune suppressed TME phenotype and poorer survival. Low m6A score was also correlated with increased tumor mutation burden (TMB) and better response to immunotherapy, and vice versa. A significant therapeutic advantage in patients with low m6A score was confirmed with an anti-PDL1 immunotherapy cohort. Conclusions m6A patterns played an important role in the diversity and complexity of TME. m6A score could be used to evaluate the m6A pattern of individual tumor to enhance our understanding of TME infiltration and guide more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Zijian Liu
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinlan He
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Han
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiangping Yang
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Wenjun Liao
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Nianyong Chen
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Bai Z, Zhou Y, Ye Z, Xiong J, Lan H, Wang F. Tumor-Infiltrating Lymphocytes in Colorectal Cancer: The Fundamental Indication and Application on Immunotherapy. Front Immunol 2022; 12:808964. [PMID: 35095898 PMCID: PMC8795622 DOI: 10.3389/fimmu.2021.808964] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/24/2021] [Indexed: 12/22/2022] Open
Abstract
The clinical success of immunotherapy has revolutionized the treatment of cancer patients, bringing renewed attention to tumor-infiltrating lymphocytes (TILs) of various cancer types. Immune checkpoint blockade is effective in patients with mismatched repair defects and high microsatellite instability (dMMR-MSI-H) in metastatic colorectal cancer (CRC), leading the FDA to accelerate the approval of two programmed cell death 1 (PD-1) blocking antibodies, pembrolizumab and nivolumab, for treatment of dMMR-MSI-H cancers. In contrast, patients with proficient mismatch repair and low levels of microsatellite stability or microsatellite instability (pMMR-MSI-L/MSS) typically have low tumor-infiltrating lymphocytes and have shown unsatisfied responses to the immune checkpoint inhibitor. Different TILs environments reflect different responses to immunotherapy, highlighting the complexity of the underlying tumor-immune interaction. Profiling of TILs fundamental Indication would shed light on the mechanisms of cancer-immune evasion, thus providing opportunities for the development of novel therapeutic strategies. In this review, we summarize phenotypic diversities of TILs and their connections with prognosis in CRC and provide insights into the subsets-specific nature of TILs with different MSI status. We also discuss current clinical immunotherapy approaches based on TILs as well as promising directions for future expansion, and highlight existing clinical data supporting its use.
Collapse
Affiliation(s)
- Ziyi Bai
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yao Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zifan Ye
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Jialong Xiong
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hongying Lan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Feng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
18
|
Han S, Tay JK, Loh CJL, Chu AJM, Yeong JPS, Lim CM, Toh HC. Epstein–Barr Virus Epithelial Cancers—A Comprehensive Understanding to Drive Novel Therapies. Front Immunol 2021; 12:734293. [PMID: 34956172 PMCID: PMC8702733 DOI: 10.3389/fimmu.2021.734293] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 11/12/2021] [Indexed: 12/19/2022] Open
Abstract
Epstein–Barr virus (EBV) is a ubiquitous oncovirus associated with specific epithelial and lymphoid cancers. Among the epithelial cancers, nasopharyngeal carcinoma (NPC), lymphoepithelioma-like carcinoma (LELC), and EBV-associated gastric cancers (EBVaGC) are the most common. The role of EBV in the pathogenesis of NPC and in the modulation of its tumour immune microenvironment (TIME) has been increasingly well described. Much less is known about the pathogenesis and tumour–microenvironment interactions in other EBV-associated epithelial cancers. Despite the expression of EBV-related viral oncoproteins and a generally immune-inflamed cancer subtype, EBV-associated epithelial cancers have limited systemic therapeutic options beyond conventional chemotherapy. Immune checkpoint inhibitors are effective only in a minority of these patients and even less efficacious with molecular targeting drugs. Here, we examine the key similarities and differences of NPC, LELC, and EBVaGC and comprehensively describe the clinical, pathological, and molecular characteristics of these cancers. A deeper comparative understanding of these EBV-driven cancers can potentially uncover targets in the tumour, TIME, and stroma, which may guide future drug development and cast light on resistance to immunotherapy.
Collapse
Affiliation(s)
- Shuting Han
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joshua K. Tay
- Department of Otolaryngology—Head & Neck Surgery, National University of Singapore, Singapore, Singapore
| | | | | | - Joe Poh Sheng Yeong
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Chwee Ming Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- *Correspondence: Han Chong Toh,
| |
Collapse
|
19
|
Multifaceted Roles of Chemokines and Chemokine Receptors in Tumor Immunity. Cancers (Basel) 2021; 13:cancers13236132. [PMID: 34885241 PMCID: PMC8656932 DOI: 10.3390/cancers13236132] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Various immune cells are involved in host immune responses to cancer. T-helper (Th) 1 cells, cytotoxic CD8+ T cells, and natural killer cells are the major effector cells in anti-tumor immunity, whereas cells such as regulatory T cells and myeloid-derived suppressor cells are negatively involved in anti-tumor immunity. Th2 cells and Th17 cells have been shown to have both pro-tumor and anti-tumor activities. The migratory properties of various immune cells are essential for their function and critically regulated by the chemokine superfamily. In this review, we summarize the roles of various immune cells in tumor immunity and their migratory regulation by the chemokine superfamily. We also assess the therapeutic possibilities of targeting chemokines and chemokine receptors in cancer immunotherapy. Abstract Various immune cells are involved in host tumor immune responses. In particular, there are many T cell subsets with different roles in tumor immunity. T-helper (Th) 1 cells are involved in cellular immunity and thus play the major role in host anti-tumor immunity by inducing and activating cytotoxic T lymphocytes (CTLs). On the other hand, Th2 cells are involved in humoral immunity and suppressive to Th1 responses. Regulatory T (Treg) cells negatively regulate immune responses and contribute to immune evasion of tumor cells. Th17 cells are involved in inflammatory responses and may play a role in tumor progression. However, recent studies have also shown that Th17 cells are capable of directly inducting CTLs and thus may promote anti-tumor immunity. Besides these T cell subsets, there are many other innate immune cells such as dendritic cells (DCs), natural killer (NK) cells, and myeloid-derived suppressor cells (MDSCs) that are involved in host immune responses to cancer. The migratory properties of various immune cells are critical for their functions and largely regulated by the chemokine superfamily. Thus, chemokines and chemokine receptors play vital roles in the orchestration of host immune responses to cancer. In this review, we overview the various immune cells involved in host responses to cancer and their migratory properties regulated by the chemokine superfamily. Understanding the roles of chemokines and chemokine receptors in host immune responses to cancer may provide new therapeutic opportunities for cancer immunotherapy.
Collapse
|
20
|
Lisnawati L, Billianti YD, Manatar AF. Association between Foxp3 Tumor Infiltrating Lymphocyte Expression and Response After Chemoradiation in Nasopharyngeal Carcinoma. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND: Nasopharyngeal carcinoma (NPC) is a carcinoma originating from the surface epithelium of the nasopharynx with the highest incidence in China and South East Asia. Currently, many researchers are developing tumor microenvironment which can be assessed by tumor-infiltrating lymphochyte, and its association with treatment response in several tumors, including NPC. Foxp3, known as a regulatory T cell (Treg) marker, plays a role in the immunoregulatory environment of tumor cells and can be used as a prognostic factor. The relationship between Foxp3 expression and treatment response is considered as one of the factors affecting the prognosis of NPC.
AIM: This study aims to determine the relationship between Foxp3 expression and treatment response in NPC.
MATERIALS AND METHODS: A cross-sectional study was done to analyze the association between Foxp3 and treatment response in NPC. This study included 60 samples who were diagnosed with non-keratinizing NPC at the Department of Anatomical Pathology, Faculty of Medicine Universitas Indonesia/Cipto Mangunkusumo Hospital from January 2018 until December 2020. Immunohistochemistry was done to evaluate the expression of Foxp3. Foxp3 expression was evaluated in the intratumoral and peritumoral areas.
RESULTS: Among 60 patients, the number of males were more than females (66.7%, 33.3%, respectively) with a ratio of 2:1. There was statistically significant difference between intratumoral and total Foxp3 expression and treatment response (p < 0.05, p = 0.001, respectively); however, no significant differences found between peritumoral Foxp3 expression and treatment response (p = 0.114).
CONCLUSION: Foxp3 expression had a statistically significant relationship with response therapy after chemoradiation.
Collapse
|
21
|
Yang L, Liu G, Li Y, Pan Y. The emergence of tumor-infiltrating lymphocytes in nasopharyngeal carcinoma: Predictive value and immunotherapy implications. Genes Dis 2021; 9:1208-1219. [PMID: 35873027 PMCID: PMC9293699 DOI: 10.1016/j.gendis.2021.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 11/09/2022] Open
Abstract
The clinical study of nasopharyngeal carcinoma (NPC) often reveals a large number of lymphocytes infiltrating the primary tumor site. As an important part of the tumor microenvironment, tumor-infiltrating lymphocytes (TILs) do not exist alone but as a complex multicellular population with high heterogeneity. TILs play an extremely significant role in the occurrence, development, invasion and metastasis of NPC. The latest research shows that they participate in tumorigenesis and treatment, and the composition, quantity, functional status and distribution of TILs subsets have good predictive value for the prognosis of NPC patients. TILs are an independent prognostic factor for TNM stage and significantly correlated with better prognosis. Additionally, adoptive immunotherapy using anti-tumor TILs has achieved good results in a variety of solid tumors including NPC. This review evaluates recent clinical and preclinical studies of NPC, summarizes the role of TILs in promoting and inhibiting tumor growth, evaluates the predictive value of TILs, and explores the potential benefits of TILs-based immunotherapy in the treatment of NPC.
Collapse
|
22
|
Berele BA, Cai Y, Yang G. Prognostic Value of Tumor Infiltrating Lymphocytes in Nasopharyngeal Carcinoma Patients: Meta-Analysis. Technol Cancer Res Treat 2021; 20:15330338211034265. [PMID: 34323154 PMCID: PMC8330464 DOI: 10.1177/15330338211034265] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective: To evaluate the prognostic value of tumor infiltrating lymphocytes (TILs) in nasopharyngeal carcinoma (NPC) patients. Method: Meta-analysis was performed on eligible studies that was identified by systematic searching of Google scholar, MEDLINE, CNKI, Scopus, PubMed, PMC, Embase and Web of Science databases. The study protocol was registered in International Platform of Registered Systematic Review and Meta-Analysis Protocols-INPLASY (registration number: INPLASY202160014). Databases were searched from inception to January 20, 2020 to identify eligible studies. Those studies that evaluated survival in the form of hazard ratio (HR) in TILs of NPC patients was analyzed. All statistical analysis was performed by using STATA version 16.0 software. Result: Fourteen studies with a total of 3025 patients was analyzed. The pooled result showed that high TILs was significantly associated with favorable overall survival (OS) (HR = 0.55; 95%CI = 0.39-0.77; P = 0.001) and disease free survival (DFS) (HR = 0.60; 95%CI = 0.44-0.81; P = 0.04). Interestingly, high intratumoral TILs had relatively better OS (HR = 0.45; 95%CI = 0.35-0.58; P = 0.006) than stromal TILs (HR = 0.59; 95%CI = 0.36-0.97; P = 0.03). Moreover, an increased level of CD4+ cells infiltration was correlated with favorable OS (HR = 0.4; 95%CI = 0.18-0.85; P = 0.01). CD3+, CD8+ and FoxP3+ lymphocyte’s better prognosis was not statistically significant for OS (P = 0.09; P = 0.07; P = 0.52) and for DFS (P = 0.13; P = 0.29) respectively. However, subgroup analysis of intratumoral CD3+ (HR = 0.48; 95%CI = 0.33-0.70; P = 0.05) and intratumoral CD8+ (HR = 0.32; 95%CI = 0.16-0.62; P = 0.001) was significantly associated with improved OS, but not significant in stromal CD3+ (HR = 0.66; 95%CI = 0.20-2.20; P = 0.62). Conclusion: TILs were variably correlated with better prognosis depending on their microanatomic location and subset of TILs in NPC patients. CD4+, intratumoral CD3+ and intratumoral CD8+ lymphocytes could predict favorable patient outcome which suggest that their role in mediating antitumor immune response could potentially be exploited in the treatment of NPC patients. Future large study on the prognostic value of microanatomic location of TILs is needed for confirmation.
Collapse
Affiliation(s)
| | - Yuxiang Cai
- Department of Pathology, 89674Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Guifang Yang
- Department of Pathology, 89674Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Yeo MK, Sun P, Chung C, Park JH, Kang SH, Moon HS, Sung JK, Jeong HY, Kim JS. Clinical Significance of Composition Changes in T-cell Subpopulations After Chemotherapy in Patients With Gastric Cancer. In Vivo 2021; 35:2417-2424. [PMID: 34182525 DOI: 10.21873/invivo.12519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND/AIM New therapeutic agents and prognostic biomarkers for gastric cancer are needed. We analyzed the composition of peripheral blood T-cell subpopulations in response to chemotherapy in patients with gastric cancer. PATIENTS AND METHODS Peripheral blood samples were collected from patients diagnosed with gastric cancer before and after chemotherapy (FOLFOX; oxaliplatin, 5-fluorouracil, and leucovorin). Peripheral blood mononuclear cells were isolated. Patients were divided into responder (n=5) and non-responder groups (n=2) based on their chemotherapy outcomes. RESULTS Non-responders showed lower numbers of CD4+/total cells and CD8+/total cells after chemotherapy compared to the responder group, but the difference was not significant (p=0.905, p=0.095). Naïve T, central memory T, effector memory T and effector T-cell counts differed in both groups after chemotherapy. CONCLUSION Changes in peripheral T-cell subpopulations after chemotherapy were confirmed in patients with gastric cancer, which may be a prognostic predictor and development of therapeutic agents.
Collapse
Affiliation(s)
- Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Pureum Sun
- Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Ho Park
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sun Hyung Kang
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hee Seok Moon
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Jae Kyu Sung
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hyun Yong Jeong
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Ju Seok Kim
- Department of Internal Medicine, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
24
|
Liu W, Chen G, Zhang C, Liao X, Xie J, Liang T, Liao W, Song L, Zhang X. Prognostic significance of tumor-infiltrating lymphocytes and macrophages in nasopharyngeal carcinoma: a systematic review and meta-analysis. Eur Arch Otorhinolaryngol 2021; 279:25-35. [PMID: 34027599 DOI: 10.1007/s00405-021-06879-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/11/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Many studies have investigated the prognostic value of tumor-infiltrating lymphocytes (TILs) and tumor-infiltrating macrophages (TIMs) in patients with nasopharyngeal carcinoma (NPC), but the results remain controversial. Here, we performed a meta-analysis to evaluate the prognostic significance of TILs/TIMs in patients with NPC METHODS: The study was registered with PROSPERO (CRD42021234078). PubMed, Embase, and Web of Science databases were searched up to Dec 30, 2020. We reviewed studies that evaluated the relationship between TILs/TIMs and overall survival (OS), disease-free survival (DFS), or progression-free survival (PFS) in NPC. For TILs, CD3, CD4, CD8, and FOXP3 were searched as T-cell markers, CD19 and CD20 as B-cell markers, and CD56 as a natural killer cell marker. For TIMs, CD68 and CD163 were searched as total and M2 macrophage markers, respectively. RESULTS In total, 19 studies with 3708 NPC were included in this meta-analysis. We found that high total numbers of TILs were significantly associated with favorable OS [hazard ratio (HR) 0.46, 95% confidence interval (CI) 0.38-0.57 and PFS (HR 0.48, 95% CI 0.38-0.62)]. In contrast, tumor infiltration by CD3+ T cells (HR 0.55, 95% CI 0.39-0.76), CD4+ T cells (HR 0.40, 95% CI 0.18-0.85), and CD8+ T cells (HR 0.56, 95% CI 0.34-0.93) correlated positively with OS. No significant relationship was found between survival and tumor infiltration by FOXP3+ T cells, CD68+ macrophages, or CD163+ macrophages. CONCLUSION Our findings revealed that tumor infiltration by CD3+ , CD4+ , and CD8+ T cells could be prognostic biomarkers in NPC.
Collapse
Affiliation(s)
- Weixing Liu
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Gui Chen
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Chunyi Zhang
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Xiao Liao
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Junyang Xie
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Tianhao Liang
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Wenjing Liao
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Lijuan Song
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China
| | - Xiaowen Zhang
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiangxi Road, Guangzhou, 510120, China.
| |
Collapse
|
25
|
Qi Z, Liu Y, Mints M, Mullins R, Sample R, Law T, Barrett T, Mazul AL, Jackson RS, Kang SY, Pipkorn P, Parikh AS, Tirosh I, Dougherty J, Puram SV. Single-Cell Deconvolution of Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:1230. [PMID: 33799782 PMCID: PMC7999850 DOI: 10.3390/cancers13061230] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 02/26/2021] [Indexed: 12/24/2022] Open
Abstract
Complexities in cell-type composition have rightfully led to skepticism and caution in the interpretation of bulk transcriptomic analyses. Recent studies have shown that deconvolution algorithms can be utilized to computationally estimate cell-type proportions from the gene expression data of bulk blood samples, but their performance when applied to tumor tissues, including those from head and neck, remains poorly characterized. Here, we use single-cell data (~6000 single cells) collected from 21 head and neck squamous cell carcinoma (HNSCC) samples to generate cell-type-specific gene expression signatures. We leverage bulk RNA-seq data from >500 HNSCC samples profiled by The Cancer Genome Atlas (TCGA), and using single-cell data as a reference, apply two newly developed deconvolution algorithms (CIBERSORTx and MuSiC) to the bulk transcriptome data to quantitatively estimate cell-type proportions for each tumor in TCGA. We show that these two algorithms produce similar estimates of constituent/major cell-type proportions and that a high T-cell fraction correlates with improved survival. By further characterizing T-cell subpopulations, we identify that regulatory T-cells (Tregs) were the major contributor to this improved survival. Lastly, we assessed gene expression, specifically in the Treg population, and found that TNFRSF4 (Tumor Necrosis Factor Receptor Superfamily Member 4) was differentially expressed in the core Treg subpopulation. Moreover, higher TNFRSF4 expression was associated with greater survival, suggesting that TNFRSF4 could play a key role in mechanisms underlying the contribution of Treg in HNSCC outcomes.
Collapse
Affiliation(s)
- Zongtai Qi
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Yating Liu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Michael Mints
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (M.M.); (I.T.)
| | - Riley Mullins
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Reilly Sample
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
- Clinical Research Training Center, Washington University School of Medicine, St Louis, MO 63110, USA
| | - Travis Law
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Thomas Barrett
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Angela L. Mazul
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Ryan S. Jackson
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Stephen Y. Kang
- Division of Head and Neck Oncology, Department of Otolaryngology—Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; (S.Y.K.); (A.S.P.)
| | - Patrik Pipkorn
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
| | - Anuraag S. Parikh
- Division of Head and Neck Oncology, Department of Otolaryngology—Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; (S.Y.K.); (A.S.P.)
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Otolaryngology, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel; (M.M.); (I.T.)
| | - Joseph Dougherty
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
- Department of Otolaryngology, Harvard Medical School, Boston, MA 02114, USA
| | - Sidharth V. Puram
- Department of Otolaryngology-Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA; (Z.Q.); (R.M.); (R.S.); (T.L.); (T.B.); (A.L.M.); (R.S.J.); (P.P.)
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA;
| |
Collapse
|
26
|
Gong L, Kwong DLW, Dai W, Wu P, Li S, Yan Q, Zhang Y, Zhang B, Fang X, Liu L, Luo M, Liu B, Chow LKY, Chen Q, Huang J, Lee VHF, Lam KO, Lo AWI, Chen Z, Wang Y, Lee AWM, Guan XY. Comprehensive single-cell sequencing reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of nasopharyngeal carcinoma. Nat Commun 2021; 12:1540. [PMID: 33750785 PMCID: PMC7943808 DOI: 10.1038/s41467-021-21795-z] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
The tumor microenvironment (TME) of nasopharyngeal carcinoma (NPC) harbors a heterogeneous and dynamic stromal population. A comprehensive understanding of this tumor-specific ecosystem is necessary to enhance cancer diagnosis, therapeutics, and prognosis. However, recent advances based on bulk RNA sequencing remain insufficient to construct an in-depth landscape of infiltrating stromal cells in NPC. Here we apply single-cell RNA sequencing to 66,627 cells from 14 patients, integrated with clonotype identification on T and B cells. We identify and characterize five major stromal clusters and 36 distinct subpopulations based on genetic profiling. By comparing with the infiltrating cells in the non-malignant microenvironment, we report highly representative features in the TME, including phenotypic abundance, genetic alternations, immune dynamics, clonal expansion, developmental trajectory, and molecular interactions that profoundly influence patient prognosis and therapeutic outcome. The key findings are further independently validated in two single-cell RNA sequencing cohorts and two bulk RNA-sequencing cohorts. In the present study, we reveal the correlation between NPC-specific characteristics and progression-free survival. Together, these data facilitate the understanding of the stromal landscape and immune dynamics in NPC patients and provides deeper insights into the development of prognostic biomarkers and therapeutic targets in the TME.
Collapse
Affiliation(s)
- Lanqi Gong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wei Dai
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Pingan Wu
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Shanshan Li
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Qian Yan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yu Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Baifeng Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiaona Fang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li Liu
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- The AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Disease, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Min Luo
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Beilei Liu
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Larry Ka-Yue Chow
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qingyun Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jinlin Huang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor Ho-Fun Lee
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ka-On Lam
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Anthony Wing-Ip Lo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zhiwei Chen
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- The AIDS Institute, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Emerging Infectious Disease, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan Wang
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Anne Wing-Mui Lee
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Oncology in Southern China, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|
27
|
Zhang Y, Wang D, Peng M, Tang L, Ouyang J, Xiong F, Guo C, Tang Y, Zhou Y, Liao Q, Wu X, Wang H, Yu J, Li Y, Li X, Li G, Zeng Z, Tan Y, Xiong W. Single-cell RNA sequencing in cancer research. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:81. [PMID: 33648534 PMCID: PMC7919320 DOI: 10.1186/s13046-021-01874-1] [Citation(s) in RCA: 149] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq), a technology that analyzes transcriptomes of complex tissues at single-cell levels, can identify differential gene expression and epigenetic factors caused by mutations in unicellular genomes, as well as new cell-specific markers and cell types. scRNA-seq plays an important role in various aspects of tumor research. It reveals the heterogeneity of tumor cells and monitors the progress of tumor development, thereby preventing further cellular deterioration. Furthermore, the transcriptome analysis of immune cells in tumor tissue can be used to classify immune cells, their immune escape mechanisms and drug resistance mechanisms, and to develop effective clinical targeted therapies combined with immunotherapy. Moreover, this method enables the study of intercellular communication and the interaction of tumor cells and non-malignant cells to reveal their role in carcinogenesis. scRNA-seq provides new technical means for further development of tumor research and is expected to make significant breakthroughs in this field. This review focuses on the principles of scRNA-seq, with an emphasis on the application of scRNA-seq in tumor heterogeneity, pathogenesis, and treatment.
Collapse
Affiliation(s)
- Yijie Zhang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dan Wang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Miao Peng
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Le Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jiawei Ouyang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Fang Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Can Guo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yanyan Tang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Yujuan Zhou
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Qianjin Liao
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Xu Wu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Hui Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Jianjun Yu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China
| | - Yong Li
- Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas, USA
| | - Xiaoling Li
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Guiyuan Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yixin Tan
- Department of Dermatology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, Changsha, Hunan, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China. .,Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Disease Genome Research Center, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
28
|
Liu Y, He S, Wang XL, Peng W, Chen QY, Chi DM, Chen JR, Han BW, Lin GW, Li YQ, Wang QY, Peng RJ, Wei PP, Guo X, Li B, Xia X, Mai HQ, Hu XD, Zhang Z, Zeng YX, Bei JX. Tumour heterogeneity and intercellular networks of nasopharyngeal carcinoma at single cell resolution. Nat Commun 2021; 12:741. [PMID: 33531485 PMCID: PMC7854640 DOI: 10.1038/s41467-021-21043-4] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
The heterogeneous nature of tumour microenvironment (TME) underlying diverse treatment responses remains unclear in nasopharyngeal carcinoma (NPC). Here, we profile 176,447 cells from 10 NPC tumour-blood pairs, using single-cell transcriptome coupled with T cell receptor sequencing. Our analyses reveal 53 cell subtypes, including tumour-infiltrating CD8+ T, regulatory T (Treg), and dendritic cells (DCs), as well as malignant cells with different Epstein-Barr virus infection status. Trajectory analyses reveal exhausted CD8+ T and immune-suppressive TNFRSF4+ Treg cells in tumours might derive from peripheral CX3CR1+CD8+ T and naïve Treg cells, respectively. Moreover, we identify immune-regulatory and tolerogenic LAMP3+ DCs. Noteworthily, we observe intensive inter-cell interactions among LAMP3+ DCs, Treg, exhausted CD8+ T, and malignant cells, suggesting potential cross-talks to foster an immune-suppressive niche for the TME. Collectively, our study uncovers the heterogeneity and interacting molecules of the TME in NPC at single-cell resolution, which provide insights into the mechanisms underlying NPC progression and the development of precise therapies for NPC.
Collapse
Affiliation(s)
- Yang Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Shuai He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, People's Republic of China
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Xi-Liang Wang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Wan Peng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Qiu-Yan Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Dong-Mei Chi
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
- Department of Anesthesiology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Jie-Rong Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
- Guangdong General Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Bo-Wei Han
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Guo-Wang Lin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510282, People's Republic of China
| | - Yi-Qi Li
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Qian-Yu Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Rou-Jun Peng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Pan-Pan Wei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Xiang Guo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Bo Li
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
- RNA Biomedical Institute, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Xiaojun Xia
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Hai-Qiang Mai
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China
| | - Xue-Da Hu
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, 100871, People's Republic of China
| | - Zemin Zhang
- BIOPIC, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, 100871, People's Republic of China.
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, People's Republic of China.
| | - Yi-Xin Zeng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China.
| | - Jin-Xin Bei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, 510060, People's Republic of China.
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, People's Republic of China.
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
29
|
Senovilla L, Vacchelli E, Galon J, Adjemian S, Eggermont A, Fridman WH, Sautès-Fridman C, Ma Y, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Prognostic and predictive value of the immune infiltrate in cancer. Oncoimmunology 2021; 1:1323-1343. [PMID: 23243596 PMCID: PMC3518505 DOI: 10.4161/onci.22009] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Solid tumors are constituted of a variety of cellular components, including bona fide malignant cells as well as endothelial, structural and immune cells. On one hand, the tumor stroma exerts major pro-tumorigenic and immunosuppressive functions, reflecting the capacity of cancer cells to shape the microenvironment to satisfy their own metabolic and immunological needs. On the other hand, there is a component of tumor-infiltrating leucocytes (TILs) that has been specifically recruited in the attempt to control tumor growth. Along with the recognition of the critical role played by the immune system in oncogenesis, tumor progression and response to therapy, increasing attention has been attracted by the potential prognostic and/or predictive role of the immune infiltrate in this setting. Data from large clinical studies demonstrate indeed that a robust infiltration of neoplastic lesions by specific immune cell populations, including (but not limited to) CD8+ cytotoxic T lymphocytes, Th1 and Th17 CD4+ T cells, natural killer cells, dendritic cells, and M1 macrophages constitutes an independent prognostic indicator in several types of cancer. Conversely, high levels of intratumoral CD4+CD25+FOXP3+ regulatory T cells, Th2 CD4+ T cells, myeloid-derived suppressor cells, M2 macrophages and neutrophils have frequently been associated with dismal prognosis. So far, only a few studies have addressed the true predictive potential of TILs in cancer patients, generally comforting the notion that—at least in some clinical settings—the immune infiltrate can reliably predict if a specific patient will respond to therapy or not. In this Trial Watch, we will summarize the results of clinical trials that have evaluated/are evaluating the prognostic and predictive value of the immune infiltrate in the context of solid malignancies.
Collapse
Affiliation(s)
- Laura Senovilla
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Orsay, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Elsharkawy SS, Elrheem MA, Elrheem SA. The Tumor Infiltrating Lymphocytes (TILs): Did We Find the Missed Piece of the Huge Puzzle? OPEN JOURNAL OF OBSTETRICS AND GYNECOLOGY 2021; 11:146-161. [DOI: 10.4236/ojog.2021.112017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
31
|
Characterization and Differentiation of the Tumor Microenvironment (TME) of Orthotopic and Subcutaneously Grown Head and Neck Squamous Cell Carcinoma (HNSCC) in Immunocompetent Mice. Int J Mol Sci 2020; 22:ijms22010247. [PMID: 33383676 PMCID: PMC7796118 DOI: 10.3390/ijms22010247] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/20/2020] [Accepted: 12/24/2020] [Indexed: 12/12/2022] Open
Abstract
For the development and evaluation of new head and neck squamous cell carcinoma (HNSCC) therapeutics, suitable, well-characterized animal models are needed. Thus, by analyzing orthotopic versus subcutaneous models of HNSCC in immunocompetent mice, we evaluated the existence of adenosine-related immunosuppressive B- and T lymphocyte populations within the tumor microenvironment (TME). Applying the SCC VII model for the induction of HNSCC in immunocompetent C3H/HeN mice, the cellular TME was characterized after tumor initiation over time by flow cytometry. The TME in orthotopic grown tumors revealed a larger population of tumor-infiltrating lymphocytes (TIL) with more B cells and CD4+ T cells than the subcutaneously grown tumors. Immune cell populations in the blood and bone marrow showed a rather distinct reaction toward tumor induction and tumor location compared to the spleen, lymph nodes, or thymus. In addition, large numbers of immunosuppressive B- and T cells were identified within the TME but also in secondary lymphoid organs, independently of the tumor initiation site. The altered immunogenic TME may influence the response to any treatment attempt. Moreover, when analyzing the TME and other lymphoid organs of tumor-bearing mice, we observed conditions reflecting largely those of patients suffering from HNSCC suggesting the C3H/HeN mouse model as a suitable tool for studies aiming to target immunosuppression to improve anti-cancer therapies.
Collapse
|
32
|
Cho JH, Lim YC. Prognostic impact of regulatory T cell in head and neck squamous cell carcinoma: A systematic review and meta-analysis. Oral Oncol 2020; 112:105084. [PMID: 33181417 DOI: 10.1016/j.oraloncology.2020.105084] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The impact of regulatory T (Treg) cells as a prognostic factor of survival in head and neck squamous cell carcinoma (HNSCC) remains controversial. We aimed to evaluate the prognostic value of Treg cells in patients with HNSCC through a meta-analysis. MATERIALS AND METHODS Through a literature search in PubMed, Embase, and Cochrane, we included 11 articles in this meta-analysis and investigated the effect of Treg cell level on the survival of patients with HNSCC. Also, we performed a subgroup analysis according to the study sample (blood vs. tumor tissue), primary tumor site, HPV infectivity, or Treg cell marker. RESULTS High levels of circulating Treg cells in the peripheral blood of patients with HNSCC can significantly increase the disease specific survival rate of patients. Moreover, subgroup analysis showed that high levels of Treg in peripheral blood were significantly associated with better disease specific survival in patients with oral cancer, a subsite of HNSCC, but not in those with other head and neck subsite. Positivity of HPV infection did not influence the prognosis of patients with HNSCC. CONCLUSION Increase in the levels of circulating Treg cells in peripheral blood can be a prognostic factor of survival in patients with oral cancer.
Collapse
Affiliation(s)
- Jae Hoon Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Young Chang Lim
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
33
|
Zou Z, Ha Y, Liu S, Huang B. Identification of tumor-infiltrating immune cells and microenvironment-relevant genes in nasopharyngeal carcinoma based on gene expression profiling. Life Sci 2020; 263:118620. [PMID: 33096113 DOI: 10.1016/j.lfs.2020.118620] [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: 06/09/2020] [Revised: 09/30/2020] [Accepted: 10/11/2020] [Indexed: 12/24/2022]
Abstract
AIMS The purpose of this study was to investigate the prognostic significance of tumor-infiltrating immune cells and microenvironment-relevant genes in nasopharyngeal carcinoma (NPC) and their correlations. MATERIALS AND METHODS The "xCell" algorithm was used to calculate the enrichment scores for 33 immune cells in the samples of GSE12452, GSE40290, GSE53819, GSE68799, and GSE102349. The difference of immune cells between NPC group and non-cancerous group and the prognostic value of the immune cells were analyzed. Besides, based on the Microenvironment scores, the differentially expressed genes (DEGs) between the high- and low-score groups were screened to identify the microenvironment-relevant hub genes. Furthermore, the DEGs were used to establish a risk score model for predicting progression-free survival (PFS) via LASSO penalized Cox regression. KEY FINDINGS The scores of B-cells and Memory B-cells of NPC were significantly lower than those of non-cancerous tissues, and they were positively associated with PFS. Moreover, 10 hub genes (PTPRC, CD19, CD79B, BTK, CD79A, SELL, MS4A1, CD38, CD52, and CD22) were identified and positively correlated with B-cells, Memory B-cells, and Microenvironment scores in GSE12452, GSE68799, and GSE102349. High expression levels of CD22, CD38, CD79B, MS4A1, SELL, and PTPRC were associated with longer PFS. Besides, a risk score model composed of DARC, IL33, IGHG1, and SLC6A8 was established with a good performance for PFS prediction. SIGNIFICANCE These results enhance our understanding of the composition and prognostic significance of tumor-infiltrating immune cells in NPC lesions, and provide potential targets for prognostication and immunotherapy for NPC patients.
Collapse
Affiliation(s)
- Zhenning Zou
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Yanping Ha
- Department of Pathology, Guangdong Medical University, Zhanjiang, China
| | - Shuguang Liu
- Department of Pathology, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Bowan Huang
- Department of Anesthesiology, Zhanjiang Central Hospital, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
34
|
Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-infiltrating lymphocytes: Warriors fight against tumors powerfully. Biomed Pharmacother 2020; 132:110873. [PMID: 33068926 DOI: 10.1016/j.biopha.2020.110873] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are infiltrating lymphocytes in tumor tissues. After isolation, screening and amplification in vitro, they will be implanted into patients and play a specific killing effect on tumors. Since TILs have not been genetically modified and come from the body of patients, there will be relatively few adverse reactions. This is also the advantage of TIL treatment. In recent years, its curative effect on solid tumors began to show its sharpness. However, due to the limitations of the immune microenvironment and the mutation of antigens, TIL's development was slowed down. This article reviews the research progress, biological characteristics, preparation and methods of enhancing the therapeutic effect of tumor-infiltrating lymphocytes, their roles in different tumors and prognosis, and emphasizes the important value of tumor-infiltrating lymphocytes in anti-tumor.
Collapse
Affiliation(s)
- Baisheng Lin
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Likun Du
- First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Hongmei Li
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xiao Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China.
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
35
|
Predictive Value of CD8 Expression and FoxP3 Methylation in Nasopharyngeal Carcinoma Patients Treated with Chemoradiotherapy in a Non-endemic Area. Pathol Oncol Res 2020; 26:2459-2467. [PMID: 32564263 DOI: 10.1007/s12253-020-00859-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 06/16/2020] [Indexed: 10/24/2022]
Abstract
Undifferentiated Nasopharyngeal Carcinoma (UNPC) is associated with Epstein-Barr Virus (EBV) and characterized by an abundant immune infiltrate potentially influencing the prognosis. Thus, we retrospectively assessed the significance of immunosuppression in the UNPC microenvironment as prognostic biomarker of treatment failure in a non-endemic area, and monitored the variation of systemic EBV-specific immunity before and after chemoradiotherapy (CRT). DNA and RNA were extracted from diagnostic biopsies obtained by tumor and adjacent mucosa from 63 consecutive EBV+ UNPC patients who underwent radical CRT. Among these patients 11 relapsed within 2 years. The expression of the EBV-derived UNPC-specific BARF1 gene and several immune-related genes was monitored through quantitative RT-PCR and methylation-specific PCR analyses. Peripheral T cell responses against EBV and BARF1 were measured in 14 patients (7 relapses) through IFN-γ ELISPOT assay. We found significantly higher expression levels of BARF1, CD8, IFN-γ, IDO, PD-L1, and PD-1 in UNPC samples compared to healthy tissues. CD8 expression was significantly reduced in both tumor and healthy tissues in UNPC patients who relapsed within two years. We observed a hypomethylated FOXP3 intron 1 exclusively in relapsed UNPC patients. Finally, we noticed a significant decrease in EBV- and BARF1-specific T-cells after CRT only in relapsing patients. Our data suggest that a high level of immunosuppression (low CD8, hypomethylated FoxP3) in UNPC microenvironment may predict treatment failure and may allow an early identification of patients who could benefit from the addition of immune modulating strategies to improve first line CRT.
Collapse
|
36
|
Liu WN, Fong SY, Tan WWS, Tan SY, Liu M, Cheng JY, Lim S, Suteja L, Huang EK, Chan JKY, Iyer NG, Yeong JPS, Lim DWT, Chen Q. Establishment and Characterization of Humanized Mouse NPC-PDX Model for Testing Immunotherapy. Cancers (Basel) 2020; 12:cancers12041025. [PMID: 32331230 PMCID: PMC7225949 DOI: 10.3390/cancers12041025] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/13/2020] [Accepted: 04/17/2020] [Indexed: 12/25/2022] Open
Abstract
Immune checkpoint blockade (ICB) monotherapy shows early promise for the treatment of nasopharyngeal carcinoma (NPC) in patients. Nevertheless, limited representative NPC models hamper preclinical studies to evaluate the efficacy of novel ICB and combination regimens. In the present study, we engrafted NPC biopsies in non-obese diabetic-severe combined immunodeficiency interleukin-2 receptor gamma chain-null (NSG) mice and established humanized mouse NPC-patient-derived xenograft (NPC-PDX) model successfully. Epstein–Barr virus was detected in the NPC in both NSG and humanized mice as revealed by Epstein–Barr virus-encoded small RNA (EBER) in situ hybridization (ISH) and immunohistochemical (IHC) staining. In the NPC-bearing humanized mice, the percentage of tumor-infiltrating CD8+ cytotoxic T cells was lowered, and the T cells expressed higher levels of various inhibitory receptors, such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) than those in blood. The mice were then treated with nivolumab and ipilimumab, and the anti-tumor efficacy of combination immunotherapy was examined. In line with paired clinical data, the NPC-PDX did not respond to the treatment in terms of tumor burden, whilst an immunomodulatory response was elicited in the humanized mice. From our results, human proinflammatory cytokines, such as interferon-gamma (IFN-γ) and interleukin-6 (IL-6) were significantly upregulated in plasma. After treatment, there was a decrease in CD4/CD8 ratio in the NPC-PDX, which also simulated the modulation of intratumoral CD4/CD8 profile from the corresponding donor. In addition, tumor-infiltrating T cells were re-activated and secreted more IFN-γ towards ex vivo stimulation, suggesting that other factors, including soluble mediators and metabolic milieu in tumor microenvironment may counteract the effect of ICB treatment and contribute to the tumor progression in the mice. Taken together, we have established and characterized a novel humanized mouse NPC-PDX model, which plausibly serves as a robust platform to test for the efficacy of immunotherapy and may predict clinical outcomes in NPC patients.
Collapse
Affiliation(s)
- Wai Nam Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Shin Yie Fong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Wilson Wei Sheng Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Sue Yee Tan
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Min Liu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Jia Ying Cheng
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Sherlly Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Lisda Suteja
- Division of Medical Oncology, National Cancer Centre, Singapore 169610, Singapore; (L.S.); (N.G.I.)
| | - Edwin Kunxiang Huang
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore 229899, Singapore; (E.K.H.); (J.K.Y.C.)
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore 229899, Singapore; (E.K.H.); (J.K.Y.C.)
- Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
| | | | - Joe Poh Sheng Yeong
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
| | - Darren Wan-Teck Lim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
- Division of Medical Oncology, National Cancer Centre, Singapore 169610, Singapore; (L.S.); (N.G.I.)
- Correspondence: (D.W.-T.L.); (Q.C.); Tel.: +65-6586-9873 (Q.C.)
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138673, Singapore; (W.N.L.); (S.Y.F.); (W.W.S.T.); (S.Y.T.); (M.L.); (J.Y.C.); (S.L.); (J.P.S.Y.)
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- Correspondence: (D.W.-T.L.); (Q.C.); Tel.: +65-6586-9873 (Q.C.)
| |
Collapse
|
37
|
Kuen DS, Kim BS, Chung Y. IL-17-Producing Cells in Tumor Immunity: Friends or Foes? Immune Netw 2020; 20:e6. [PMID: 32158594 PMCID: PMC7049578 DOI: 10.4110/in.2020.20.e6] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/25/2020] [Accepted: 01/26/2020] [Indexed: 02/07/2023] Open
Abstract
IL-17 is produced by RAR-related orphan receptor gamma t (RORγt)-expressing cells including Th17 cells, subsets of γδT cells and innate lymphoid cells (ILCs). The biological significance of IL-17-producing cells is well-studied in contexts of inflammation, autoimmunity and host defense against infection. While most of available studies in tumor immunity mainly focused on the role of T-bet-expressing cells, including cytotoxic CD8+ T cells and NK cells, and their exhaustion status, the role of IL-17-producing cells remains poorly understood. While IL-17-producing T-cells were shown to be anti-tumorigenic in adoptive T-cell therapy settings, mice deficient in type 17 genes suggest a protumorigenic potential of IL-17-producing cells. This review discusses the features of IL-17-producing cells, of both lymphocytic and myeloid origins, as well as their suggested pro- and/or anti-tumorigenic functions in an organ-dependent context. Potential therapeutic approaches targeting these cells in the tumor microenvironment will also be discussed.
Collapse
Affiliation(s)
- Da-Sol Kuen
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.,BK21 Plus Program, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Byung-Seok Kim
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yeonseok Chung
- Laboratory of Immune Regulation, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea.,BK21 Plus Program, Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
38
|
Khorrami M, Prasanna P, Gupta A, Patil P, Velu PD, Thawani R, Corredor G, Alilou M, Bera K, Fu P, Feldman M, Velcheti V, Madabhushi A. Changes in CT Radiomic Features Associated with Lymphocyte Distribution Predict Overall Survival and Response to Immunotherapy in Non-Small Cell Lung Cancer. Cancer Immunol Res 2020; 8:108-119. [PMID: 31719058 PMCID: PMC7718609 DOI: 10.1158/2326-6066.cir-19-0476] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/04/2019] [Accepted: 11/05/2019] [Indexed: 12/26/2022]
Abstract
No predictive biomarkers can robustly identify patients with non-small cell lung cancer (NSCLC) who will benefit from immune checkpoint inhibitor (ICI) therapies. Here, in a machine learning setting, we compared changes ("delta") in the radiomic texture (DelRADx) of CT patterns both within and outside tumor nodules before and after two to three cycles of ICI therapy. We found that DelRADx patterns could predict response to ICI therapy and overall survival (OS) for patients with NSCLC. We retrospectively analyzed data acquired from 139 patients with NSCLC at two institutions, who were divided into a discovery set (D1 = 50) and two independent validation sets (D2 = 62, D3 = 27). Intranodular and perinodular texture descriptors were extracted, and the relative differences were computed. A linear discriminant analysis (LDA) classifier was trained with 8 DelRADx features to predict RECIST-derived response. Association of delta-radiomic risk score (DRS) with OS was determined. The association of DelRADx features with tumor-infiltrating lymphocyte (TIL) density on the diagnostic biopsies (n = 36) was also evaluated. The LDA classifier yielded an AUC of 0.88 ± 0.08 in distinguishing responders from nonresponders in D1, and 0.85 and 0.81 in D2 and D3 DRS was associated with OS [HR: 1.64; 95% confidence interval (CI), 1.22-2.21; P = 0.0011; C-index = 0.72). Peritumoral Gabor features were associated with the density of TILs on diagnostic biopsy samples. Our results show that DelRADx could be used to identify early functional responses in patients with NSCLC.
Collapse
Affiliation(s)
- Mohammadhadi Khorrami
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Prateek Prasanna
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Amit Gupta
- Department of Radiology-Cardiothoracic Imaging, University Hospitals, Cleveland, Ohio
| | - Pradnya Patil
- Department of Solid Tumor Oncology, Cleveland Clinic, Cleveland, Ohio
| | - Priya D Velu
- Pathology and Laboratory Medicine, Weill Cornell Medicine Physicians, New York, New York
| | - Rajat Thawani
- Department of Internal Medicine, Maimonides Medical Center, Brooklyn, New York
| | - German Corredor
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Mehdi Alilou
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Kaustav Bera
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, CWRU, Cleveland, Ohio
| | - Michael Feldman
- Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Vamsidhar Velcheti
- Department of Hematology and Oncology, NYU Langone Health, New York, New York
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio.
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, Ohio
| |
Collapse
|
39
|
Armstrong D, Chang CY, Lazarus DR, Corry D, Kheradmand F. Lung Cancer Heterogeneity in Modulation of Th17/IL17A Responses. Front Oncol 2019; 9:1384. [PMID: 31921642 PMCID: PMC6914699 DOI: 10.3389/fonc.2019.01384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
The interplay between tumors and their immune microenvironment is critical for cancer development and progression. The discovery of tumor heterogeneity has provided a window into a complex interplay between tumors, their secreted products, and host immune responses at the cellular and molecular levels. Tumor heterogeneity can also act as a driving force in promoting treatment resistance and correlates with distinct tumor-mediated acquired immune responses. A prevailing question is how genetic aberrations in solid tumors can shape the immune landscape, resulting in pro-tumor or anti-tumor activities. Here we review evidence for clinical and pathophysiological mechanisms that underlie different types of non-small cell lung cancer (NSCLC) and provide new insights for future immunomodulatory-based therapies. Some of the more common driver mutations in NSCLC heterogeneity includes the opposing immune responses in oncogenic mutations in K-ras vs. non-K-ras models and their pro-inflammatory cytokines such as interleukin (IL)17A. We will discuss possible molecular and metabolic mechanisms that may govern the opposing immune responses observed in distinct genetic models of NSCLCs. A deeper understanding of how tumor heterogeneity modulates immune response can improve current therapeutic strategies and provide precise treatment to individual lung cancer patients.
Collapse
Affiliation(s)
- Dominique Armstrong
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Cheng-Yen Chang
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Donald R Lazarus
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs, Houston, TX, United States
| | - David Corry
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States.,Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
| | - Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States.,Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States.,Biology of Inflammation Center, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
40
|
Lin C, Lin S, Guo QJ, Zong JF, Lu TZ, Lin N, Lin SJ, Pan JJ. Systemic immune-inflammation index as a prognostic marker in patients with newly diagnosed metastatic nasopharyngeal carcinoma: a propensity score-matched study. Transl Cancer Res 2019; 8:2089-2098. [PMID: 35116958 PMCID: PMC8797649 DOI: 10.21037/tcr.2019.09.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 09/04/2019] [Indexed: 01/21/2023]
Abstract
Background Systemic immune-inflammation index (SII) is significantly associated with poor survival in variety of cancers. However, SII has not yet been investigated in patients with newly diagnosed metastatic nasopharyngeal carcinoma (mNPC). Thus, our aim is to explore the role of SII in metastatic Nasopharyngeal Carcinoma. Methods Two hundred and forty-three patients with newly diagnosed mNPC were retrospectively enrolled. The Kaplan-Meier analysis and Cox regression analysis was performed to evaluate the prognostic value of SII in overall survival (OS) and progression-free survival (PFS). Heterogeneity of factors was balanced by using propensity score-matched (PSM) analysis (1:1 for high SII versus low SII). Results Kaplan-Meier analysis showed that patients with high SII were associated with poor median OS (18.0 vs. 36.0 m, P<0.001) and PFS (10.0 vs. 22.0 m, P<0.001) in mNPC. The Cox regression analysis suggested that high SII was a prognostic factor for OS (HR 1.75, 95% CI: 1.22–2.52, P=0.001) and PFS (HR 1.69, 95% CI: 1.22–2.35, P=0.002). PSM analysis still confirmed that SII was an independent marker for OS (HR 1.86, 95% CI: 1.22–2.83, P=0.004) and PFS (HR 1.84, 95% CI: 1.23–2.77, P=0.003). Conclusions SII is an independent prognostic biomarker for poor OS and PFS in patients with newly diagnosed mNPC and might be a promising tool for guiding treatment strategy decisions.
Collapse
Affiliation(s)
- Cheng Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Sheng Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Qiao-Juan Guo
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Jing-Feng Zong
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Tian-Zhu Lu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Na Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Shao-Jun Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| | - Jian-Ji Pan
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou 350000, China
| |
Collapse
|
41
|
Chow JC, Ngan RK, Cheung KM, Cho WC. Immunotherapeutic approaches in nasopharyngeal carcinoma. Expert Opin Biol Ther 2019; 19:1165-1172. [PMID: 31361154 DOI: 10.1080/14712598.2019.1650910] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Nasopharyngeal carcinoma (NPC) is endemic in Southern China and Southeast Asia. Epstein-Barr virus (EBV) represents a unique etiological culprit in the poorly differentiated nonkeratinizing and undifferentiated subtypes. EBV antigens are expressed on tumor cells albeit in a restricted manner. Treatment options for recurrent or metastatic disease are limited. Nevertheless, emerging data from immunotherapy studies in NPC have shed light into their potential antitumor efficacy. Areas covered: This article reviews existing clinical evidence for different immunotherapeutic approaches for NPC, including adoptive cellular therapy, therapeutic cancer vaccines, and immune checkpoint inhibitors. Expert opinion: There is a growing understanding on EBV virology and the immune evasion mechanisms in NPC. Immunotherapeutic strategies leveraging these properties have shown encouraging efficacy and safety results in early-phase clinical studies. Moving forward, areas to be addressed include appropriate patient selection, optimal incorporation into standard treatment paradigms, biomarker identification, as well as the development of scalable and reproducible immune product generation processes.
Collapse
Affiliation(s)
- James Ch Chow
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR , China
| | - Roger Kc Ngan
- Department of Clinical Oncology, Gleneagles Hong Kong Hospital , Hong Kong SAR, China
| | - K M Cheung
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR , China
| | - William Cs Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital , Hong Kong SAR , China
| |
Collapse
|
42
|
CT Evaluation of Squamous Cell Carcinoma of the Nasopharynx. CURRENT HEALTH SCIENCES JOURNAL 2019; 45:79-86. [PMID: 31297267 PMCID: PMC6592665 DOI: 10.12865/chsj.45.01.11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/10/2019] [Indexed: 12/11/2022]
Abstract
Squamous cell carcinoma is the most common type of neoplasia which affects the mucosa of the upper aero-digestive tract. Nasopharyngeal carcinoma is a unique disease with clinical behavior, epidemiology, and histopathology that is different from that of squamous cell carcinomas of the head and neck. For malignant tumors such as SCC, rapid growth may occur even though there are no previous clinical signs. Enlargement of a cervical lymph node, as the first presenting feature of neoplasia, is not uncommon, particularly with certain “silent” site, such as nasopharynx. Therefore, clinical examination must be complemented by radiological examination for the assessment of size, thickness and depth of the tumor, the degree of bone tissue invasion, and to detect the presence of enlarged lymph nodes. A total of 16 cases of patients were studied using CT, all diagnosed and operated with squamous cancer with localization in the nasopharynx.
Collapse
|
43
|
Xue W, Li W, Zhang T, Li Z, Wang Y, Qiu Y, Wang Y, Chen C, Fu D, Zhang M. Anti-PD1 up-regulates PD-L1 expression and inhibits T-cell lymphoma progression: possible involvement of an IFN-γ-associated JAK-STAT pathway. Onco Targets Ther 2019; 12:2079-2088. [PMID: 30962691 PMCID: PMC6433108 DOI: 10.2147/ott.s187280] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose NK/T-cell neoplasms are rare, highly aggressive, and insensitive to chemotherapy. These lymphomas have a poor prognosis, with patients being vulnerable to relapse. Hence, there is a need for alternative treatments. The purpose of this study is to investigate whether anti-PD1 takes effect on NK/T cell lymphoma. Methods The expression of PD-L1 in NK/T cell lines was investigated by flow cytometry and by Western blot. In vivo, overall survival and median survival time of mice bearing an NK/T cell line tumor was assessed. Tumor-infiltrating T cells and monocyte-derived suppressor cells were evaluated by flow cytometry. Levels of PD-L1 and components of the JAK-STAT pathway were assessed in tumor tissues by immunohistochemistry. Results NK/T cell lines had greater expression of PD-L1 than normal peripheral blood human NK cells. In vivo, anti-PD1 treatment improved overall survival and median survival time of mice bearing an NK/T cell line. Furthermore, anti-PD1 treatment increased levels of PD-L1. Cultured tumor-infiltrating lymphocytes from mice treated with anti-PD1 had greater levels of IFN-γ than cultured lymphocytes from untreated animals. Further, levels of JAK2 and STAT1 were greater in mice treated with anti-PD1. Conclusion In vivo, anti-PD1 inhibited the progression of an NK/T-cell lymphoma and up-regulated PD-L1 expression. This up-regulation may be through the IFN-γ-associated JAK-STAT pathway.
Collapse
Affiliation(s)
- Weili Xue
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Weiming Li
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Tiantian Zhang
- Toni Stephenson Lymphoma Center, City of Hope, Duarte, CA, USA
| | - Zhaoming Li
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Yingjun Wang
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Yajuan Qiu
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Yuanyuan Wang
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Changying Chen
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| | - Dongjun Fu
- New Drug Research and Development Center, School of Pharmaceutical Sciences of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Mingzhi Zhang
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Erqi District, Zhengzhou, Henan Province, China,
| |
Collapse
|
44
|
Kara İ, Çağlı S, Vural A, Yüce İ, Gündoğ M, Deniz K, Kökoğlu K. The effect of FoxP3 on tumour stage, treatment response, recurrence and survivalability in nasopharynx cancer patients. Clin Otolaryngol 2019; 44:349-355. [PMID: 30756505 DOI: 10.1111/coa.13311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/27/2019] [Accepted: 02/05/2019] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To investigate the relationship between the cell percentage of T regulator (Treg) cells of patients' specimens and disease severity, survivability, recurrence and metastasis in patients who were diagnosed with nasopharyngeal carcinoma (NPC). DESIGN, SETTING AND PARTICIPANTS Sixty patients who were diagnosed as NPC and treated by the same protocol were enrolled to the study. Patient files were reviewed retrospectively and their clinical and pathological results were recorded. Deparaffinized samples of nasopharyngeal carcinoma patients were stained immunohistochemically with anti-FoxP3 monoclonal antibody. All patients's Anti-FoxP3 stained slides were evaluated by the same pathologist. Stained Treg lymphocytes around the tumoral foci were investigated. Patients were divided into two groups according to the total anti-FoxP3-stained Treg cell counts of the specimens; that is, less than 20% of the total or more than 20% of the total. These groups were compared statistically. MAIN OUTCOME MEASURES Intensity of FoxP3 which is related to negative tumor response was the main outcome measure. It was evaluated in terms of stage, survival, recurrence and metastasis. RESULTS The study group consisted of 42 male patients (70%) and 18 female patients (30%). The mean age was 47 ± 14.9. NPC subtypes among the patients were undifferentiated non-keratinized type in 54 patients (90%), differentiated non-keratinized type in 4 patients (6.66%) and keratinized type squamous cell carcinoma (SCC) in 2 patients (3.33%). When the two groups were compared in terms of pathological subtype, there was no significant variation between the two groups. There was also no significant variation between the two groups when compared on the basis of tumor stage (P = 0.36 for T phase, P = 0.122 for N phase), early stage, late phase (P = 0.15), survival rate (P = 0.69 for general survival), recurrence (P = 0.2 for local recurrence, P = 0.37 for regional recurrence) and distant metastasis (P = 0.3). CONCLUSION There was no significant relationship between the concentration of these cells in the stained specimens and the disease stage, survival rate, recurrence and distant metastasis discovered.
Collapse
Affiliation(s)
- İrfan Kara
- ENT Clinic, Besni State Hospital, Adıyaman, Turkey
| | - Sedat Çağlı
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Alperen Vural
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - İmdat Yüce
- Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Mete Gündoğ
- Department of Radiation Oncology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Kemal Deniz
- Department of Pathology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Kerem Kökoğlu
- ENT Clinic, Develi HMK State Hospital, Kayseri, Turkey
| |
Collapse
|
45
|
Wu L, Chung YL. Tumor-Infiltrating T Cell Receptor-Beta Repertoires are Linked to the Risk of Late Chemoradiation-Induced Temporal Lobe Necrosis in Locally Advanced Nasopharyngeal Carcinoma. Int J Radiat Oncol Biol Phys 2019; 104:165-176. [PMID: 30654091 DOI: 10.1016/j.ijrobp.2019.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/18/2018] [Accepted: 01/05/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Temporal lobe necrosis (TLN), a late complication of nasopharyngeal carcinoma (NPC) after concurrent chemoradiotherapy (CCRT), causes permanent neurologic deficits. We aimed to investigate the risk factors for the development of CCRT-induced TLN in locally advanced NPC patients. METHODS AND MATERIALS The incidence of CCRT-induced TLN was assessed in consecutive patients with NPC initially staged with T3-4N0-3M0 receiving curative intensity modulated radiation therapy (IMRT) and cisplatin-based chemotherapy with long-term follow-up. The TLN risk was evaluated with radiation dose-volume histograms (a dosimetric risk indicator of organ injury) and the dynamics of blood circulating neutrophil-to-lymphocyte ratios (a clinical indicator of systemic inflammation) by linear and logistic regression models. High-throughput unbiased T cell receptor-beta (TCRbeta) sequencing was performed to correlate the different TCRbeta repertoires of NPC-infiltrating lymphocytes (a biological factor of the immune microenvironment) with TLN incidence. RESULTS In the era of modern IMRT-based CCRT, radiation doses of up to 74 Gy achieved local control rates of more than 90% in both T3 and T4 diseases but still induced a remarkably higher incidence of TLN in the T4 patients (30.14%) compared with the rare incidence of TLN observed in the T3 patients (2.78%) (P < .0001). We found that in the T4 NPC patients, univariate and multivariate analyses showed the radiation tolerance dose-volume effect was not an absolutely independent factor influencing TLN occurrence. However, increased TLN risk was observed in association with higher pre-CCRT baseline and post-CCRT neutrophil-to-lymphocyte ratios. There was also a link between intratumoral TCRbeta repertoire subtypes and TLN incidence. Combining the inherent TCRbeta genomic susceptibility with the clinical variable neutrophil-to-lymphocyte ratio better predicted the risk of TLN for T4 NPC patients after CCRT. CONCLUSIONS The associations of tumor-infiltrating lymphocyte repertoires and blood circulating neutrophil-to-lymphocyte ratios with TLN occurrence in T4 NPC patients suggest that the immune and inflammatory milieus play roles in the late brain damage caused by CCRT. Modulated or provoked by CCRT locally and systemically, the reciprocal interactions of neutrophils and lymphocytes in the intracranial NPC-associated immune microenvironment could be a key driver of chronic TLN pathogenesis.
Collapse
Affiliation(s)
- LiFu Wu
- Department of Radiation Oncology, Koo Foundation Sun-Yat-Sen Cancer Center, Taipei, Taiwan; Medical Physics and Informatics Laboratory of Electronics Engineering, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan
| | - Yih-Lin Chung
- Department of Radiation Oncology, Koo Foundation Sun-Yat-Sen Cancer Center, Taipei, Taiwan.
| |
Collapse
|
46
|
Ye J, Zou MM, Li P, Lin XJ, Jiang QW, Yang Y, Huang JR, Yuan ML, Xing ZH, Wei MN, Li Y, Shi Z, Liu H. Oxymatrine and Cisplatin Synergistically Enhance Anti-tumor Immunity of CD8 + T Cells in Non-small Cell Lung Cancer. Front Oncol 2018; 8:631. [PMID: 30619765 PMCID: PMC6305450 DOI: 10.3389/fonc.2018.00631] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/04/2018] [Indexed: 12/31/2022] Open
Abstract
Oxymatrine (OMT) has shown broad antitumor activities for the treatment of several types of cancers. However, little is known about its effect on anti-tumor immunity. Combination therapy is a potentially promising strategy of cancer to enhance anticancer activity, overcome drug resistance, and lower treatment failure rate. In the present study, we demonstrated that the combination of OMT with cisplatin (DDP) synergistically inhibited non-small cell lung cancer (NSCLC) cells growth when co-cultured with peripheral blood mononuclear cells in vitro. Furthermore, the combination of OMT with DDP significantly inhibited the growth of Lewis lung cancer (LLC) mouse xenograft tumors. Flow cytometry analysis revealed that OMT and DDP synergistically increase the CD8+/ regulatory T cells ratio and enhanced more CD8+ T cells secreted cytokines of IFN-γ, TNF-α, and IL-2 in vivo. Mechanistically, upregulation of miR-155 and downregulation of suppressor of cytokine signaling-1 (SOCS1) were confirmed as a target signaling pathway to positively regulate the anti-tumor response of CD8+ T cells. Overall, OMT in combination with DDP showed outstanding synergistic anti-tumor immunity, suggesting that this beneficial combination may offer a potential immunotherapy for NSCLC patients.
Collapse
Affiliation(s)
- Jin Ye
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Man-Man Zou
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Pei Li
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xi-Jun Lin
- Department of Otolaryngology-Head and Neck Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qi-Wei Jiang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yang Yang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jia-Rong Huang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Meng-Ling Yuan
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zi-Hao Xing
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Meng-Ning Wei
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yao Li
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhi Shi
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Hui Liu
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
47
|
Lazarus J, Maj T, Smith JJ, Perusina Lanfranca M, Rao A, D'Angelica MI, Delrosario L, Girgis A, Schukow C, Shia J, Kryczek I, Shi J, Wasserman I, Crawford H, Nathan H, Pasca Di Magliano M, Zou W, Frankel TL. Spatial and phenotypic immune profiling of metastatic colon cancer. JCI Insight 2018; 3:121932. [PMID: 30429368 DOI: 10.1172/jci.insight.121932] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 10/11/2018] [Indexed: 12/14/2022] Open
Abstract
Paramount to the efficacy of immune checkpoint inhibitors is proper selection of patients with adequate tumor immunogenicity and a robust but suppressed immune infiltrate. In colon cancer, immune-based therapies are approved for patients with DNA mismatch repair (MMR) deficiencies, in whom accumulation of genetic mutations results in increased neoantigen expression, triggering an immune response that is suppressed by the PD-L1/PD-1 pathway. Here, we report that characterization of the microenvironment of MMR-deficient metastatic colorectal cancer using multiplex fluorescent immunohistochemistry (mfIHC) identified increased infiltration of cytotoxic T lymphocytes (CTLs), which were more often engaged with epithelial cells (ECs) and improved overall survival. A subset of patients with intact MMR but a similar immune microenvironment to MMR-deficient patients was identified and found to universally express high levels of PD-L1, suggesting that they may represent a currently untreated, checkpoint inhibitor-responsive population. Further, PD-L1 expression on antigen-presenting cells (APCs) in the tumor microenvironment (TME) resulted in impaired CTL/EC engagement and enhanced infiltration and engagement of Tregs. Characterization of the TME by mfIHC highlights the interconnection between immunity and immunosuppression in metastatic colon cancer and may better stratify patients for receipt of immunotherapies.
Collapse
Affiliation(s)
- Jenny Lazarus
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Tomasz Maj
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - J Joshua Smith
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Arvind Rao
- Department of Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael I D'Angelica
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Alexander Girgis
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Casey Schukow
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Isaac Wasserman
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Howard Crawford
- Department of Molecular and Cellular Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Hari Nathan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Weiping Zou
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Pathology and
| | - Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
48
|
Chraa D, Naim A, Olive D, Badou A. T lymphocyte subsets in cancer immunity: Friends or foes. J Leukoc Biol 2018; 105:243-255. [DOI: 10.1002/jlb.mr0318-097r] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/15/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Dounia Chraa
- Cellular and Molecular Pathology LaboratoryFaculty of Medicine and Pharmacy of CasablancaHassan II University Casablanca Morocco
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS, UMR7258Institut Paoli‐CalmettesAix‐Marseille University, UM 105 Marseille France
| | - Asmaa Naim
- Cellular and Molecular Pathology LaboratoryFaculty of Medicine and Pharmacy of CasablancaHassan II University Casablanca Morocco
- University Mohammed VI for Health ScienceCheick Khalifa Hospital Casablanca Morocco
| | - Daniel Olive
- Team Immunity and Cancer, Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, CNRS, UMR7258Institut Paoli‐CalmettesAix‐Marseille University, UM 105 Marseille France
| | - Abdallah Badou
- Cellular and Molecular Pathology LaboratoryFaculty of Medicine and Pharmacy of CasablancaHassan II University Casablanca Morocco
| |
Collapse
|
49
|
Jin YB, Luo W, Zhang GY, Lin KR, Cui JH, Chen XP, Pan YM, Mao XF, Tang J, Wang YJ. TCR repertoire profiling of tumors, adjacent normal tissues, and peripheral blood predicts survival in nasopharyngeal carcinoma. Cancer Immunol Immunother 2018; 67:1719-1730. [PMID: 30155576 PMCID: PMC11028245 DOI: 10.1007/s00262-018-2237-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/23/2018] [Indexed: 12/29/2022]
Abstract
The T-cell immune responses in nasopharyngeal carcinoma patients have been extensively investigated recently for designing adoptive immunotherapy or immune checkpoint blockade therapy. However, the distribution characteristics of T cells associated with NPC pathogenesis are largely unknown. We performed deep sequencing for TCR repertoire profiling on matched tumor/adjacent normal tissue from 15 NPC patients and peripheral blood from 39 NPC patients, 39 patients with other nasopharyngeal diseases, and 33 healthy controls. We found that a lower diversity of TCR repertoire in tumors than paired tissues or a low similarity between the paired tissues was associated with a poor prognosis in NPC. A more diverse TCR repertoire was identified in the peripheral blood of NPC patients relative to the controls; this was related to a significant decrease in the proportion of high-frequency TCR clones in NPC. Higher diversity in peripheral blood of NPC patients was associated with a worse prognosis. Due to the peculiarity of the Vβ gene usage patterns in the peripheral blood of NPC patients, 15 Vβ genes were selected to distinguish NPC patients from controls by the least absolute shrinkage and selection operator analysis. We identified 11 clonotypes shared by tumors and peripheral blood samples from different NPC patients, defined as "NPC-associated" that might have value in adoptive immunotherapy. In conclusion, we here report the systematic and overall characteristics of the TCR repertoire in tumors, adjacent normal tissues, and peripheral blood of NPC patients. The data obtained may be relevant to future clinical studies in the setting of immunotherapy for NPC patients.
Collapse
Affiliation(s)
- Ya-Bin Jin
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Wei Luo
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China.
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China.
| | - Guo-Yi Zhang
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Cancer Center, Foshan Hospital, Sun Yat-sen University, Foshan, 528000, Guangdong, China
| | - Kai-Rong Lin
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Jin-Huan Cui
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Xiang-Ping Chen
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Ying-Ming Pan
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Xiao-Fan Mao
- Foshan Hospital, Clinical Research Institute, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
| | - Jun Tang
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China
- Otolaryngology Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, Foshan, 528000, Guangdong, China
| | - Yue-Jian Wang
- Head and Neck Cancer Research, Department of Otolaryngology-Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, #81, North of Lingnan Ave, Foshan, 528000, Guangdong, China.
- Otolaryngology Head and Neck Surgery, Foshan Hospital, Sun Yat-sen University, Foshan, 528000, Guangdong, China.
| |
Collapse
|
50
|
The Microenvironment in Epstein-Barr Virus-Associated Malignancies. Pathogens 2018; 7:pathogens7020040. [PMID: 29652813 PMCID: PMC6027429 DOI: 10.3390/pathogens7020040] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 12/27/2022] Open
Abstract
The Epstein–Barr virus (EBV) can cause a wide variety of cancers upon infection of different cell types and induces a highly variable composition of the tumor microenvironment (TME). This TME consists of both innate and adaptive immune cells and is not merely an aspecific reaction to the tumor cells. In fact, latent EBV-infected tumor cells utilize several specific mechanisms to form and shape the TME to their own benefit. These mechanisms have been studied largely in the context of EBV+ Hodgkin lymphoma, undifferentiated nasopharyngeal carcinoma, and EBV+ gastric cancer. This review describes the composition, immune escape mechanisms, and tumor cell promoting properties of the TME in these three malignancies. Mechanisms of susceptibility which regularly involve genes related to immune system function are also discussed, as only a small proportion of EBV-infected individuals develops an EBV-associated malignancy.
Collapse
|