1
|
Zhao Z, Ma X, Cai Z. The potential role of CD8+ cytotoxic T lymphocytes and one branch connected with tissue-resident memory in non-luminal breast cancer. PeerJ 2024; 12:e17667. [PMID: 39006029 PMCID: PMC11246025 DOI: 10.7717/peerj.17667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/11/2024] [Indexed: 07/16/2024] Open
Abstract
Advances in understanding the pathological mechanisms of breast cancer have resulted in the emergence of novel therapeutic strategies. However, triple-negative breast cancer (TNBC), a molecular subtype of breast cancer with a poor prognosis, lacks classical and general therapeutic targets, hindering the clinical application of several therapies to breast cancer. As insights into the unique immunity and molecular mechanisms of TNBC have become more extensive, immunotherapy has gradually become a valuable complementary approach to classical radiotherapy and chemotherapy. CD8+ cells are significant actors in the tumor immunity cycle; thus, research on TNBC immunotherapy is increasingly focused in this direction. Recently, CD8+ tissue-resident memory (TRM) cells, a subpopulation of CD8+ cells, have been explored in relation to breast cancer and found to seemingly play an undeniably important role in tumor surveillance and lymphocytic infiltration. In this review, we summarize the recent advances in the mechanisms and relative targets of CD8+ T cells, and discuss the features and potential applications of CD8+ TRM cells in non-luminal breast cancer immunotherapy.
Collapse
Affiliation(s)
- Ziqi Zhao
- Department of Breast Cancer, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Xinyu Ma
- Department of Breast Cancer, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Zhengang Cai
- Department of Breast Cancer, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
2
|
Shahjahan, Dey JK, Dey SK. Translational bioinformatics approach to combat cardiovascular disease and cancers. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 139:221-261. [PMID: 38448136 DOI: 10.1016/bs.apcsb.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Bioinformatics is an interconnected subject of science dealing with diverse fields including biology, chemistry, physics, statistics, mathematics, and computer science as the key fields to answer complicated physiological problems. Key intention of bioinformatics is to store, analyze, organize, and retrieve essential information about genome, proteome, transcriptome, metabolome, as well as organisms to investigate the biological system along with its dynamics, if any. The outcome of bioinformatics depends on the type, quantity, and quality of the raw data provided and the algorithm employed to analyze the same. Despite several approved medicines available, cardiovascular disorders (CVDs) and cancers comprises of the two leading causes of human deaths. Understanding the unknown facts of both these non-communicable disorders is inevitable to discover new pathways, find new drug targets, and eventually newer drugs to combat them successfully. Since, all these goals involve complex investigation and handling of various types of macro- and small- molecules of the human body, bioinformatics plays a key role in such processes. Results from such investigation has direct human application and thus we call this filed as translational bioinformatics. Current book chapter thus deals with diverse scope and applications of this translational bioinformatics to find cure, diagnosis, and understanding the mechanisms of CVDs and cancers. Developing complex yet small or long algorithms to address such problems is very common in translational bioinformatics. Structure-based drug discovery or AI-guided invention of novel antibodies that too with super-high accuracy, speed, and involvement of considerably low amount of investment are some of the astonishing features of the translational bioinformatics and its applications in the fields of CVDs and cancers.
Collapse
Affiliation(s)
- Shahjahan
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India
| | - Joy Kumar Dey
- Central Council for Research in Homoeopathy, Ministry of Ayush, Govt. of India, New Delhi, Delhi, India
| | - Sanjay Kumar Dey
- Laboratory for Structural Biology of Membrane Proteins, Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, Delhi, India.
| |
Collapse
|
3
|
Yang J, Qiu L, Wang X, Chen X, Cao P, Yang Z, Wen Q. Liquid biopsy biomarkers to guide immunotherapy in breast cancer. Front Immunol 2023; 14:1303491. [PMID: 38077355 PMCID: PMC10701691 DOI: 10.3389/fimmu.2023.1303491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) therapy has emerged as a promising treatment strategy for breast cancer (BC). However, current reliance on immunohistochemical (IHC) detection of PD-L1 expression alone has limited predictive capability, resulting in suboptimal efficacy of ICIs for some BC patients. Hence, developing novel predictive biomarkers is indispensable to enhance patient selection for immunotherapy. In this context, utilizing liquid biopsy (LB) can provide supplementary or alternative value to PD-L1 IHC testing for identifying patients most likely to benefit from immunotherapy and exhibit favorable responses. This review discusses the predictive and prognostic value of LB in breast cancer immunotherapy, as well as its limitations and future directions. We aim to promote the individualization and precision of immunotherapy in BC by elucidating the role of LB in clinical practice.
Collapse
Affiliation(s)
- Jinghan Yang
- Department of Biological Science, Vanderbilt University, Nashville, TN, United States
| | - Liang Qiu
- Department of Radiation Oncology, Stanford University, Palo Alto, CA, United States
| | - Xi Wang
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xi Chen
- Department of Human Resource, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Pingdong Cao
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhe Yang
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiang Wen
- Department of Radiation Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
4
|
Cao B, Zhang Z, Wang C, Lv X. Prognostic relevance of tumor‑infiltrating lymphocytes in residual tumor tissue from patients with triple‑negative breast cancer following neoadjuvant chemotherapy: A systematic review and meta‑analysis. Oncol Lett 2023; 26:441. [PMID: 37664648 PMCID: PMC10472026 DOI: 10.3892/ol.2023.14028] [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: 06/25/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
Further adjuvant chemotherapy treatment can provide benefits to certain patients with triple-negative breast cancer (TNBC) that fail to achieve pathological complete response (pCR) after the administration of a neoadjuvant chemotherapy (NAC) regimen. However, biomarkers suitable for identifying patients likely to experience poor prognostic outcomes after undergoing additional adjuvant chemotherapy are currently lacking. Accordingly, the present meta-analysis was conducted to explore the relationship between tumor-infiltrating lymphocytes (TILs) or TIL subtypes (CD4+ or CD8+) in residual tumor (RT) tissue following NAC and TNBC patient prognosis. Relevant studies published through March 2023 were identified in Pubmed, The Cochrane Library, Embase and Web of Science databases. After excluding irrelevant studies, data were extracted from the remaining reports, while study quality was analyzed with the Newcastle-Ottawa Scale. Subsequent analyses were performed with Stata 14.0 and Review Manager 5.3. In total, seven relevant studies incorporating 1,202 patients were identified, all of which were retrospective cohort studies. Pooled analyses demonstrated that those patients exhibiting higher levels of RT TIL infiltration following NAC exhibited significantly improved recurrence-free, metastasis-free and event-free survival (RFS/MFS/EFS) compared with patients with lower RT TIL infiltration levels, together with an improved distant recurrence-free interval (DRFI) [hazard ratio (HR)=0.52; 95% confidence interval (CI)=0.39-0.69; P<0.00001]. In addition, patients exhibiting high RT TIL infiltration exhibited improved overall survival (OS) and breast cancer-specific survival (BCSS; HR=0.49; 95% CI=0.38-0.65; P<0.00001). Additional subgroup analyses revealed that patients with higher TIL infiltration levels or TIL subtype (CD4+ or CD8+) infiltration exhibited improved RFS/MFS/EFS/DRFI as compared with patients with lower levels of overall TIL or TIL subtype (CD4+ or CD8+) infiltration in RT tissue (HR=0.35, 95% CI=0.20-0.59, P<0.0001; HR=0.49, 95% CI=0.33-0.71, P=0.0002). Consistently, the OS/BCSS of patients exhibiting high levels of overall TIL or TIL subtype (CD4+ or CD8+) infiltration was increased compared with patients with lower levels of such infiltration (HR=0.33, 95% CI=0.19-0.59, P=0.0002; HR=0.55, 95% CI=0.41-0.76, P=0.0002). These data thus demonstrate that levels of overall TIL infiltration or infiltration by CD4+ or CD8+ TILs in RT following NAC can be used as a biomarker to reliably predict prognostic outcomes in patients with TNBC, in addition to highlighting possible targets that may guide the further immunotherapeutic management of these patients.
Collapse
Affiliation(s)
- Bo Cao
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children's Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Ziran Zhang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children's Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Chaoxian Wang
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children's Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiang Lv
- Department of Breast Diseases, Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children's Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
5
|
Tyagi T, Jain K, Yarovinsky TO, Chiorazzi M, Du J, Castro C, Griffin J, Korde A, Martin KA, Takyar SS, Flavell RA, Patel AA, Hwa J. Platelet-derived TLT-1 promotes tumor progression by suppressing CD8+ T cells. J Exp Med 2023; 220:e20212218. [PMID: 36305874 DOI: 10.1084/jem.20212218] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/25/2022] [Accepted: 10/05/2022] [Indexed: 01/19/2023] Open
Abstract
Current understanding of tumor immunosuppressive mechanisms forms the basis for modern day immunotherapies. Immunoregulatory role of platelets in cancer remains largely elusive. Platelets from non-small cell lung cancer (NSCLC) patients revealed a distinct activation phenotype. TREM-like transcript 1 (TLT-1), a platelet protein, was increased along with enhanced extracellular release from NSCLC platelets. The increased platelet TLT-1 was also evident in humanized mice with patient-derived tumors. In immunocompetent mice with syngeneic tumors, TLT-1 binding to T cells, in vivo, led to suppression of CD8 T cells, promoting tumor growth. We identified direct interaction between TLT-1 and CD3ε on T cells, implicating the NF-κB pathway in CD8 T cell suppression. Anti-TLT-1 antibody rescued patients' T cells from platelet-induced suppression ex vivo and reduced tumors in mice in vivo. Clinically, higher TLT-1 correlated with reduced survival of NSCLC patients. Our findings thus identify TLT-1 as a platelet-derived immunosuppressor that suppresses CD8 T cells and demonstrate its therapeutic and prognostic significance in cancer.
Collapse
Affiliation(s)
- Tarun Tyagi
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Kanika Jain
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Timur O Yarovinsky
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Michael Chiorazzi
- Department of Immunobiology, Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT
- Yale Cancer Center, New Haven, CT
| | - Jing Du
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Cecilia Castro
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Jules Griffin
- Department of Biochemistry, Cambridge University, Cambridge, UK
| | - Asawari Korde
- Pulmonary Critical Care, Yale Internal Medicine, New Haven, CT
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Shervin S Takyar
- Pulmonary Critical Care, Yale Internal Medicine, New Haven, CT
- Yale Cancer Center, New Haven, CT
| | - Richard A Flavell
- Department of Immunobiology, Howard Hughes Medical Institute, Yale School of Medicine, New Haven, CT
- Yale Cancer Center, New Haven, CT
| | - Abhijit A Patel
- Yale Therapeutic Radiology, Yale Cancer Center, New Haven, CT
- Yale Cancer Center, New Haven, CT
| | - John Hwa
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Yale Cancer Center, New Haven, CT
| |
Collapse
|
6
|
Muraro E, Del Ben F, Turetta M, Cesselli D, Bulfoni M, Zamarchi R, Rossi E, Spazzapan S, Dolcetti R, Steffan A, Brisotto G. Clinical relevance of the combined analysis of circulating tumor cells and anti-tumor T-cell immunity in metastatic breast cancer patients. Front Oncol 2022; 12:983887. [PMID: 36081561 PMCID: PMC9446887 DOI: 10.3389/fonc.2022.983887] [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: 07/01/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Background Metastatic breast cancer (mBC) is a heterogeneous disease with varying responses to treatments and clinical outcomes, still requiring the identification of reliable predictive biomarkers. In this context, liquid biopsy has emerged as a powerful tool to assess in real-time the evolving landscape of cancer, which is both orchestrated by the metastatic process and immune-surveillance mechanisms. Thus, we investigated circulating tumor cells (CTCs) coupled with peripheral T-cell immunity to uncover their potential clinical relevance in mBC. Methods A cohort of 20 mBC patients was evaluated, before and one month after starting therapy, through the following liquid biopsy approaches: CTCs enumerated by a metabolism-based assay, T-cell responses against tumor-associated antigens (TAA) characterized by interferon-γ enzyme-linked immunosorbent spot (ELISpot), and the T-cell receptor (TCR) repertoire investigated by a targeted next-generation sequencing technique. TCR repertoire features were characterized by the Morisita’s overlap and the Productive Simpson Clonality indexes, and the TCR richness. Differences between groups were calculated by Fisher’s, Mann-Whitney or Kruskal-Wallis test, as appropriate. Prognostic data analysis was estimated by Kaplan-Meier method. Results Stratifying patients for their prognostic level of 6 CTCs before therapy, TAA specific T-cell responses were detected only in patients with a low CTC level. By analyzing the TCR repertoire, the highest TCR clonality was observed in the case of CTCs under the cut-off and a positive ELISpot response (p=0.03). Whereas, at follow-up, patients showing a good clinical response coupled with a low number of CTCs were characterized by the most elevated TCR clonality (p<0.05). The detection of CTCs≥6 in at least one time-point was associated with a lower TCR clonality (p=0.02). Intriguingly, by combining overall survival analysis with TCR repertoire, we highlighted a potential prognostic role of the TCR clonality measured at follow-up (p=0.03). Conclusion These data, whether validated in a larger cohort of patients, suggest that the combined analysis of CTCs and circulating anti-tumor T-cell immunity could represent a valuable immune-oncological biomarker for the liquid biopsy field. The clinical application of this promising tool could improve the management of mBC patients, especially in the setting of immunotherapy, a rising approach for BC treatment requiring reliable predictive biomarkers.
Collapse
Affiliation(s)
- Elena Muraro
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
- *Correspondence: Elena Muraro,
| | - Fabio Del Ben
- Department of Medicine, University of Udine, Udine, Italy
| | - Matteo Turetta
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Daniela Cesselli
- Department of Medicine, University of Udine, Udine, Italy
- Institute of Pathology, University Hospital of Udine (Azienda sanitaria universitaria Friuli Centrale, ASUFC), Udine, Italy
| | - Michela Bulfoni
- Institute of Pathology, University Hospital of Udine (Azienda sanitaria universitaria Friuli Centrale, ASUFC), Udine, Italy
| | - Rita Zamarchi
- Department of Surgery, Oncology & Gastroenterology, University of Padova, Padua, Italy
| | - Elisabetta Rossi
- Department of Surgery, Oncology & Gastroenterology, University of Padova, Padua, Italy
- Veneto Institute of Oncology IOV - Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Simon Spazzapan
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Riccardo Dolcetti
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
- The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| | - Giulia Brisotto
- Immunopathology and Cancer Biomarkers Units, Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico, Aviano, Italy
| |
Collapse
|
7
|
Wang Z, Zhong Y, Zhang Z, Zhou K, Huang Z, Yu H, Liu L, Liu S, Yang H, Zhou J, Fan J, Wu L, Sun Y. Characteristics and Clinical Significance of T-Cell Receptor Repertoire in Hepatocellular Carcinoma. Front Immunol 2022; 13:847263. [PMID: 35371059 PMCID: PMC8965762 DOI: 10.3389/fimmu.2022.847263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Several studies have demonstrated that the T-cell receptor (TCR) repertoire is associated with prognosis and immune therapy response in several types of cancer. However, the comprehensive features of TCR repertoire in tumor-infiltrating and circulating T cells, as well as its clinical significance of diagnosis in hepatocellular carcinoma (HCC) patients, are still unknown. In this study, we perform paired tumor/peritumoral tissues and peripheral blood samples from 58 patients with HCC and sequenced them with high-throughput TCR to comprehensively analyze the characteristics of TCR and the clinical significance of peripheral TCR sequence. By exploring the abundance and diversity of TCR repertoires, we observe that there was a significantly higher TCR diversity in peripheral blood than in tumoral and peritumoral tissues, while tumoral and peritumoral tissues showed similar TCR diversity. A substantial difference in the usage frequencies of several Vβ, Jβ genes, and TCRβ VJ pairings was found among three types of tissues. Moreover, we reveal that HCC patients have a unique profile of TCR repertoire in peripheral blood in contrast to healthy individuals. We further establish an HCC diagnostic model based on TCRβ VJ pairing usage in peripheral blood, which yields a best-fit area under the curve (AUC) of 0.9746 ± 0.0481 (sensitivity = 0.9675 ± 0.0603, specificity = 0.9998 ± 0.0007, average of 100 repeats) in the test set. Our study describes the characteristics of tissue infiltration and circulating T-cell bank in patients with HCC and shows the potential of using circulating TCR sequence as a biomarker for the non-invasive diagnosis of patients with HCC.
Collapse
Affiliation(s)
- Zifei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
| | - Yu Zhong
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
| | - Zefan Zhang
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Kaiqian Zhou
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Zhihao Huang
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
| | - Hao Yu
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
| | - Longqi Liu
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| | - Shiping Liu
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Liang Wu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Beijing Genomics Institute at Shenzhen, Shenzhen, China
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen, China
| | - Yunfan Sun
- Zhong-Hua Precision Medical Center, Zhongshan Hospital, Fudan University-BGI, Shanghai, China
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| |
Collapse
|
8
|
Zhang J, Pan S, Jian C, Hao L, Dong J, Sun Q, Jin H, Han X. Immunostimulatory Properties of Chemotherapy in Breast Cancer: From Immunogenic Modulation Mechanisms to Clinical Practice. Front Immunol 2022; 12:819405. [PMID: 35069604 PMCID: PMC8766762 DOI: 10.3389/fimmu.2021.819405] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer (BC) is the most common malignancy among females. Chemotherapy drugs remain the cornerstone of treatment of BC and undergo significant shifts over the past 100 years. The advent of immunotherapy presents promising opportunities and constitutes a significant complementary to existing therapeutic strategies for BC. Chemotherapy as a cytotoxic treatment that targets proliferation malignant cells has recently been shown as an effective immune-stimulus in multiple ways. Chemotherapeutic drugs can cause the release of damage-associated molecular patterns (DAMPs) from dying tumor cells, which result in long-lasting antitumor immunity by the key process of immunogenic cell death (ICD). Furthermore, Off-target effects of chemotherapy on immune cell subsets mainly involve activation of immune effector cells including natural killer (NK) cells, dendritic cells (DCs), and cytotoxic T cells, and depletion of immunosuppressive cells including Treg cells, M2 macrophages and myeloid-derived suppressor cells (MDSCs). Current mini-review summarized recent large clinical trials regarding the combination of chemotherapy and immunotherapy in BC and addressed the molecular mechanisms of immunostimulatory properties of chemotherapy in BC. The purpose of our work was to explore the immune-stimulating effects of chemotherapy at the molecular level based on the evidence from clinical trials, which might be a rationale for combinations of chemotherapy and immunotherapy in BC.
Collapse
Affiliation(s)
- Jinguo Zhang
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Shuaikang Pan
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Chen Jian
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Li Hao
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Jie Dong
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Qingqing Sun
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongwei Jin
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Xinghua Han
- Department of Medical Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
9
|
Cai G, Guan Z, Jin Y, Su Z, Chen X, Liu Q, Wang C, Yin X, Zhang L, Ye G, Luo W. Circulating T-Cell Repertoires Correlate With the Tumor Response in Patients With Breast Cancer Receiving Neoadjuvant Chemotherapy. JCO Precis Oncol 2022; 6:e2100120. [PMID: 35025620 PMCID: PMC8769146 DOI: 10.1200/po.21.00120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 10/11/2021] [Accepted: 12/10/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Neoadjuvant chemotherapy (NAC) has been widely used in patients with breast cancer to minish tumor burden and increase resection rate of cancer. T-cell repertoire has been believed to be able to monitor antitumor immune responses. This study aimed to explore the dynamic change of T-cell repertoire and its clinical value in evaluating the tumor response in patients with breast cancer receiving NAC. MATERIALS AND METHODS Ninety-four patients who underwent NAC before surgery were recruited, and peripheral blood samples were collected at multiple time points during NAC. High-throughput T-cell receptor (TCR)-β sequencing was used to characterize the T-cell repertoire of every sample and analyzed the changes in circulating T-cell repertoire during NAC. RESULTS We found that the diversity of TCR repertoires was associated with age and clinical stage of the patients with breast cancer. The distribution of Vβ and Jβ genes in TCR repertoires was skewed in patients with human epidermal growth factor receptor 2-positive (HER2+) breast cancer. Vβ20.1 and Vβ30 expression levels before NAC correlate with tumor response after all cycles of NAC in HER2- and HER2+ patients, respectively. Some CDR3 motifs that correlated with clinical response in either HER2+ or HER2- patients were identified. Besides, TCR repertoire evolved during NAC and the diversity of TCR repertoire decreased more after two cycles of NAC in patients with good tumor response after all cycles of NAC (P = .0061). CONCLUSION Our results demonstrated that TCR repertoire correlated with the characteristics of the tumor, such as the expression status of HER2. Moreover, some characteristics of TCR repertoires that correlated with clinical response were identified and they might provide useful information to tailor therapeutic regimens at the early cycle of NAC.
Collapse
Affiliation(s)
- Gengxi Cai
- The First People's Hospital of Foshan, Foshan, China
| | - Zhanwen Guan
- The First People's Hospital of Foshan, Foshan, China
| | - Yabin Jin
- The First People's Hospital of Foshan, Foshan, China
| | - Zuhui Su
- The First People's Hospital of Foshan, Foshan, China
| | | | - Qing Liu
- The First People's Hospital of Foshan, Foshan, China
| | | | - Xiaoxia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou, China
| | - Lifang Zhang
- The First People's Hospital of Foshan, Foshan, China
| | - Guolin Ye
- The First People's Hospital of Foshan, Foshan, China
| | - Wei Luo
- The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
10
|
Su Z, Jin Y, Zhang Y, Guan Z, Li H, Chen X, Xie C, Zhang C, Liu X, Li P, Ye P, Zhang L, Kong Y, Luo W. The Diagnostic and Prognostic Potential of the B-Cell Repertoire in Membranous Nephropathy. Front Immunol 2021; 12:635326. [PMID: 34122405 PMCID: PMC8190383 DOI: 10.3389/fimmu.2021.635326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
Membranous nephropathy (MN), an autoimmune glomerular disease, is one of the most common causes of nephrotic syndrome in adults. In current clinical practice, the diagnosis is dependent on renal tissue biopsy. A new method for diagnosis and prognosis surveillance is urgently needed for patients. In the present study, we recruited 66 MN patients before any treatment and 11 healthy control (HC) and analyzed multiple aspects of the immunoglobulin heavy chain (IGH) repertoire of these samples using high-throughput sequencing. We found that the abnormalities of CDR-H3 length, hydrophobicity, somatic hypermutation (SHM), and germ line index were progressively more prominent in patients with MN, and the frequency of IGHV3-66 in post-therapy patients was significantly lower than that in pre-therapy patients. Moreover, we found that the IGHV3-38 gene was significantly related to PLA2R, which is the most commonly used biomarker. The most important discovery was that several IGHV, IGHD transcripts, CDR-H3 length, and SHM rate in pre-therapy patients had the potential to predict the therapeutic effect. Our study further demonstrated that the IGH repertoire could be a potential biomarker for prognosis prediction of MN. The landscape of circulating B-lymphocyte repertoires sheds new light on the detection and surveillance of MN.
Collapse
Affiliation(s)
- Zuhui Su
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Yabin Jin
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Yu Zhang
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Zhanwen Guan
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Huishi Li
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Xiangping Chen
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Chao Xie
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Chuling Zhang
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Xiaofen Liu
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Peixian Li
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Peiyi Ye
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Lifang Zhang
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Yaozhong Kong
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
11
|
Yin XX, Jin Y, Gao M, Hadjiloucas S. Artificial Intelligence in Breast MRI Radiogenomics: Towards Accurate Prediction of Neoadjuvant Chemotherapy Responses. Curr Med Imaging 2021; 17:452-458. [PMID: 32842944 DOI: 10.2174/1573405616666200825161921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/03/2020] [Accepted: 07/17/2020] [Indexed: 11/22/2022]
Abstract
Neoadjuvant Chemotherapy (NAC) in breast cancer patients has considerable prognostic and treatment potential and can be tailored to individual patients as part of precision medicine protocols. This work reviews recent advances in artificial intelligence so as to enable the use of radiogenomics for accurate NAC analysis and prediction. The work addresses a new problem in radiogenomics mining: How to combine structural radiomics information and non-structural genomics information for accurate NAC prediction. This requires the automated extraction of parameters from structural breast radiomics data, and finding non-structural feature vectors with diagnostic value, which then are combined with genomics data acquired from exocrine bodies in blood samples from a cohort of cancer patients to enable accurate NAC prediction. A self-attention-based deep learning approach, along with an effective multi-channel tumour image reconstruction algorithm of high dimensionality, is proposed. The aim was to generate non-structural feature vectors for accurate prediction of the NAC responses by combining imaging datasets with exocrine body related genomics analysis.
Collapse
Affiliation(s)
- Xiao-Xia Yin
- Cyberspace Institute of Advanced Technology, Guangzhou University, Guangzhou 510006, China
| | - Yabin Jin
- The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China
| | - Mingyong Gao
- The First People's Hospital of FoShan (Affiliated FoShan Hospital of Sun Yat-sen University), Foshan 528000, China
| | - Sillas Hadjiloucas
- Department of Biomedical Engineering, The University of Reading, RG6 6AY, United Kingdom
| |
Collapse
|
12
|
Chobrutskiy BI, Chobrutskiy A, Zaman S, Yeagley M, Huda TI, Blanck G. High-throughput, sliding-window algorithm for assessing chemical complementarity between immune receptor CDR3 domains and cancer mutant peptides: TRG-PIK3CA interactions and breast cancer. Mol Immunol 2021; 135:247-253. [PMID: 33933816 DOI: 10.1016/j.molimm.2021.02.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/03/2021] [Accepted: 02/24/2021] [Indexed: 01/25/2023]
Abstract
Physicochemical assessments of a vast accumulation of adaptive immune receptor (IR) recombinations have led to correlations of those properties with sub-divisions of various diseases. In the cancer setting, such assessments, particularly for the complementarity determining region-3 (CDR3) immune receptor domain, have been used to establish chemical complementarity matches to mutant amino acids (AA). These matches, in some cases, over very large numbers of tumor samples, have correlated with survival and gene expression distinctions. For example, in melanoma, electrostatic charge based, T-cell receptor CDR3-DNAH9 mutant AA complementarity represents better survival over multiple datasets that represent tumor tissue, T-cell receptor CDR3s. In this report, the complementarity approach has been expanded to include a more comprehensive representation of the interaction of T-cell receptor CDR3s and mutant AAs by incorporating the impact of the wild-type AAs surrounding the mutant AA. This "sliding window" approach was benchmarked against two large datasets of empirically determined CDR3-epitope pairs; showed more significant patient subdivisions; revealed a novel, TRG CDR3-mutant PIK3CA linkage in breast cancer; and was particularly suited to use with big data collections using only modest and widely-available processors. Thus, the algorithm should support more rapid and convenient indications (or prescreens) of CDR3-mutant peptide interactions for more focused studies and more efficient development of patient immunology-related prognostic tools and therapies.
Collapse
Affiliation(s)
- Boris I Chobrutskiy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, FL, United States
| | - Andrea Chobrutskiy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, FL, United States
| | - Saif Zaman
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, FL, United States
| | - Michelle Yeagley
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, FL, United States
| | - Taha I Huda
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, FL, United States
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, FL, United States; Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, United States.
| |
Collapse
|
13
|
Qiao M, Jiang T, Liu X, Mao S, Zhou F, Li X, Zhao C, Chen X, Su C, Ren S, Zhou C. Immune Checkpoint Inhibitors in EGFR-Mutated NSCLC: Dusk or Dawn? J Thorac Oncol 2021; 16:1267-1288. [PMID: 33915248 DOI: 10.1016/j.jtho.2021.04.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
Although immune checkpoint inhibitors (ICIs) that target programmed cell death protein-1/programmed cell death ligand-1 axis have significantly shifted the treatment paradigm in advanced NSCLC, clinical benefits of these agents are limited in patients with EGFR-mutated NSCLC. Several predictive biomarkers (e.g., programmed cell death ligand-1 expression, tumor mutation burden), which have been validated in EGFR-wild type NSCLC, however, are not efficacious in EGFR-mutated tumors, suggesting the unique characteristics of tumor microenvironment of EGFR-mutated NSCLC. Here, we first summarized the clinical evidence on the efficacy of ICIs in patients with EGFR-mutated NSCLC. Then, the cancer immunogram features of EGFR-mutated NSCLC was depicted to visualize the state of cancer-immune system interactions, including tumor foreignness, tumor sensitivity to immune effectors, metabolism, general immune status, immune cell infiltration, cytokines, and soluble molecules. We further discussed the potential subpopulations with EGFR mutations that could benefit from ICI treatment. Lastly, we put forward future strategies to adequately maximize the efficacy of ICI treatment in patients with EGFR-mutated NSCLC in the upcoming era of combination immunotherapies.
Collapse
Affiliation(s)
- Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Tao Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xinyu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xiaoxia Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
14
|
Batalha S, Ferreira S, Brito C. The Peripheral Immune Landscape of Breast Cancer: Clinical Findings and In Vitro Models for Biomarker Discovery. Cancers (Basel) 2021; 13:1305. [PMID: 33804027 PMCID: PMC8001103 DOI: 10.3390/cancers13061305] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/09/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the deadliest female malignancy worldwide and, while much is known about phenotype and function of infiltrating immune cells, the same attention has not been paid to the peripheral immune compartment of breast cancer patients. To obtain faster, cheaper, and more precise monitoring of patients' status, it is crucial to define and analyze circulating immune profiles. This review compiles and summarizes the disperse knowledge on the peripheral immune profile of breast cancer patients, how it departs from healthy individuals and how it changes with disease progression. We propose this data to be used as a starting point for validation of clinically relevant biomarkers of disease progression and therapy response, which warrants more thorough investigation in patient cohorts of specific breast cancer subtypes. Relevant clinical findings may also be explored experimentally using advanced 3D cellular models of human cancer-immune system interactions, which are under intensive development. We review the latest findings and discuss the strengths and limitations of such models, as well as the future perspectives. Together, the scientific advancement of peripheral biomarker discovery and cancer-immune crosstalk in breast cancer will be instrumental to uncover molecular mechanisms and putative biomarkers and drug targets in an all-human setting.
Collapse
Affiliation(s)
- Sofia Batalha
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal;
- Instituto de Tecnologia Química e Biológica António Xavier, University Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Sofia Ferreira
- Instituto Português de Oncologia de Lisboa Francisco Gentil, Rua Prof Lima Basto, 1099-023 Lisboa, Portugal;
| | - Catarina Brito
- Instituto de Biologia Experimental e Tecnológica (iBET), Apartado 12, 2781-901 Oeiras, Portugal;
- Instituto de Tecnologia Química e Biológica António Xavier, University Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
15
|
Arana Echarri A, Beresford M, Campbell JP, Jones RH, Butler R, Gollob KJ, Brum PC, Thompson D, Turner JE. A Phenomic Perspective on Factors Influencing Breast Cancer Treatment: Integrating Aging and Lifestyle in Blood and Tissue Biomarker Profiling. Front Immunol 2021; 11:616188. [PMID: 33597950 PMCID: PMC7882710 DOI: 10.3389/fimmu.2020.616188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/11/2020] [Indexed: 01/10/2023] Open
Abstract
Breast cancer is the most common malignancy among women worldwide. Over the last four decades, diagnostic and therapeutic procedures have improved substantially, giving patients with localized disease a better chance of cure, and those with more advanced cancer, longer periods of disease control and survival. However, understanding and managing heterogeneity in the clinical response exhibited by patients remains a challenge. For some treatments, biomarkers are available to inform therapeutic options, assess pathological response and predict clinical outcomes. Nevertheless, some measurements are not employed universally and lack sensitivity and specificity, which might be influenced by tissue-specific alterations associated with aging and lifestyle. The first part of this article summarizes available and emerging biomarkers for clinical use, such as measurements that can be made in tumor biopsies or blood samples, including so-called liquid biopsies. The second part of this article outlines underappreciated factors that could influence the interpretation of these clinical measurements and affect treatment outcomes. For example, it has been shown that both adiposity and physical activity can modify the characteristics of tumors and surrounding tissues. In addition, evidence shows that inflammaging and immunosenescence interact with treatment and clinical outcomes and could be considered prognostic and predictive factors independently. In summary, changes to blood and tissues that reflect aging and patient characteristics, including lifestyle, are not commonly considered clinically or in research, either for practical reasons or because the supporting evidence base is developing. Thus, an aim of this article is to encourage an integrative phenomic approach in oncology research and clinical management.
Collapse
Affiliation(s)
| | - Mark Beresford
- Department of Oncology and Haematology, Royal United Hospitals Bath NHS Trust, Bath, United Kingdom
| | | | - Robert H. Jones
- Department of Medical Oncology, Velindre Cancer Centre, Cardiff, United Kingdom
- Department of Cancer and Genetics, Cardiff University, Cardiff, United Kingdom
| | - Rachel Butler
- South West Genomics Laboratory Hub, North Bristol NHS Trust, Bristol, United Kingdom
| | - Kenneth J. Gollob
- International Center for Research, A.C.Camargo Cancer Center, São Paulo, Brazil
| | - Patricia C. Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Dylan Thompson
- Department for Health, University of Bath, Bath, United Kingdom
| | - James E. Turner
- Department for Health, University of Bath, Bath, United Kingdom
| |
Collapse
|
16
|
Prognostic value of tumor-infiltrating lymphocyte subtypes in residual tumors of patients with triple-negative breast cancer after neoadjuvant chemotherapy. Chin Med J (Engl) 2020; 133:552-560. [PMID: 32044815 PMCID: PMC7065871 DOI: 10.1097/cm9.0000000000000656] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background: After neoadjuvant chemotherapy (NAC), non-pathological complete response of breast cancer patients can benefit from tailored adjuvant chemotherapy. However, it is difficult to select patients with poorer prognosis for additional adjuvant chemotherapy to maximize the benefits. Our study aimed to explore whether the subtypes of tumor-infiltrating lymphocytes (TILs) in residual tumors (RT) is related to the prognosis of triple-negative breast cancer (TNBC) after NAC. Methods: Data from patients with primary TNBC consecutively diagnosed at the Breast Disease Center of Peking University First Hospital from 2008 to 2014 were retrieved, and the cases with RT in the breast after NAC were enrolled. TILs subtypes in RT were observed by double-staining immunohistochemistry, and counted with the median TILs value per square millimeter as the cut-off to define high versus low TILs density in each subtype. The relationships between the TIL density of each subgroup and the clinicopathological characteristics of the RT after NAC patients were analyzed by Fisher exact test. Disease-free survival (DFS) and overall survival (OS) were analyzed by the Kaplan-Meier method and log-rank statistics. Results: A total of 37 eligible patients were included in this study, and the median follow-up period was 50 months (range 17–106 months). There was no significant correlation between the infiltrate density of CD4+, CD8+, CD20+, and CD68+ lymphocytes and clinic-pathological characteristics. Significantly better prognosis was observed in patients with high CD4+-TILs (DFS: P = 0.005, OS: P = 0.021) and high CD8+-TILs (DFS: P = 0.018) and low CD20+-TILs (OS: P = 0.042). Further analysis showed that patients with CD4+/CD20+ ratio greater than 1 (DFS: P = 0.001, OS: P = 0.002) or CD8+/CD20+ ratio greater than 1 (DFS: P = 0.009, OS: P = 0.022) had a better prognosis. Conclusions: Subtypes of TILs in RT is a potential predictive biomarker of survival in TNBC patients after NAC.
Collapse
|
17
|
Zhang M, Lu X, Wei C, Li X. Association between αβ and γδ T-cell subsets and clinicopathological characteristics in patients with breast cancer. Oncol Lett 2020; 20:325. [PMID: 33123241 PMCID: PMC7584013 DOI: 10.3892/ol.2020.12188] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/14/2020] [Indexed: 12/30/2022] Open
Abstract
The aim of the present study was to discuss the effect of surgery on the T-lymphocyte subsets of patients with breast cancer (BC) and investigate the association between peripheral blood αβ and γδ T-cell counts and the clinicopathological characteristics of BC. The CD3+, CD4+, CD8+ and γδ T-cell subsets in the peripheral blood of healthy volunteers and Patients with BC before and after surgery were determined using flow cytometry. The association between αβ and γδ T-cell counts in the peripheral blood and clinicopathological characteristics was analyzed by comparing the differences in the αβ and γδ T-cell counts in the peripheral blood of Patients with BC before and after surgery with those of healthy volunteers and combining with clinicopathological data. The CD3+, CD4+ and γδ T-cell counts in the peripheral blood of Patients with BC were lower compared with those in healthy volunteers (P=0.0077, 0.0116 and 0.0003, respectively), whereas the number of CD8+ cells was higher (P=0.0241). The CD3+, CD4+ and γδ T-cell counts and the CD4+/CD8+ ratio after surgery were significantly higher compared with those before surgery (P=0.0109, 0.0031, 0.0165 and 0.018, respectively). There was no significant difference between the number of CD8+ cells before and after surgery (P=0.0053), but the number of CD8+ cells was higher in healthy volunteers compared with that in Patients with BC (P<0.05). Moreover, the CD3+ cell number was higher in patients with TNM stage II/III compared with those with TNM stage I disease (P=0.187 and 0.022, respectively), and the peripheral blood CD4+/CD8+ ratio and number of γδ T cells were lower in stage III compared with stage I Patients with BC (P=0.0065 and 0.0176, respectively). Histological grading demonstrated that the CD4+/CD8+ ratio and number of γδ T cells in patients with stage III BC were lower compared with those with stage I BC (P=0.02 and 0.0128, respectively). The γδ T-cell count in patients with luminal A and B subtypes was significantly higher compared with that in patients with basal-like subtype (P=0.004 and 0.0104, respectively). The CD3+, CD4+ and γδ T-cell counts were significantly lower in patients with lymph node (LN) metastasis compared with those without LN metastasis, and the CD8+ cell number was lower in patients without LN metastasis compared with that in patients with >10 LN metastases (P=0.0086, 0.0000 and 0.00468, respectively). The CD8+ cell count in patients without LN metastasis was lower compared with that in patients with 4–9 and >10 LN metastases (P=0.0435 and 0.0283, respectively). Surgery affects the T-lymphocyte subpopulations in patients with BC, and αβ and γδ T-cell counts may increase following mastectomy. Therefore, measurement of peripheral blood lymphocyte subsets is crucial for understanding the immune function status of Patients with BC with differences in TNM stage, histological grade, cell subtypes and LN metastases, and may provide a basis for the application of T-cell subsets in the comprehensive treatment of BC.
Collapse
Affiliation(s)
- Meng Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Xueling Lu
- Department of Nuclear Medicine, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Changran Wei
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Xiangqi Li
- Department of Breast Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shandong 271000, P.R. China
| |
Collapse
|
18
|
Keup C, Kimmig R, Kasimir-Bauer S. Liquid Biopsies to Evaluate Immunogenicity of Gynecological/Breast Tumors: On the Way to Blood-Based Biomarkers for Immunotherapies. Breast Care (Basel) 2020; 15:470-480. [PMID: 33223990 PMCID: PMC7650128 DOI: 10.1159/000510509] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Despite the assumption of breast cancer (BC) as a cold, non-immunogenic tumor, immune checkpoint inhibitor (ICI) therapy is favorable for a subgroup of patients. Immunohistochemical assessment of the programmed cell death ligand 1 (PD-L1) is the only approved companion diagnostic for anti-PD-L1 therapy in metastatic triple-negative BC; however, challenges regarding the standardization of PD-L1 scoring in tumor tissue still remain. Consequently, to select patients most likely to respond to ICI, blood-based biomarkers are urgently needed. SUMMARY AND KEY MESSAGES Liquid biopsy, comprising circulating immune cells, circulating tumor cells and extracellular vesicles, as well as their surface proteins, is of high potential, and these analytes were already shown to be molecular correlates or regulators of the evasion from antitumoral immune reaction. Liquid biopsy, also enabling the evaluation of tumor mutational burden (TMB), microsatellite instability, and the T-cell receptor repertoire, allows serial sampling for monitoring purposes and reflects intra-tumoral heterogeneity which qualifies as marker for immunogenicity. Only a very few studies have already elucidated the potential of these analytes as biomarkers under ICI therapy. Nonetheless, the topic is of growing interest and has high relevance for the future. However, for clinical implementation, these multifarious analytes first need to pass robust standardization and validation procedures.
Collapse
Affiliation(s)
| | | | - Sabine Kasimir-Bauer
- Department of Gynecology and Obstetrics, University Hospital Essen, Essen, Germany
| |
Collapse
|
19
|
Oliver AJ, Darcy PK, Trapani JA, Kershaw MH, Slaney CY. Cross-talk between tumors at anatomically distinct sites. FEBS J 2020; 288:81-90. [PMID: 32248616 DOI: 10.1111/febs.15316] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/27/2020] [Indexed: 12/15/2022]
Abstract
Cancer tissue is not homogenous, and individual metastases at different anatomical locations can differ from the primary tumor and from one another in both their morphology and cellular composition, even within an individual patient. Tumors are composed of cancer cells and a range of other cell types, which, together with a variety of secreted molecules, collectively comprise the tumor microenvironment (TME). Cells of the TME can communicate with each other and with distant tissues in a form of molecular cross-talk to influence their growth and function. Cross-talk between cancer cells and local immune cells is well described and can lead to the induction of local immunosuppression. Recently, it has become apparent that tumors located remotely from each other, can engage in cross-talk that can influence their responsiveness to various therapies, including immunotherapy. In this article, we review studies that describe how tumors systemically communicate with distant tissues through motile cells, extracellular vesicles, and secreted molecules that can affect their function. In addition, we summarize evidence from mouse studies and the clinic that indicate an ability of some tumors to influence the progression and therapeutic responses of other tumors in different anatomical locations.
Collapse
Affiliation(s)
- Amanda J Oliver
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Joseph A Trapani
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| |
Collapse
|
20
|
Zhang Y, Jin Y, Guan Z, Li H, Su Z, Xie C, Chen X, Liu X, Pan Y, Ye P, Zhang L, Kong Y, Luo W. The Landscape and Prognosis Potential of the T-Cell Repertoire in Membranous Nephropathy. Front Immunol 2020; 11:387. [PMID: 32210970 PMCID: PMC7076165 DOI: 10.3389/fimmu.2020.00387] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 02/18/2020] [Indexed: 12/29/2022] Open
Abstract
Membranous nephropathy (MN), a common pathological type of adult nephrotic syndrome, is an antibody-mediated kidney disease. It is widely accepted now that MN is an immune-related disease that involves the whole immune system. In this study, we analyzed the T-cell receptor beta chain (TCRβ) repertoire of the circulating T lymphocytes of MN patients and healthy controls using high-throughput sequencing. We compared multiple aspects of the TCRβ repertoire, including diversity and the Vβ and Jβ genes between MN patients and healthy controls, and we found that the diversities within the VJ cassette combination in the peripheral blood of MN patients were lower than in the healthy controls. We also found the TCRβ repertoire similarity between pre- and post-therapy could reflect the clinical outcome, and two Vβ genes in pre-therapy had the potential to predict the therapeutic effect. These findings indicated the potential of the TCRβ repertoire as non-invasive biomarkers for the prognosis prediction of MN. The characteristics of circulating T-lymphocyte repertoires shed light on MN detection, treatment, and surveillance.
Collapse
Affiliation(s)
- Yu Zhang
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Yabin Jin
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Zhanwen Guan
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Huishi Li
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Zuhui Su
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Chao Xie
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Xiangping Chen
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Xiaofen Liu
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Yingming Pan
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Peiyi Ye
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Lifang Zhang
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| | - Yaozhong Kong
- Nephrology Department, The First People's Hospital of Foshan, Foshan, China
| | - Wei Luo
- Clinical Research Institute, The First People's Hospital of Foshan, Foshan, China
| |
Collapse
|
21
|
Chobrutskiy BI, Yeagley M, Diviney A, Zaman S, Gozlan EC, Tipping P, Koohestani DM, Roca AM, Blanck G. A scoring system for the electrostatic complementarities of T-cell receptors and cancer-mutant amino acids: multi-cancer analyses of associated survival rates. Immunology 2020; 159:373-383. [PMID: 31821535 DOI: 10.1111/imm.13165] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
The anti-tumor immune response is considered to be due to the T-cell receptor (TCR) binding to tumor antigens, which can be either wild-type, early stem cell proteins, presumably foreign to a developed immune system; or mutant peptides, foreign to the immune system because of a mutant amino acid (aa) or otherwise somatically altered aa sequence. Recently, very large numbers of TCR complementarity-determining region-3 (CDR3) aa sequences obtained from tumor specimens have become available. We developed a novel algorithm for assessing the complementarity of tumor mutant peptides and TCR CDR3s, based on the retrieval of TCR CDR3 aa sequences from both tumor specimen and patient blood exomes and by using an automated process of assessing CDR3 and mutant aa electrical charges. Results indicated many instances where high electrostatic complementarity was associated with a higher survival rate. In particular, our approach led to the identification of specific genes contributing significantly to the complementary, TCR CDR3-mutant aa. These results suggest a novel approach to tumor immunoscoring and may lead to the identification of high-priority neo-antigen, peptide vaccines; or to the identification of ex vivo stimulants of tumor-infiltrating lymphocytes.
Collapse
Affiliation(s)
- Boris I Chobrutskiy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Michelle Yeagley
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Andrea Diviney
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Saif Zaman
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Etienne C Gozlan
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Price Tipping
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Darush M Koohestani
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Andrea M Roca
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - George Blanck
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.,Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
22
|
Abstract
Our immune system plays a key role in health and disease as it is capable of responding to foreign antigens as well as acquired antigens from cancer cells. Latter are caused by somatic mutations, the so-called neoepitopes, and might be recognized by T cells if they are presented by HLA molecules on the surface of cancer cells. Personalized mutanome vaccines are a class of customized immunotherapies, which is dependent on the detection of individual cancer-specific tumor mutations and neoepitope (i.e., prediction, followed by a rational vaccine design, before on-demand production. The development of next generation sequencing (NGS) technologies and bioinformatic tools allows a large-scale analysis of each parameter involved in this process. Here, we provide an overview of the bioinformatic aspects involved in the design of personalized, neoantigen-based vaccines, including the detection of mutations and the subsequent prediction of potential epitopes, as well as methods for associated biomarker research, such as high-throughput sequencing of T-cell receptors (TCRs), followed by data analysis and the bioinformatics quantification of immune cell infiltration in cancer samples.
Collapse
Affiliation(s)
- Christoph Holtsträter
- TRON-Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz gemeinnützige GmbH, Freiligrathstraße, Mainz, Germany
| | - Barbara Schrörs
- TRON-Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz gemeinnützige GmbH, Freiligrathstraße, Mainz, Germany
| | - Thomas Bukur
- TRON-Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz gemeinnützige GmbH, Freiligrathstraße, Mainz, Germany
| | - Martin Löwer
- TRON-Translationale Onkologie an der Universitätsmedizin der Johannes Gutenberg-Universität Mainz gemeinnützige GmbH, Freiligrathstraße, Mainz, Germany.
| |
Collapse
|
23
|
Cowell LG. The Diagnostic, Prognostic, and Therapeutic Potential of Adaptive Immune Receptor Repertoire Profiling in Cancer. Cancer Res 2019; 80:643-654. [PMID: 31888887 DOI: 10.1158/0008-5472.can-19-1457] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/14/2019] [Accepted: 12/17/2019] [Indexed: 11/16/2022]
Abstract
Lymphocytes play a critical role in antitumor immune responses. They are directly targeted by some therapies, and the composition and spatial organization of intratumor T-cell populations is prognostic in some cancer types. A better understanding of lymphocyte population dynamics over the course of disease and in response to therapy is urgently needed to guide therapy decisions and to develop new therapy targets. Deep sequencing of the repertoire of antigen receptor-encoding genes expressed in a lymphocyte population has become a widely used approach for profiling the population's immune status. Lymphocyte antigen receptor repertoire deep sequencing data can be used to assess the clonal richness and diversity of lymphocyte populations; to track clone members over time, between tissues, and across lymphocyte subsets; to detect clonal expansion; and to detect the recruitment of new clones into a tissue. Repertoire sequencing is thus a critical complement to other methods of lymphocyte and immune profiling in cancer. This review describes the current state of knowledge based on repertoire sequencing studies conducted on human cancer patients, with a focus on studies of the T-cell receptor beta chain locus. The review then outlines important questions left unanswered and suggests future directions for the field.
Collapse
Affiliation(s)
- Lindsay G Cowell
- Department of Population and Data Sciences, Department of Immunology, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
24
|
Abdel-Aziz AK, Saadeldin MK, D'Amico P, Orecchioni S, Bertolini F, Curigliano G, Minucci S. Preclinical models of breast cancer: Two-way shuttles for immune checkpoint inhibitors from and to patient bedside. Eur J Cancer 2019; 122:22-41. [PMID: 31606656 DOI: 10.1016/j.ejca.2019.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 08/17/2019] [Indexed: 12/18/2022]
Abstract
The Food and Drug Administration has lately approved atezolizumab, anti-programmed death ligand 1 (PD-L1), to be used together with nanoparticle albumin-bound (nab) paclitaxel in treating patients with triple negative breast cancer (BC) expressing PD-L1. Nonetheless, immune checkpoint inhibitors (ICIs) are still challenged by the resistance and immune-related adverse effects evident in a considerable subset of treated patients without conclusive comprehension of the underlying molecular basis, biomarkers and tolerable therapeutic regimens capable of unleashing the anti-tumour immune responses. Stepping back to preclinical models is thus inevitable to address these inquiries. Herein, we comprehensively review diverse preclinical models of BC exploited in investigating ICIs underscoring their pros and cons as well as the learnt and awaited lessons to allow full exploitation of ICIs in BC therapy.
Collapse
Affiliation(s)
- Amal Kamal Abdel-Aziz
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Mona Kamal Saadeldin
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Faculty of Biotechnology, October University for Modern Sciences and Arts, 6th October City, Cairo, Egypt
| | - Paolo D'Amico
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Stefania Orecchioni
- Laboratory of Hematology-Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy.
| | - Saverio Minucci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy.
| |
Collapse
|