1
|
Georgiev P, Han S, Huang AY, Nguyen TH, Drijvers JM, Creasey H, Pereira JA, Yao CH, Park JS, Conway TS, Fung ME, Liang D, Peluso M, Joshi S, Rowe JH, Miller BC, Freeman GJ, Sharpe AH, Haigis MC, Ringel AE. Age-Associated Contraction of Tumor-Specific T Cells Impairs Antitumor Immunity. Cancer Immunol Res 2024; 12:1525-1541. [PMID: 39186561 PMCID: PMC11532741 DOI: 10.1158/2326-6066.cir-24-0463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/14/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Progressive decline of the adaptive immune system with increasing age coincides with a sharp increase in cancer incidence. In this study, we set out to understand whether deficits in antitumor immunity with advanced age promote tumor progression and/or drive resistance to immunotherapy. We found that multiple syngeneic cancers grew more rapidly in aged versus young adult mice, driven by dysfunctional CD8+ T-cell responses. By systematically mapping immune cell profiles within tumors, we identified loss of tumor antigen-specific CD8+ T cells as a primary feature accelerating the growth of tumors in aged mice and driving resistance to immunotherapy. When antigen-specific T cells from young adult mice were administered to aged mice, tumor outgrowth was delayed and the aged animals became sensitive to PD-1 blockade. These studies reveal how age-associated CD8+ T-cell dysfunction may license tumorigenesis in elderly patients and have important implications for the use of aged mice as preclinical models of aging and cancer.
Collapse
Affiliation(s)
- Peter Georgiev
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - SeongJun Han
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amy Y. Huang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Thao H. Nguyen
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jefte M. Drijvers
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hannah Creasey
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joseph A. Pereira
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Joon Seok Park
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Thomas S. Conway
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Megan E. Fung
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dan Liang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michael Peluso
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jared H. Rowe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Brian C. Miller
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Arlene H. Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Marcia C. Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
- Gene Lay Institute of Immunology and Inflammation of Brigham and Women’s Hospital, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Alison E. Ringel
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
2
|
Chen S, Shen X, Cao P, Chen Q, Zhong R, Cao Y. METTL3 affects the biological function of lung adenocarcinoma through the FGF2/PI3K/AKT /mTOR pathway. Front Oncol 2024; 14:1474701. [PMID: 39497721 PMCID: PMC11532124 DOI: 10.3389/fonc.2024.1474701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/24/2024] [Indexed: 11/07/2024] Open
Abstract
Introduction This study aims to investigate the role of the m6A regulatory factor METTL3 in LUAD. Methods By examining the expression of METTL3 in LUAD and conducting cellular functional experiments, the biological functions of METTL3 were discussed. mRNA-seq and MeRIP-qPCR were used to identify downstream target genes and pathways. Results The expression level of METTL3 in LUAD is lower than that in the control group. The downregulation of METTL3 promoted the proliferation, migration, and invasion of LUAD cells, while overexpression of METTL3 results in the opposite effects. Furthermore, we found that FGF2 was negatively regulated by METTL3. Inhibiting FGF2 reversed the tumor-promoting effects caused by METTL3 downregulation in LUAD cells. Silencing METTL3 enhanced the stability of FGF2 mRNA. Silencing FGF2 resulted in reduced activity of the PI3K/AKT/mTOR signaling pathway in METTL3 knockdown LUAD cells. Discussion In summary, our findings unveil an intricate signaling network involving METTL3/FGF2/PI3K/AKT/mTOR in LUAD and provide valuable insights into the molecular mechanisms underlying tumor progression, thus holding significant implications for targeted therapy and advancing LUAD research.
Collapse
Affiliation(s)
- Shaoting Chen
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Xiuqing Shen
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Pengju Cao
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Qianshun Chen
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Rongxin Zhong
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, China
| | - Yingping Cao
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
3
|
Bai M, Wang WX, Deng T, Duan JJ, Ba Y. Feasibility and Safety of PD-1 Blockades Among Elderly Patients with Metastatic Esophageal Squamous Cell Carcinoma: A Real-World Study. Drug Des Devel Ther 2024; 18:4135-4151. [PMID: 39308693 PMCID: PMC11414636 DOI: 10.2147/dddt.s476457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024] Open
Abstract
Objective This study aimed to identify the effectiveness and safety of PD-1 blockades among elderly patients with metastatic esophageal squamous cell carcinoma (ESCC) clinically. Methods A total of 78 elderly patients with previously treated metastatic ESCC aged ≥65 years who received PD-1 blockades monotherapy were included retrospectively. Demographic characteristics, therapeutic effectiveness and adverse reactions of the elderly patients who underwent PD-1 blockade therapy were recorded. Regular follow-up was conducted for all patients. The analysis aimed to identify potential risk factors for OS by examining the correlation between prognosis and subgroups based on baseline characteristics. Results The median age of the 78 elderly patients was 73 years, ranging from 65 to 87 years. Among the 78 patients, 18 cases showed partial response, 26 cases had stable disease, 29 cases experienced progressive disease and 5 cases were not assessable for response, yielding an ORR of 23.1%, a DCR of 56.4%. The prognostic outcomes indicated that among the 78 patients with metastatic ESCC who received PD-1 blockades, the median PFS was 3.1 months [95% confidence interval (CI): 1.64-4.56], and the median OS was 10.9 months (95% CI: 6.02-15.78), 24-month OS rate was 22.7% (95% CI: 12.8-34.2%). In terms of the safety profile, among the 78 patients with metastatic ESCC during PD-1 blockades single-agent treatment, a total of 61 patients (78.2%) experienced any grade adverse reactions and the incidence of grade ≥3 adverse reactions were 20.5%. Briefly, the common adverse reactions manifested as fatigue (32.1%), gastrointestinal reaction (24.4%), diarrhea (19.2%), anemia (17.9%) and rash (16.7%). Overall tolerability of PD-1 blockade monotherapy in elderly patients with metastatic ESCC was acceptable and manageable. Conclusion PD-1 blockades single agent demonstrated encouraging effectiveness and acceptable safety profile for elderly patients with previously treated metastatic ESCC in clinical practice. Prospective study should be performed to elucidate the conclusion in this study subsequently.
Collapse
Affiliation(s)
- Ming Bai
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, 300060, People’s Republic of China
| | - Wei-Xue Wang
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, 300060, People’s Republic of China
- Aerospace Center Hospital, Beijing, 100049, People’s Republic of China
| | - Ting Deng
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, 300060, People’s Republic of China
| | - Jing-Jing Duan
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, 300060, People’s Republic of China
| | - Yi Ba
- Department of Gastrointestinal Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, Tianjin, 300060, People’s Republic of China
- Peking Union Medical College Hospital, Beijing, 100730, People’s Republic of China
| |
Collapse
|
4
|
Santos TPMD, Hicks WL, Magner WJ, Al Afif A, Kirkwood KL. Metabolic and Aging Influence on Anticancer Immunity in Oral Cancer. J Dent Res 2024; 103:953-961. [PMID: 39185914 DOI: 10.1177/00220345241264728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024] Open
Abstract
The average age and obesity prevalence are increasing globally. Both aging and metabolic disease burden increase the risk of oral squamous cell carcinoma (OSCC) through profound effects on the immunological and metabolic characteristics within the OSCC tumor microenvironment. While the mechanisms that link aging and obesity to OSCC remain unclear, there is evidence that the antitumor responses are diminished in both conditions. Remarkably, however, immune checkpoint blockade, a form of cancer immunotherapy, remains intact despite the enhanced immunosuppressive tumor microenvironment in the context of either aging or obesity. Herein, we review the current knowledge of how aging and systemic metabolic changes affect antitumor immunity with an emphasis on the role of tumor-associated macrophages that greatly contribute to tumor immunosuppression. Key aspects discussed include the mechanisms of angiogenesis, cytokine release, phagocytosis attenuation, and immune cell recruitment during obesity and aging that create an immune-suppressive tumor microenvironment by recruitment and repolarization of tumor-associated macrophages. Through a deeper appreciation of these mechanisms, the development of novel therapeutic approaches to control OSCC will provide more refined management of the tumor microenvironment in the context of aging and obesity.
Collapse
Affiliation(s)
- T P M D Santos
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
- Department of Head & Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - W L Hicks
- Department of Head & Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - W J Magner
- Department of Head & Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - A Al Afif
- Department of Head & Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - K L Kirkwood
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY, USA
- Department of Head & Neck/Plastic and Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
5
|
Ding X, Zhang L, Fan M, Li L. TME-NET: an interpretable deep neural network for predicting pan-cancer immune checkpoint inhibitor responses. Brief Bioinform 2024; 25:bbae410. [PMID: 39167797 PMCID: PMC11337220 DOI: 10.1093/bib/bbae410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 08/23/2024] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICIs) is increasingly used to treat various tumor types. Determining patient responses to ICIs presents a significant clinical challenge. Although components of the tumor microenvironment (TME) are used to predict patient outcomes, comprehensive assessments of the TME are frequently overlooked. Using a top-down approach, the TME was divided into five layers-outcome, immune role, cell, cellular component, and gene. Using this structure, a neural network called TME-NET was developed to predict responses to ICIs. Model parameter weights and cell ablation studies were used to investigate the influence of TME components. The model was developed and evaluated using a pan-cancer cohort of 948 patients across four cancer types, with Area Under the Curve (AUC) and accuracy as performance metrics. Results show that TME-NET surpasses established models such as support vector machine and k-nearest neighbors in AUC and accuracy. Visualization of model parameter weights showed that at the cellular layer, Th1 cells enhance immune responses, whereas myeloid-derived suppressor cells and M2 macrophages show strong immunosuppressive effects. Cell ablation studies further confirmed the impact of these cells. At the gene layer, the transcription factors STAT4 in Th1 cells and IRF4 in M2 macrophages significantly affect TME dynamics. Additionally, the cytokine-encoding genes IFNG from Th1 cells and ARG1 from M2 macrophages are crucial for modulating immune responses within the TME. Survival data from immunotherapy cohorts confirmed the prognostic ability of these markers, with p-values <0.01. In summary, TME-NET performs well in predicting immunotherapy responses and offers interpretable insights into the immunotherapy process. It can be customized at https://immbal.shinyapps.io/TME-NET.
Collapse
Affiliation(s)
- Xiaobao Ding
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
- Institute of Big Data and Artificial Intelligence in Medicine, School of Electronics and Information Engineering, Taizhou University, Taizhou 318000, Zhejiang, China
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Lin Zhang
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, Zhejiang, China
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, China
| |
Collapse
|
6
|
Kinoshita R, Nakao M, Kiyotoshi H, Sugihara M, Kuriyama M, Takeda N, Muramatsu H. Geriatric Nutritional Risk Index as Prognostic Marker for Elderly Patients With Small Cell Lung Cancer. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:482-488. [PMID: 38962547 PMCID: PMC11215438 DOI: 10.21873/cdp.10352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/12/2024] [Indexed: 07/05/2024]
Abstract
Background/Aim The Geriatric Nutritional Risk Index (GNRI) indicates nutritional status based on serum albumin concentration and ideal body weight. Pretreatment GNRI has been suggested as a prognostic factor for various malignancies. However, little is known about the clinical value of GNRI for small-cell lung cancer (SCLC), especially in elderly patients. Patients and Methods We retrospectively analyzed 53 elderly (≥71) patients with extensive-disease (ED) SCLC treated with first-line platinum-doublet chemotherapy in relation to the pretreatment GNRI level in a real-world setting. Results Thirty-six patients with a low GNRI (<92) had statistically poorer progression-free survival (PFS) and overall survival (OS) than 17 patients with a high GNRI (≥92) (median PFS=80 days vs. 133 days, respectively; p=0.002; median OS=123 days vs. 274 days, respectively; p=0.004). In a multivariate analysis, a low GNRI was also an independent poor prognostic factor for PFS [hazard ratio (HR)=0.396; 95% confidence interval (CI)=0.199-0.789; p=0.008] and OS (HR=0.295; 95%CI=0.143-0.608; p<0.001). Conclusion The GNRI might be a predictive and prognostic marker in elderly patients with ED-SCLC treated with platinum-doublet chemotherapy.
Collapse
Affiliation(s)
- Ryosuke Kinoshita
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| | - Makoto Nakao
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| | - Hiroko Kiyotoshi
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| | - Masahiro Sugihara
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| | - Mamiko Kuriyama
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| | - Norihisa Takeda
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| | - Hideki Muramatsu
- Department of Respiratory Medicine, Kainan Hospital Aichi Prefectural Welfare Federation of Agricultural Cooperatives, Yatomi, Japan
| |
Collapse
|
7
|
Kosumi K, Baba Y, Hara Y, Wang H, Nomoto D, Toihata T, Ohuchi M, Harada K, Eto K, Ogawa K, Ishimoto T, Iwatsuki M, Iwagami S, Miyamoto Y, Yoshida N, Baba H. Body Composition and Clinical Outcomes in Esophageal Cancer Patients Treated with Immune Checkpoint Inhibitors. Ann Surg Oncol 2024; 31:3839-3849. [PMID: 38421531 DOI: 10.1245/s10434-024-15093-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/08/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Obesity is associated with increased mortality in various cancers, but the relationship between obesity and clinical outcomes in unresectable or recurrent esophageal cancer who receive immune checkpoint inhibitors (ICIs) remains unknown. This study investigated the association between body composition and clinical outcomes in patients with unresectable or recurrent esophageal cancer who received ICIs. METHODS Utilizing an unbiased database of 111 unresectable or recurrent esophageal cancers, we evaluated the relationships between body composition (body mass index, waist circumference, psoas major muscle volume, and subcutaneous and visceral fat areas) at the initiation of ICI treatment and clinical outcomes including the disease control rate and progression-free survival (PFS). RESULTS Waist circumference was significantly associated with the disease control rate at the first assessment (P = 0.0008). A high waist circumference was significantly associated with favorable PFS in patients treated with nivolumab. In an univariable model, for 5-cm increase of waist circumference in the outcome category of PFS, univariable hazard ratio (HR) was 0.73 (95% confidence interval [CI], 0.61-0.87; P = 0.0002). A multivariable model controlling for potential confounders yielded a similar finding (multivariable HR, 0.56; 95% CI, 0.33-0.94; P = 0.027). We observed the similar finding in esophageal cancer patients treated with pembrolizumab+CDDP+5-FU (P = 0.048). In addition, waist circumference was significantly associated with the prognostic nutritional index (P = 0.0073). CONCLUSIONS A high waist circumference was associated with favorable clinical outcomes in ICI-treated patients with unresectable or recurrent esophageal cancer, providing a platform for further investigations on the relationships among body composition, nutrition, and the immune status.
Collapse
Affiliation(s)
- Keisuke Kosumi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Next-Generation Surgical Therapy Development, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshifumi Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Department of Next-Generation Surgical Therapy Development, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshihiro Hara
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Haolin Wang
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Daichi Nomoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tasuku Toihata
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Mayuko Ohuchi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuto Harada
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kojiro Eto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Katsuhiro Ogawa
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takatsugu Ishimoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Masaaki Iwatsuki
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shiro Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuji Miyamoto
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Naoya Yoshida
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
8
|
Durand A, Bonilla N, Level T, Ginestet Z, Lombès A, Guichard V, Germain M, Jacques S, Letourneur F, Do Cruzeiro M, Marchiol C, Renault G, Le Gall M, Charvet C, Le Bon A, Martin B, Auffray C, Lucas B. Type 1 interferons and Foxo1 down-regulation play a key role in age-related T-cell exhaustion in mice. Nat Commun 2024; 15:1718. [PMID: 38409097 PMCID: PMC10897180 DOI: 10.1038/s41467-024-45984-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/05/2024] [Indexed: 02/28/2024] Open
Abstract
Foxo family transcription factors are critically involved in multiple processes, such as metabolism, quiescence, cell survival and cell differentiation. Although continuous, high activity of Foxo transcription factors extends the life span of some species, the involvement of Foxo proteins in mammalian aging remains to be determined. Here, we show that Foxo1 is down-regulated with age in mouse T cells. This down-regulation of Foxo1 in T cells may contribute to the disruption of naive T-cell homeostasis with age, leading to an increase in the number of memory T cells. Foxo1 down-regulation is also associated with the up-regulation of co-inhibitory receptors by memory T cells and exhaustion in aged mice. Using adoptive transfer experiments, we show that the age-dependent down-regulation of Foxo1 in T cells is mediated by T-cell-extrinsic cues, including type 1 interferons. Taken together, our data suggest that type 1 interferon-induced Foxo1 down-regulation is likely to contribute significantly to T-cell dysfunction in aged mice.
Collapse
Affiliation(s)
- Aurélie Durand
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Nelly Bonilla
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Théo Level
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Zoé Ginestet
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Amélie Lombès
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Vincent Guichard
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Mathieu Germain
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Sébastien Jacques
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Franck Letourneur
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Marcio Do Cruzeiro
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Carmen Marchiol
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Gilles Renault
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Morgane Le Gall
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Céline Charvet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Illkirch, France
- CNRS UMR7104, Illkirch, France
- INSERM U1258, Illkirch, France
- Université de Strasbourg, Strasbourg, France
| | - Agnès Le Bon
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Bruno Martin
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Cédric Auffray
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France
| | - Bruno Lucas
- Université Paris-Cité, Institut Cochin, Centre National de la Recherche Scientifique (CNRS) UMR8104, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, 75014, Paris, France.
| |
Collapse
|
9
|
Sivasankar C, Hewawaduge C, Muthuramalingam P, Lee JH. Tumor-targeted delivery of lnc antisense RNA against RCAS1 by live-attenuated tryptophan-auxotrophic Salmonella inhibited 4T1 breast tumors and metastasis in mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102053. [PMID: 37941832 PMCID: PMC10628790 DOI: 10.1016/j.omtn.2023.102053] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Emerging chemo- and radiotherapy resistance exacerbated the cancer risk and necessitated novel treatment strategies. Although RNA therapeutics against pro-oncogenic genes are highly effective, tumor-specific delivery remains a barrier to the implementation of this valuable tool. In this study, we report a tryptophan-auxotrophic Salmonella typhimurium strain as an onco-therapeutic delivery system with tumor-targeting ability using 4T1 mice breast-cancer model. The receptor-binding cancer antigen expressed on SiSo cell (RCAS1) is a cancer-specific protein that induces the apoptosis of peripheral lymphocytes and confers tumor immune evasion. We designed a long non-coding antisense-RNA against RCAS1 (asRCAS1) and delivered by Salmonella using a non-antibiotic, auxotrophic-selective, eukaryotic expression plasmid, pJHL204. After in vivo tumor-to-tumor passaging, the JOL2888 (ΔtrpA, ΔtrpE, Δasd + asRCAS1) strain exhibited high sustainability in tumors, but did not last in healthy organs, thereby demonstrating tumor specificity and safety. RCAS1 inhibition in the tumor was confirmed by western blotting and qPCR. In mice, JOL2888 treatment reduced tumor-associated macrophages, improved the T cell population, elicited cell-mediated immunity, and suppressed cancer-promoting genes. Consequently, the JOL2888 treatment significantly decreased the tumor volume by 80%, decreased splenomegaly by 30%, and completely arrested lung metastasis. These findings highlight the intrinsic tumor-targeting ability of tryptophan-auxotrophic Salmonella for delivering onco-therapeutic macromolecules.
Collapse
Affiliation(s)
- Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| | | | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| |
Collapse
|
10
|
McKenzie ND, Ahmad S, Awada A, Kuhn TM, Recio FO, Holloway RW. Prognostic features of the tumor microenvironment in high-grade serous ovarian cancer and dietary immunomodulation. Life Sci 2023; 333:122178. [PMID: 37839778 DOI: 10.1016/j.lfs.2023.122178] [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: 09/18/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/17/2023]
Abstract
High-grade serous ovarian cancer (HGSOC) is a particularly lethal malignancy that is prognostically influenced by the immune profile of the tumor microenvironment (TME). TME immune profiles have been sub-categorized according to features associated with both survival outcomes as well as response to systemic therapies. Five suggested immune phenotypes have been described and correlated with overall survival outcomes. Phenotypes associated with shorter overall survival rates appear to have prominent immunosuppressive features within their TME. The opportunity to triage patients according to their prognostic TME profile might allow selection of individual patients with poor prognostic features who could most benefit from innovative immunomodulatory treatment strategies. Two potential strategies to indirectly manipulate the TME (and oncologic outcomes) are alteration of the gut microbiome composition and alteration of TME metabolism through dietary interventions. Experimental dietary modifications in humans designed for influencing cancer outcomes are only beginning to be studied in a prospective fashion. Herein we summarize prognostic TME features in HGSOC and potential opportunities for immunomodulation via dietary and gut microbial interventions.
Collapse
Affiliation(s)
- Nathalie D McKenzie
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA.
| | - Sarfraz Ahmad
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA.
| | - Ahmad Awada
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Theresa M Kuhn
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Fernando O Recio
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| | - Robert W Holloway
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL 32804, USA
| |
Collapse
|
11
|
Noll JH, Levine BL, June CH, Fraietta JA. Beyond youth: Understanding CAR T cell fitness in the context of immunological aging. Semin Immunol 2023; 70:101840. [PMID: 37729825 DOI: 10.1016/j.smim.2023.101840] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/22/2023]
Abstract
Population aging, a pervasive global demographic trend, is anticipated to challenge health and social systems worldwide. This phenomenon is due to medical advancements enabling longer lifespans, with 20% of the US population soon to be over 65 years old. Consequently, there will be a surge in age-related diseases. Senescence, characterized by the loss of biological maintenance and homeostasis at molecular and cellular levels, either correlates with or directly causes age-related phenotypic changes. Decline of the immune system is a critical factor in the senescence process, with cancer being a primary cause of death in elderly populations. Chimeric antigen receptor (CAR) T cell therapy, an innovative approach, has demonstrated success mainly in pediatric and young adult hematological malignancies but remains largely ineffective for diseases affecting older populations, such as late-in-life B cell malignancies and most solid tumor indications. This limitation arises because CAR T cell efficacy heavily relies on the fitness of the patient-derived starting T cell material. Numerous studies suggest that T cell senescence may be a key driver of CAR T cell deficiency. This review examines correlates and underlying factors associated with favorable CAR T cell outcomes and explores potential experimental and clinically actionable strategies for T cell rejuvenation.
Collapse
Affiliation(s)
- Julia Han Noll
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Binder AF, Walker CJ, Mark TM, Baljevic M. Impacting T-cell fitness in multiple myeloma: potential roles for selinexor and XPO1 inhibitors. Front Immunol 2023; 14:1275329. [PMID: 37954586 PMCID: PMC10637355 DOI: 10.3389/fimmu.2023.1275329] [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: 08/09/2023] [Accepted: 10/05/2023] [Indexed: 11/14/2023] Open
Abstract
Competent T-cells with sufficient levels of fitness combat cancer formation and progression. In multiple myeloma (MM), T-cell exhaustion is caused by several factors including tumor burden, constant immune activation due to chronic disease, age, nutritional status, and certain MM treatments such as alkylating agents and proteasome inhibitors. Many currently used therapies, including bispecific T-cell engagers, anti-CD38 antibodies, proteasome inhibitors, and CART-cells, directly or indirectly depend on the anti-cancer activity of T-cells. Reduced T-cell fitness not only diminishes immune defenses, increasing patient susceptibility to opportunistic infections, but can impact effectiveness MM therapy effectiveness, bringing into focus sequencing strategies that could modulate T-cell fitness and potentially optimize overall benefit and clinical outcomes. Certain targeted agents used to treat MM, such as selective inhibitors of nuclear export (SINE) compounds, have the potential to mitigate T-cell exhaustion. Herein referred to as XPO1 inhibitors, SINE compounds inhibit the nuclear export protein exportin 1 (XPO1), which leads to nuclear retention and activation of tumor suppressor proteins and downregulation of oncoprotein expression. The XPO1 inhibitors selinexor and eltanexor reduced T-cell exhaustion in cell lines and animal models, suggesting their potential role in revitalizating these key effector cells. Additional clinical studies are needed to understand how T-cell fitness is impacted by diseases and therapeutic factors in MM, to potentially facilitate the optimal use of available treatments that depend on, and impact, T-cell function. This review summarizes the importance of T-cell fitness and the potential to optimize treatment using T-cell engaging therapies with a focus on XPO1 inhibitors.
Collapse
Affiliation(s)
- Adam F. Binder
- Department of Medical Oncology, Division of Hematopoietic Stem Cell Transplant and Hematologic Malignancies, Thomas Jefferson University, Philadelphia, PA, United States
| | - Christopher J. Walker
- Department of Translational Research, Karyopharm Therapeutics, Inc, Newton, MA, United States
| | - Tomer M. Mark
- Department of Translational Research, Karyopharm Therapeutics, Inc, Newton, MA, United States
| | - Muhamed Baljevic
- Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
13
|
Sun R, Xu H, Liu F, Zhou B, Li M, Sun X. Unveiling the intricate causal nexus between pancreatic cancer and peripheral metabolites through a comprehensive bidirectional two-sample Mendelian randomization analysis. Front Mol Biosci 2023; 10:1279157. [PMID: 37954977 PMCID: PMC10634252 DOI: 10.3389/fmolb.2023.1279157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Aim: Pancreatic cancer (PC) is a devastating malignancy characterized by its aggressive nature and poor prognosis. However, the relationship of PC with peripheral metabolites remains not fully investigated. The study aimed to explore the causal linkage between PC and peripheral metabolite profiles. Methods: Employing publicly accessible genome-wide association studies (GWAS) data, we conducted a bidirectional two-sample Mendelian randomization (MR) analysis. The primary analysis employed the inverse-variance weighted (IVW) method. To address potential concerns about horizontal pleiotropy, we also employed supplementary methods such as maximum likelihood, weighted median, MR-Egger regression, and MR pleiotropy residual sum and outlier (MR-PRESSO). Results: We ascertained 20 genetically determined peripheral metabolites with causal linkages to PC while high-density lipoprotein (HDL) and very low-density lipoprotein (VLDL) particles accounted for the vast majority. Specifically, HDL particles exhibited an elevated PC risk while VLDL particles displayed an opposing pattern. The converse MR analysis underscored a notable alteration in 17 peripheral metabolites due to PC, including branch chain amino acids and derivatives of glycerophospholipid. Cross-referencing the bidirectional MR results revealed a reciprocal causation of PC and X-02269 which might form a self-perpetuating loop in PC development. Additionally, 1-arachidonoylglycerophosphocholine indicated a reduced PC risk and an increase under PC influence, possibly serving as a negative feedback regulator. Conclusion: Our findings suggest a complex interplay between pancreatic cancer and peripheral metabolites, with potential implications for understanding the etiology of pancreatic cancer and identifying novel early diagnosis and therapeutic targets. Moreover, X-02269 may hold a pivotal role in PC onset and progression.
Collapse
Affiliation(s)
| | | | | | | | - Minli Li
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiangdong Sun
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Qiao W, Hu C, Ma J, Dong X, Dalangood S, Li H, Yuan C, Lu B, Gao WQ, Wen Z, Yin W, Gui J. Low-dose metronomic chemotherapy triggers oxidized mtDNA sensing inside tumor cells to potentiate CD8 +T anti-tumor immunity. Cancer Lett 2023; 573:216370. [PMID: 37660883 DOI: 10.1016/j.canlet.2023.216370] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/08/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Low-dose metronomic (LDM) chemotherapy, the frequent and continuous use of low doses of conventional chemotherapeutics, is emerging as a promising form of chemotherapy utilization. LDM chemotherapy exerts immunomodulatory effects. However, the underlying mechanism is not fully understood. Here we found that suppressing tumor growth by LDM chemotherapy was dependent on the activation of CD8+T cells. LDM chemotherapy potentiated the cytotoxic function of CD8+T cells by stimulating cancer-cell autonomous type I interferon (IFN) induction. Mechanistically, LDM chemotherapy evoked mitochondrial dysfunction and increased reactive oxygen species (ROS) production. ROS triggered the oxidation of cytosolic mtDNA, which was sensed by cGAS-STING, consequently inducing type I IFN production in the cancer cells. Moreover, the cGAS-STING-IFN axis increased PD-L1 expression and predicted favorable clinical responses to chemoimmunotherapy. Antioxidant N-acetylcysteine inhibited oxidized mtDNA-induced type I IFN production and attenuated the efficacy of combination therapy with LDM chemotherapy and PD-L1 blockade. This study elucidates the critical role of intratumoral oxidized mtDNA sensing in LDM chemotherapy-mediated activation of CD8+T cell immune response. These findings may provide new insights for designing combinatorial immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Wen Qiao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Cegui Hu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jiayi Ma
- Department of Breast Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xinrui Dong
- Department of Breast Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Sumiya Dalangood
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hanjun Li
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Chenwei Yuan
- Department of Breast Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Binbin Lu
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China; School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China.
| | - Wenjin Yin
- Department of Breast Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Jun Gui
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
15
|
Vick LV, Collins CP, Khuat LT, Wang Z, Dunai C, Aguilar EG, Stoffel K, Yendamuri S, Smith R, Mukherjee S, Barbi J, Canter RJ, Monjazeb AM, Murphy WJ. Aging augments obesity-induced thymic involution and peripheral T cell exhaustion altering the "obesity paradox". Front Immunol 2023; 13:1012016. [PMID: 36776393 PMCID: PMC9910174 DOI: 10.3389/fimmu.2022.1012016] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/07/2022] [Indexed: 01/28/2023] Open
Abstract
Introduction The incidence of obesity, a condition characterized by systemic chronic inflammation, has reached pandemic proportions and is a poor prognostic factor in many pathologic states. However, its role on immune parameters has been diverse and at times contradictory. We have previously demonstrated that obesity can result in what has been called the "obesity paradox" which results in increased T cell exhaustion, but also greater efficacy of immune checkpoint blockade in cancer treatment. Methods The role of obesity, particularly in the context of aging, has not been robustly explored using preclinical models. We therefore evaluated how age impacts the immune environment on T cell development and function using diet-induced obese (DIO) mice. Results We observed that DIO mice initially displayed greater thymopoiesis but then developed greater thymic involution over time compared to their lean counterparts. Both aging and obesity resulted in increased T cell memory conversion combined with increased expression of T cell exhaustion markers and Treg expansion. This increased T cell immunosuppression with age then resulted in a loss of anti-tumor efficacy by immune checkpoint inhibitors (ICIs) in older DIO mice compared to the younger DIO counterparts. Discussion These results suggest that both aging and obesity contribute to T cell dysfunction resulting in increased thymic involution. This combined with increased T cell exhaustion and immunosuppressive parameters affects immunotherapy efficacy reducing the advantage of obesity in cancer immunotherapy responses.
Collapse
Affiliation(s)
- Logan V. Vick
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Craig P. Collins
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Lam T. Khuat
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ziming Wang
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Cordelia Dunai
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Ethan G. Aguilar
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Kevin Stoffel
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Sai Yendamuri
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Randall Smith
- Department of Immunology Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Sarbajit Mukherjee
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Joseph Barbi
- Department of Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Department of Immunology Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Robert J. Canter
- Division of Surgical Oncology, Department of Surgery, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Arta M. Monjazeb
- Department of Radiation Oncology, University of California Davis Comprehensive Cancer Center, University of California School of Medicine, Sacramento, CA, United States
| | - William J. Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
16
|
López-Otín C, Pietrocola F, Roiz-Valle D, Galluzzi L, Kroemer G. Meta-hallmarks of aging and cancer. Cell Metab 2023; 35:12-35. [PMID: 36599298 DOI: 10.1016/j.cmet.2022.11.001] [Citation(s) in RCA: 134] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/11/2022] [Accepted: 11/07/2022] [Indexed: 01/05/2023]
Abstract
Both aging and cancer are characterized by a series of partially overlapping "hallmarks" that we subject here to a meta-analysis. Several hallmarks of aging (i.e., genomic instability, epigenetic alterations, chronic inflammation, and dysbiosis) are very similar to specific cancer hallmarks and hence constitute common "meta-hallmarks," while other features of aging (i.e., telomere attrition and stem cell exhaustion) act likely to suppress oncogenesis and hence can be viewed as preponderantly "antagonistic hallmarks." Disabled macroautophagy and cellular senescence are two hallmarks of aging that exert context-dependent oncosuppressive and pro-tumorigenic effects. Similarly, the equivalence or antagonism between aging-associated deregulated nutrient-sensing and cancer-relevant alterations of cellular metabolism is complex. The agonistic and antagonistic relationship between the processes that drive aging and cancer has bearings for the age-related increase and oldest age-related decrease of cancer morbidity and mortality, as well as for the therapeutic management of malignant disease in the elderly.
Collapse
Affiliation(s)
- Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain; Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Federico Pietrocola
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - David Roiz-Valle
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy, Villejuif, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
17
|
Deycmar S, Gomes B, Charo J, Ceppi M, Cline JM. Spontaneous, naturally occurring cancers in non-human primates as a translational model for cancer immunotherapy. J Immunother Cancer 2023; 11:e005514. [PMID: 36593067 PMCID: PMC9808758 DOI: 10.1136/jitc-2022-005514] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 01/03/2023] Open
Abstract
The complexity of cancer immunotherapy (CIT) demands reliable preclinical models to successfully translate study findings to the clinics. Non-human primates (NHPs; here referring to rhesus and cynomolgus macaques) share broad similarities with humans including physiology, genetic homology, and importantly also immune cell populations, immune regulatory mechanisms, and protein targets for CIT. Furthermore, NHP naturally develop cancers such as colorectal and breast cancer with an incidence, pathology, and age pattern comparable to humans. Thus, these tumor-bearing monkeys (TBMs) have the potential to bridge the experimental gap between early preclinical cancer models and patients with human cancer.This review presents our current knowledge of NHP immunology, the incidence and features of naturally-occurring cancers in NHP, and recent TBM trials investigating CIT to provide a scientific rationale for this unique model for human cancer.
Collapse
Affiliation(s)
- Simon Deycmar
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Bruno Gomes
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - Jehad Charo
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Maurizio Ceppi
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
- iTeos Therapeutics Inc, Watertown, Massachusetts, USA
| | - J Mark Cline
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
18
|
Kim D, Kiprov DD, Luellen C, Lieb M, Liu C, Watanabe E, Mei X, Cassaleto K, Kramer J, Conboy MJ, Conboy IM. Old plasma dilution reduces human biological age: a clinical study. GeroScience 2022; 44:2701-2720. [PMID: 35999337 PMCID: PMC9398900 DOI: 10.1007/s11357-022-00645-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/10/2022] [Indexed: 01/07/2023] Open
Abstract
This work extrapolates to humans the previous animal studies on blood heterochronicity and establishes a novel direct measurement of biological age. Our results support the hypothesis that, similar to mice, human aging is driven by age-imposed systemic molecular excess, the attenuation of which reverses biological age, defined in our work as a deregulation (noise) of 10 novel protein biomarkers. The results on biological age are strongly supported by the data, which demonstrates that rounds of therapeutic plasma exchange (TPE) promote a global shift to a younger systemic proteome, including youthfully restored pro-regenerative, anticancer, and apoptotic regulators and a youthful profile of myeloid/lymphoid markers in circulating cells, which have reduced cellular senescence and lower DNA damage. Mechanistically, the circulatory regulators of the JAK-STAT, MAPK, TGF-beta, NF-κB, and Toll-like receptor signaling pathways become more youthfully balanced through normalization of TLR4, which we define as a nodal point of this molecular rejuvenation. The significance of our findings is confirmed through big-data gene expression studies.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | | | - Connor Luellen
- Biophysics, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Michael Lieb
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Etsuko Watanabe
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Xiaoyue Mei
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | | | - Joel Kramer
- Brain Aging Center, UCSF, San Francisco, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
19
|
Lansdorp PM. Telomeres, Telomerase and Cancer. Arch Med Res 2022; 53:741-746. [PMID: 36334946 DOI: 10.1016/j.arcmed.2022.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Telomeres and telomerase play a crucial role in human aging and cancer. Three "drivers" of human aging can be identified. The developmental program encoded in DNA is the primary determinant of lifespan. Faithful execution of the developmental program requires stability of the (epi-)genome which is challenged throughout life by damage to DNA as well as epigenetic 'scars' from error-free DNA repair and stochastic errors made during the establishment and maintenance of the "epigenome". Over time (epi-)mutations accumulate, compromising cellular function and causing (pre-)malignant alterations. Damage to the genome and epigenome can be considered the second "driver" of aging. A third driver of the aging process, important to suppress tumors in long-lived animals, is caused by progressive loss of telomeric DNA. Telomere erosion protects against cancer early in life but limits cell renewal late in life, in agreement with the Antagonistic Pleiotropy theory on the evolutionary origin of aging. Malignant tumors arise when mutations and/or epimutations in cells (clock 2) corrupt the developmental program (clock 1) as well as tumor suppression by telomere erosion (clock 3). In cancer cells clock 3 is typically inactivated by loss of p53 as well as increased expression of telomerase. Taken together, aging in humans can be described by the ticking of three clocks: the clock that directs development, the accumulation of (epi-)mutations over time and the telomere clock that limits the number of cell divisions in normal stem and immune cells.
Collapse
Affiliation(s)
- Peter M Lansdorp
- Terry Fox Laboratory, BC Cancer Research Institute, Vancouver, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
20
|
Park JH, Kang I, Kim HC, Lee Y, Lee SK, Lee HK. Obesity enhances antiviral immunity in the genital mucosa through a microbiota-mediated effect on γδ T cells. Cell Rep 2022; 41:111594. [DOI: 10.1016/j.celrep.2022.111594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/11/2022] [Accepted: 10/12/2022] [Indexed: 11/09/2022] Open
|
21
|
Ren Z, Yang F, Yao S, Bi L, Jiang G, Huang J, Tang Y. Effects of low molecular weight peptides from monkfish (Lophius litulon) roe on immune response in immunosuppressed mice. Front Nutr 2022; 9:929105. [PMID: 36211506 PMCID: PMC9532971 DOI: 10.3389/fnut.2022.929105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate the immunomodulatory activation of low-molecular-weight peptides from monkfish (Lophius litulon) roe (named MRP) on cyclophosphamide (CTX)-induced immunosuppressed mice. Our results indicated that MRP (100 mg/kg/d BW) could significantly increase the body weight and immune organ index, and improve the morphological changes in the spleen and thymus of mice. These effects subsequently enhance the serum levels of interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, and immunoglobulin (Ig) A, IgM, and IgG. Furthermore, MRP could also improve CTX-induced oxidative stress, and activate the NF-κB and MAPK pathways in the spleen tissues. The findings reported herein indicate that MRP has a good immunomodulatory activation toward immunosuppressed mice, hence can potentially be developed as an immune adjuvant or functional food.
Collapse
Affiliation(s)
- Zhexin Ren
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Fei Yang
- Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Neonatal Intensive Care Unit, Hangzhou, China
| | - Sijia Yao
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Lijun Bi
- College of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, China
| | - Guanqin Jiang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
| | - Ju Huang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
- Key Laboratory of Health Risk Factors for Seafood of Zhejiang Province, Zhejiang Ocean University, Zhoushan, China
- *Correspondence: Ju Huang
| | - Yunping Tang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
- Yunping Tang
| |
Collapse
|
22
|
Effectiveness and Safety of PD-1 Inhibitor Monotherapy for Elderly Patients with Advanced Non-Small Cell Lung Cancer: A Real-World Exploratory Study. JOURNAL OF ONCOLOGY 2022; 2022:1710272. [PMID: 35909903 PMCID: PMC9337937 DOI: 10.1155/2022/1710272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 06/29/2022] [Indexed: 12/17/2022]
Abstract
Background Immunotherapy represented by PD-1 blockades had become the standard of care for advanced non-small cell lung cancer (NSCLC) gradually. Unfortunately, several PD-1 inhibitor-related studies excluded elderly patients with NSCLC over 75 years of age, resulting in relatively limited evidence regarding the efficacy and safety of PD-1 in elderly patients with NSCLC clinically. Objective This study aimed to identify the effectiveness and safety of PD-1 blockade monotherapy among elderly patients with advanced NSCLC. Methods Elderly patients with advanced NSCLC (≥65 years) who received PD-1 blockade monotherapy from September 2018 to December 2021 were screened retrospectively, and a total of 68 elderly patients with NSCLC were eligible for inclusion ultimately. The PD-1 blockades in the study were the available PD-1 monoclonal antibodies that had been approved for marketing in China, including camrelizumab, sintilimab, pembrolizumab, and nivolumab. The effectiveness and safety of the patients was collected retrospectively. Additionally, the correlation between prognosis and baseline characteristic subgroups was analyzed to identify the potential risk factors for progression-free survival (PFS). Results The median age of the 68 elderly patients with advanced NSCLC was 73 years (range: 65–82 years). Best overall response during PD-1 blockade administration suggested that no patients were found with complete response, partial response was found in 14 patients, stable disease was noted in 29 patients, and 25 patients had progressive disease, yielding an objective response rate (ORR) of 20.6% (95%CI: 11.7%–32.1%) and a disease control rate (DCR) of 63.2% (95%CI: 50.7%–74.6%). Furthermore, prognostic analysis exhibited that the median progression-free survival (PFS) of the 68 patients with advanced NSCLC was 3.5 months (95%CI: 2.4–4.6) and the median overall survival (OS) was 10.5 months (95%CI: 6.3–14.7). Additionally, a total of 48 patients were observed with the treatment-related adverse reaction (70.6%) of the 68 elderly patients with NSCLC, and the incidence of grade 3 or above adverse reactions was 16.2%. Specifically, the most common adverse reactions were fatigue, diarrhea, rash, and abnormal liver function with the incidence of 25.0%, 22.1%, 16.2%, and 14.7%, respectively. Exploratory analysis between PFS and baseline characteristic subgroups suggested that ECOG performance status and number of metastatic lesions might be independent factors for PFS. Conclusion PD-1 blockade monotherapy exhibited potential effectiveness and acceptable toxicity for elderly patients with NSCLC. ECOG performance status and number of metastatic lesions might be potential risk factors to predict the PFS of elderly patients with advanced NSCLC.
Collapse
|
23
|
Zhang H, Yao Y, Wu J, Zhou J, Zhao C, He J, Xu B. Comprehensive Analysis Identifies and Validates the Tumor Microenvironment Subtypes to Predict Anti-Tumor Therapy Efficacy in Hepatocellular Carcinoma. Front Immunol 2022; 13:838374. [PMID: 35924239 PMCID: PMC9339643 DOI: 10.3389/fimmu.2022.838374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 03/07/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThe objective of this study was to explore and verify the subtypes in hepatocellular carcinoma based on the immune (lymphocyte and myeloid cells), stem, and stromal cells in the tumor microenvironment and analyze the biological characteristics and potential relevance of each cluster.MethodsWe used the xCell algorithm to calculate cell scores and got subtypes by k-means clustering. In the external validation sets, we verified the conclusion stability by a neural network model. Simultaneously, we speculated the inner connection between clusters by pseudotime trajectory analysis and confirmed it by pathway enrichment, TMB, CNV, etc., analysis.ResultAccording to the results of the consensus cluster, we chose k = 4 as the optimal value and got four different subtypes (C1, C2, C3, and C4) with different biological characteristics based on infiltrating levels of 48 cells in TME. In univariable Cox regression, the hazard ratio (HR) value of C3 versus C1 was 2.881 (95% CI: 1.572–5.279); in multivariable Cox regression, we corrected the age and TNM stage, and the HR value of C3 versus C1 was 2.510 (95% CI: 1.339–4.706). C1 and C2 belonged to the immune-active type, C3 and C4 related to the immune-insensitive type and the potential conversion relationships between clusters. We established a neural network model, and the area under the curves of the neural network model was 0.949 in the testing cohort; the same survival results were also observed in the external validation set. We compared the differences in cell infiltration, immune function, pathway enrichment, TMB, and CNV of four clusters and speculated that C1 and C2 were more likely to benefit from immunotherapy and C3 may benefit from FGF inhibitors.DiscussionOur analysis provides a new approach for the identification of four tumor microenvironment clusters in patients with liver cancer and identifies the biological differences and predicts the immunotherapy efficacy between the four subtypes.
Collapse
Affiliation(s)
- Haohan Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- Hubei Provincial Research Center for Precision Medicine of Cancer, Wuhan, China
| | - Jie Wu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jin Zhou
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chen Zhao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Junju He
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Bin Xu,
| |
Collapse
|
24
|
Howard FHN, Kwan A, Winder N, Mughal A, Collado-Rojas C, Muthana M. Understanding Immune Responses to Viruses-Do Underlying Th1/Th2 Cell Biases Predict Outcome? Viruses 2022; 14:1493. [PMID: 35891472 PMCID: PMC9324514 DOI: 10.3390/v14071493] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/04/2022] [Accepted: 07/06/2022] [Indexed: 12/15/2022] Open
Abstract
Emerging and re-emerging viral diseases have increased in number and geographical extent during the last decades. Examples include the current COVID-19 pandemic and the recent epidemics of the Chikungunya, Ebola, and Zika viruses. Immune responses to viruses have been well-characterised within the innate and adaptive immunity pathways with the outcome following viral infection predominantly attributed to properties of the virus and circumstances of the infection. Perhaps the belief that the immune system is often considered as a reactive component of host defence, springing into action when a threat is detected, has contributed to a poorer understanding of the inherent differences in an individual's immune system in the absence of any pathology. In this review, we focus on how these host factors (age, ethnicity, underlying pathologies) may skew the T helper cell response, thereby influencing the outcome following viral infection but also whether we can use these inherent biases to predict patients at risk of a deviant response and apply strategies to avoid or overcome them.
Collapse
Affiliation(s)
- Faith H. N. Howard
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK; (A.K.); (N.W.); (A.M.); (C.C.-R.); (M.M.)
| | | | | | | | | | | |
Collapse
|
25
|
Huang ZL, Xu B, Li TT, Xu YH, Huang XY, Huang XY. Integrative Analysis Identifies Cell-Type-Specific Genes Within Tumor Microenvironment as Prognostic Indicators in Hepatocellular Carcinoma. Front Oncol 2022; 12:878923. [PMID: 35707353 PMCID: PMC9190278 DOI: 10.3389/fonc.2022.878923] [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: 02/18/2022] [Accepted: 04/13/2022] [Indexed: 11/13/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, but effective early detection and prognostication methods are lacking. Methods The Cox regression model was built to stratify the HCC patients. The single-cell RNA sequencing data analysis and gene set enrichment analysis were employed to investigate the biological function of identified markers. PLCB1 gain- or loss-of-function experiments were performed, and obtained HCC samples were analyzed using quantitative real-time PCR and immunohistochemistry assay to validate the biological function of identified markers. Results In this study, we developed a model using optimized markers for HCC recurrence prediction. Specifically, we screened out 8 genes through a series of data analyses, and built a multivariable Cox model based on their expression. The risk stratifications using the Eight-Gene Cox (EGC) model were closely associated with the recurrence-free survivals (RFS) in both training and three validation cohorts. We further demonstrated that this risk stratification could serve as an independent predictor in predicting HCC recurrence, and that the EGC model could outperform other models. Moreover, we also investigated the cell-type-specific expression patterns of the eight recurrence-related genes in tumor microenvironment using single-cell RNA sequencing data, and interpreted their functional roles from correlation and gene set enrichment analyses, in vitro and in vivo experiments. Particularly, PLCB1 and SLC22A7 were predominantly expressed in malignant cells, and they were predicted to promote angiogenesis and to help maintain normal metabolism in liver, respectively. In contrast, both FASLG and IL2RB were specifically expressed in T cells, and were highly correlated with T cell marker genes, suggesting that these two genes might assist in maintaining normal function of T cell-mediated immune response in tumor tissues. Conclusion In conclusion, the EGC model and eight identified marker genes could not only facilitate the accurate prediction of HCC recurrence, but also improve our understanding of the mechanisms behind HCC recurrence.
Collapse
Affiliation(s)
- Zi-Li Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Radiology, Xuhui District Central Hospital of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bin Xu
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of General Surgery, The Tenth People's Hospital of Tongji University, Shanghai, China
| | - Ting-Ting Li
- Department of Infectious Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yong-Hua Xu
- Department of Radiology, Xuhui District Central Hospital of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin-Yu Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiu-Yan Huang
- Department of General Surgery, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
26
|
Song P, Zhou S, Qi X, Jiao Y, Gong Y, Zhao J, Yang H, Qian Z, Qian J, Tang L. RNA modification writers influence tumor microenvironment in gastric cancer and prospects of targeted drug therapy. J Bioinform Comput Biol 2022; 20:2250004. [PMID: 35287562 DOI: 10.1142/s0219720022500044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Background: RNA adenosine modifications are crucial for regulating RNA levels. N6-methyladenosine (m6A), N1-methyladenosine (m1A), adenosine-to-inosine RNA editing, and alternative polyadenylation (APA) are four major RNA modification types. Methods: We evaluated the altered mRNA expression profiles of 27 RNA modification enzymes and compared the differences in tumor microenvironment (TME) and clinical prognosis between two RNA modification patterns using unsupervised clustering. Then, we constructed a scoring system, WM_score, and quantified the RNA modifications in patients of gastric cancer (GC), associating WM_score with TME, clinical outcomes, and effectiveness of targeted therapies. Results: RNA adenosine modifications strongly correlated with TME and could predict the degree of TME cell infiltration, genetic variation, and clinical prognosis. Two modification patterns were identified according to high and low WM_scores. Tumors in the WM_score-high subgroup were closely linked with survival advantage, CD4[Formula: see text] T-cell infiltration, high tumor mutation burden, and cell cycle signaling pathways, whereas those in the WM_score-low subgroup showed strong infiltration of inflammatory cells and poor survival. Regarding the immunotherapy response, a high WM_score showed a significant correlation with PD-L1 expression, predicting the effect of PD-L1 blockade therapy. Conclusion: The WM_scoring system could facilitate scoring and prediction of GC prognosis.
Collapse
Affiliation(s)
- Peng Song
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Sheng Zhou
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Xiaoyang Qi
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Yuwen Jiao
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Yu Gong
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Jie Zhao
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Haojun Yang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Zhifen Qian
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Jun Qian
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| | - Liming Tang
- Department of Gastrointestinal Surgery, The Affiliated Changzhou, No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, Jiangsu Province, P. R. China
| |
Collapse
|
27
|
Lansdorp PM. Telomeres, aging, and cancer: the big picture. Blood 2022; 139:813-821. [PMID: 35142846 PMCID: PMC8832478 DOI: 10.1182/blood.2021014299] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The role of telomeres in human health and disease is yet to be fully understood. The limitations of mouse models for the study of human telomere biology and difficulties in accurately measuring the length of telomere repeats in chromosomes and cells have diverted attention from many important and relevant observations. The goal of this perspective is to summarize some of these observations and to discuss the antagonistic role of telomere loss in aging and cancer in the context of developmental biology, cell turnover, and evolution. It is proposed that both damage to DNA and replicative loss of telomeric DNA contribute to aging in humans, with the differences in leukocyte telomere length between humans being linked to the risk of developing specific diseases. These ideas are captured in the Telomere Erosion in Disposable Soma theory of aging proposed herein.
Collapse
Affiliation(s)
- Peter M Lansdorp
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada; and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
28
|
Ligotti ME, Aiello A, Accardi G, Aprile S, Bonura F, Bulati M, Gervasi F, Giammanco GM, Pojero F, Zareian N, Caruso C, Farzaneh F, Candore G. Analysis of T and NK cell subsets in the Sicilian population from young to supercentenarian: The role of age and gender. Clin Exp Immunol 2021; 205:198-212. [PMID: 33866541 PMCID: PMC8274165 DOI: 10.1111/cei.13606] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 01/07/2023] Open
Abstract
Ageing dramatically affects number and function of both innate and adaptive arms of immune system, particularly T cell subsets, contributing to reduced vaccination efficacy, decreased resistance to infections and increased prevalence of cancer in older people. In the present paper, we analysed the age-related changes in the absolute number of lymphocytes in 214 Sicilian subjects, and in the percentages of T and natural killer (NK) cells in a subcohort of donors. We compared these results with the immunophenotype of the oldest living Italian supercentenarian (aged 111 years). The results were also sorted by gender. The correlation between number/percentage of cells and age in all individuals. and separately in males and females, was examined using a simple linear regression analysis. We did not record the increase in the rate of inversion of the CD4/CD8 ratio, frequently reported as being associated with ageing in literature. Our observation was the direct consequence of a flat average trend of CD4+ and CD8+ T cell percentages in ageing donors, even when gender differences were included. Our results also suggest that CD4+ and CD8+ subsets are not affected equally by age comparing females with males, and we speculated that gender may affect the response to cytomegalovirus (CMV) infection. The supercentenarian showed a unique immunophenotypic signature regarding the relative percentages of her T cell subsets, with CD4+ and CD8+ T cell percentages and CD4+ naive T cell values in line with those recorded for the octogenarian subjects. This suggests that the supercentenarian has a naive 'younger' T cell profile comparable to that of a >80-year-old female.
Collapse
Affiliation(s)
- Mattia Emanuela Ligotti
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
- School of Cancer and Pharmaceutical SciencesKing’s College LondonThe Rayne InstituteLondonUK
| | - Anna Aiello
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | - Giulia Accardi
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | - Stefano Aprile
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
- Unit of Transfusion MedicineSan Giovanni di Dio HospitalAgrigentoItaly
| | - Floriana Bonura
- Department of Health Promotion, Mother and Child CareInternal Medicine and Medical Specialties, Microbiology SectionUniversity of PalermoPalermoItaly
| | - Matteo Bulati
- Research DepartmentMediterranean Institute for Transplantation and Advanced Specialized Therapies (IRCCS ISMETT)PalermoItaly
| | - Francesco Gervasi
- Specialistic Oncology Laboratory UnitARNAS Hospitals CivicoDi Cristina e BenfratelliPalermoItaly
| | - Giovanni M. Giammanco
- Department of Health Promotion, Mother and Child CareInternal Medicine and Medical Specialties, Microbiology SectionUniversity of PalermoPalermoItaly
| | - Fanny Pojero
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | - Nahid Zareian
- School of Cancer and Pharmaceutical SciencesKing’s College LondonThe Rayne InstituteLondonUK
| | - Calogero Caruso
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| | - Farzin Farzaneh
- School of Cancer and Pharmaceutical SciencesKing’s College LondonThe Rayne InstituteLondonUK
| | - Giuseppina Candore
- Laboratory of Immunopathology and ImmunosenescenceDepartment of Biomedicine, Neuroscience and Advanced DiagnosticsUniversity of PalermoPalermoItaly
| |
Collapse
|
29
|
Traba J, Sack MN, Waldmann TA, Anton OM. Immunometabolism at the Nexus of Cancer Therapeutic Efficacy and Resistance. Front Immunol 2021; 12:657293. [PMID: 34079545 PMCID: PMC8166297 DOI: 10.3389/fimmu.2021.657293] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
Constitutive activity of the immune surveillance system detects and kills cancerous cells, although many cancers have developed strategies to avoid detection and to resist their destruction. Cancer immunotherapy entails the manipulation of components of the endogenous immune system as targeted approaches to control and destroy cancer cells. Since one of the major limitations for the antitumor activity of immune cells is the immunosuppressive tumor microenvironment (TME), boosting the immune system to overcome the inhibition provided by the TME is a critical component of oncotherapeutics. In this article, we discuss the main effects of the TME on the metabolism and function of immune cells, and review emerging strategies to potentiate immune cell metabolism to promote antitumor effects either as monotherapeutics or in combination with conventional chemotherapy to optimize cancer management.
Collapse
Affiliation(s)
- Javier Traba
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Michael N. Sack
- Cardiovascular Branch, National Heart, Lung and Blood Institute, NIH, Bethesda, MD, United States
| | - Thomas A. Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Olga M. Anton
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
30
|
Collet L, Delrieu L, Bouhamama A, Crochet H, Swalduz A, Nerot A, Marchal T, Chabaud S, Heudel PE. Association between Body Mass Index and Survival Outcome in Metastatic Cancer Patients Treated by Immunotherapy: Analysis of a French Retrospective Cohort. Cancers (Basel) 2021; 13:cancers13092200. [PMID: 34063692 PMCID: PMC8124396 DOI: 10.3390/cancers13092200] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022] Open
Abstract
The response to immunotherapy has been little investigated in overweight and obese cancer patients. We evaluated the relationships between BMI, toxicity, and survival in patients treated by immunotherapy for metastatic cancer. We included metastatic cancer patients treated by immunotherapy between January 2017 and June 2020 at the Centre Léon Bérard. In total, 272 patients were included: 64% men and 36% women, with a median age of 61.4 years. BMI ≥ 25 in 34.2% and 50% had non-small cell lung cancer (n = 136). Most received monotherapy, with nivolumab in 41.9% and pembrolizumab in 37.9%. Toxicity, mostly dysthyroiditis, occurred in 41%. Median overall survival (OS), estimated by Kaplan-Meier analysis, was significantly longer for patients with a BMI ≥ 25 than for those with a BMI < 25 (24.8 versus 13.7 months HR = 0.63; 95% CI 0.44-0.92, p = 0.015), and for patients experiencing toxicity than for those without toxicity (NR versus 7.8 months, HR = 0.22; 95% CI 0.15-0.33, p < 0.001). Adjusted OS was associated with toxicity, and the occurrence of toxicity was associated with sex and histological features but not with BMI. Thus, being overweight and experiencing toxicity was associated with longer overall survival in patients treated by immunotherapy. More attention should be paid to body composition in the care of cancer patients.
Collapse
Affiliation(s)
- Laetitia Collet
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France; (L.C.); (A.S.)
| | - Lidia Delrieu
- Department of Prevention Cancer Environment, Léon Bérard Cancer Centre, 69008 Lyon, France;
- Residual Tumor & Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, Paris University, 75005 Paris, France
| | - Amine Bouhamama
- Radiology Department, Centre Léon Bérard, 69008 Lyon, France; (A.B.); (A.N.)
| | - Hugo Crochet
- Data and Artificial Intelligence Team, Centre Léon Bérard, 69008 Lyon, France;
| | - Aurélie Swalduz
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France; (L.C.); (A.S.)
| | - Alexandre Nerot
- Radiology Department, Centre Léon Bérard, 69008 Lyon, France; (A.B.); (A.N.)
| | - Timothée Marchal
- Department of Supportive Care, Institut Curie, 75005 Paris, France;
| | - Sylvie Chabaud
- Department of Clinical Research and Innovation, Léon Bérard Cancer Centre, 69008 Lyon, France;
| | - Pierre Etienne Heudel
- Department of Medical Oncology, Centre Léon Bérard, 69008 Lyon, France; (L.C.); (A.S.)
- Correspondence: ; Tel.: +33-0478782958
| |
Collapse
|
31
|
Wallace K, Nahhas GJ, Bookhout C, Lewin DN, Paulos CM, Nikolaishvili-Feinberg N, Cohen SM, Guglietta S, Bakhtiari A, Camp ER, Hill EG, Baron JA, Wu JD, Alekseyenko AV. Preinvasive Colorectal Lesions of African Americans Display an Immunosuppressive Signature Compared to Caucasian Americans. Front Oncol 2021; 11:659036. [PMID: 33987094 PMCID: PMC8112239 DOI: 10.3389/fonc.2021.659036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/29/2021] [Indexed: 12/31/2022] Open
Abstract
Background African Americans (AAs) have higher colorectal cancer (CRC) incidence and mortality rate than Caucasian Americans (CAs). Recent studies suggest that immune responses within CRCs contribute to the disparities. If racially distinct immune signatures are present in the early phases of carcinogenesis, they could be used to develop interventions to prevent or slow disease. Methods We selected a convenience sample of 95 patients (48 CAs, 47 AAs) with preinvasive colorectal adenomas from the surgical pathology laboratory at the Medical University of South Carolina. Using immunofluorescent-conjugated antibodies on tissue slides from the lesions, we quantified specific immune cell populations: mast cells (CD117+), Th17 cells (CD4+RORC+), and NK cell ligand (MICA/B) and inflammatory cytokines, including IL-6, IL-17A, and IFN-γ. We compared the mean density counts (MDCs) and density rate ratios (RR) and 95% CI of immune markers between AAs to CAs using negative binomial regression analysis. We adjusted our models for age, sex, clinicopathologic characteristics (histology, location, dysplasia), and batch. Results We observed no racial differences in age or sex at the baseline endoscopic exam. AAs compared to CAs had a higher prevalence of proximal adenomas (66% vs. 40%) and a lower prevalence of rectal adenomas (11% vs. 23%) (p =0.04) but no other differences in pathologic characteristics. In age, sex, and batch adjusted models, AAs vs. CAs had lower RRs for cells labeled with IFNγ (RR 0.50 (95% CI 0.32-0.81); p=0.004) and NK cell ligand (RR 0.67 (0.43-1.04); p=0.07). In models adjusted for age, sex, and clinicopathologic variables, AAs had reduced RRs relative to CAs for CD4 (p=0.02), NK cell ligands (p=0.01), Th17 (p=0.005), mast cells (p=0.04) and IFN-γ (p< 0.0001). Conclusions Overall, the lower RRs in AAs vs. CAs suggests reduced effector response capacity and an immunosuppressive (‘cold’) tumor environment. Our results also highlight the importance of colonic location of adenoma in influencing these differences; the reduced immune responses in AAs relative to CAs may indicate impaired immune surveillance in early carcinogenesis. Future studies are needed to understand the role of risk factors (such as obesity) in influencing differences in immune responses by race.
Collapse
Affiliation(s)
- Kristin Wallace
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States.,Department of Public Health Sciences, MUSC, Charleston, SC, United States
| | - Georges J Nahhas
- Department of Public Health Sciences, MUSC, Charleston, SC, United States.,Department of Psychiatry and Behavioral Sciences, MUSC, Charleston, SC, United States
| | - Christine Bookhout
- Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - David N Lewin
- Department of Pathology and Laboratory Medicine, MUSC, Charleston, SC, United States
| | - Chrystal M Paulos
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States.,Department of Microbiology/Immunology, Emory University School of Medicine, Atlanta, GA, United States.,Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States
| | | | - Stephanie M Cohen
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Silvia Guglietta
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| | - Ali Bakhtiari
- Department of Public Health Sciences, MUSC, Charleston, SC, United States
| | - E Ramsay Camp
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States
| | - Elizabeth G Hill
- Hollings Cancer Center, Medical University of South Carolina (MUSC), Charleston, SC, United States.,Department of Public Health Sciences, MUSC, Charleston, SC, United States
| | - John A Baron
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| | - Jennifer D Wu
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alexander V Alekseyenko
- Department of Public Health Sciences, MUSC, Charleston, SC, United States.,Bioinformatics Center, MUSC, Charleston, SC, United States.,Department of Oral Health Sciences, MUSC, Charleston, SC, United States.,Department of Healthcare Leadership and Management, MUSC, Charleston, SC, United States
| |
Collapse
|
32
|
Metabolic Classification and Intervention Opportunities for Tumor Energy Dysfunction. Metabolites 2021; 11:metabo11050264. [PMID: 33922558 PMCID: PMC8146396 DOI: 10.3390/metabo11050264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 04/21/2021] [Accepted: 04/21/2021] [Indexed: 12/13/2022] Open
Abstract
A comprehensive view of cell metabolism provides a new vision of cancer, conceptualized as tissue with cellular-altered metabolism and energetic dysfunction, which can shed light on pathophysiological mechanisms. Cancer is now considered a heterogeneous ecosystem, formed by tumor cells and the microenvironment, which is molecularly, phenotypically, and metabolically reprogrammable. A wealth of evidence confirms metabolic reprogramming activity as the minimum common denominator of cancer, grouping together a wide variety of aberrations that can affect any of the different metabolic pathways involved in cell physiology. This forms the basis for a new proposed classification of cancer according to the altered metabolic pathway(s) and degree of energy dysfunction. Enhanced understanding of the metabolic reprogramming pathways of fatty acids, amino acids, carbohydrates, hypoxia, and acidosis can bring about new therapeutic intervention possibilities from a metabolic perspective of cancer.
Collapse
|
33
|
Drijvers JM, Gillis JE, Muijlwijk T, Nguyen TH, Gaudiano EF, Harris IS, LaFleur MW, Ringel AE, Yao CH, Kurmi K, Juneja VR, Trombley JD, Haigis MC, Sharpe AH. Pharmacologic Screening Identifies Metabolic Vulnerabilities of CD8 + T Cells. Cancer Immunol Res 2021; 9:184-199. [PMID: 33277233 PMCID: PMC7864883 DOI: 10.1158/2326-6066.cir-20-0384] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/19/2020] [Accepted: 11/24/2020] [Indexed: 11/16/2022]
Abstract
Metabolic constraints in the tumor microenvironment constitute a barrier to effective antitumor immunity and similarities in the metabolic properties of T cells and cancer cells impede the specific therapeutic targeting of metabolism in either population. To identify distinct metabolic vulnerabilities of CD8+ T cells and cancer cells, we developed a high-throughput in vitro pharmacologic screening platform and used it to measure the cell type-specific sensitivities of activated CD8+ T cells and B16 melanoma cells to a wide array of metabolic perturbations during antigen-specific killing of cancer cells by CD8+ T cells. We illustrated the applicability of this screening platform by showing that CD8+ T cells were more sensitive to ferroptosis induction by inhibitors of glutathione peroxidase 4 (GPX4) than B16 and MC38 cancer cells. Overexpression of ferroptosis suppressor protein 1 (FSP1) or cytosolic GPX4 yielded ferroptosis-resistant CD8+ T cells without compromising their function, while genetic deletion of the ferroptosis sensitivity-promoting enzyme acyl-CoA synthetase long-chain family member 4 (ACSL4) protected CD8+ T cells from ferroptosis but impaired antitumor CD8+ T-cell responses. Our screen also revealed high T cell-specific vulnerabilities for compounds targeting NAD+ metabolism or autophagy and endoplasmic reticulum (ER) stress pathways. We focused the current screening effort on metabolic agents. However, this in vitro screening platform may also be valuable for rapid testing of other types of compounds to identify regulators of antitumor CD8+ T-cell function and potential therapeutic targets.
Collapse
Affiliation(s)
- Jefte M Drijvers
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Jacob E Gillis
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Tara Muijlwijk
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Thao H Nguyen
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Emily F Gaudiano
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Isaac S Harris
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Martin W LaFleur
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Alison E Ringel
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Kiran Kurmi
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
| | - Vikram R Juneja
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Justin D Trombley
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, Massachusetts.
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
34
|
Jensen AWP, Carnaz Simões AM, thor Straten P, Holmen Olofsson G. Adrenergic Signaling in Immunotherapy of Cancer: Friend or Foe? Cancers (Basel) 2021; 13:cancers13030394. [PMID: 33494360 PMCID: PMC7866099 DOI: 10.3390/cancers13030394] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Exercise is associated with many aspects of a healthy lifestyle. Among these, exercise leads to the secretion of adrenaline and noradrenaline, which mobilize cells of the immune system, a process which is suggested to possess therapeutic value in cancer therapy, alone or in combination with immunotherapy. Strikingly, administration of β-blockers—which block the effect of adrenaline/noradrenaline—are also suggested to be useful in cancer therapy alone or in combination with immunotherapy. Herein we discuss the question of whether exercise and the administration of β-blockers could potentially be useful in cancer therapy. Abstract The incidence of cancer is increasing worldwide, which is to a large extent related to the population’s increasing lifespan. However, lifestyle changes in the Western world are causative as well. Exercise is intrinsically associated with what one could call a “healthy life”, and physical activity is associated with a lower risk of various types of cancer. Mouse models of exercise have shown therapeutic efficacy across numerous cancer models, at least in part due to the secretion of adrenaline, which mobilizes cells of the immune system, i.e., cytotoxic T and natural killer (NK) cells, through signaling of the β-2 adrenergic receptor (β2AR). Clinical trials aiming to investigate the clinical value of exercise are ongoing. Strikingly, however, the use of β-blockers—antagonists of the very same signaling pathway—also shows signs of clinical potential in cancer therapy. Cancer cells also express β-adrenergic receptors (βARs) and signaling of the receptor is oncogenic. Moreover, there are data to suggest that β2AR signaling in T cells renders the cell functionally suppressed. In this paper, we discuss these seemingly opposing mechanisms of cancer therapy—exercise, which leads to increased β2AR signaling, and β-blocker treatment, which antagonizes that same signaling—and suggest potential mechanisms and possibilities for their combination.
Collapse
Affiliation(s)
- Agnete Witness Praest Jensen
- National Center for Cancer Immune Therapy (DK-CCIT), Department of Oncology, University Hospital, DK-2730 Herlev, Denmark; (A.W.P.J.); (A.M.C.S.)
| | - Ana Micaela Carnaz Simões
- National Center for Cancer Immune Therapy (DK-CCIT), Department of Oncology, University Hospital, DK-2730 Herlev, Denmark; (A.W.P.J.); (A.M.C.S.)
| | - Per thor Straten
- National Center for Cancer Immune Therapy (DK-CCIT), Department of Oncology, University Hospital, DK-2730 Herlev, Denmark; (A.W.P.J.); (A.M.C.S.)
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: (P.t.S.); (G.H.O.); Tel.: +45-3868-2675 (P.t.S.); +45-3868-6418 (G.H.O.)
| | - Gitte Holmen Olofsson
- National Center for Cancer Immune Therapy (DK-CCIT), Department of Oncology, University Hospital, DK-2730 Herlev, Denmark; (A.W.P.J.); (A.M.C.S.)
- Correspondence: (P.t.S.); (G.H.O.); Tel.: +45-3868-2675 (P.t.S.); +45-3868-6418 (G.H.O.)
| |
Collapse
|
35
|
Zhao B, Wang W, Chen Z, Jin S. Apolipoprotein E deficiency enhances the anti-tumor immunity via repressing T cell exhaustion. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.2001382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Bian Zhao
- Department of Pharmacy, Luohe Medical College, Luohe, People’s Republic of China
| | - Wenbao Wang
- Department of Pharmacy, Luohe Medical College, Luohe, People’s Republic of China
| | - Zhijun Chen
- Department of Pharmacy, Luohe Hospital of Traditional Chinese Medicine, Luohe, People’s Republic of China
| | - Shaoju Jin
- Department of Pharmacy, Luohe Medical College, Luohe, People’s Republic of China
- Tumor Occurrence and Prevention Research Innovation Team of Henan, Luohe, People’s Republic of China
| |
Collapse
|
36
|
Ringel AE, Drijvers JM, Baker GJ, Catozzi A, García-Cañaveras JC, Gassaway BM, Miller BC, Juneja VR, Nguyen TH, Joshi S, Yao CH, Yoon H, Sage PT, LaFleur MW, Trombley JD, Jacobson CA, Maliga Z, Gygi SP, Sorger PK, Rabinowitz JD, Sharpe AH, Haigis MC. Obesity Shapes Metabolism in the Tumor Microenvironment to Suppress Anti-Tumor Immunity. Cell 2020; 183:1848-1866.e26. [PMID: 33301708 DOI: 10.1016/j.cell.2020.11.009] [Citation(s) in RCA: 373] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 07/27/2020] [Accepted: 11/04/2020] [Indexed: 01/12/2023]
Abstract
Obesity is a major cancer risk factor, but how differences in systemic metabolism change the tumor microenvironment (TME) and impact anti-tumor immunity is not understood. Here, we demonstrate that high-fat diet (HFD)-induced obesity impairs CD8+ T cell function in the murine TME, accelerating tumor growth. We generate a single-cell resolution atlas of cellular metabolism in the TME, detailing how it changes with diet-induced obesity. We find that tumor and CD8+ T cells display distinct metabolic adaptations to obesity. Tumor cells increase fat uptake with HFD, whereas tumor-infiltrating CD8+ T cells do not. These differential adaptations lead to altered fatty acid partitioning in HFD tumors, impairing CD8+ T cell infiltration and function. Blocking metabolic reprogramming by tumor cells in obese mice improves anti-tumor immunity. Analysis of human cancers reveals similar transcriptional changes in CD8+ T cell markers, suggesting interventions that exploit metabolism to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Alison E Ringel
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jefte M Drijvers
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Gregory J Baker
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Alessia Catozzi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Juan C García-Cañaveras
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Biomarkers and Precision Medicine Unit, Instituto de Investigación Sanitaria Fundación Hospital La Fe, València 46026, Spain
| | - Brandon M Gassaway
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Brian C Miller
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vikram R Juneja
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Thao H Nguyen
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Shakchhi Joshi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Cong-Hui Yao
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Haejin Yoon
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Peter T Sage
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Martin W LaFleur
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Justin D Trombley
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Connor A Jacobson
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Zoltan Maliga
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua D Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA; Biomarkers and Precision Medicine Unit, Instituto de Investigación Sanitaria Fundación Hospital La Fe, València 46026, Spain
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|