1
|
Ucgul AN, Bora H, Yaz Aydin G, Gulbahar O, Koken UH. Immune Modulation Through Stereotactic Radiotherapy: The Role of TBX21, GATA-3, FoxP3, and RORɣt. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:717. [PMID: 40283008 PMCID: PMC12028933 DOI: 10.3390/medicina61040717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Background and Objectives: Stereotactic radiotherapy enhances local tumor control by delivering high doses directly to the tumor. It is thought to activate the immune system via T-cells, possibly creating a systemic response. This study aims to evaluate stereotactic body radiotherapy's (SBRT) impact on the immune system by measuring T-cell transcription factors, such as TBX21, GATA-3, FoxP3, and RORɣt. Materials and Methods: Peripheral blood samples were collected from 103 patients before SBRT and from 66 patients two months post-treatment. We measured transcription factors TBX21, GATA-3, FOXP3, and RORγt using ELISA, and performed a complete blood count and C-reactive protein analysis to rule out infections. Statistical analyses included paired t-tests and correlation analyses to assess changes before and after treatment. Results: Post-treatment, significant reductions were observed in TBX21 (Th1), GATA-3 (Th2), and FOXP3 (Treg), while RORɣt (Th17) remained stable but trended higher in lung cancer patients. No correlations were found with demographic factors. However, TBX21 levels were significantly related to the planning target volume (PTV) and biologically effective dose (BED10) in the lung region. Larger PTVs (≥16.5 cc) and higher BED10 doses (≥100 Gy) were linked to smaller reductions in TBX21 (p = 0.008, p = 0.04) and increased RORɣt levels (p = 0.01). Conclusions: Stereotactic radiotherapy reduces immunosuppressive markers like FOXP3 and GATA-3, indicating its potential to boost immune activation by suppressing Treg and Th2 cells. Larger target volumes and higher BED10 values may enhance Th1 responses through TBX21. These findings suggest that SBRT activates the immune system, and its combination with immunotherapy could be promising.
Collapse
Affiliation(s)
- Aybala Nur Ucgul
- Department of Radiation Oncology, Gulhane Research and Training Hospital, Ankara 06010, Turkey
| | - Huseyin Bora
- Department of Radiation Oncology, Faculty of Medicine, Gazi University, Ankara 06830, Turkey
| | - Gizem Yaz Aydin
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara 06830, Turkey
| | - Ozlem Gulbahar
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara 06830, Turkey
| | - Ummu Habibe Koken
- Department of Radiation Oncology, Faculty of Medicine, Gazi University, Ankara 06830, Turkey
| |
Collapse
|
2
|
Luo C, Xiang H, Tan J. The efficacy of transcatheter arterial chemoembolization for hepatocellular carcinoma: is the alteration of the inflammation index important? Front Med (Lausanne) 2025; 12:1543903. [PMID: 40160321 PMCID: PMC11949957 DOI: 10.3389/fmed.2025.1543903] [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: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Transcatheter arterial chemoembolization (TACE) is widely applied for locoregional malignant lesions control in intermediate and selected advanced hepatocellular carcinoma (HCC). Various inflammation indices, such as neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), platelet-to-lymphocyte ratio (PLR), systemic immune inflammatory index (SII), and Lymphocyte-to-C Reactive Protein Ratio (LCR) have been explored as tools for predicting the efficacy of TACE. However, the role and predictive value for dynamic changes of peripheral inflammatory indicators pre- and post-TACE remains unclear. Objective To explore the association between the alteration in inflammatory index and the efficacy and prognosis of TACE and to provide more evidence for early prediction of the efficacy of TACE. Methods This was a retrospective single-center study. HCC patients who received TACE as initial treatment were enrolled. The relationship between the alteration of inflammation indices (calculated as post-TACE minus pre-TACE measurements) and TACE efficacy and prognosis was investigated. Progression-free survival (PFS) was the primary endpoint, and treatment efficacy was evaluated based on mRECIST criteria. Results Before propensity score matching (PSM), the change in LMR was significantly associated with treatment effective rate, with the unelevated ΔLMR group achieving a 79.4% treatment effective rate compared to 36.4% in the elevated group (p < 0.001). The estimated median PFS was 9.7 months and 4.5 months in the unelevated and elevated group, with a significant difference (p = 0.016). After PSM, the treatment effective rate was 48.7 and 38.5% (p = 0.214), and the estimated median PFS was 8.9 and 5.5 months (p = 0.173) for the unelevated and elevated group, respectively. Conclusion Our study demonstrated that alteration of indices of peripheral inflammation, including ΔNLR, ΔLMR, ΔSII, and ΔPLR, may not be valuable prognostic markers for HCC patients who received TACE.
Collapse
Affiliation(s)
- Chao Luo
- Department of Interventional Vascular Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| | - Hua Xiang
- Department of Interventional Vascular Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
- Department of Interventional Vascular Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Tan
- Department of Interventional Vascular Surgery, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, China
| |
Collapse
|
3
|
Magkouta S, Markaki E, Evangelou K, Petty R, Verginis P, Gorgoulis V. Decoding T cell senescence in cancer: Is revisiting required? Semin Cancer Biol 2025; 108:33-47. [PMID: 39615809 DOI: 10.1016/j.semcancer.2024.11.003] [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: 09/28/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/14/2024]
Abstract
Senescence is an inherent cellular mechanism triggered as a response to stressful insults. It associates with several aspects of cancer progression and therapy. Senescent cells constitute a highly heterogeneous cellular population and their identification can be very challenging. In fact, the term "senescence" has been often misused. This is also true in the case of immune cells. While several studies indicate the presence of senescent-like features (mainly in T cells), senescent immune cells are poorly described. Under this prism, we herein review the current literature on what has been characterized as T cell senescence and provide insights on how to accurately discriminate senescent cells against exhausted or anergic ones. We also summarize the major metabolic and epigenetic modifications associated with T cell senescence and underline the role of senescent T cells in the tumor microenvironment (TME). Moreover, we discuss how these cells associate with standard clinical therapeutic interventions and how they impact their efficacy. Finally, we underline the importance of precise identification and thorough characterization of "truly" senescent T cells in order to design successful therapeutic manipulations that would delay cancer incidence and maximize efficacy of immunotherapy.
Collapse
Affiliation(s)
- Sophia Magkouta
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece; Marianthi Simou and G.P. Livanos Labs, 1st Department of Critical Care and Pulmonary Services, School of Medicine, National & Kapodistrian University of Athens, "Evangelismos" Hospital, Athens 10676, Greece; Ninewells Hospital and Medical School, University of Dundee, Dundee DD19SY, UK
| | - Efrosyni Markaki
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, University of Crete Medical School, Heraklion 70013, Greece
| | - Konstantinos Evangelou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Russell Petty
- Ninewells Hospital and Medical School, University of Dundee, Dundee DD19SY, UK
| | - Panayotis Verginis
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, University of Crete Medical School, Heraklion 70013, Greece; Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Heraklion 70013, Greece
| | - Vassilis Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece; Ninewells Hospital and Medical School, University of Dundee, Dundee DD19SY, UK; Biomedical Research Foundation, Academy of Athens, Athens 11527, Greece; Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester M20 4GJ, UK.
| |
Collapse
|
4
|
Wang S, Mu X, Wang X, Chen L, Lu C, Song L. Peripheral Blood CD8 + CD28 + T Cells as an Independent Predictor of Treatment Response and Survival After Concurrent Chemoradiotherapy in Pediatric High-Grade Glioma Patients. Clin Med Insights Oncol 2024; 18:11795549241227421. [PMID: 38322666 PMCID: PMC10845990 DOI: 10.1177/11795549241227421] [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: 05/11/2023] [Accepted: 01/01/2024] [Indexed: 02/08/2024] Open
Abstract
Backgroud The tumor immune microenvironment influences the efficiency of concurrent chemoradiotherapy (CCRT) in high-grade glioma (HGG). This study investigated peripheral blood T lymphocyte subsets as clinical indicators of therapeutic response and prognosis in pediatric high-grade glioma (pHGG). Methods This retrospective study included 77 patients with postoperative pHGG who were treated concurrently with temozolomide and external beam radiotherapy between January 1, 2012, and December 31, 2018. The median follow-up was 26 (range: 5-106) months. Peripheral venous blood samples were collected before and after CCRT. The proportions of peripheral blood T lymphocytes and their association with treatment outcome and survival were determined. Results Sixty-four (83.1%) patients achieved complete remission, partial remission, and stable disease, and 13 (16.9%) patients had progressive disease. Higher CD3+ T cell, CD4+ T cell, and CD8+ CD28+ T cell ratios were predictive of better response, while a higher CD8+ CD28- T cell ratio was predictive of poorer response. Binary logistic regression analysis showed that the CD8+ CD28+ T cell ratio was a significant independent predictor of CCRT response (odds ratio [OR] = 53.521, 95% confidence interval [CI] = 4.294-667.119, P = .002). Univariate and multivariate analysis of prognostic factors associated with survival showed that the CD8+ CD28+ T lymphocyte ratio was a significant independent predictor of progression-free survival (hazard ratio [HR] = 1.80, 95% CI = 1.06-3.08, P = .03), but none of the subsets were significantly associated with overall survival. Conclusion Peripheral blood T lymphocytes have potential as predictors of CCRT response and prognosis in pHGG.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaofeng Mu
- Department of Radiotherapy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Li Chen
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Changyu Lu
- Department of Neurosurgery, Peking University International Hospital, Beijing, China
| | - Linan Song
- Department of Radiotherapy, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Wang H, Li Y, Hu P, Zhang J. The Correlation Between Low-Dose Radiotherapy Area of the Mediastinum and CD8+T Cells and the Efficacy of Radiotherapy for Non-Small Cell Lung Cancer. Cancer Manag Res 2024; 16:23-35. [PMID: 38230351 PMCID: PMC10790660 DOI: 10.2147/cmar.s438440] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/08/2024] [Indexed: 01/18/2024] Open
Abstract
Background Radiation therapy (RT) can cause changes in peripheral blood immune cells. The relationship between the efficacy of radiation therapy for non-small cell lung cancer (NSCLC) and immune cell changes and the study of how mediastinal radiation dose parameters affect immune cell changes is still unclear. This study aims to analyze the relationship between immune cell changes induced by radiotherapy and the efficacy of NSCLC radiotherapy, as well as the relationship between radiotherapy dose parameters and immune cell changes. Materials and Methods We retrospectively analyzed the data of NSCLC patients receiving mediastinal radiation therapy from 2020 to 2022. Collect lymphocytes and circulating immune cells within one week before and after radiotherapy and collect the dose-volume parameters of the whole mediastinum in the patient's RT planning system. Analyze the changes in lymphocytes and radiotherapy effects after radiotherapy, and explore the relationship between radiotherapy dose parameters and immune cell changes. Results A total of 72 patients were enrolled. Compared with before radiotherapy, the proportion of CD3+T cells, CD8+T cells, and CD8/Treg in peripheral blood significantly increased after radiotherapy (P<0.05). The increase in CD8+T cells and CD8/Treg after radiotherapy was correlated with Objective response rate (ORR) (P<0.05). Based on binary logistic univariate and multivariate regression analysis, an increase in CD8+T cells after radiotherapy is an independent predictor of objective tumor response after radiotherapy (OR=12.71, 95% CI=3.64-44.64, P=0.01), and Volume of 200 cGy irradiation (V2) is an independent positive predictor of an increase in CD8+T lymphocyte ratio after radiotherapy (high group, OR=3.40, 95% CI=1.13-10.36, P=0.03). Conclusion The increase in CD8+T cells after radiotherapy can positively predict the short-term efficacy of radiotherapy. Mediastinal low-dose radiation therapy can increase CD8+T cells, thereby improving the short-term efficacy of radiotherapy. These potentially related mechanisms are worth further verification and exploration.
Collapse
Affiliation(s)
- Hang Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| | - Yang Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| | - Pingping Hu
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| | - Jiandong Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong Province, 250000, People’s Republic of China
- Shandong Lung Cancer Institute, Jinan, Shandong Province, 25000, People’s Republic of China
| |
Collapse
|
6
|
Xu Y, Kong Y, Ma Y, Xu M, Yang J, Zhang J, Chen R, Chen G, Hong Z, Zhao X, Zhang C, Xing P, Zhang L, Zhao P. Phase I/II Clinical Study of PRaG Regimen Combined With Intraperitoneal Infusion of PD-1 Inhibitor for Advanced Refractory Solid Tumors With Cancerous Ascites (PRaG4.0P Study). Technol Cancer Res Treat 2024; 23:15330338241264169. [PMID: 39051686 PMCID: PMC11273709 DOI: 10.1177/15330338241264169] [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: 01/06/2024] [Revised: 04/23/2024] [Accepted: 05/29/2024] [Indexed: 07/27/2024] Open
Abstract
Objective: The prognosis of malignant tumors with peritoneal metastases and cancerous ascites has generally been poor, with limited treatment options. The PRaG regimen, which comprised of hypofractionated radiotherapy, programmed cell death-1 (PD-1) inhibitor, and granulocyte-macrophage colony-stimulating factor (GM-CSF), showed a survival advantage in patients with advanced solid tumors who failed at least the first line of standard systemic treatment. Intraperitoneal infusion of PD-1 inhibitors may be a novel therapeutic strategy for managing malignant ascites. Integrating the PRaG regimen with intraperitoneal perfusion of a PD-1 inhibitor might control malignant ascites and provide further survival benefits in these patients. This proposed study aims to investigate the safety and efficacy of intraperitoneal infusion of serplulimab in combination with the PRaG regimen in patients with simultaneous advanced solid tumors and cancerous ascites who fail at least the first-line treatment. Methods: This proposed study is a prospective, single-arm, open-label, multicenter clinical trial. All eligible patients will receive 2 cycles of intensive treatment, a combination of PRaG regimen with an intraperitoneal infusion of PD-1 inhibitor. The patients who are beneficially treated with intensive treatment will receive consolidation treatment every 2 weeks until ascites disappear, disease progression occurs, intolerable toxicity occurs, or for up to 1 year. Phase I of this study will be conducted using a modified 3 + 3 design. The dose of intraperitoneal infusion of PD-1 inhibitor for phase II will be determined according to dose-limiting toxicity evaluation in the phase I study. Conclusion: This prospective, open-label, multicenter study will potentially lead to intraperitoneal perfusion of a PD-1 inhibitor being a new strategy for malignant ascites patients and provide a meaningful efficacy and safety of the combination of PRaG regimen with an intraperitoneal infusion of PD-1 inhibitor for these patients.
Collapse
Affiliation(s)
- Yingying Xu
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuehong Kong
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yifu Ma
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Meiling Xu
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiabao Yang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Junjun Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Rongzheng Chen
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Guangqiang Chen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihui Hong
- Department of Nuclear Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangrong Zhao
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenyang Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Pengfei Xing
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liyuan Zhang
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Peifeng Zhao
- Center for Cancer Diagnosis and Treatment, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institution of Radiotherapy and Oncology, Soochow University, Suzhou, China
- Laboratory for Combined Radiotherapy and Immunotherapy of Cancer, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
7
|
Geng R, Tang H, You T, Xu X, Li S, Li Z, Liu Y, Qiu W, Zhou N, Li N, Ge Y, Guo F, Sun Y, Wang Y, Li T, Bai C. Peripheral CD8+CD28+ T lymphocytes predict the efficacy and safety of PD-1/PD-L1 inhibitors in cancer patients. Front Immunol 2023; 14:1125876. [PMID: 36969245 PMCID: PMC10038730 DOI: 10.3389/fimmu.2023.1125876] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundProgrammed cell death protein-1/programmed cell death ligand-1 (PD-1/PD-L1) inhibitors works by reactivating immune cells. Considering the accessibility of noninvasive liquid biopsies, it is advisable to employ peripheral blood lymphocyte subsets to predict immunotherapy outcomes.MethodsWe retrospectively enrolled 87 patients with available baseline circulating lymphocyte subset data who received first-line PD-1/PD-L1 inhibitors at Peking Union Medical College Hospital between May 2018 and April 2022. Immune cell counts were determined by flow cytometry.ResultsPatients who responded to PD-1/PD-L1 inhibitors had significantly higher circulating CD8+CD28+ T-cell counts (median [range] count: 236 [30-536] versus 138 [36-460]/μL, p < 0.001). Using 190/μL as the cutoff value, the sensitivity and specificity of CD8+CD28+ T cells for predicting immunotherapy response were 0.689 and 0.714, respectively. Furthermore, the median progression-free survival (PFS, not reached versus 8.7 months, p < 0.001) and overall survival (OS, not reached versus 16.2 months, p < 0.001) were significantly longer in the patients with higher CD8+CD28+ T-cell counts. However, the CD8+CD28+ T-cell level was also associated with the incidence of grade 3-4 immune-related adverse events (irAEs). The sensitivity and specificity of CD8+CD28+ T cells for predicting irAEs of grade 3-4 were 0.846 and 0.667, respectively, at the threshold of CD8+CD28+ T cells ≥ 309/μL.ConclusionsHigh circulating CD8+CD28+ T-cell levels is a potential biomarker for immunotherapy response and better prognosis, while excessive CD8+CD28+ T cells (≥ 309/μL) may also indicate the emergence of severe irAEs.
Collapse
Affiliation(s)
- Ruixuan Geng
- Department of International Medical Services, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hui Tang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tingting You
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuxiu Xu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Sijian Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Obstetric and Gynecologic Diseases, Beijing, China
| | - Zepeng Li
- Department of Laboratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan Liu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Qiu
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ningning Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fuping Guo
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhong Sun
- Department of Radiation Oncology, Dandong First Hospital, Dandong, Liaoning, China
| | - Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yingyi Wang, ; Taisheng Li,
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Yingyi Wang, ; Taisheng Li,
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Wang Z, Ge Y, Li H, Fei G, Wang S, Wei P. Identification and validation of a genomic mutation signature as a predictor for immunotherapy in NSCLC. Biosci Rep 2022; 42:BSR20220892. [PMID: 36305643 PMCID: PMC9702799 DOI: 10.1042/bsr20220892] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/05/2022] [Accepted: 10/27/2022] [Indexed: 08/28/2023] Open
Abstract
Currently, the benefits of immune checkpoint inhibitor (ICI) therapy prediction via emerging biomarkers have been identified, and the association between genomic mutation signatures (GMS) and immunotherapy benefits has been widely recognized as well. However, the evidence about non-small cell lung cancer (NSCLC) remains limited. We analyzed 310 immunotherapy patients with NSCLC from the Memorial Sloan Kettering Cancer Center (MSKCC) cohort. Lasso Cox regression was used to construct a GMS, and the prognostic value of GMS could be able to verify in the Rizvi cohort (N=240) and Hellmann cohort (N=75). We further conducted immunotherapy-related characteristics analysis in The Cancer Genome Atlas (TCGA) cohort (N=1052). A total of seven genes (ZFHX3, NTRK3, EPHA7, MGA, STK11, EPHA5, TP53) were identified for GMS model construction. Compared with GMS-high patients, patients with GMS-low had longer overall survival (OS; P<0.001) in the MSKCC cohort and progression-free survival (PFS; P<0.001) in the validation cohort. Multivariate Cox analysis revealed that GMS was an independent predictive factor for NSCLC patients in both the MSKCC and validation cohort. Meanwhile, we found that GMS-low patients reflected enhanced antitumor immunity in TCGA cohort. The results indicated that GMS had not only potential predictive value for the benefit of immunotherapy but also may serve as a potential biomarker to guide clinical ICI treatment decisions for NSCLC.
Collapse
Affiliation(s)
- Zemin Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - You Ge
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Han Li
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Gaoqiang Fei
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Shuai Wang
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Pingmin Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Wei H, Li Y, Guo Z, Ma X, Li Y, Wei X, Han D, Zhang T, Chen X, Yan C, Zhou J, Pang Q, Wang P, Zhang W. Comparison of dynamic changes in the peripheral CD8 + T cells function and differentiation in ESCC patients treated with radiotherapy combined with anti-PD-1 antibody or concurrent chemoradiotherapy. Front Immunol 2022; 13:1060695. [PMID: 36479110 PMCID: PMC9720318 DOI: 10.3389/fimmu.2022.1060695] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/07/2022] [Indexed: 11/22/2022] Open
Abstract
Objective The systematic immune status of cancer patients undergoing immunotherapy is little known. We prospectively identified the function and differentiation traits of peripheral CD8+ T cells based on our phase 1b clinical trial (NCT03222440) of radiotherapy combined with camrelizumab in patients with locally advanced esophageal squamous cell carcinoma (ESCC) and compared it with concurrent chemoradiotherapy (CCRT). Methods 19 and 18 patients were included in the cohort of radiotherapy plus camrelizumab and cohort of CCRT treatment. By using flow cytometry, we evaluated the expression levels of PD-1, Eomes, T-bet and IFN-γ (function), CD38 and HLA-DR (activation), and differentiation subsets classified according to the expression levels of CD45RA and CD62L in peripheral CD8+ T cells before and during treatment. Results Effective binding of anti-PD-1 antibody camrelizumab with PD-1 on CD8+ T cells was detected during treatment. Both two treatments elevated the expression levels of activation molecules CD38 and HLA-DR on CD8+ T cells. PD-1+CD8+ T cells had more activation features than PD-1-CD8+ T cells in two groups and the treatments did not alter these differences. The two treatments activated both PD-1+ and PD-1- CD8+ T cells. PD-1+CD8+ T cells had less Naïve and TEMRA but more Tcm and Tem than PD-1-CD8+ T cells in two groups and both two treatments changed the ratio of memory T cells in PD-1+ and PD-1- cells. RT plus camrelizumab treatment reduced Naïve T cells and TEMRA subsets both in PD-1+ and PD-1- CD8+ T cells while elevated Tcm subset in PD-1+CD8+ T cells and Tem subset in PD-1-CD8+ T cells. CCRT elevated Tcm subset and reduced TEMRA subset in PD-1-CD8+ T cells while did not change any subset in PD-1+CD8+ T cells. Furthermore, patients undergoing radiotherapy plus immunotherapy were found to obtain better prognosis than those receiving CCRT. Conclusions This study identified the dynamic changes of systematic immune status of patients undergoing treatment. The two treatments had similar activation effects on peripheral CD8+ T cells with different PD-1 properties but had different effects on their differentiation status. These results provided potential clues to the reasons underlying the difference in prognosis of the two treatments.
Collapse
Affiliation(s)
- Hui Wei
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yanqi Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Zhoubo Guo
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoxue Ma
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yang Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xiaoying Wei
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Dong Han
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jiahuan Zhou
- Department of Clinical Research and Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,*Correspondence: Qingsong Pang, ; Ping Wang, ; Wencheng Zhang,
| |
Collapse
|
10
|
Expression of CD28 in Hepatocellular Carcinoma and Its Prognostic Value. HEPATITIS MONTHLY 2022. [DOI: 10.5812/hepatmon.118605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
Background: CD28 expression is correlated with malignancy development in long-term survivors after liver transplantation. Immune cell activation is mediated by the interaction of CD28 with CD4 and CD8. Objectives: In this study, we attempted to investigate the expression level and prognostic value of CD28 in hepatocellular carcinoma (HCC). Methods: A total of 54 HCC patients with complete clinical information were examined. The expression level of CD28 in HCC tissues was detected by immunohistochemistry. The correlations of CD28 expression with clinical characteristics, CD4+/CD8+ T-cells, and prognosis in HCC were analyzed. The expression profile of CD28 and survival time of HCC patients were retrieved from the TCGA database, followed by survival analysis. Results: The positive expression rate of CD28 in HCC tissues was 70.73%. The CD28 expression was significantly higher in the positive expression group (area: 659174.9 ± 670060, IOD: 123348.3 ± 106348.6) than in the negative expression group (area: 8405.7 ± 9983.3, IOD: 1959.6 ± 2117.7) (P < 0.01). The CD4+ and CD8+ cell counts were 526.13 ± 258.17 cells/µL and 383.93 ± 223.39 cells/µL, respectively. The expression level of CD28 was significantly related to the degree of differentiation and the number of CD4+ and CD8+ T-cells (P < 0.05). The survival time of patients was longer in the positive CD28 expression group than in the negative expression group. Based on the CD28 expression profiles of 406 HCC patients retrieved from the TCGA database, patients with high CD28 expression showed a better prognosis than those with low expression (P < 0.05). Conclusions: CD28 may play a vital role in the occurrence, development, and prognosis of HCC by interacting with CD4+ and CD8+ T-cells. Thus, CD28 could be suggested as the immune checkpoint target for HCC treatment.
Collapse
|
11
|
Larionova I, Rakina M, Ivanyuk E, Trushchuk Y, Chernyshova A, Denisov E. Radiotherapy resistance: identifying universal biomarkers for various human cancers. J Cancer Res Clin Oncol 2022; 148:1015-1031. [PMID: 35113235 DOI: 10.1007/s00432-022-03923-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 01/12/2022] [Indexed: 12/16/2022]
Abstract
Radiotherapy (RT) is considered as a standard in the treatment of most solid cancers, including glioblastoma, lung, breast, rectal, prostate, colorectal, cervical, esophageal, and head and neck cancers. The main challenge in RT is tumor cell radioresistance associated with a high risk of locoregional relapse and distant metastasis. Despite significant progress in understanding mechanisms of radioresistance, its prediction and overcoming remain unresolved. This review presents the state-of-the-art for the potential universal biomarkers correlated to the radioresistance and poor outcome in different cancers. We describe radioresistance biomarkers functionally attributed to DNA repair, signal transduction, hypoxia, and angiogenesis. We also focus on high throughput genetic and proteomic studies, which revealed a set of molecular biomarkers related to radioresistance. In conclusion, we discuss biomarkers which are overlapped in most several cancers.
Collapse
Affiliation(s)
- Irina Larionova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia.
| | - Militsa Rakina
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, 634050, Tomsk, Russia
| | - Elena Ivanyuk
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| | - Yulia Trushchuk
- Department of Gynecologic Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| | - Alena Chernyshova
- Department of Gynecologic Oncology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| | - Evgeny Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634009, Tomsk, Russia
| |
Collapse
|
12
|
Ruiz A, Flores-Gonzalez J, Buendia-Roldan I, Chavez-Galan L. Telomere Shortening and Its Association with Cell Dysfunction in Lung Diseases. Int J Mol Sci 2021; 23:425. [PMID: 35008850 PMCID: PMC8745057 DOI: 10.3390/ijms23010425] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 01/10/2023] Open
Abstract
Telomeres are localized at the end of chromosomes to provide genome stability; however, the telomere length tends to be shortened with each cell division inducing a progressive telomere shortening (TS). In addition to age, other factors, such as exposure to pollutants, diet, stress, and disruptions in the shelterin protein complex or genes associated with telomerase induce TS. This phenomenon favors cellular senescence and genotoxic stress, which increases the risk of the development and progression of lung diseases such as idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease, SARS-CoV-2 infection, and lung cancer. In an infectious environment, immune cells that exhibit TS are associated with severe lymphopenia and death, whereas in a noninfectious context, naïve T cells that exhibit TS are related to cancer progression and enhanced inflammatory processes. In this review, we discuss how TS modifies the function of the immune system cells, making them inefficient in maintaining homeostasis in the lung. Finally, we discuss the advances in drug and gene therapy for lung diseases where TS could be used as a target for future treatments.
Collapse
Affiliation(s)
| | | | | | - Leslie Chavez-Galan
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City 14080, Mexico; (A.R.); (J.F.-G.); (I.B.-R.)
| |
Collapse
|
13
|
Zhao X, Zhang Y, Gao Z, Han Y. Prognostic value of peripheral naive CD8 + T cells in oligometastatic non-small-cell lung cancer. Future Oncol 2021; 18:55-65. [PMID: 34608815 DOI: 10.2217/fon-2021-0728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aim: This study aimed to investigate the prognostic value of peripheral naive and memory CD8+ and CD4+ T cells and other immune cells in patients with oligometastatic non-small-cell lung cancer (NSCLC) undergoing radiotherapy (RT). Methods: A total of 142 patients with oligometastatic NSCLC treated with RT were enrolled, and their blood samples were collected within 3 days before RT. Immune cells were identified by flow cytometry. Results: Patients with high levels of naive CD8+ T cells had longer overall survival (p = 0.004) and progression-free survival (p = 0.001) than those with low levels of naive CD8+ T cells. Multivariate analyses revealed that naive CD8+ T cells were independently correlated with overall survival (p = 0.019) and progression-free survival (p = 0.024). Conclusion: The results suggest that peripheral naive CD8+ T cells may be an independent prognostic indicator for patients with oligometastatic NSCLC undergoing RT.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Oncology, Hebei Medical University, Shijiazhuang 050017, PR China
| | - Yan Zhang
- Department of Oncology, Hebei Medical University, Shijiazhuang 050017, PR China.,Department of Oncology, Shijiazhuang People's Hospital, Shijiazhuang 050030, PR China
| | - Zhenlin Gao
- Department of Oncology, Shijiazhuang People's Hospital, Shijiazhuang 050030, PR China
| | - Yaguang Han
- Department of Oncology, Shijiazhuang People's Hospital, Shijiazhuang 050030, PR China
| |
Collapse
|
14
|
Yang D, Wang X, Zhou X, Zhao J, Yang H, Wang S, Morse MA, Wu J, Yuan Y, Li S, Hobeika A, Lyerly HK, Ren J. Blood microbiota diversity determines response of advanced colorectal cancer to chemotherapy combined with adoptive T cell immunotherapy. Oncoimmunology 2021; 10:1976953. [PMID: 34595059 PMCID: PMC8477924 DOI: 10.1080/2162402x.2021.1976953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/18/2021] [Accepted: 09/01/2021] [Indexed: 12/17/2022] Open
Abstract
Human microbiota influence the response of malignancies to treatment with immune checkpoint blockade; however, their impact on other forms of immunotherapy is poorly understood. This study explored the effect of blood microbiota on clinical efficacy, represented by progression-free survival (PFS) and overall survival (OS), of combined chemotherapy and adoptive cellular therapy (ACT) in advanced colon cancer patients. Plasma was collected from colorectal cancer patients (CRC) treated with either chemotherapy alone (oxaliplatin and capecitabine) (XELOX CT alone group, n = 19), or ACT with a mixed dendritic cell/cytokine-induced killer cell product (DC-CIK) + XELOX (ICT group, n = 20). Circulating microbiota analysis was performed by PCR amplification and next-generation sequencing of variable regions V3~V4 of bacterial 16S rRNA genes. The association of the blood microbial diversity with clinical response to the therapy as measured by RECIST1.1 and OS was evaluated. The baseline Chao index of blood microbial diversity predicted prolonged PFS and OS of DC/CIK immunotherapy. More diverse blood microbiota that included Bifidobacterium, Lactobacillus, and Enterococcus were identified among responders to DC/CIK compared with non-responders. The plasma bacterial DNA copy number is inversely correlated with the CD3-/CD16+/CD56+ NK cells in circulation and decreased following DC-CIK; however, the Chao index of plasma microbiota significantly increased after administration of the DC-CIK product and this subsequent change was correlated with the number of CD3-/CD16+/CD56+ and CD8+/CD28+ cells infused. The diversity of the blood microbiome is a promising predictive marker for clinical responses to chemotherapy combined with DC-CIK. Cellular immunotherapy can affect the plasma microbiota's diversity in a manner favorable to clinical responses.
Collapse
Affiliation(s)
- Duo Yang
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Xiaoli Wang
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Xinna Zhou
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Jing Zhao
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Huabing Yang
- Department of Medical Oncology, Fudan University Pudong Medical Center, Shanghai, China
| | - Shuo Wang
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Michael A. Morse
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Jiangping Wu
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Yanhua Yuan
- Department of Medical Oncology, Fudan University Pudong Medical Center, Shanghai, China
| | - Sha Li
- Department of Therapeutic Cancer Vaccines and Medical OncologyBeijing Shijitan Hospital, Capital Medical University, Beijing Key Laboratory for Therapeutic Cancer Vaccines, Beijing, China
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | - Jun Ren
- Department of Medical Oncology, Fudan University Pudong Medical Center, Shanghai, China
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
15
|
Identification and Prognostic Value Exploration of Radiotherapy Sensitivity-Associated Genes in Non-Small-Cell Lung Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5963868. [PMID: 34518802 PMCID: PMC8433590 DOI: 10.1155/2021/5963868] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/16/2021] [Accepted: 08/17/2021] [Indexed: 11/30/2022]
Abstract
Background Non-small-cell lung cancer (NSCLC) is a prevalent malignancy with high mortality and poor prognosis. The radiotherapy is one of the most common treatments of NSCLC, and the radiotherapy sensitivity of patients could affect the individual prognosis of NSCLC. However, the prognostic signatures related to radiotherapy response still remain limited. Here, we explored the radiosensitivity-associated genes and constructed the prognostically predictive model of NSCLC cases. Methods The NSCLC samples with radiotherapy records were obtained from The Cancer Genome Atlas database, and the mRNA expression profiles of NSCLC patients from the GSE30219 and GSE31210 datasets were obtained from the Gene Expression Omnibus database. The Weighted Gene Coexpression Network Analysis (WGCNA), univariate, least absolute shrinkage and selection operator (LASSO), multivariate Cox regression analysis, and nomogram were conducted to identify and validate the radiotherapy sensitivity-related signature. Results WGCNA revealed that 365 genes were significantly correlated with radiotherapy response. LASSO Cox regression analysis identified 8 genes, including FOLR3, SLC6A11, ALPP, IGFN1, KCNJ12, RPS4XP22, HIST1H2BH, and BLACAT1. The overall survival (OS) of the low-risk group was better than that of the high-risk group separated by the Risk Score based on these 8 genes for the NSCLC patients. Furthermore, the immune infiltration analysis showed that monocytes and activated memory CD4 T cells had different relative proportions in the low-risk group compared with the high-risk group. The Risk Score was correlated with immune checkpoints, including CTLA4, PDL1, LAG3, and TIGIT. Conclusion We identified 365 genes potentially correlated with the radiotherapy response of NSCLC patients. The Risk Score model based on the identified 8 genes can predict the prognosis of NSCLC patients.
Collapse
|
16
|
Wang Y, Zhou N, Zhu R, Li X, Sun Z, Gao Y, Liu W, Meng C, Ge Y, Bai C, Li T, Liu H. Circulating activated immune cells as a potential blood biomarkers of non-small cell lung cancer occurrence and progression. BMC Pulm Med 2021; 21:282. [PMID: 34488711 PMCID: PMC8420051 DOI: 10.1186/s12890-021-01636-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/10/2021] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Treatment for non-small cell lung cancer (NSCLC) has greatly improved in recent years. However, noninvasive early screening for carcinogenesis and progression unclear. The aim of this study was to explore the predictive value of peripheral blood immune cells in untreated NSCLC patients. METHODS We retrospectively enrolled 305 untreated NSCLC patients and 132 healthy participants from February 2016 to August 2019 in Peking Union Medical College Hospital. Immune cell levels were determined by flow cytometry and routine blood tests. RESULTS NSCLC patients had lower levels of T lymphocytes, NK cells, CD8+ T cells, naïve CD4+/CD4+, naïve CD4+ T cells and higher levels of CD4+ T cells, memory CD4+/CD4+ T cells, memory CD4+ T cells, CD4+CD28+/CD4+ T cells, CD4+CD28+ T cells, CD8+CD28+/CD8+ T cells, CD8+HLA-DR+/CD8+ T cells, CD8+HLA-DR+ T cells T cells, CD8+CD38+/CD8+ T cells, CD8+CD38+ T cells and CD4+/CD8+ T cells than those in controls. The percentages of specific lymphocyte subtypes were significantly different in cancer patients versus healthy individuals. For instance, cancer patients had lower levels of B cells, CD4+ T cells, naïve CD4+/CD4+ T cells, naïve CD4+ T cells, CD4+CD28+ T cells, CD8+CD28+ T cells and higher levels of NK cells, white blood cells (WBC), monocytes, neutrophils, eosinophils, basophils, monocytes to lymphocyte ratio (MLR), neutrophils to lymphocyte ratio (NLR), eosinophil to lymphocyte ratio (ELR), basophil to lymphocyte ratio (BLR), and blood platelet to lymphocyte ratio (PLR). CONCLUSIONS Abnormal T cell levels can be used as an independent predictive biomarker for noninvasive early screening in NSCLC occurrence and progression.
Collapse
Affiliation(s)
- Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Zhu
- Department of Medical Record, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyuan Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhao Sun
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Gao
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Liu
- Department of Intervention Group of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changting Meng
- Institute for Systems Biology, Seattle University, Seattle, WA, 98109, USA
| | - Yuping Ge
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Taisheng Li
- Department of Infectious Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China.
| | - Hongsheng Liu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
17
|
Liu Q, Zhao C, Jiang P, Liu D. Circulating tumor cells counts are associated with CD8+ T cell levels in programmed death-ligand 1-negative non-small cell lung cancer patients after radiotherapy: A retrospective study. Medicine (Baltimore) 2021; 100:e26674. [PMID: 34398034 PMCID: PMC8294890 DOI: 10.1097/md.0000000000026674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/03/2021] [Indexed: 01/04/2023] Open
Abstract
This study aimed to explore the dynamics of circulating tumor cells (CTCs) and CD8+ T cells in stage II-III non-small cell lung cancer patients with CTCs in different programmed death-ligand 1 (PD-L1) status treated with radiotherapy and evaluate the correlation between CTCs and CD8+ T cells.This study was a retrospective study which reviewed 69 stage II-III non-small cell lung cancer patients underwent postoperative radiotherapy and peripheral blood tests of CTCs and T lymphocyte were available before radiation, 1 week after radiation and 1 month after radiation.In this study, 25 patients had PD-L1 positive CTCs and 44 patients had PD-L1 negative CTCs. The CTCs count was significantly decreased compared with baseline in patients with different PD-L1 status CTCs at 1 week and 1 month after radiotherapy. The proportion of CD8+ T cells was significantly increased at 1 month after radiotherapy compared with baseline in the total population (mean change, 7.24 ± 2.12; P < .05) and patients with PD-L1 negative CTCs (mean change, 7.17 ± 2.65; P < .05). One month after radiotherapy, the proportion of CD8+ T cells was negatively correlated with the CTCs count in the total population (r = -0.255, P = .034) and PD-L1 negative patients (r = -0.330, P = .029). In patients with PD-L1 negative CTCs, the CTCs count 1 week after radiotherapy (hazard ratio, 0.150 [95% confidence intervals., 0.027-0.840], P = .031) and the proportion of CD8+ T cells 1 month after radiotherapy (hazard ratio, 7.961 [95% confidence intervals, 1.028-61.68], P = .047) were independent prognostic factors for disease recurrence.After radiotherapy, only PD-L1-negative patients had a significant increase in the CD8+ T cell levels, while it was negatively correlated with CTCs count and was an independent prognostic factors of disease recurrence.
Collapse
Affiliation(s)
- Qingyun Liu
- The Third Affiliated Hospital of Shandong First Medical University, Jinan, Shandong Province, China
| | - Chaoren Zhao
- Shandong Provincial Chest Hospital, Jinan, Shandong Province, China
| | - Penghui Jiang
- Shandong Provincial Chest Hospital, Jinan, Shandong Province, China
| | - Dawei Liu
- Shandong Provincial Chest Hospital, Jinan, Shandong Province, China
| |
Collapse
|
18
|
Senescent T cells: a potential biomarker and target for cancer therapy. EBioMedicine 2021; 68:103409. [PMID: 34049248 PMCID: PMC8170103 DOI: 10.1016/j.ebiom.2021.103409] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/09/2021] [Accepted: 05/06/2021] [Indexed: 12/27/2022] Open
Abstract
The failure of T cells to eradicate tumour cells in the tumour microenvironment is mainly due to the dysfunction of T cells. Senescent T cells, with defects in proliferation and effector functions, accumulate in ageing, chronic viral infections, and autoimmune disorders where antigen stimulation persists. Increasing evidence suggests that inducing T cell senescence is a key strategy used by malignant tumours to evade immune surveillance. In this review, we summarize the general features, functional regulation, and signalling network of senescent T cells in tumour development and highlight their potential as prognostic biomarkers in multiple cancer treatments, including chemotherapy, radiotherapy, and immunotherapy. Moreover, we discuss possible therapeutic strategies for preventing or rejuvenating senescence in tumour-specific T cells. Understanding these critical issues may provide novel strategies to enhance cancer immunotherapy.
Collapse
|
19
|
Yang H, Wang Y, Jia Z, Wang Y, Yang X, Wu P, Song Y, Xu H, Gu D, Chen R, Xia X, Bing Z, Gao C, Cao L, Li S, Cao Z, Liang N. Characteristics of T-Cell Receptor Repertoire and Correlation With EGFR Mutations in All Stages of Lung Cancer. Front Oncol 2021; 11:537735. [PMID: 33777727 PMCID: PMC7991722 DOI: 10.3389/fonc.2021.537735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 01/26/2021] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide, and its occurrence is related to the accumulation of gene mutations and immune escape of the tumor. Sequencing of the T-cell receptor (TCR) repertoire can reveal the immunosurveillance status of the tumor microenvironment, which is related to tumor escape and immunotherapy. This study aimed to determine the characteristics and clinical significance of the TCR repertoire in lung cancer. To comprehensively profile the TCR repertoire, results from high-throughput sequencing of samples from 93 Chinese patients with lung cancer were analyzed. We found that the TCR clonality of tissues was related to smoking, with higher clonality in patients who had quit smoking for less than 1 year. As expected, TCR clonality was correlated with stages: patients with stage IV disease showed higher clonality than others. The correlation between TCR repertoire and epidermal growth factor receptor (EGFR) status was also investigated. Patients with EGFR non-L858R mutations showed higher clonality and a lower Shannon index than other groups, including patients with EGFR L858R mutation and wild-type EGFR. Furthermore, we analyzed the TCR similarity metrics—that is, the TCR shared between postoperative peripheral blood and tissue of patients with non-distant metastasis of lung cancer. A similar trend was found, in which patients with EGFR L858R mutations had lower overlap index (OLI) and Morisita index (MOI) scores. Moreover, the OLI showed a positive correlation with several clinical characteristics, including the tumor mutational burden of tissues and the maximum somatic allele frequency of blood; OLI showed a negative correlation with the ratio of CD4+CD28+ in CD4+ cells and the ratio of CD8+CD28+ in CD8+ cells. In conclusion, TCR clonality and TCR similarity metrics correlated with clinical characteristics of patients with lung cancer. Differences in TCR clonality, Shannon index, and OLI across EGFR subtypes provide information to improve understanding about varied responses to immunotherapy in patients with different EGFR mutations.
Collapse
Affiliation(s)
- Huaxia Yang
- Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yadong Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziqi Jia
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yanyu Wang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoying Yang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Pancheng Wu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.,Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Song
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Huihui Xu
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Dejian Gu
- Medical Center, Geneplus-Beijing Institute, Beijing, China
| | - Rongrong Chen
- Medical Center, Geneplus-Beijing Institute, Beijing, China
| | - Xuefeng Xia
- Medical Center, Geneplus-Beijing Institute, Beijing, China
| | - Zhongxing Bing
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Chao Gao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shanqing Li
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhili Cao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Naixin Liang
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
20
|
Keam S, Gill S, Ebert MA, Nowak AK, Cook AM. Enhancing the efficacy of immunotherapy using radiotherapy. Clin Transl Immunology 2020; 9:e1169. [PMID: 32994997 PMCID: PMC7507442 DOI: 10.1002/cti2.1169] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/04/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
Recent clinical breakthroughs in cancer immunotherapy, especially with immune checkpoint blockade, offer great hope for cancer sufferers - and have greatly changed the landscape of cancer treatment. However, whilst many patients achieve clinical responses, others experience minimal benefit or do not respond to immune checkpoint blockade at all. Researchers are therefore exploring multimodal approaches by combining immune checkpoint blockade with conventional cancer therapies to enhance the efficacy of treatment. A growing body of evidence from both preclinical studies and clinical observations indicates that radiotherapy could be a powerful driver to augment the efficacy of immune checkpoint blockade, because of its ability to activate the antitumor immune response and potentially overcome resistance. In this review, we describe how radiotherapy induces DNA damage and apoptosis, generates immunogenic cell death and alters the characteristics of key immune cells in the tumor microenvironment. We also discuss recent preclinical work and clinical trials combining radiotherapy and immune checkpoint blockade in thoracic and other cancers. Finally, we discuss the scheduling of immune checkpoint blockade and radiotherapy, biomarkers predicting responses to combination therapy, and how these novel data may be translated into the clinic.
Collapse
Affiliation(s)
- Synat Keam
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
| | - Suki Gill
- Department of Radiation OncologySir Charles Gairdner HospitalPerthWAAustralia
| | - Martin A Ebert
- Department of Radiation OncologySir Charles Gairdner HospitalPerthWAAustralia
- School of Physics, Mathematics and ComputingThe University of Western AustraliaPerthWAAustralia
| | - Anna K Nowak
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
- Department of Medical OncologySir Charles Gairdner HospitalNedlands, PerthWAAustralia
| | - Alistair M Cook
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
| |
Collapse
|
21
|
Maibach F, Sadozai H, Seyed Jafari SM, Hunger RE, Schenk M. Tumor-Infiltrating Lymphocytes and Their Prognostic Value in Cutaneous Melanoma. Front Immunol 2020; 11:2105. [PMID: 33013886 PMCID: PMC7511547 DOI: 10.3389/fimmu.2020.02105] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/04/2020] [Indexed: 12/17/2022] Open
Abstract
Recent breakthroughs in tumor immunotherapy such as immune checkpoint blockade (ICB) antibodies, have demonstrated the capacity of the immune system to fight cancer in a number of malignancies such as melanoma and lung cancer. The numbers, localization and phenotypes of tumor-infiltrating lymphocytes (TIL) are not only predictive of response to immunotherapy but also key modulators of disease progression. In this review, we focus on TIL profiling in cutaneous melanoma using histopathological approaches and highlight the observed prognostic value of the primary TIL subsets. The quantification of TIL in formalin-fixed tumor samples ranges from visual scoring of lymphocytic infiltrates in H&E to multiplex immunohistochemistry and immunofluorescence followed by enumeration using image analysis software. Nevertheless, TIL enumeration in the current literature primarily relies upon single marker immunohistochemistry analyses of major lymphocyte subsets such as conventional T cells (CD3, CD4, CD8), regulatory T cells (FOXP3) and B cells (CD20). We review key studies in the literature on associations between TIL subsets and patient survival. We also cover recent findings with respect to the existence of ectopic lymphoid aggregates found in the TME which are termed tertiary lymphoid structures (TLS) and are generally a positive prognostic feature. In addition to their prognostic significance, the existence of various TIL sub-populations has also been reported to predict a patient's response to ICB. Thus, the literature on the predictive potential of TIL subsets in melanoma patients receiving ICB has also been discussed. Finally, we describe recently developed state-of-the-art profiling approaches for tumor infiltrating immune cells such as digital pathology scoring algorithms (e.g., Immunoscore) and multiplex proteomics-based immunophenotyping platforms (e.g., imaging mass cytometry). Translating these novel technologies have the potential to revolutionize tumor immunopathology leading to altering our current understanding of cancer immunology and dramatically improving outcomes for patients.
Collapse
Affiliation(s)
- Fabienne Maibach
- Institute of Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| | - Hassan Sadozai
- Institute of Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| | | | - Robert E. Hunger
- Department of Dermatology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Mirjam Schenk
- Institute of Pathology, Experimental Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
22
|
Merhi M, Raza A, Inchakalody VP, Siveen KS, Kumar D, Sahir F, Mestiri S, Hydrose S, Allahverdi N, Jalis M, Relecom A, Al Zaidan L, Hamid MSE, Mostafa M, Gul ARZ, Uddin S, Al Homsi M, Dermime S. Persistent anti-NY-ESO-1-specific T cells and expression of differential biomarkers in a patient with metastatic gastric cancer benefiting from combined radioimmunotherapy treatment: a case report. J Immunother Cancer 2020; 8:e001278. [PMID: 32913031 PMCID: PMC7484873 DOI: 10.1136/jitc-2020-001278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
Combined radioimmunotherapy is currently being investigated to treat patients with cancer. Anti-programmed cell death-1 (PD-1) immunotherapy offers the prospect of long-term disease control in solid tumors. Radiotherapy has the ability to promote immunogenic cell death leading to the release of tumor antigens, increasing infiltration and activation of T cells. New York esophageal squamous cell carcinoma-1 (NY-ESO-1) is a cancer-testis antigen expressed in 20% of advanced gastric cancers and known to induce humoral and cellular immune responses in patients with cancer. We report on the dynamic immune response to the NY-ESO-1 antigen and important immune-related biomarkers in a patient with metastatic gastric cancer treated with radiotherapy combined with anti-PD-1 pembrolizumab antibody.Our patient was an 81-year-old man diagnosed with locally advanced unresectable mismatch repair-deficient gastric cancer having progressed to a metastatic state under a second line of systemic treatment consisting of an anti-PD-1 pembrolizumab antibody. The patient was subsequently treated with local radiotherapy administered concomitantly with anti-PD-1, with a complete response on follow-up radiologic assessment. Disease control was sustained with no further therapy for a period of 12 months before relapse. We have identified an NY-ESO-1-specific interferon-γ (IFN-γ) secretion from the patients' T cells that was significantly increased at response (****p˂0.0001). A novel promiscuous immunogenic NY-ESO-1 peptide P39 (P153-167) restricted to the four patient's HLA-DQ and HLA-DP alleles was identified. Interestingly, this peptide contained the known NY-ESO-1-derived HLA-A2-02:01(P157-165) immunogenic epitope. We have also identified a CD107+ cytotoxic T cell subset within a specific CD8+/HLA-A2-NY-ESO-1 T cell population that was low at disease progression, markedly increased at disease resolution and significantly decreased again at disease re-progression. Finally, we identified two groups of cytokines/chemokines. Group 1 contains five cytokines (IFN-γ, tumor necrosis factor-α, interleukin-2 (IL-2), IL-5 and IL-6) that were present at disease progression, significantly downregulated at disease resolution and dramatically upregulated again at disease re-progression. Group 2 contains four biomarkers (perforin, soluble FAS, macrophage inflammatory protein-3α and C-X-C motif chemokine 11/Interferon-inducible T Cell Alpha Chemoattractant that were present at disease progression, significantly upregulated at disease resolution and dramatically downregulated again at disease re-progression. Combined radioimmunotherapy can enhance specific T cell responses to the NY-ESO-1 antigen that correlates with beneficial clinical outcome of the patient.
Collapse
Affiliation(s)
- Maysaloun Merhi
- Medical Oncology, Hamad Medical Corporation, Doha, Ad Dawhah, Qatar
| | - Afsheen Raza
- Medical Oncology, Hamad Medical Corporation, Doha, Ad Dawhah, Qatar
| | | | | | - Deepak Kumar
- Computational Biology, Carnegie Mellon University - Qatar Campus, Doha, Ad Dawhah, Qatar
| | | | | | | | | | - Munir Jalis
- Hamad Medical Corporation, Doha, Ad Dawhah, Qatar
| | | | | | | | - Mai Mostafa
- Hamad Medical Corporation, Doha, Ad Dawhah, Qatar
| | | | - Shahab Uddin
- Hamad Medical Corporation, Doha, Ad Dawhah, Qatar
| | | | - Said Dermime
- Medical Oncology, National Center for Cancer Care and Research, Doha, Qatar
| |
Collapse
|
23
|
Sasaki A, Nakamura Y, Togashi Y, Kuno H, Hojo H, Kageyama S, Nakamura N, Takashima K, Kadota T, Yoda Y, Mishima S, Sawada K, Kotani D, Kawazoe A, Kuboki Y, Taniguchi H, Kojima T, Doi T, Yoshino T, Yano T, Kobayashi T, Akimoto T, Nishikawa H, Shitara K. Enhanced tumor response to radiotherapy after PD-1 blockade in metastatic gastric cancer. Gastric Cancer 2020; 23:893-903. [PMID: 32180056 DOI: 10.1007/s10120-020-01058-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/06/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors may enhance the efficacy of radiotherapy (RT) in cancer treatment but the effect remains unknown in metastatic gastric cancer (mGC). This study aimed to compare the tumor shrinkage by palliative RT for mGC patients with or without previous exposure to anti-PD-1 therapy. METHODS Data of 36 mGC patients who had received palliative RT from April 2013 to May 2019 were analyzed. Primary tumor responses were evaluated through a volumetric measurement-based method using computed tomography (CT) and endoscopic responses were evaluated in patients who underwent endoscopy before and after RT. Tumor microenvironment (TME) immune status was investigated by analyzing tumor-infiltrating lymphocytes by flow cytometry. RESULTS Among 36 patients, 18 had previous exposure to anti-PD-1 before RT showing no significant differences in baseline characteristics with the other 18 patients without exposure to anti-PD-1 treatment. Tumor responses were observed in 28% (5/18) and none (0/18) in the anti-PD-1-exposed vs. naïve group, respectively (P = 0.045). Five out of eight patients in the anti-PD-1-exposed group, who underwent endoscopy after RT showed partial response, but none in the anti-PD-1-naïve patients showed response (P = 0.026). Increase in the CD8+ T cell/effector regulatory T cell ratio in TILs after anti-PD-1 therapy was noted in three responders to RT, but not in the other three non-responders. CONCLUSIONS Prior exposure to anti-PD-1 therapy increases tumor response to RT. Immune profiling suggests that anti-PD-1 therapy may enhance the efficacy of RT by immunoactivation in the TME.
Collapse
Affiliation(s)
- Akinori Sasaki
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.,Courses of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yosuke Togashi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Centre, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hirofumi Kuno
- Department of Diagnostic Radiology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hidehiro Hojo
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shunichiro Kageyama
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoki Nakamura
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kenji Takashima
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tomohiro Kadota
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yusuke Yoda
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Saori Mishima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Kentaro Sawada
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Daisuke Kotani
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Akihito Kawazoe
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Yasutoshi Kuboki
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Hiroya Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Toshihiko Doi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| | - Tomonori Yano
- Department of Gastroenterology and Endoscopy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tatsushi Kobayashi
- Department of Diagnostic Radiology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuo Akimoto
- Courses of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research and Clinical Trial Centre, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kohei Shitara
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
24
|
Correlative studies investigating effects of PI3K inhibition on peripheral leukocytes in metastatic breast cancer: potential implications for immunotherapy. Breast Cancer Res Treat 2020; 184:357-364. [PMID: 32767201 DOI: 10.1007/s10549-020-05846-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 07/30/2020] [Indexed: 01/03/2023]
Abstract
PURPOSE Patients with localized breast cancer have a 5-year survival rate > 99% compared to patients with metastatic breast cancer (MBC) that have a 5-year survival rate of ~ 27%. Unregulated PI3K/AKT signaling is a common characteristic of MBC, making it a desirable therapeutic target for tumors with activating mutations in this pathway. Interestingly, inhibition of the PI3K/AKT pathway can affect signaling in immune cells, which could potentially alter the immune phenotype of patients undergoing therapy with these drugs. The purpose of this study is to evaluate how PI3K inhibition affects the immune cells of MBC patients during treatment. METHODS We investigated the effects of PI3K inhibition on the immune cell populations in peripheral blood of MBC patients enrolled in 4 different clinical trials utilizing PI3K inhibitors. Peripheral blood was drawn at different points in patient treatment cycles to record immune cell fluctuations in response to therapy. RESULTS MBC patients who responded to treatment with a positive fold-change in cytotoxic T cell population, had an average duration of treatment response of 31.4 months. In contrast, MBC patients who responded to treatment with a negative fold-change in cytotoxic T-cell population, had an average duration of therapeutic response of 5 months. These data suggest that patients with a more robust, initial anti-tumor T cell response may have a longer therapeutic response compared to patients who do not have a robust, initial anti-tumor T cell response. CONCLUSIONS These results highlight the potential for PI3K inhibition to sensitize tumors to immune checkpoint inhibitors, thus providing additional therapeutic options for patients with MBC.
Collapse
|
25
|
Shen Y, Wang H, Chen X, Li W, Chen J. Prognostic significance of lymphocyte-to-monocyte ratio and platelet-to-lymphocyte ratio in patients with hepatocellular carcinoma undergoing transcatheter arterial chemoembolization and radiofrequency ablation. Onco Targets Ther 2019; 12:7129-7137. [PMID: 31564897 PMCID: PMC6731523 DOI: 10.2147/ott.s217935] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/23/2019] [Indexed: 12/23/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) ranks fifth among malignancies globally. Previous studies have shown that systemic inflammatory response, platelet-to-lymphocyte ratio (PLR), and lymphocyte-to-monocyte ratio (LMR) are associated with poor prognosis of various types of cancer. Materials and methods Radiofrequency ablation (RFA) was performed using an internal cooling electrode with a 2- or 3-cm exposed tip. The LMR was calculated as the ratio of lymphocytes to monocytes. In order to explore the influence of pretreatment with PLR and LMR on survival of HCC patients undergoing transcatheter arterial chemoembolization (TACE) and RFA, 204 cases with HCC which accepted RFA and TACE were retrospectively analyzed and assigned into 2 groups based on optimal cutoff values for LMR (low: ≤2.13 or high: >2.13) and PLR (low: ≤95.65 or high: >95.65). Results Patients with a lower PLR had a longer overall survival (OS) compared to those with a higher PLR (median OS, 20 versus 13 months), and patients with a higher LMR had a longer OS than those with a lower LMR (OS, 22 versus 10 months). Multivariate logistic regression analysis was performed using Cox proportional hazards regression analysis for multiple prognostic factors and identified PLR and LMR as prognostic factors for OS of HCC cases. Conclusion We conclude that PLR and LMR, whose detection is generally available and affordable, may be novel noninvasive circulating markers to potentially assist doctors assess the prognosis of patients.
Collapse
Affiliation(s)
- Yanjun Shen
- Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, People's Republic of China
| | - Huige Wang
- Department of Gynecology,Wangjing Hospital of Chinese Academy of Chinese Medical, Beijing 100102, People's Republic of China
| | - Xiangmei Chen
- Department of Pathology, Beijing Ditan Hospital, Capital Medical Uiversity, Beijing 100015, People's Republic of China
| | - Wendong Li
- Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, People's Republic of China
| | - Jinglong Chen
- Department of Oncology, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, People's Republic of China
| |
Collapse
|