1
|
Zhang Y, Li Z, Huang Y, Xu Y, Zou B. Advancements in immunotherapy for advanced esophageal squamous cell carcinoma: a comprehensive review of current strategies and future directions. Expert Rev Clin Immunol 2024; 20:971-984. [PMID: 38884604 DOI: 10.1080/1744666x.2024.2368194] [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: 12/19/2023] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Esophageal cancer (EC), particularly esophageal squamous cell carcinoma (ESCC), is characterized by high incidence and poor prognosis worldwide, necessitating novel therapeutic approaches like immunotherapy. This review explores the impact of immune checkpoint inhibitors (ICIs) on ESCC, especially focusing on PD-1/PD-L1 and CTLA-4 inhibitors. Our literature search, conducted across databases including PubMed, Web of Science, and EMBASE, from January 2010 to December 2023, aimed at identifying advancements, challenges, and future directions in the use of immunotherapy for ESCC. AREAS COVERED We provide a detailed analysis of clinical trials evaluating the efficacy of ICIs as monotherapy and in combination with chemotherapy, radiotherapy, and targeted therapy for locally advanced ESCC. Our findings highlight the significant survival benefits offered by ICIs, albeit with varying efficacy across patient populations, emphasizing the need for precise biomarkers to tailor treatment strategies. EXPERT OPINION The integration of immunotherapy into the ESCC treatment paradigm represents a significant shift, improving survival outcomes. Future research should focus on optimizing combination therapies and novel immunotherapeutic agents, incorporating genetic and tumor microenvironment analyses to enhance patient selection and treatment efficacy.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Zheng Li
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Ying Huang
- College of Management, Sichuan Agricultural University, Chengdu, P.R. China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
2
|
Chen W, Cao K, Zhang L, Zhao X, Chen B, Li W, Shang R, Sun L, Jiang Z, Wang J, Xue W. Efficacy and safety evaluation of frontline immunotherapy combinations in advanced esophageal squamous cell carcinoma: a network meta-analysis highlighting the value of PD-L1 expression positivity scores. Front Immunol 2024; 15:1414753. [PMID: 39050848 PMCID: PMC11266001 DOI: 10.3389/fimmu.2024.1414753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction The systematic review and network meta-analysis (NMA) consolidate all relevant randomized controlled trials (RCTs) related to initial immunotherapy treatments for advanced esophageal squamous cell carcinoma (ESCC). Our goal is to thoroughly assess the effectiveness and safety of various immunotherapy methods, focusing on overall survival (OS) and progression-free survival (PFS) among patients with advanced ESCC positive for PD-L1. Methods We conducted a systematic search of the PubMed, Embase, Cochrane Library, and Web of Science databases, covering all records from their inception until January 22, 2024. The inclusion criteria targeted patients with advanced ESCC undergoing first-line immunotherapy or chemotherapy, limiting the study selection to randomized controlled trials (RCTs) exclusively. The study upholds the values of openness, originality, and dependability, as evidenced by its enrollment in the Prospective Register of Systematic Reviews (CRD42024504992). Results Our analysis encompasses 7 RCTs, totaling 4688 patients, and evaluates 8 distinct immunotherapy combinations. In advanced ESCC patients irrespective of PD-L1 expression, both sintilimab-chemotherapy and toripalimab-chemotherapy regimens demonstrated comparable OS benefits (HR=0.92, 95% CI: 0.64-1.33). The most pronounced PFS advantages were seen with sintilimab-chemotherapy and camrelizumab-chemotherapy as compared to standard chemotherapy (HR=0.56, 95% CI: 0.46-0.58). Notably, camrelizumab-chemotherapy (HR=0.83, 95% CI: 0.59-1.16) and nivolumab-ipilimumab (HR=0.84, 95% CI: 0.60-1.17) demonstrated significant safety profiles over chemotherapy alone. Subgroup analysis based on PD-L1 expression revealed nivolumab-chemotherapy to yield the highest OS benefit (HR=0.54, 95% CI: 0.37-0.79) in ESCC patients with PD-L1 expression ≥1%. Furthermore, camrelizumab-chemotherapy (HR=0.51, 95% CI: 0.39-0.67) exhibited superior PFS benefits. Among patients with PD-L1 expression ≥10%, camrelizumab-chemotherapy (HR=0.52, 95% CI: 0.35-0.78) emerged as the most efficacious in improving OS, while serplulimab-chemotherapy (HR=0.48, 95% CI: 0.34-0.68) was associated with the longest PFS benefit. Conclusion The integration of immune checkpoint inhibitors (ICIs) with chemotherapy appears to significantly enhance survival outcomes in patients with advanced ESCC compared to chemotherapy alone. Sintilimab-chemotherapy is potentially the optimal regimen for patients without PD-L1 expression. In contrast, nivolumab-chemotherapy and camrelizumab-chemotherapy are likely to offer the best OS and PFS benefits, respectively, in patients with PD-L1 expression ≥1%. Among those with PD-L1 expression ≥10%, camrelizumab-chemotherapy is projected to provide the greatest OS advantage, whereas serplulimab-chemotherapy is anticipated to offer the most prolonged PFS benefit. Since most of the patients in this study originated from Asia, the above findings are more applicable to the Asian population. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024504992.
Collapse
Affiliation(s)
- Wei Chen
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Keming Cao
- School of Pharmacy, North China University of Science and Technology, Heibei, China
| | - Lili Zhang
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Xingyu Zhao
- School of Pharmaceutical Sciences, Capital Medical University School, Beijing, China
| | - Bixiao Chen
- Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Wei Li
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Rongguo Shang
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| | - Lichaoyue Sun
- Pharmacy Department, Aerospace Center Hospital, Beijing, China
| | - Ze Jiang
- Pharmaceutical Department, TongRen Hospital, Capital Medical University, Beijing, China
| | - Jingxin Wang
- School of Pharmacy, North China University of Science and Technology, Heibei, China
| | - Wenxin Xue
- Department of Pharmacy, Emergency General Hospital, Beijing, China
| |
Collapse
|
3
|
Sun M, Yang P, Wang W, Yu Y, Yang D, Ping Y, Zhu B. Advancements in the research of immune checkpoint inhibitors for the treatment of advanced esophageal squamous cell carcinoma. Am J Cancer Res 2024; 14:1981-1998. [PMID: 38859835 PMCID: PMC11162652 DOI: 10.62347/xuwc6412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/27/2024] [Indexed: 06/12/2024] Open
Abstract
Esophageal cancer (EC) has a high mortality rate and poor prognosis. Most patients are diagnosed at an advanced stage or with distant metastasis, making surgery impossible. Traditional curative radiotherapy and chemotherapy have limited efficacy. In recent years, with the development of clinical trials, immune checkpoint inhibitors (ICIs) have shown promising results in treating advanced and metastatic esophageal squamous cell carcinoma (ESCC) patients. ICIs have gradually become a primary therapeutic approach for EC. This review summarizes and provides an overview of the current research status and progress of ICIs in the treatment of advanced ESCC patients.
Collapse
Affiliation(s)
- Mengfei Sun
- College of Pharmacy, Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| | - Pengjie Yang
- Department of Thoracic Surgery, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| | - Weisong Wang
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| | - Yongjun Yu
- Department of Thoracic Surgery, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| | - Dongdong Yang
- Department of Pharmacy, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| | - Yaodong Ping
- Department of Pharmacy, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
- Department of Pharmacy, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and InstituteBeijing, China
| | - Benben Zhu
- Department of Pharmacy, Peking University Cancer Hospital (Inner Mongolia Campus)/Affiliated Cancer Hospital of Inner Mongolia Medical UniversityHohhot, Inner Mongolia Autonomous Region, China
| |
Collapse
|
4
|
Yang Y, Yi Y, Wang Z, Li S, Zhang B, Sang Z, Zhang L, Cao Q, Li B. A combined nomogram based on radiomics and hematology to predict the pathological complete response of neoadjuvant immunochemotherapy in esophageal squamous cell carcinoma. BMC Cancer 2024; 24:460. [PMID: 38609892 PMCID: PMC11015586 DOI: 10.1186/s12885-024-12239-0] [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: 10/17/2023] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND To predict pathological complete response (pCR) in patients receiving neoadjuvant immunochemotherapy (nICT) for esophageal squamous cell carcinoma (ESCC), we explored the factors that influence pCR after nICT and established a combined nomogram model. METHODS We retrospectively included 164 ESCC patients treated with nICT. The radiomics signature and hematology model were constructed utilizing least absolute shrinkage and selection operator (LASSO) regression, and the radiomics score (radScore) and hematology score (hemScore) were determined for each patient. Using the radScore, hemScore, and independent influencing factors obtained through univariate and multivariate analyses, a combined nomogram was established. The consistency and prediction ability of the nomogram were assessed utilizing calibration curve and the area under the receiver operating factor curve (AUC), and the clinical benefits were assessed utilizing decision curve analysis (DCA). RESULTS We constructed three predictive models.The AUC values of the radiomics signature and hematology model reached 0.874 (95% CI: 0.819-0.928) and 0.772 (95% CI: 0.699-0.845), respectively. Tumor length, cN stage, the radScore, and the hemScore were found to be independent factors influencing pCR according to univariate and multivariate analyses (P < 0.05). A combined nomogram was constructed from these factors, and AUC reached 0.934 (95% CI: 0.896-0.972). DCA demonstrated that the clinical benefits brought by the nomogram for patients across an extensive range were greater than those of other individual models. CONCLUSIONS By combining CT radiomics, hematological factors, and clinicopathological characteristics before treatment, we developed a nomogram model that effectively predicted whether ESCC patients would achieve pCR after nICT, thus identifying patients who are sensitive to nICT and assisting in clinical treatment decision-making.
Collapse
Affiliation(s)
- Yu Yang
- Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Yan Yi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhongtang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shanshan Li
- Department of Oncology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Bin Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zheng Sang
- Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lili Zhang
- Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Qiang Cao
- Shandong Medical Imaging and Radiotherapy Engineering Center (SMIREC), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
5
|
Gao D, Ou J, Tan BG, Yu ZY, Li KY, Li R, Zhang XM, Chen TW, Zhou HY. A novel quantitative model based on gross tumor volume corresponding to anatomical distribution measured with multidetector computed tomography to determine the resectability of non‑distant metastatic esophageal squamous cell carcinoma. Oncol Lett 2023; 26:485. [PMID: 37818136 PMCID: PMC10561156 DOI: 10.3892/ol.2023.14072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
It is important to accurately determine the resectability of thoracic esophageal squamous cell carcinoma (ESCC) for treatment decision-making. Previous studies have revealed that the CT-derived gross tumor volume (GTV) is associated with the staging of ESCC. The present study aimed to explore whether the anatomical distribution-based GTV of non-distant metastatic thoracic ESCC measured using multidetector computed tomography (MDCT) could quantitatively determine the resectability. For this purpose, 473 consecutive patients with biopsy-confirmed non-distant metastatic thoracic ESCC who underwent contrast-enhanced CT were randomly divided into a training cohort (TC; 376 patients) and validation cohort (VC; 97 patients). GTV was retrospectively measured using MDCT. Univariate and multivariate analyses were performed to identify the determinants of the resectability of ESCC in the TC. Receiver operating characteristic (ROC) analysis was performed to clarify whether anatomical distribution-based GTV could help quantitatively determinate resectability. Unweighted Cohen's Kappa tests in VC were used to assess the performance of the previous models. Univariate analysis demonstrated that sex, anatomic distribution, cT stage, cN stage and GTV were related to the resectability of ESCC in the TC (all P<0.05). Multivariate analysis revealed that GTV [P<0.001; odds ratio (OR) 1.158] and anatomic distribution (P=0.027; OR, 1.924) were independent determinants of resectability. ROC analysis revealed that the GTV cut-offs for the determination of the resectability of the upper, middle and lower thoracic portions were 23.57, 22.89 and 22.58 cm3, respectively, with areas under the ROC curves of >0.9. Unweighted Cohen's Kappa tests revealed an excellent performance of the ROC models in the upper, middle and lower thoracic portions with Cohen k-values of 0.913, 0.879 and 0.871, respectively. On the whole, the present study demonstrated that GTV and the anatomic distribution of non-distant metastatic thoracic ESCC may be independent determinants of resectability, and anatomical distribution-based GTV can effectively be used to quantitatively determine resectability.
Collapse
Affiliation(s)
- Dan Gao
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- Department of Radiology, Qionglai Medical Center Hospital, Chengdu, Sichuan 611530, P.R. China
| | - Jing Ou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Bang-Guo Tan
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, Sichuan 617067, P.R. China
| | - Zi-Yi Yu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Ke-Ying Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Rui Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Tian-Wu Chen
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Hai-Ying Zhou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
6
|
Cheng M, Xin Q, Ma S, Ge M, Wang F, Yan X, Jiang B. Advances in the Theranostics of Oesophageal Squamous Carcinoma. ADVANCED THERAPEUTICS 2023; 6. [DOI: 10.1002/adtp.202200251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Indexed: 01/04/2025]
Abstract
AbstractOesophageal squamous carcinoma (ESCC) is one of the most lethal human malignancies, and it is a more aggressive form of oesophageal cancer (EC) that comprises over 90% of all EC cases in China compared with oesophageal adenocarcinoma (EAC). The high mortality of ESCC is attributed to the late‐stage diagnosis, chemoradiotherapy resistance, and lack of appropriate therapeutic targets and corresponding therapeutic formulations. Recently, emerging clinical and translational investigations have involved genome analyses, diagnostic biomarkers, and targeted therapy for ESCC, and these studies provide a new horizon for improving the clinical outcomes of patients with ESCC. Here, the latest research advances in the theranostics of ESCC are reviewed and the unique features of ESCC (including differences from EAC, genomic alterations, and microbe infections), tissue and circulating biomarkers, chemoradiotherapy resistance, clinical targeted therapy for ESCC, identification of novel therapeutic targets, and designation of nanotherapeutic systems for ESCC are particularly focused on. Finally, the perspectives for future clinical and translational theranostic research of ESCC are discussed and the obstacles that must be overcome in ESCC theranostics are described.
Collapse
Affiliation(s)
- Miaomiao Cheng
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Qi Xin
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Saiyu Ma
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Mengyue Ge
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
| | - Feng Wang
- Oncology Department The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450000 China
| | - Xiyun Yan
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
- CAS Engineering Laboratory for Nanozyme Key Laboratory of Protein and Peptide Pharmaceuticals Institute of Biophysics Chinese Academy of Sciences Beijing 100101 China
| | - Bing Jiang
- Nanozyme Medical Center School of Basic Medical Sciences Zhengzhou University Zhengzhou 450001 China
- State Key Laboratory of Esophageal Cancer Prevention &Treatment Zhengzhou Henan 450001 China
| |
Collapse
|
7
|
Zhen H, Tian J, Li G, Zhao P, Zhang Y, Che J, Cao B. Raltitrexed enhanced antitumor effect of anlotinib in human esophageal squamous carcinoma cells on proliferation, invasiveness, and apoptosis. BMC Cancer 2023; 23:207. [PMID: 36870981 PMCID: PMC9985835 DOI: 10.1186/s12885-023-10691-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
BACKGROUND Anlotinib is a multi-targeted receptor tyrosine kinase inhibitor (TKI) which has exhibited encouraging clinical activity in advanced non-small cell lung cancer (NSCLC) and soft tissue sarcoma. Raltitrexed is well known to be effective in the treatment of colorectal cancer in China. The present study aims to investigate the combinatory antitumor effect of anlotinib and raltitrexed on human esophageal squamous carcinoma cells and further explore the molecular mechanisms in vitro. METHODS Human esophageal squamous cell lines KYSE-30 and TE-1 were treated with anlotinib or raltitrexed, or both, then cell proliferation was measured by MTS and colony formation assay; cell migration and invasion were detected by wound-healing and transwell assays; cell apoptosis rate was studied by flow cytometry and the transcription of apoptosis-associated proteins were monitored by quantitative polymerase chain reaction (qPCR) analysis. Finally, western blot was performed to check phosphorylation of apoptotic proteins after treatment. RESULTS Treatment with raltitrexed and anlotinib showed enhanced inhibitory effects on cell proliferation, migration and invasiveness compared with raltitrexed or anlotinib monotherapy. Meanwhile, raltitrexed combined with anlotinib strongly increased cell apoptosis percentage. Moreover, the combined treatment down-regulated mRNA level of the anti-apoptotic protein Bcl-2 and invasiveness-associated protein matrix metalloproteinases-9 (MMP-9), while up-regulated pro-apoptotic Bax and caspase-3 transcription. Western blotting showed that the combination of raltitrexed and anlotinib could inhibit the expression of phosphorylated Akt (p-Akt), Erk (p-Erk) and MMP-9. CONCLUSIONS This study indicated that raltitrexed enhanced the antitumor effects of anlotinib on human ESCC cells by down-regulating phosphorylation of Akt and Erk, providing a novel treatment option for patients with esophageal squamous cell carcinoma (ESCC).
Collapse
Affiliation(s)
- Hongchao Zhen
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Jizheng Tian
- Department of Oncology, Beijing Shunyi District Hospital, Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, China
| | - Guangxin Li
- Radiation Oncology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Pengfei Zhao
- Department of Radiotherapy, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Ying Zhang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Juanjuan Che
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, #95 Yong An Road, Xicheng District, Beijing, 100050, China.
| |
Collapse
|
8
|
Chen W, Li D, Bian X, Wu Y, Xu M, Wu M, Tao M. Peripheral Blood Markers Predictive of Progression-Free Survival in Advanced Esophageal Squamous Cell Carcinoma Patients Treated With PD-1 Inhibitors Plus Chemotherapy as First-Line Therapy. Nutr Cancer 2022; 75:207-218. [PMID: 36190755 DOI: 10.1080/01635581.2022.2123533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Aim: To determine the prognostic value of peripheral blood markers in advanced esophageal squamous cell carcinoma (ESCC) patients receiving programmed cell death protein 1 inhibitors plus chemotherapy as first-line therapy. Methods: A retrospective analysis of 54 patients with advanced ESCC was performed to assess 12 blood markers involving inflammation, nutrition, and tumor burden. Analysis of variance or Kruskal-Wallis tests were used to explore the difference in markers among different response to therapy. Survival curves were constructed using the Kaplan-Meier method. Multivariate Cox models were applied to identify independent predictors of outcome. Results: Patients who achieved response had significantly higher prealbumin, increased BMI, and lower hs-CRP levels at baseline compared with those who experienced disease progression. In the univariate analysis, ALI > 23.55, PNI > 45.175, NLR ≤ 5, and hs-CRP ≤ 6.7 mg/L were significantly associated with a better progression-free survival. Cox regression analysis revealed that ALI >23.55 (P = 0.037) and hs-CRP ≤6.7 mg/L (P = 0.043) were independently associated with superior PFS. Increased tumor abnormal protein (TAP) levels post two cycles was significantly associated with a worse prognosis (P = 0.004). Conclusions: A baseline signature of low ALI and high hs-CRP as well as an early increase in TAP in ESCC appear to be predictive of inferior PFS.
Collapse
Affiliation(s)
- Wei Chen
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dapeng Li
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xuyu Bian
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Wu
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengdan Xu
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mengyao Wu
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Min Tao
- Department of Oncology, the First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Oncology, DuShu Lake Hospital Affiliated to Soochow University, Suzhou, China
| |
Collapse
|
9
|
Fang P, Zhou J, Liang Z, Yang Y, Luan S, Xiao X, Li X, Zhang H, Shang Q, Zeng X, Yuan Y. Immunotherapy resistance in esophageal cancer: Possible mechanisms and clinical implications. Front Immunol 2022; 13:975986. [PMID: 36119033 PMCID: PMC9478443 DOI: 10.3389/fimmu.2022.975986] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/12/2022] [Indexed: 11/23/2022] Open
Abstract
Esophageal cancer (EC) is a common malignant gastrointestinal (GI) cancer in adults. Although surgical technology combined with neoadjuvant chemoradiotherapy has advanced rapidly, patients with EC are often diagnosed at an advanced stage and the five-year survival rate remains unsatisfactory. The poor prognosis and high mortality in patients with EC indicate that effective and validated therapy is of great necessity. Recently, immunotherapy has been successfully used in the clinic as a novel therapy for treating solid tumors, bringing new hope to cancer patients. Several immunotherapies, such as immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy, and tumor vaccines, have achieved significant breakthroughs in EC treatment. However, the overall response rate (ORR) of immunotherapy in patients with EC is lower than 30%, and most patients initially treated with immunotherapy are likely to develop acquired resistance (AR) over time. Immunosuppression greatly weakens the durability and efficiency of immunotherapy. Because of the heterogeneity within the immune microenvironment and the highly disparate oncological characteristics in different EC individuals, the exact mechanism of immunotherapy resistance in EC remains elusive. In this review, we provide an overview of immunotherapy resistance in EC, mainly focusing on current immunotherapies and potential molecular mechanisms underlying immunosuppression and drug resistance in immunotherapy. Additionally, we discuss prospective biomarkers and novel methods for enhancing the effect of immunotherapy to provide a clear insight into EC immunotherapy.
Collapse
Affiliation(s)
- Pinhao Fang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianfeng Zhou
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Zhiwen Liang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Luan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiao
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaokun Li
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Hanlu Zhang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Qixin Shang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery and Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Yong Yuan,
| |
Collapse
|
10
|
Comprehensive Curative Effect of Targeting PD-1 or Traditional Single-Agent Chemotherapy in Second-Line Therapy for Terminal or Metastatic Esophageal Cancer: A Systematic Review and Meta-Analysis. JOURNAL OF ONCOLOGY 2022; 2022:4033863. [PMID: 36059800 PMCID: PMC9436578 DOI: 10.1155/2022/4033863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022]
Abstract
The number of programmed cell death protein 1 (PD-1) inhibitors is gradually increasing; this study aimed to comprehensively and systematically evaluate the impact of PD-1 inhibitors as second-line therapy for terminal or metastatic esophageal cancer (EC) on patient survival and the occurrence of adverse events. Suitable randomized controlled trials (RCTs) were retrieved from PubMed, Web of Science, Embase, and Cochrane Library databases. Moreover, we searched for conference abstracts from the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO) to compare the comprehensive curative effects of PD-1 inhibitors or single-agent therapy in terminal or metastatic EC. The primary outcome indicators were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). Treatment-related adverse events (TRAEs) were the secondary outcome indicators. We compared the previously mentioned indicators of the two treatment modalities using Stata software (version 12.0). We compared the long-term survival rates of both treatment groups and analyzed the possible factors affecting OS. We selected five RCTs with 2197 patients as study subjects. Compared with conventional single-agent chemotherapy, PD-1 inhibitors greatly improved the patients' OS (HR = 0.77, 95% CI 0.70–0.85, P < 0.001), but PFS (HR = 0.93, 95% CI 0.77–1.12, P=0.431) and DCR (RR = 0.93, 95% CI 0.71–1.22, P=0.609) were not greatly improved. Moreover, PD-1 inhibitors improved ORR (RR = 1.83, 95% CI 1.16–2.89, P=0.009) and decreased TRAEs (RR = 0.76, 95% CI 0.61–0.95, P < 0.001) and serious TRAEs (RR = 0.40, 95% CI 0.32–0.49, P < 0.001). Further analysis demonstrated that OS was affected by age, sex, region, smoking history, and the number of organ and lymph node metastases. Compared with the traditional single chemotherapy drugs, PD-1 inhibitors can achieve higher OS and ORR, fewer and more serious TRAEs, and better efficacy and safety for second-line therapy of terminal or metastatic EC.
Collapse
|
11
|
Qiu G, Deng Y. ZFP64 transcriptionally activates PD-1 and CTLA-4 and plays an oncogenic role in esophageal cancer. Biochem Biophys Res Commun 2022; 622:72-78. [DOI: 10.1016/j.bbrc.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/05/2022] [Indexed: 11/02/2022]
|
12
|
Naimi A, Mohammed RN, Raji A, Chupradit S, Yumashev AV, Suksatan W, Shalaby MN, Thangavelu L, Kamrava S, Shomali N, Sohrabi AD, Adili A, Noroozi-Aghideh A, Razeghian E. Tumor immunotherapies by immune checkpoint inhibitors (ICIs); the pros and cons. Cell Commun Signal 2022; 20:44. [PMID: 35392976 PMCID: PMC8991803 DOI: 10.1186/s12964-022-00854-y] [Citation(s) in RCA: 204] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
The main breakthrough in tumor immunotherapy was the discovery of immune checkpoint (IC) proteins, which act as a potent suppressor of the immune system by a myriad of mechanisms. After that, scientists focused on the immune checkpoint molecules mainly. Thereby, much effort was spent to progress novel strategies for suppressing these inhibitory axes, resulting in the evolution of immune checkpoint inhibitors (ICIs). Then, ICIs have become a promising approach and shaped a paradigm shift in tumor immunotherapies. CTLA-4 plays an influential role in attenuation of the induction of naïve and memory T cells by engagement with its responding ligands like B7-1 (CD80) and B7-2 (CD86). Besides, PD-1 is predominantly implicated in adjusting T cell function in peripheral tissues through its interaction with programmed death-ligand 1 (PD-L1) and PD-L2. Given their suppressive effects on anti-tumor immunity, it has firmly been documented that ICIs based therapies can be practical and rational therapeutic approaches to treat cancer patients. Nonetheless, tumor inherent or acquired resistance to ICI and some treatment-related toxicities restrict their application in the clinic. The current review will deliver a comprehensive overview of the ICI application to treat human tumors alone or in combination with other modalities to support more desired outcomes and lower toxicities in cancer patients. Video Abstract.
Collapse
Affiliation(s)
- Adel Naimi
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Rebar N. Mohammed
- Medical Laboratory Analysis Department, Cihan University Sulaimaniya, Sulaymaniyah, 46001 Kurdistan Region Iraq
- College of Veterinary Medicine, University of Sulaimani, Suleimanyah, Iraq
| | - Ahmed Raji
- College of Medicine, University of Babylon, Department of Pathology, Babylon, Iraq
| | - Supat Chupradit
- Department of Occupational Therapy, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, 50200 Thailand
| | | | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, 10210 Thailand
| | - Mohammed Nader Shalaby
- Associate Professor of Biological Sciences and Sports Health Department, Faculty of Physical Education, Suez Canal University, Ismailia, Egypt
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, India
| | - Siavash Kamrava
- Department of Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Shomali
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Armin D. Sohrabi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Adili
- Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Noroozi-Aghideh
- Department of Hematology, Faculty of Paramedicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
13
|
Xu J, Li Y, Fan Q, Shu Y, Yang L, Cui T, Gu K, Tao M, Wang X, Cui C, Xu N, Xiao J, Gao Q, Liu Y, Zhang T, Bai Y, Li W, Zhang Y, Dai G, Ma D, Zhang J, Bai C, Huang Y, Liao W, Wu L, Chen X, Yang Y, Wang J, Ji S, Zhou H, Wang Y, Ma Z, Wang Y, Peng B, Sun J, Mancao C. Clinical and biomarker analyses of sintilimab versus chemotherapy as second-line therapy for advanced or metastatic esophageal squamous cell carcinoma: a randomized, open-label phase 2 study (ORIENT-2). Nat Commun 2022; 13:857. [PMID: 35165274 PMCID: PMC8844279 DOI: 10.1038/s41467-022-28408-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
This randomized, open-label, multi-center phase 2 study (NCT03116152) assessed sintilimab, a PD-1 inhibitor, versus chemotherapy in patients with esophageal squamous cell carcinoma after first-line chemotherapy. The primary endpoint was overall survival (OS), while exploratory endpoint was the association of biomarkers with efficacy. The median OS in the sintilimab group was significantly improved compared with the chemotherapy group (median OS 7.2 vs.6.2 months; P = 0.032; HR = 0.70; 95% CI, 0.50-0.97). Incidence of treatment-related adverse events of grade 3-5 was lower with sintilimab than with chemotherapy (20.2 vs. 39.1%). Patients with high T-cell receptor (TCR) clonality and low molecular tumor burden index (mTBI) showed the longest median OS (15.0 months). Patients with NLR < 3 at 6 weeks post-treatment had a significantly prolonged median OS (16.6 months) compared with NLR ≥ 3. The results demonstrate a significant improvement in OS of sintilimab compared to chemotherapy as second-line treatment for advanced or metastatic ESCC.
Collapse
Affiliation(s)
- Jianming Xu
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Yi Li
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Yang
- Department of Radiotherapy, Nantong Tumor Hospital, Nantong, China
| | - Tongjian Cui
- Department of Medical Oncology, Fujian Provincial Hospital, Fuzhou, China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiuwen Wang
- Department of Chemotherapy, Qilu Hospital of Shandong University, Jinan, China
| | - Chengxu Cui
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Juxiang Xiao
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Quanli Gao
- Department of Immunotherapy, Henan Cancer Hospital, Zhengzhou, China
| | - Yunpeng Liu
- Department of Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Tao Zhang
- Cancer Center, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yuxian Bai
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wei Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Yiping Zhang
- Department of Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Guanghai Dai
- Department of Oncology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Dong Ma
- Department of Gastrointestinal Oncology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Jingdong Zhang
- Department of Gastroenterology, Liaoning Cancer Hospital, Shenyang, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Yunnan Cancer Hospital, Kunming, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lin Wu
- Departmentof Thoracic Medical Oncology, Hunan Cancer Hospital, Changsha, China
| | - Xi Chen
- Department of Oncology, No. 900 Hospital of The Joint Logistic Support Force, Fuzhou, China
| | - Yan Yang
- Department of Gastrointestinal Oncology, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Junye Wang
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Shoujian Ji
- Department of Gastrointestinal Oncology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui Zhou
- Innovent Biologics, Inc., Suzhou, China
| | - Yan Wang
- Innovent Biologics, Inc., Suzhou, China
| | - Zhuo Ma
- Innovent Biologics, Inc., Suzhou, China
| | | | - Bo Peng
- Innovent Biologics, Inc., Suzhou, China
| | - Jiya Sun
- Innovent Biologics, Inc., Suzhou, China
| | | |
Collapse
|
14
|
Janjua KA, Shahzad R, Shehzad A. Development of Novel Cancer Biomarkers for Diagnosis and Prognosis. CANCER BIOMARKERS IN DIAGNOSIS AND THERAPEUTICS 2022:277-343. [DOI: 10.1007/978-981-16-5759-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
15
|
The Functionalities and Clinical Significance of Tumor-Infiltrating Immune Cells in Esophageal Squamous Cell Carcinoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8635381. [PMID: 34616847 PMCID: PMC8490060 DOI: 10.1155/2021/8635381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 11/18/2022]
Abstract
Tumor-infiltrating immune cells have been implicated in the tumorigenesis and progression of esophageal squamous cell carcinoma (ESCC). However, the functionalities and clinical significance of immune cells remain largely unveiled. In this study, the gene expression data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were extracted. The relative infiltrating levels were estimated by single-sample gene set enrichment analysis. Some cytotoxic immune cells were attenuated, and resting cytotoxic immune cells were accumulated in ESCC. Remarkably, we also observed that infiltrating levels of macrophage M2 and resting natural killer (NK) cells were increased in nonresponders of CRT, and T cells that had anticancer activities such as activated memory CD4 and T helper 2 (Th2) cells were significantly reduced in ESCC tissues of the nonresponders. Moreover, the high infiltrations of the resting natural killer (NK) and dendritic cell (DC) were observed to result in a shorter overall survival in ESCC. Consistently, high expression of immune checkpoint genes, CTLA4 and HAVCR2, was associated with poor prognosis. Furthermore, STAT5B, a key transcription factor, as well as its target genes, involved in the regulation of T cells, was significantly downregulated in ESCC, especially subgroup I, indicating that downregulation of STAT5B might be associated with reduced T cell-mediated anticancer activity. In conclusion, the present study significantly improved our understanding of the regulatory roles of immune cells in ESCC.
Collapse
|
16
|
Zhang L, Wang X, Li Y, Han J, Gao X, Li S, Wang F. c-Myb facilitates immune escape of esophageal adenocarcinoma cells through the miR-145-5p/SPOP/PD-L1 axis. Clin Transl Med 2021; 11:e464. [PMID: 34586738 PMCID: PMC8473478 DOI: 10.1002/ctm2.464] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 05/24/2021] [Accepted: 05/30/2021] [Indexed: 02/03/2023] Open
Abstract
Esophageal adenocarcinoma (EAC), a subtype of esophageal carcinoma, is a severe health problem associated with high death rate and poor prognosis. Immunotherapy has proven to be effective in many solid tumors, including EAC, but immune escape blocks its effectiveness. Thus, we explored the mechanisms and functional role of c-Myb in immune escape of EAC cells. Clinical EAC tissues were collected for determining the expression of c-Myb, speckled POZ protein (SPOP), and miR-145-5p. Functional assays were then performed to detect the interactions between c-Myb and SPOP as well as between SPOP and miR-145-5p. EAC cell invasion and migration were assessed. Next, T cells were sorted and cocultured with EAC cells with different treatments followed by detection of T-cell viability. In addition, a mouse model of EAC was constructed for relevant in vivo assays. c-Myb and miR-145-5p were highly expressed and SPOP had low expressions in EAC. c-Myb activated the transcription of miR-145-5p, which in turn targeted SPOP. Further, SPOP accelerated the ubiquitination of PD-L1 to enhance its expression. Overexpression of PD-L1 suppressed T-cell functions and promoted proliferative and migrative abilities of EAC cells to induce immune escape. The above findings were also confirmed in the ECA mouse model in vivo. Our findings uncovered that c-Myb can promote the immune escape of EAC cells by favoring the transcription of miR-145-5p and inhibiting SPOP-dependent ubiquitination and degradation of PD-L1, thus, presenting new target for EAC adjunct therapy.
Collapse
Affiliation(s)
- Lan Zhang
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
| | - Xiaohui Wang
- Department of PathologyThe First Affiliated Hospital of Xinxiang Medical UniversityZhengzhouP. R. China
| | - Yunfei Li
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
| | - Jing Han
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
| | - Xianzheng Gao
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
| | - Shenglei Li
- Department of PathologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
| | - Feng Wang
- Department of OncologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouP. R. China
| |
Collapse
|
17
|
Sun Y, Tan J, Miao Y, Zhang Q. The role of PD-L1 in the immune dysfunction that mediates hypoxia-induced multiple organ injury. Cell Commun Signal 2021; 19:76. [PMID: 34256773 PMCID: PMC8276205 DOI: 10.1186/s12964-021-00742-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
Hypoxia is a pathological condition common to many diseases, although multiple organ injuries induced by hypoxia are often overlooked. There is increasing evidence to suggest that the hypoxic environment may activate innate immune cells and suppress adaptive immunity, further stimulating inflammation and inhibiting immunosurveillance. We found that dysfunctional immune regulation may aggravate hypoxia-induced tissue damage and contribute to secondary injury. Among the diverse mechanisms of hypoxia-induced immune dysfunction identified to date, the role of programmed death-ligand 1 (PD-L1) has recently attracted much attention. Besides leading to tumour immune evasion, PD-L1 has also been found to participate in the progression of the immune dysfunction which mediates hypoxia-induced multiple organ injury. In this review, we aimed to summarise the role of immune dysfunction in hypoxia-induced multiple organ injury, the effects of hypoxia on the cellular expression of PD-L1, and the effects of upregulated PD-L1 expression on immune regulation. Furthermore, we summarise the latest information pertaining to the involvement, diagnostic value, and therapeutic potential of immunosuppression induced by PD-L1 in various types of hypoxia-related diseases, including cancers, ischemic stroke, acute kidney injury, and obstructive sleep apnoea. Video Abstract.
Collapse
Affiliation(s)
- Yang Sun
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052 China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052 China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Anshan Road NO.154, Tianjin, 300052 China
| |
Collapse
|
18
|
Mahmoudian RA, Mozhgani S, Abbaszadegan MR, Mokhlessi L, Montazer M, Gholamin M. Correlation between the immune checkpoints and EMT genes proposes potential prognostic and therapeutic targets in ESCC. J Mol Histol 2021; 52:597-609. [PMID: 33884540 DOI: 10.1007/s10735-021-09971-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/27/2021] [Indexed: 12/15/2022]
Abstract
PD-1, PD-L1, CTLA-4, TIM-3, and LAG-3, crucial immune checkpoint molecules in the tumor microenvironment, identify as key targets for cancer immunotherapy. There is a correlation between immune cells and epithelial-mesenchymal transition (EMT)-related genes expression in varies human cancers. In this study, we aimed to investigate the probable association between expression of immune checkpoints and EMT in esophageal squamous cell carcinoma (ESCC) with clinical treats for providing the new therapeutic targets and prognostic value for the disease. Quantitative real-time PCR was used to investigate the gene expression profile of immune checkpoints (PD-1, PD-L1, CTLA-4, TIM-3, and LAG-3) and EMT (TWIST1 and MMP-13) genes based on the mRNA expression levels in 51 ESCC tissues. The upregulation of CTLA-4, PD-1, PD-L1, TIM-3, LAG-3, MMP-13, and TWIST1 were observed in 31.37%, 29.41%, 21.56%, 39.21%, 25.49%, 60.78%, and 56.86% of ESCC cases at the mRNA level, respectively. Dysregulation of immune checkpoints was related to lymph node involvement, stage of tumor progression, and depth of tumor invasion (P < 0.05). While overexpression of MMP-13 and TWIST1 was associated with lymph node involvement, stage of tumor progression, and grade of tumor differentiation (P < 0.05). The mRNA expression of immune checkpoint genes was significantly correlated to each other's (P = 0.000). Of importance, the data explored the significant association between the concomitant expression of immune checkpoints and EMT-related genes with each other in a variety of clinicopathological traits (P < 0.05). Consequently, immune checkpoints were positively correlated with EMT status in ESCC. The correlation between tumor immune microenvironment with the elevation of multiple immune checkpoints and EMT status may help to identify potential biomarkers for the simultaneous clinical use of multiple immune checkpoints blockade and other immunotherapies approaches for advanced ESCC patients.
Collapse
Affiliation(s)
| | - Sahar Mozhgani
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Leila Mokhlessi
- School of Pharmacy, Goethe-University, Frankfurt a. M., Germany
| | - Mehdi Montazer
- Department of Pathology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehran Gholamin
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, P.O.Box, 345-91357, Mashhad, Iran.
| |
Collapse
|
19
|
Seo I, Lee HW, Byun SJ, Park JY, Min H, Lee SH, Lee JS, Kim S, Bae SU. Neoadjuvant chemoradiation alters biomarkers of anticancer immunotherapy responses in locally advanced rectal cancer. J Immunother Cancer 2021; 9:jitc-2020-001610. [PMID: 33692216 PMCID: PMC7949478 DOI: 10.1136/jitc-2020-001610] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Neoadjuvant chemoradiation therapy (CRT) is a widely used preoperative treatment strategy for locally advanced rectal cancer (LARC). However, a few studies have evaluated the molecular changes caused by neoadjuvant CRT in these cancer tissues. Here, we aimed to investigate changes in immunotherapy-related immunogenic effects in response to preoperative CRT in LARC. METHODS We analyzed 60 pairs of human LARC tissues before and after irradiation from three independent LARC cohorts, including a LARC patient RNA sequencing (RNA-seq) dataset from our cohort and GSE15781 and GSE94104 datasets. RESULTS Gene ontology analysis showed that preoperative CRT significantly enriched the immune response in LARC tissues. Moreover, gene set enrichment analysis revealed six significantly enriched Kyoto Encyclopedia of Genes and Genomes pathways associated with downregulated genes, including mismatch repair (MMR) genes, in LARC tissues after CRT in all three cohorts. Radiation also induced apoptosis and downregulated various MMR system-related genes in three colorectal cancer cells. One patient with LARC showed a change in microsatellite instability (MSI) status after CRT, as demonstrated by the loss of MMR protein and PCR for MSI. Moreover, CRT significantly increased tumor mutational burden in LARC tissues. CIBERSORT analysis revealed that the proportions of M2 macrophages and CD8 T cells were significantly increased after CRT in both the RNA-seq dataset and GSE94104. Notably, preoperative CRT increased various immune biomarker scores, such as the interferon-γ signature, the cytolytic activity and the immune signature. CONCLUSIONS Taken together, our findings demonstrated that neoadjuvant CRT modulated the immune-related characteristics of LARC, suggesting that neoadjuvant CRT may enhance the responsiveness of LARC to immunotherapy.
Collapse
Affiliation(s)
- Incheol Seo
- Department of Microbiology, Dongguk University College of Medicine, Gyeongju, Gyeongsangbuk-do, Korea (the Republic of)
| | - Hye Won Lee
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea (the Republic of).,Institute for Cancer Research, Keimyung University, Daegu, Korea (the Republic of)
| | - Sang Jun Byun
- Department of Radiation Oncology, Keimyung University School of Medicine, Daegu, Korea (the Republic of)
| | - Jee Young Park
- Department of Pathology, Keimyung University Dongsan Medical Center, Daegu, Korea (the Republic of).,Department of Immunology, Keimyung University School of Medicine, Daegu, Korea (the Republic of)
| | - Hyeonji Min
- Department of Immunology, Keimyung University School of Medicine, Daegu, Korea (the Republic of)
| | - Sung Hwan Lee
- Department of Surgery, CHA University - Bundang Campus, Seongnam, Gyeonggi-do, Korea (the Republic of).,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ju-Seog Lee
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shin Kim
- Department of Immunology, Keimyung University School of Medicine, Daegu, Korea (the Republic of) .,Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Institute of Medical Science & Institute for Cancer Research, Keimyung University, Daegu, Korea (the Republic of)
| | - Sung Uk Bae
- Institute of Medical Science & Institute for Cancer Research, Keimyung University, Daegu, Korea (the Republic of) .,Department of Surgery, Keimyung University Dongsan Medical Center, Daegu, Korea (the Republic of)
| |
Collapse
|
20
|
Zhu T, Ma Z, Wang H, Wei D, Wang B, Zhang C, Fu L, Li Z, Yu G. Immune-Related Long Non-coding RNA Signature and Clinical Nomogram to Evaluate Survival of Patients Suffering Esophageal Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:641960. [PMID: 33748133 PMCID: PMC7969885 DOI: 10.3389/fcell.2021.641960] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) turns out to be one of the most prevalent cancer types, leading to a relatively high mortality among worldwide sufferers. In this study, gene microarray data of ESCC patients were obtained from the GEO database, with the samples involved divided into a training set and a validation set. Based on the immune-related differential long non-coding RNAs (lncRNAs) we identified, a prognostic eight-lncRNA-based risk signature was constructed following regression analyses. Then, the predictive capacity of the model was evaluated in the training set and validation set using survival curves and receiver operation characteristic curves. In addition, univariate and multivariate regression analyses based on clinical information and the model-based risk score also demonstrated the ability of the risk score in independently determining the prognosis of patients. Besides, based on the CIBERSORT tool, the abundance of immune infiltrates in tumor samples was scored, and a significant difference was presented between the high- and low- risk groups. Correlation analysis with immune checkpoints (PD1, PDL1, and CTLA4) indicated that the eight-lncRNA signature–based risk score was negatively correlated with PD1 expression, suggesting that the eight-lncRNA signature may have an effect in immunotherapy for ESCC. Finally, GO annotation was performed for the differential mRNAs that were co-expressed with the eight lncRNAs, and it was uncovered that they were remarkably enriched in immune-related biological functions. These results suggested that the eight-lncRNA signature–based risk model could be employed as an independent biomarker for ESCC prognosis and might play a part in evaluating the response of ESCC to immunotherapy with immune checkpoint blockade.
Collapse
Affiliation(s)
- Ting Zhu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Zhifeng Ma
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Haiyong Wang
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Desheng Wei
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Bin Wang
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Chu Zhang
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Linhai Fu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Zhupeng Li
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People's Hospital, Shaoxing, China
| |
Collapse
|
21
|
Zhang N, Shi J, Shi X, Chen W, Liu J. Mutational Characterization and Potential Prognostic Biomarkers of Chinese Patients with Esophageal Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:12797-12809. [PMID: 33363385 PMCID: PMC7751839 DOI: 10.2147/ott.s275688] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/12/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal cancer in China and the 5-year mortality rate is up to 70%. Studies on the ESCC genetic landscape are needed to further explore clinical therapeutic strategies. In this study, we evaluated the genetic landscape of ESCC to aid the search for clinical therapeutic strategies. Patients and Methods A total of 225 ESCC patients were enrolled in this study. Deep sequencing of 450 cancer genes was performed on formalin-fixed paraffin-embedded tumor biopsies and matched blood samples from patients. Tumor mutational burden (TMB) was calculated using an algorithm developed in-house. Results Our results showed that the most commonly mutated genes in ESCC were TP53 (96%), CCND1 (46%), FGF4 (44%), FGF19 (44%), FGF3 (44%), CDKN2A (31%), PIK3CA (26%), NOTCH1 (24%), KMT2D (18%), FAT1 (16%), and LRP1B (16%). We found that TMB correlated with patient drinking status. We identified mutations associated with sex, early ESCC, high TMB, and metastasis lymph nodes. KMT2D mutations associated with sex (P = 0.035), tumor stage (P = 0.016), high TMB (P = 0.0072), and overall survival of patients (P = 0.0026). SPEN mutations associated with high TMB (P = 0.0016) and metastasis-positive lymph nodes (P = 0.027). These results suggested that SPEN and KMT2D could be potential prognosis biomarkers for Chinese patients with ESCC. We also found that the number of positive lymph nodes was associated with disease-free survival. Clinical target gene analysis indicated that nearly half of Chinese ESCC patients might benefit from treatment with gene-specific target drugs. Conclusion Our study revealed the ESCC mutational landscape in 225 Chinese patients and uncovered the potential prognosis biomarker for Chinese patients with ESCC.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Junping Shi
- Department of Medicine, OrigiMed Co. Ltd, Shanghai, People's Republic of China
| | - Xiaoliang Shi
- Department of Medicine, OrigiMed Co. Ltd, Shanghai, People's Republic of China
| | - Wenting Chen
- Department of Medicine, OrigiMed Co. Ltd, Shanghai, People's Republic of China
| | - Junfeng Liu
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
22
|
Yang D, Xu F, Lai X, Li Y, Gao H, Xu Y, Chen R, Ma D. Combined treatment with anlotinib and chemotherapy for advanced esophageal squamous cell carcinoma improved patient survival: a case report. Am J Transl Res 2020; 12:6578-6583. [PMID: 33194054 PMCID: PMC7653628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 06/03/2020] [Indexed: 06/11/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the predominant type of esophageal cancer in Eastern Asia. Historically, advanced ESCC treatments have had low efficacy and new treatments, including immunotherapy or combination therapies, are emerging. Here, we report a special case of recurrent ESCC after surgery. The patient had a failed immunotherapy course, but benefited from anlotinib combined with chemotherapy for a fourth-line therapy. Survival after the combined therapy was greater than 19 months, and the overall patient survival was greater than 32 months.
Collapse
Affiliation(s)
- Dongyang Yang
- Department of Gastrointestinal Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, China
| | - Fei Xu
- Department of Gastrointestinal Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, China
| | - Xiaorong Lai
- Department of Gastrointestinal Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, China
| | - Ying Li
- Department of Gastrointestinal Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, China
| | - Huibing Gao
- Department of Gastrointestinal Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, China
| | - Ying Xu
- Department of Computer Science and Technology, School of Electronic and Information Engineering, Xi’an Jiaotong UniversityXi’an 710049, Shaanxi, China
| | | | - Dong Ma
- Department of Gastrointestinal Cancer, Cancer Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical SciencesGuangzhou 510080, China
| |
Collapse
|
23
|
Baba Y, Nomoto D, Okadome K, Ishimoto T, Iwatsuki M, Miyamoto Y, Yoshida N, Baba H. Tumor immune microenvironment and immune checkpoint inhibitors in esophageal squamous cell carcinoma. Cancer Sci 2020; 111:3132-3141. [PMID: 32579769 PMCID: PMC7469863 DOI: 10.1111/cas.14541] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 05/18/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the main prevalent histological type of esophageal cancer, predominantly constituting 90% of cases worldwide. Despite the development of multidisciplinary therapeutic approaches, its prognosis remains unfavorable. Recently, the development of monoclonal antibodies inhibiting programmed death 1 (PD-1) or programmed death-ligand 1 (PD-L1) has led to marked therapeutic responses among multiple malignancies including ESCC. However, only a few patients achieved clinical benefits due to resistance. Therefore, precise and accurate predictive biomarkers should be identified for personalized immunotherapy in clinical settings. Because the tumor immune microenvironment can potentially influence the patient's response to immune checkpoint inhibitors, tumor immunity, such as PD-L1 expression on tumors, tumor-infiltrating lymphocytes, tumor-associated macrophages, and myeloid-derived suppressor cells, in ESCC should be further investigated. In this review, accumulated evidence regarding the tumor immune microenvironment and immune checkpoint inhibitors in ESCC are summarized.
Collapse
Affiliation(s)
- Yoshifumi Baba
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
- Department of Next‐Generation Surgical Therapy DevelopmentGraduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Daichi Nomoto
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| | - Kazuo Okadome
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| | - Takatsugu Ishimoto
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| | - Masaaki Iwatsuki
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| | - Yuji Miyamoto
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| | - Naoya Yoshida
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| | - Hideo Baba
- Department of Gastroenterological SurgeryGraduate School of Medical ScienceKumamoto UniversityKumamotoJapan
| |
Collapse
|
24
|
Zhang W, Wang P, Pang Q. Immune checkpoint inhibitors for esophageal squamous cell carcinoma: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1193. [PMID: 33241042 PMCID: PMC7576075 DOI: 10.21037/atm-20-4625] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Esophageal cancer (EC) has the seventh highest incidence and the sixth highest mortality rate of any type of cancer worldwide. In China, esophageal squamous cell carcinoma (ESCC) accounts for more than 95% of EC patients. The main treatment for EC patients is surgery and/or chemoradiotherapy (CRT). A large proportion of EC patients are already at an advanced stage of the disease by the time they are diagnosed. In these cases, CRT is left as the only treatment choice, and the treatment outcome is poor. Immune checkpoint inhibitors (ICIs) can improve clinical response and patient survival of patient with many types of tumors through reactivating antitumor immune response. The study of ICIs in ESCC is relative delayed compared with that in other solid tumors. Recent results from clinical trials have demonstrated the safety and efficacy of ICIs either alone or combined with chemotherapy or chemoradiotherapy in ESCC patients. Accumulated evidences also have shown the improved treatment outcome was associated with PD-L1 expression, tumor DNA instability-induced tumor mutational burden (TMB), and drawing lymphocytes into the tumor. Based on these findings, ICIs combined with CRT or radiotherapy (RT) are the focus of ongoing studies. This review will summarize the recent progress in this field, especially the mechanism of ICIs used in ESCC, their clinical efficacy and toxicities, and potential biomarkers.
Collapse
Affiliation(s)
- Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital/National Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital/National Clinical Research Center for Cancer, Tianjin, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital/National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
25
|
Zheng S, Liu Q, Liu T, Yang L, Zhang Q, Shen T, Zhang X, Han X, Lu X. NME4 modulates PD-L1 expression via the STAT3 signaling pathway in squamous cell carcinoma. Biochem Biophys Res Commun 2020; 526:29-34. [PMID: 32192776 DOI: 10.1016/j.bbrc.2020.03.055] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/05/2020] [Indexed: 02/04/2023]
Abstract
NME4, also named Nm23-H4, is a contraction of NME/NM23 Nucleoside Diphosphate Kinase 4, whose major role is the synthesis of nucleoside triphosphates. However, its association with programmed death ligand 1 (PD-L1) remains far from understood. Herein, it was discovered that silencing NME4 can lead to the marked downregulation of PD-L1, with phosphorylated STAT3 at the 705th serine being inactivated in vitro in esophageal squamous cell carcinoma (ESCC) cell lines. To further validate the association between NME4 and PD-L1 that was observed in cell lines, Pearson correlation analysis was performed on the data regarding the transcriptomic RNA sequencing of NME4 and PD-L1 in cervical squamous cell carcinoma (CSCC), which pathologically highly resembles ESCC in terms of tumor origin, obtained from the GEPIA database. It was demonstrated that their correlation was significant but negative between NME4 and PD-L1 in CSCC. To the best of our knowledge, this is the first report describing a modulation exerted by NME4 over PD-L1 in the background of squamous cell carcinoma, strongly suggestive of the underlying role of NME4 working to exclude CD8 T cells from infiltrating into the squamous cell carcinoma microenvironment.
Collapse
Affiliation(s)
- Shutao Zheng
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Qing Liu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Tao Liu
- Health Management Center, Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Lifei Yang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Qiqi Zhang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Tongxue Shen
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Xiao Zhang
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Xiujuan Han
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China
| | - Xiaomei Lu
- Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, PR China; State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Xinjiang Uygur Autonomous Region, Urumqi, PR China.
| |
Collapse
|