1
|
Chi Z, Peng L, Karamchandani DM, Xu J. PD-L1 (22C3) expression and prognostic implications in esophageal squamous cell carcinoma. Ann Diagn Pathol 2025; 74:152394. [PMID: 39549525 DOI: 10.1016/j.anndiagpath.2024.152394] [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: 10/10/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/18/2024]
Abstract
Programmed cell death-ligand 1 (PD-L1) clone 22C3 is the only Food and Drug Administration-approved companion diagnostic test for pembrolizumab for the treatment of esophageal squamous cell carcinoma (ESCC). However, prior studies conducted in Asia and Europe have used various PD-L1 antibody clones other than 22C3. We aimed to study the expression profile of PD-L1, specifically of clone 22C3, in ESCC and its significance with regards to histological features, clinical parameters, and overall survival in a case series from two large US hospital systems. PD-L1 (22C3) immunohistochemistry was performed on 82 specimens obtained from 75 patients. Electronic medical records were reviewed to obtain the clinical and follow-up data. Of these specimens, 39 % (32/82) were negative for PD-L1 (22C3) expression (combined positive score (CPS) of 0). The remaining 50 specimens were positive, with CPSs ranging from 1 to 100. Treated specimens showed decreased PD-L1 (22C3) expression compared to untreated specimens. In the multivariate Cox proportional hazards regression model, PD-L1 (22C3) expression was shown to be a favorable prognostic factor for overall survival (p = 0.03, hazard ratio 0.16) only when the CPSs were ≥ 25, independent of surgery, definitive chemotherapy and/or radiotherapy, immunotherapy, and initial clinical stages. We performed a comprehensive study to investigate the expression profile of PD-L1 clone 22C3 in the US patients with ESCC. Our analysis showed that PD-L1 (22C3) expression decreased in treated specimens, and a CPS of ≥25 was associated with a favorable prognosis.
Collapse
Affiliation(s)
- Zhikai Chi
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Lan Peng
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Dipti M Karamchandani
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jing Xu
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
2
|
He Y, Zhu M, Lai X, Zhang H, Jiang W. The roles of PD-L1 in the various stages of tumor metastasis. Cancer Metastasis Rev 2024; 43:1475-1488. [PMID: 38733457 DOI: 10.1007/s10555-024-10189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
The interaction between tumor programmed death ligand 1 (PD-L1) and T-cell programmed cell death 1 (PD-1) has long been acknowledged as a mechanism for evading immune surveillance. Recent studies, however, have unveiled a more nuanced role of tumor-intrinsic PD-L1 in reprograming tumoral phenotypes. Preclinical models emphasize the synchronized effects of both intracellular and extracellular PD-L1 in promoting metastasis, with intricate interactions with the immune system. This review aims to summarize recent findings to elucidate the spatiotemporal heterogeneity of PD-L1 expression and the pro-metastatic roles of PD-L1 in the entire process of tumor metastasis. For example, PD-L1 regulates the epithelial-to-mesenchymal transition (EMT) process, facilitates the survival of circulating tumor cells, and induces the formation of immunosuppressive environments at pre-metastatic niches and metastatic sites. And the complexed and dynamic regulation process of PD-L1 for tumor metastasis is related to the spatiotemporal heterogeneity of PD-L1 expression and functions from tumor primary sites to various metastatic sites. This review extends the current understandings for the roles of PD-L1 in mediating tumor metastasis and provides new insights into therapeutic decisions in clinical practice.
Collapse
Affiliation(s)
- Yinjun He
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, 310009, China
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Ming Zhu
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Xuan Lai
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China.
| | - Weiqin Jiang
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, 310009, China.
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China.
| |
Collapse
|
3
|
Yu Q, Li J, Mao W, Li Z, Li X, Li B. The Prognostic Value of Serum Soluble Programmed Death 1 (sPD-1) and Programmed Death Ligand 1 (sPD-L1) in Esophageal Squamous Cell Carcinoma: A Systematic Review and Meta-Analysis About Cohort Studies. Health Sci Rep 2024; 7:e70178. [PMID: 39587995 PMCID: PMC11586633 DOI: 10.1002/hsr2.70178] [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: 01/19/2024] [Revised: 10/01/2024] [Accepted: 10/16/2024] [Indexed: 11/27/2024] Open
Abstract
Background and Aims There are still no useful biomarkers for the prognosis of esophageal squamous cell carcinoma (ESCC). In the prognosis of some kinds of cancer, soluble programmed death 1 (sPD-1) and programmed death ligand 1 (sPD-L1) have demonstrated statistical significance, but the prognostic value of serum sPD-L1 and sPD-1 remains unclear in ESCC. Methods Here, a meta-analysis was performed to estimate the prognostic value of sPD-L1 and sPD-1 in ESCC. To obtain eligible studies, we searched mainstream databases (PubMed, Cochrane, Embase, Web of Science, Wanfang Data, and CNKI), and the survival data including hazard ratios (HR) and its 95% confidence intervals (95% CI) from included literature were extracted. Results Six articles were included, including 645 patients with ESCC. The statistical result of this meta-analysis indicated that serum sPD-1 had no significant correlation with overall survival (OS) of patients with ESCC (p > 0.05). Patients with ESCC with high concentrations of serum sPD-L1 demonstrated a significantly poor prognosis (HR = 1.73, 95% CI: 1.42-2.11, p < 0.001). Conclusion Higher levels of serum sPD-L1 may predict poor OS in ESCC patients, which may be a promising and credible prognostic biomarker for esophageal cancer.
Collapse
Affiliation(s)
- Qiyao Yu
- Department of Plastic SurgeryBOE HospitalChengduChina
- Department of Thoracic SurgerySecond Hospital of Lanzhou University, Second Clinical Medical College of Lanzhou UniversityChina
| | - Jie Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Key Laboratory of Environmental OncologyLanzhouChina
| | - Wenjie Mao
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Key Laboratory of Environmental OncologyLanzhouChina
| | - Zheng Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Key Laboratory of Environmental OncologyLanzhouChina
| | - Xuan Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Key Laboratory of Environmental OncologyLanzhouChina
| | - Bin Li
- Department of Thoracic Surgery, The Second Hospital & Clinical Medical SchoolLanzhou UniversityLanzhouChina
- Gansu Province Key Laboratory of Environmental OncologyLanzhouChina
| |
Collapse
|
4
|
Zhao H, Luo K, Liu M, Cai Y, Liu S, Li S, Zhao Y, Zhang H. Immune regulation and prognostic prediction model establishment and validation of PSMB6 in lung adenocarcinoma. Front Genet 2024; 15:1458047. [PMID: 39507618 PMCID: PMC11538069 DOI: 10.3389/fgene.2024.1458047] [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: 07/01/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Lung cancer is one of the most common malignant tumors, and patients are often diagnosed at an advanced stage, posing a substantial risk to human health, so it is crucial to establish a model to forecast the prognosis of patients with lung cancer. Recent research has indicated that proteasome 20S subunit 6 (PSMB6) may be closely associated with anti-apoptotic pathways, and proliferation transduction signals in tumor cells of different tumors. However, the precise role of PSMB6 in the immunoregulatory processes within lung adenocarcinoma (LUAD) is yet to be elucidated. By analyzing the TCGA database, we discovered a positive correlation between the expression of PSMB6 and tumor growth trends, and lung adenocarcinoma patients with elevated PSMB6 expression levels had a worse prognosis. Our findings suggest a close correlation between PSMB6 expression levels, immune cell infiltration and immune checkpoint gene expression, which suggests that PSMB6 may become a new independent prognostic indicator. In addition, we developed a prognostic model of PSMB6-regulated immune infiltration-associated genes by analyzing the link between PSMB6 and the immune microenvironment. This model can not only predict the prognosis of lung adenocarcinoma but also forecasts the patient's reaction to immunotherapy. The validity of this research outcome has been confirmed by the GSE31210 and IMvigor210 cohorts. Analysis of the Kaplan-Meier Plotter database indicates that individuals with elevated levels of PSMB6 expression exhibit a poorer prognosis. Additionally, in vitro experiments demonstrated that knockdown of PSMB6 inhibits the proliferation, migration, and invasion of lung adenocarcinoma cells while promoting their apoptosis. Overall, our findings suggest that PSMB6 could remarkably influence the management and treatment of lung adenocarcinoma, opening new avenues for targeted immunotherapeutic strategies.
Collapse
Affiliation(s)
- Haiyang Zhao
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
| | - Kexin Luo
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
| | - Meihan Liu
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuanze Cai
- North Sichuan Medical College, Nanchong, China
| | - Siman Liu
- North Sichuan Medical College, Nanchong, China
| | - Shijuan Li
- Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Yongsheng Zhao
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hongpan Zhang
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Therapeutic Proteins Key Laboratory of Sichuan Province, Nanchong, China
| |
Collapse
|
5
|
Groeger S, Meyle J. The role of programmed death receptor (PD-)1/PD-ligand (L)1 in periodontitis and cancer. Periodontol 2000 2024; 96:150-169. [PMID: 38351432 PMCID: PMC11579837 DOI: 10.1111/prd.12548] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 11/22/2024]
Abstract
The programmed-death-ligand-1 (PD-L1) is an immune-modulating molecule that is constitutively expressed on various immune cells, different epithelial cells and a multitude of cancer cells. It is a costimulatory molecule that may impair T-cell mediated immune response. Ligation to the programmed-death-receptor (PD)-1, on activated T-cells and further triggering of the related signaling pathways can induce T-cells apoptosis or anergy. The upregulation of PD-L1 in various cancer types, including oral squamous cell carcinomas, was demonstrated and has been linked to immune escape of tumors and poor prognosis. A bidirectional relationship exists between the increased PD-L1 expression and periodontitis as well as the epithelial-mesenchymal transition (EMT), a process of interconversion of epithelial cells to mesenchymal cells that may induce immune escape of tumors. Interaction between exosomal PD-L1 and PD-1 on T-cells may cause immunosuppression by blocking the activation and proliferation of T-cells. The efficacy and importance of treatment with PD-1/PD-L1 checkpoint inhibitors and their prognostic influence on human cancers was demonstrated. Regarding PD-1/PD-L1 checkpoint inhibitors, resistances exist or may develop, basing on various factors. Further investigations of the underlying mechanisms will help to overcome the therapeutic limitations that result from resistances and to develop new strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
- Department of Orthodontics, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| | - Joerg Meyle
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| |
Collapse
|
6
|
Hu J, Toyozumi T, Murakami K, Endo S, Matsumoto Y, Otsuka R, Shiraishi T, Iida S, Morishita H, Makiyama T, Nishioka Y, Uesato M, Hayano K, Nakano A, Matsubara H. Prognostic value of tumor-infiltrating lymphocytes and PD-L1 expression in esophageal squamous cell carcinoma. Cancer Med 2024; 13:e70179. [PMID: 39264227 PMCID: PMC11391568 DOI: 10.1002/cam4.70179] [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: 04/26/2024] [Revised: 08/05/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Tumor cells (TC) participate in tumor progression by altering the immune responses in the tumor microenvironment. However, the clinical relevance and prognostic effect of PD-L1 expression and tumor-infiltrating lymphocytes (TILs) in esophageal squamous cell carcinoma (ESCC) are unknown. The purpose of this study was to investigate the interactions and clinical significance of PD-L1 expression and TILs in ESCC. METHODS Tissue specimens were collected from 126 patients with ESCC who underwent curative esophagectomy. Immunohistochemical analysis and multiplex immunofluorescence for CD4, CD8, CD25, FOXP3, and PD-L1 in the tumor were used to identify multiple tumor-infiltrating immune cells (TIIC), Tregs, and TC. RESULTS PD-L1 was expressed in tumor cells (PD-L1 TC). PD-L1 TIIC and PD-L1 TC affected the biological behavior of TC. The positive expression rate of PD-L1 TC and CD8+ TILs was 27.8% (35/126) and 31.7% (40/126), respectively. Kaplan-Meier analysis showed that overall survival (OS) was significantly associated with decreased CD8+ TILs and PD-L1 TC-positive expression, which promote ESCC progression and metastasis. CONCLUSION Tumor depth, CD8, and PD-L1 TC were independent prognostic factors in ESCC, and a predictive nomogram with these three risk factors improved the accuracy of predicting OS in patients with ESCC after surgical resection. The conjoint analysis of multiple immune-related factors is beneficial for stratifying patient survival risk.
Collapse
Affiliation(s)
- Jie Hu
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Takeshi Toyozumi
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Kentaro Murakami
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Satoshi Endo
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Yasunori Matsumoto
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Ryota Otsuka
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Tadashi Shiraishi
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Shinichiro Iida
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Hiroki Morishita
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Tenshi Makiyama
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Yuri Nishioka
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Masaya Uesato
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Koichi Hayano
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Akira Nakano
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| | - Hisahiro Matsubara
- Department of Frontier SurgeryGraduate School of Medicine, Chiba UniversityChibaJapan
| |
Collapse
|
7
|
Shahabinejad M, Bagheri Shirvan S, Fatemi G, Mohajer Tehran F. Comparative analysis of PDL1 and cluster of differentiation 68 marker expression in oral squamous cell carcinoma patients: Correlation with depth of invasion and immunofluorescence through immunohistochemistry. Dent Res J (Isfahan) 2024; 21:48. [PMID: 39376261 PMCID: PMC11457987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 04/24/2024] [Accepted: 05/05/2024] [Indexed: 10/09/2024] Open
Abstract
Background Over the past 5 years, the use of immune checkpoint inhibitors in the treatment of head-and-neck squamous cell carcinoma (HNSCC) has increased. Both programmed death-ligand 1 (PD-L1) and cluster of differentiation 68 (CD68) are overexpressed in various carcinomas. Consequently, evaluating the expression of CD68 and PD-L1 in HNSCC lesions may lead to detecting a possible marker for HNSCC. This study aimed to evaluate the expression of PDL1 and CD68 markers in a patient with oral squamous cell carcinoma (OSCC) and examine its relationship with depth of invasion (DOI) and immunofluorescence (IF) through immunohistochemistry. Materials and Methods This cross-sectional study was conducted in the School of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran, Department of Oral and Maxillofacial Pathology. Thirty-four paraffin blocks and demographic information of 15 female and 19 male OSCC patients were collected. Following sample preparations, immunohistochemical staining was performed. Subsequently, each tissue section was analyzed for tumor-infiltrating lymphocytes by CD68 marker and PD-L1 expression. Data analysis was conducted using SPSS software (version 25). Chi-square, Shapiro-Wilk, and independent t-analytical tests were employed for statistical assessments. P < 0.05 was remarked as statistically significant. Results CD68 and PDL1 expression in the squamous cell carcinoma (SCC) group was higher than the control group (P < 0.001). There was an increasing expression of PDL1 and CD68 as the grade of the disease progressed (P < 0.001 for each), as well as an increasing expression of IF and DOI. Conclusion The expression levels of CD68 and PDL1 were elevated in SCC tissues in comparison to the unaffected, healthy parts of the tissue section.
Collapse
Affiliation(s)
- Mehdi Shahabinejad
- Department of Oral and Maxillofacial Pathology, Oral and Maxillofacial Diseases Research Center, Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Siavash Bagheri Shirvan
- Department of Oral and Maxillofacial Pathology, Oral and Maxillofacial Diseases Research Center, Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Golnaz Fatemi
- Department of Oral and Maxillofacial Pathology, Oral and Maxillofacial Diseases Research Center, Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farnaz Mohajer Tehran
- Department of Oral and Maxillofacial Pathology, Oral and Maxillofacial Diseases Research Center, Faculty of Dentistry, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Pimenta J, Prada J, Pires I, Cotovio M. Programmed-cell death ligand 1 (PD-L1) expression in equine sarcoids and squamous cell carcinoma. Open Vet J 2024; 14:1476-1482. [PMID: 39055760 PMCID: PMC11268900 DOI: 10.5455/ovj.2024.v14.i6.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 05/15/2024] [Indexed: 07/27/2024] Open
Abstract
Background Sarcoids and squamous cell carcinomas (SCCs) are the most concerning equine oncological diseases. Both tumors are challenging to manage due to their invasive behavior and high prevalence of recurrences. Furthermore, SCCs have a propensity to metastasize. Programed cell-death ligand 1 (PD-L1) has been one of the main therapeutic targets for immunotherapy in various human tumors. PD-L1 research in equine tumors is scarce and more efforts are necessary to understand the potential of this biomarker as a therapeutical target. Aim Evaluate the immunohistochemical expression of PD-L1 in equine sarcoids and SCC. Methods Thirteen equine tumors (seven sarcoids and 6 SCCs) were tested by immunohistochemistry and evaluated semi quantitatively to assess the percentage of positive cells. Results None of the sarcoids presented PD-L1 expression. Regarding SCC, 2 tumors presented <10% of labeled cells; 2 tumors presented 10%-25% of labeled cells and 2 tumors presented 25%-50% of labeled cells. There were statistically significant differences between sarcoids and SCC regarding the expression of PD-L1. Conclusion Our results point to the fact that PD-L1 could be a potential therapeutic target against SCC, and also encourage in-depth studies in this area, with larger sample sizes.
Collapse
Affiliation(s)
- José Pimenta
- CECAV Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Vila Real, Portugal
- CIVG Vasco da Gama Research Center / EUVG – Vasco da Gama University School, Coimbra, Portugal
| | - Justina Prada
- CECAV Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Isabel Pires
- CECAV Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Vila Real, Portugal
- Veterinary Sciences Department, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
| | - Mário Cotovio
- CECAV Veterinary and Animal Research Center, University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Vila Real, Portugal
- Faculty of Veterinary Medicine, Lusófona University, Lisbon, Portugal
| |
Collapse
|
9
|
Yu X, Xu J. TWIST1 Drives Cytotoxic CD8+ T-Cell Exhaustion through Transcriptional Activation of CD274 (PD-L1) Expression in Breast Cancer Cells. Cancers (Basel) 2024; 16:1973. [PMID: 38893094 PMCID: PMC11171171 DOI: 10.3390/cancers16111973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
In breast cancer, epithelial-mesenchymal transition (EMT) is positively associated with programmed death ligand 1 (PD-L1) expression and immune escape, and TWIST1 silences ERα expression and induces EMT and cancer metastasis. However, how TWIST1 regulates PD-L1 and immune evasion is unknown. This study analyzed TWIST1 and PD-L1 expression in breast cancers, investigated the mechanism for TWIST1 to regulate PD-L1 transcription, and assessed the effects of TWIST1 and PD-L1 in cancer cells on cytotoxic CD8+ T cells. Interestingly, TWIST1 expression is correlated with high-level PD-L1 expression in ERα-negative breast cancer cells. The overexpression and knockdown of TWIST1 robustly upregulate and downregulate PD-L1 expression, respectively. TWIST1 binds to the PD-L1 promoter and recruits the TIP60 acetyltransferase complex in a BRD8-dependent manner to transcriptionally activate PD-L1 expression, which significantly accelerates the exhaustion and death of the cytotoxic CD8+ T cells. Accordingly, knockdown of TWIST1 or BRD8 or inhibition of PD-L1 significantly enhances the tumor antigen-specific CD8+ T cells to suppress the growth of breast cancer cells. These results demonstrate that TWIST1 directly induces PD-L1 expression in ERα-negative breast cancer cells to promote immune evasion. Targeting TWIST1, BRD8, and/or PD-L1 in ERα-negative breast cancer cells with TWIST1 expression may sensitize CD8+ T-cell-mediated immunotherapy.
Collapse
Affiliation(s)
- Xiaobin Yu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jianming Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA;
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
10
|
Cordani M, Strippoli R, Trionfetti F, Barzegar Behrooz A, Rumio C, Velasco G, Ghavami S, Marcucci F. Immune checkpoints between epithelial-mesenchymal transition and autophagy: A conflicting triangle. Cancer Lett 2024; 585:216661. [PMID: 38309613 DOI: 10.1016/j.canlet.2024.216661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/01/2024] [Accepted: 01/17/2024] [Indexed: 02/05/2024]
Abstract
Inhibitory immune checkpoint (ICP) molecules are pivotal in inhibiting innate and acquired antitumor immune responses, a mechanism frequently exploited by cancer cells to evade host immunity. These evasion strategies contribute to the complexity of cancer progression and therapeutic resistance. For this reason, ICP molecules have become targets for antitumor drugs, particularly monoclonal antibodies, collectively referred to as immune checkpoint inhibitors (ICI), that counteract such cancer-associated immune suppression and restore antitumor immune responses. Over the last decade, however, it has become clear that tumor cell-associated ICPs can also induce tumor cell-intrinsic effects, in particular epithelial-mesenchymal transition (EMT) and macroautophagy (hereafter autophagy). Both of these processes have profound implications for cancer metastasis and drug responsiveness. This article reviews the positive or negative cross-talk that tumor cell-associated ICPs undergo with autophagy and EMT. We discuss that tumor cell-associated ICPs are upregulated in response to the same stimuli that induce EMT. Moreover, ICPs themselves, when overexpressed, become an EMT-inducing stimulus. As regards the cross-talk with autophagy, ICPs have been shown to either stimulate or inhibit autophagy, while autophagy itself can either up- or downregulate the expression of ICPs. This dynamic equilibrium also extends to the autophagy-apoptosis axis, further emphasizing the complexities of cellular responses. Eventually, we delve into the intricate balance between autophagy and apoptosis, elucidating its role in the broader interplay of cellular dynamics influenced by ICPs. In the final part of this article, we speculate about the driving forces underlying the contradictory outcomes of the reciprocal, inhibitory, or stimulatory effects between ICPs, EMT, and autophagy. A conclusive identification of these driving forces may allow to achieve improved antitumor effects when using combinations of ICIs and compounds acting on EMT and/or autophagy. Prospectively, this may translate into increased and/or broadened therapeutic efficacy compared to what is currently achieved with ICI-based clinical protocols.
Collapse
Affiliation(s)
- Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; Department of Epidemiology, Preclinical Research and Advanced Diagnostics, National Institute for Infectious Diseases L., Spallanzani, IRCCS, Via Portuense, 292, 00149 Rome, Italy
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Cristiano Rumio
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3T 2N2, Canada.
| | - Fabrizio Marcucci
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Trentacoste 2, 20134 Milan, Italy.
| |
Collapse
|
11
|
Singh D, Siddique HR. Epithelial-to-mesenchymal transition in cancer progression: unraveling the immunosuppressive module driving therapy resistance. Cancer Metastasis Rev 2024; 43:155-173. [PMID: 37775641 DOI: 10.1007/s10555-023-10141-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
Cancer cells undergo phenotypic switching (cancer cell plasticity) in response to microenvironmental cues, including exposure to therapy/treatment. Phenotypic plasticity enables the cancer cells to acquire more mesenchymal traits promoting cancer cells' growth, survival, therapy resistance, and disease recurrence. A significant program in cancer cell plasticity is epithelial-to-mesenchymal transition (EMT), wherein a comprehensive reprogramming of gene expression occurs to facilitate the translational shift from epithelial-to-mesenchymal phenotypes resulting in increased invasiveness and metastasis. In addition, EMT plays a pivotal role in facilitating cancer cells' escape from the body's immune system using several mechanisms, such as the downregulation of major histocompatibility complex-mediated antigen presentation, upregulation of immune checkpoint molecules, and recruitment of immune-suppressive cells. Cancer cells' ability to undergo phenotypic switching and EMT-driven immune escape presents a formidable obstacle in cancer management, highlighting the need to unravel the intricate mechanisms underlying these processes and develop novel therapeutic strategies. This article discusses the role of EMT in promoting immune evasion and therapy resistance. We also discuss the ongoing research on developing therapeutic approaches targeting intrinsic and induced cell plasticity within the immune suppressive microenvironment. We believe this review article will update the current research status and equip researchers, clinicians, and other healthcare professionals with valuable insights enhancing their existing knowledge and shedding light on promising directions for future cancer research. This will facilitate the development of innovative strategies for managing therapy-resistant cancers and improving patient outcomes.
Collapse
Affiliation(s)
- Deepti Singh
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India
| | - Hifzur R Siddique
- Molecular Cancer Genetics & Translational Research Lab, Section of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, India.
| |
Collapse
|
12
|
Karami Z, Mortezaee K, Majidpoor J. Dual anti-PD-(L)1/TGF-β inhibitors in cancer immunotherapy - Updated. Int Immunopharmacol 2023; 122:110648. [PMID: 37459782 DOI: 10.1016/j.intimp.2023.110648] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 08/25/2023]
Abstract
Immune checkpoint inhibitor (ICI) therapy suffers from tumor resistance and relapse in majority of patients due to the suppressive tumor immune microenvironment (TIME). Advances in the field have brought about development of fusion proteins able to target two signaling simultaneously and to exert maximal anti-cancer immunity. Bispecific inhibitors of transforming growth factor (TGF)-β signaling and programmed death-1 (PD-1) or programmed death-ligand 1 (PD-L1) are developed to reduce the rate of relapse and to achieve durable anti-cancer therapy. TGF-β is well-known for its immunosuppressive activity, and it takes critical roles in promotion of all tumor hallmarks. Bispecific anti-PD-(L)1/TGF-β inhibitors reinvigorate effector activity of CD8+ T and natural killer (NK) cells, hamper regulatory T cell (Treg) expansion, and increase the density of anti-tumor type 1 macrophages (M1). Responses to the bispecific approach are higher compared with solo anti-PD-(L)1 or TGF-β targeted therapy, and are seemingly more pronounced in human papillomavirus (HPV)+ patients. High expression of PD-L1 or immune-excluded phenotype in a tumor can also be markers of better response to the bispecific strategy. Besides, anti-PD-(L)1/TGF-β inhibitor therapy can be used safely with other therapeutic modalities including vaccination, radiation and chemotherapy.
Collapse
Affiliation(s)
- Zana Karami
- Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
13
|
Zhang YC, Zhang YT, Wang Y, Zhao Y, He LJ. What role does PDL1 play in EMT changes in tumors and fibrosis? Front Immunol 2023; 14:1226038. [PMID: 37649487 PMCID: PMC10463740 DOI: 10.3389/fimmu.2023.1226038] [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: 05/20/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Epithelial-mesenchymal transformation (EMT) plays a pivotal role in embryonic development, tissue fibrosis, repair, and tumor invasiveness. Emerging studies have highlighted the close association between EMT and immune checkpoint molecules, particularly programmed cell death ligand 1 (PDL1). PDL1 exerts its influence on EMT through bidirectional regulation. EMT-associated factors, such as YB1, enhance PDL1 expression by directly binding to its promoter. Conversely, PDL1 signaling triggers downstream pathways like PI3K/AKT and MAPK, promoting EMT and facilitating cancer cell migration and invasion. Targeting PDL1 holds promise as a therapeutic strategy for EMT-related diseases, including cancer and fibrosis. Indeed, PDL1 inhibitors, such as pembrolizumab and nivolumab, have shown promising results in clinical trials for various cancers. Recent research has also indicated their potential benefit in fibrosis treatment in reducing fibroblast activation and extracellular matrix deposition, thereby addressing fibrosis. In this review, we examine the multifaceted role of PDL1 in immunomodulation, growth, and fibrosis promotion. We discuss the challenges, mechanisms, and clinical observations related to PDL1, including the limitations of the PD1/PDL1 axis in treatment and PD1-independent intrinsic PDL1 signaling. Our study highlights the dynamic changes in PDL1 expression during the EMT process across various tumor types. Through interplay between PDL1 and EMT, we uncover co-directional alterations, regulatory pathways, and diverse changes resulting from PDL1 intervention in oncology. Additionally, our findings emphasize the dual role of PDL1 in promoting fibrosis and modulating immune responses across multiple diseases, with potential implications for therapeutic approaches. We particularly investigate the therapeutic potential of targeting PDL1 in type II EMT fibrosis: strike balance between fibrosis modulation and immune response regulation. This analysis provides valuable insights into the multifaceted functions of PDL1 and contributes to our understanding of its complex mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Yun-Chao Zhang
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yu-Ting Zhang
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yi Wang
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ya Zhao
- Department of Medical Microbiology and Parasitology, Fourth Military Medical University, Xi'an, China
| | - Li-Jie He
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
14
|
Zhang Y, Li C, Du K, Pengkhun N, Huang Z, Gong M, Li Y, Liu X, Li L, Wang D, Wang C, Chen F, Li J. Comparative analysis of immune checkpoint inhibitors in first-line treatment of esophageal squamous cell carcinoma: a network meta-analysis. Immunotherapy 2023. [PMID: 37139963 DOI: 10.2217/imt-2022-0236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Aim: To evaluate the efficacy and safety of first-line immunochemotherapy in the treatment of advanced esophageal squamous cell carcinoma (CRD42021287033). Methods: PubMed, Embase, Cochrane Library and Web of Science were systematically searched to obtain randomized controlled trials, and the outcome indicators of the reports were compared and analyzed. Results: A total of 3163 patients from five reported randomized controlled trials were included in the meta-analysis. The results showed the comprehensive benefits of toripalimab combined with chemotherapy, in terms of overall survival (hazard ratio: 0.59; 95% CI: 0.43-0.81) and progression-free survival (hazard ratio: 0.58; 95% CI: 0.46-0.73). Conclusion: Toripalimab combined with chemotherapy may be a better choice for first-line immunochemotherapy, although this needs to be verified by clinical studies.
Collapse
Affiliation(s)
- Yangfeng Zhang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
- TCM Integrated Cancer Center, Southern Medical University, Guangzhou, 510315, China
| | - Chunhui Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
- TCM Integrated Cancer Center, Southern Medical University, Guangzhou, 510315, China
| | - Kunpeng Du
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Nov Pengkhun
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Zijian Huang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Min Gong
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Yanyang Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Xiang Liu
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Lilin Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Duanyu Wang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Changqian Wang
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| | - Fengsheng Chen
- TCM Integrated Cancer Center, Southern Medical University, Guangzhou, 510315, China
| | - Jiqiang Li
- Department of Radiotherapy, Oncology Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510285, China
| |
Collapse
|
15
|
Kouzu K, Kajiwara Y, Tsujimoto H, Mochizuki S, Okamoto K, Shinto E, Kishi Y, Matsukuma S, Ueno H. Prognostic impact of desmoplastic reaction in esophageal squamous cell carcinoma patients with neoadjuvant therapy. Esophagus 2023:10.1007/s10388-023-00996-z. [PMID: 36917327 DOI: 10.1007/s10388-023-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/03/2023] [Indexed: 03/16/2023]
Abstract
AIM This study aimed to examine the prognostic value of desmoplastic reaction (DR) in esophageal squamous cell carcinoma (ESCC), particularly in patients who received neoadjuvant therapy, such as chemotherapy (NAC) or chemoradiotherapy (NACRT). METHOD In total, 153 patients with pStage II/III ESCC were included in this study. Ninety-one patients received neoadjuvant therapy (NAC, 70; NACRT, 21). Patients were classified according to three DR categories based on the presence of keloid-like collagen and/or myxoid stroma. RESULTS In total, 50, 50, and 53 patients were classified as having mature, intermediate, and immature DR, respectively. The weighted kappa coefficient was 0.623 in the patients with preoperative treatments and 0.782, in those without. The 5-year disease-specific survival (DSS) rates in patients with intermediate/immature DR was significantly worse than those with mature DR (40.7% vs. 73.3%, p < 0.001). Similarly, the 5-year DSS rate in patients with intermediate/immature DR was significantly worse than those with mature DR in a study of patients who received neoadjuvant therapy (46.7% vs. 71.2%, p = 0.009). Multivariate analysis revealed that DR (hazard ratio [HR]: 3.15, 95% confidence interval [CI] 1.58-6.27, p = 0.001), along with N factors, was an independent risk factor for DSS. Moreover, multivariate analysis of patients who received neoadjuvant therapy revealed only DR (HR: 2.47, 95% CI 1.02-5.96, p = 0.045) as independent risk factors for DSS. CONCLUSION The DR classification was a valuable prognostic factor not only in the ESCC patients without neoadjuvant therapy but also in those with neoadjuvant therapy.
Collapse
Affiliation(s)
- Keita Kouzu
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Yoshiki Kajiwara
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan.
| | - Hironori Tsujimoto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Satsuki Mochizuki
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Koichi Okamoto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Eiji Shinto
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| | - Susumu Matsukuma
- Department of Pathology and Laboratory Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, Japan
| |
Collapse
|
16
|
Effect of BIM expression on the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Sci Rep 2023; 13:3943. [PMID: 36894581 PMCID: PMC9998621 DOI: 10.1038/s41598-023-30565-4] [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: 04/05/2022] [Accepted: 02/25/2023] [Indexed: 03/11/2023] Open
Abstract
The role of Programmed Cell Death Ligand 1 (PD-L1) expression in predicting epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKIs) efficacy remains controversial. Recent studies have highlighted that tumor-intrinsic PD-L1 signaling can be modulated by STAT3, AKT, MET oncogenic pathway, epithelial-mesenchymal transition, or BIM expression. This study aimed to investigate whether these underlying mechanisms affect the prognostic role of PD-L1. We retrospectively enrolled patients with EGFR mutant advanced stage NSCLC who received first-line EGFR-TKI between January 2017 and June 2019, the treatment efficacy of EGFR-TKI was assessed. Kaplan-Meier analysis of progression-free survival (PFS) revealed that patients with high BIM expression had shorter PFS, regardless of PD-L1 expression. This result was also supported by the COX proportional hazard regression analysis. In vitro, we further proved that the knockdown of BIM, instead of PDL1, induced more cell apoptosis following gefitinib treatment. Our data suggest that among the pathways affecting tumor-intrinsic PD-L1 signaling, BIM is potentially the underlying mechanism that affects the role of PD-L1 expression in predicting response to EGFR TKI and mediates cell apoptosis under treatment with gefitinib in EGFR-mutant NSCLC. Further prospective studies are required to validate these results.
Collapse
|
17
|
Haghshenas MR, Ghaderi H, Daneste H, Ghaderi A. Immunological and biological dissection of normal and tumoral salivary glands. Int Rev Immunol 2023; 42:139-155. [PMID: 34378486 DOI: 10.1080/08830185.2021.1958806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Salivary glands naturally play central roles in oral immunity. The salivary glands microenvironment inevitable may be exposed to exogenous factors consequently triggering the initiation and formation of various malignant and benign tumors. Mesenchymal stem cells are recruited into salivary gland microenvironment, interact with tumor cells, and induce inhibitory cytokines as well as cells with immunosuppressive phenotypes such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). The immune components and tumor immune responses in malignant and benign SGTs are still under investigation. Immune responses may directly play a limiting role in tumor growth and expansion, or may participate in formation of a rich milieu for tumor growth in cooperation with other cellular and regulatory molecules. Immune checkpoint molecules (e.g. PDLs, HLA-G and LAG3) are frequently expressed on tumor cells and/or tumor-infiltrating lymphocytes (TILs) in salivary gland microenvironment, and an increase in their expression is associated with T cell exhaustion, immune tolerance and tumor immune escape. Chemokines and chemokine receptors have influential roles on aggressive behaviors of SGTs, and thereby they could be candidate targets for cancer immunotherapy. To present a broad knowledge on salivary glands, this review first provides a brief description on immunological functions of normal salivary glands, and then describe the SGT's tumor microenvironment, by focusing on mesenchymal stem cells, immune cell subsets, immune checkpoint molecules, chemokines and chemokine receptors, and finally introduces immune checkpoint inhibitors as well as potential targets for cancer therapy.
Collapse
Affiliation(s)
- Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hossein Daneste
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
Goto Y, Tajiri K, Tanaka S, Murayama A, Muraishi N, Hayashi Y, Yasuda I. A ruptured sarcomatoid hepatocellular carcinoma treated with combined immunotherapy. Clin J Gastroenterol 2023; 16:244-249. [PMID: 36625992 DOI: 10.1007/s12328-023-01758-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/06/2023] [Indexed: 01/11/2023]
Abstract
Sarcomatoid hepatocellular carcinoma (sHCC) is a rare phenotype of HCC with extremely poor prognosis and no established pharmacological treatment. Interventional therapies such as radiofrequency ablation (RFA) or transcatheter arterial embolization (TAE) have been shown to limit the development of sHCC through mechanisms involving hypoxia-induced epithelial-mesenchymal transition. This report describes an 83-year-old man who developed sHCC 2 years after RFA treatment for HCC and experienced sHCC rupture. Following TAE-induced hematostasis, he was administered lenvatinib for tumor control. Although his physical status had improved, due to loss of fever and attenuation of arterial enhancement in the tumor, for 1 month after lenvatinib administration, tumor re-growth was observed 2 months after lenvatinib treatment. His general condition was preserved, and he was treated with 10 courses of atezolizumab plus bevacizumab (Atez+Bev), resulting in tumor shrinkage that was maintained for 3-8 months after Atez+Bev. Findings in this patient showed that combined immunotherapy was effective for sHCC. Further investigation in additional patients is required to maximize prognosis in patients with sHCC.
Collapse
Affiliation(s)
- Yuno Goto
- The Third Department of Internal Medicine, Toyama University Hospital, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kazuto Tajiri
- The Third Department of Internal Medicine, Toyama University Hospital, 2630 Sugitani, Toyama, 930-0194, Japan.
| | | | - Aiko Murayama
- The Third Department of Internal Medicine, Toyama University Hospital, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Nozomu Muraishi
- The Third Department of Internal Medicine, Toyama University Hospital, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Yuka Hayashi
- The Third Department of Internal Medicine, Toyama University Hospital, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Ichiro Yasuda
- The Third Department of Internal Medicine, Toyama University Hospital, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
19
|
Li Q, Liu T, Ding Z. Neoadjuvant immunotherapy for resectable esophageal cancer: A review. Front Immunol 2022; 13:1051841. [PMID: 36569908 PMCID: PMC9773255 DOI: 10.3389/fimmu.2022.1051841] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common cancers worldwide, especially in China. Despite therapeutic advances, the 5-year survival rate of EC is still dismal. For patients with resectable disease, neoadjuvant chemoradiotherapy (nCRT) in combination with esophagectomy is the mainstay of treatment. However, the pathological complete response (pCR) rate to nCRT of 29.2% to 43.2% is not satisfactory, and approximately half of the patients will develop either a locoregional recurrence or distant metastasis. It is, therefore, necessary to explore novel and effective treatment strategies to improve the clinical efficacy of treatment. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) has significantly changed the treatment paradigm for a wide variety of advanced cancers, including EC. More recently, increasing clinical evidence has demonstrated that neoadjuvant immunotherapy can potentially improve the survival of patients with resectable cancers. Furthermore, accumulating findings support the idea that chemotherapy and/or radiotherapy can activate the immune system through a variety of mechanisms, so a combination of chemotherapy and/or radiotherapy with immunotherapy can have a synergistic antitumor effect. Therefore, it is reasonable to evaluate the role of neoadjuvant immunotherapy for patients with surgically resectable EC. In this review, we discuss the rationale for neoadjuvant immunotherapy in patients with EC, summarize the current results of utilizing this strategy, review the planned and ongoing studies, and highlight the challenges and future research needs.
Collapse
Affiliation(s)
| | | | - Zhenyu Ding
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Peng M, Fan S, Li J, Zhou X, Liao Q, Tang F, Liu W. Programmed death-ligand 1 signaling and expression are reversible by lycopene via PI3K/AKT and Raf/MEK/ERK pathways in tongue squamous cell carcinoma. GENES & NUTRITION 2022; 17:3. [PMID: 35164673 PMCID: PMC8903509 DOI: 10.1186/s12263-022-00705-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/24/2022] [Indexed: 11/25/2022]
Abstract
Background Cancer therapy targeting programmed death receptor-1 (PD-1 or CD279) or programmed death-ligand 1 (PD-L1 or CD274) gives hope to Tongue Squamous Cell Carcinoma (TSCC) treatment. However, the tumor-intrinsic mechanism of PD-L1 is not fully elucidated in TSCC. On the other hand, lycopene showed antitumor effects and chemotherapy/radiotherapy-enhancing effects by mechanisms closely correlated with PD-L1. Purpose We aimed to explore whether the mechanisms of PD-L1 signaling and regulation are reversible by lycopene treatment in TSCC. Methods We collected TSCC tissues and normal tissues for assessment of PD-L1 expression by immunohistochemical technique and western blotting. We measured the expression of PD-L1 in three TSCC cell lines and constructed cell lines with knockdown and overexpression of PD-L1. Then, we measured the proliferation by CCK-8 assay, migration and invasion by Transwell assay, and apoptosis by TUNEL assay in five groups with treatment of blank control, negative control with vector transfection, PD-L1 knockdown/overexpression, 4 μM lycopene, and combined 4 μM lycopene and PD-L1 knockdown/overexpression. We also systematically analyzed the PD-L1 constitutive signaling pathways and their effect EMT pathways. In order to bring out the mechanism underlying PI3K/AKT depressing Raf/MEK/ERK, we used PI3K inhibitor LY294002. Results We detected significant PD-L1 upregulation in biopsies by western blot and immunohistochemistry. Our study demonstrated that PD-L1 upregulation elevated IGF-1R to activate the PI3K/AKT pathway but inactivated the Raf/MEK/ERK pathway in TSCC cell line CAL27, while PD-L1 knockdown decreased IGF-1R to inactivate both PI3K/AKT and Raf/MEK/ERK pathways in cell line SCC9, to increase/decrease p-FOXOs and decrease/increase p-GSK-3β, producing further changes in EMT, proliferation, migration, invasion, and apoptosis. Lycopene reversed PD-L1 signaling and expression by mechanisms opposite to PD-L1 upregulation but similar to PD-L1 knockdown. Conclusion Taken together, this study firstly confirmed PD-L1 expression and signaling are reversible by lycopene via PI3K/AKT and Raf/MEK/ERK pathways in TSCC. Our study provides a sounder basis for comprehending PD-L1 signaling and expression and prevention and treatment of TSCC.
Collapse
|
21
|
Burger GA, Nesenberend DN, Lems CM, Hille SC, Beltman JB. Bidirectional crosstalk between epithelial-mesenchymal plasticity and IFN γ-induced PD-L1 expression promotes tumour progression. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220186. [PMID: 36397970 PMCID: PMC9626257 DOI: 10.1098/rsos.220186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 10/05/2022] [Indexed: 06/16/2023]
Abstract
Epithelial-mesenchymal transition (EMT) and immunoevasion through upregulation of programmed death-ligand 1 (PD-L1) are important drivers of cancer progression. While EMT has been proposed to facilitate PD-L1-mediated immunosuppression, molecular mechanisms of their interaction remain obscure. Here, we provide insight into these mechanisms by proposing a mathematical model that describes the crosstalk between EMT and interferon gamma (IFNγ)-induced PD-L1 expression. Our model shows that via interaction with microRNA-200 (miR-200), the multi-stability of the EMT regulatory circuit is mirrored in PD-L1 levels, which are further amplified by IFNγ stimulation. This IFNγ-mediated effect is most prominent for cells in a fully mesenchymal state and less strong for those in an epithelial or partially mesenchymal state. In addition, bidirectional crosstalk between miR-200 and PD-L1 implies that IFNγ stimulation allows cells to undergo EMT for lower amounts of inducing signal, and the presence of IFNγ accelerates EMT and decelerates mesenchymal-epithelial transition (MET). Overall, our model agrees with published findings and provides insight into possible mechanisms behind EMT-mediated immune evasion, and primary, adaptive, or acquired resistance to immunotherapy. Our model can be used as a starting point to explore additional crosstalk mechanisms, as an improved understanding of these mechanisms is indispensable for developing better diagnostic and therapeutic options for cancer patients.
Collapse
Affiliation(s)
- Gerhard A. Burger
- Division of Drug Discovery and Safety, Leiden University, Leiden, The Netherlands
| | - Daphne N. Nesenberend
- Division of Drug Discovery and Safety, Leiden University, Leiden, The Netherlands
- Mathematical Institute, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Carlijn M. Lems
- Division of Drug Discovery and Safety, Leiden University, Leiden, The Netherlands
| | - Sander C. Hille
- Mathematical Institute, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Joost B. Beltman
- Division of Drug Discovery and Safety, Leiden University, Leiden, The Netherlands
| |
Collapse
|
22
|
Secinti IE, Ozgur T, Dede I. PD-L1 Expression in Colorectal Adenocarcinoma Is Associated With the Tumor Immune Microenvironment and Epithelial-Mesenchymal Transition. Am J Clin Pathol 2022; 158:506-515. [PMID: 35938631 DOI: 10.1093/ajcp/aqac077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 05/18/2022] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES Colorectal carcinomas are the third-most common tumors in the world, and colorectal cancer ranks second in cancer-related deaths. Our aim in this study was to investigate the correlation between programmed cell death ligand 1 (PD-L1) expression and clinicopathologic parameters in colorectal carcinomas and their relationship to the tumor immune microenvironment, epithelial-mesenchymal transition (EMT), and microsatellite instability. We also investigated the predictive and prognostic role of PD-L1. METHODS One hundred patients with a diagnosis of colorectal adenocarcinoma who did not receive neoadjuvant therapy were included in the study. The relationships among the altered expression of PD-L1; vimentin; E-cadherin; mismatch repair status; and pathologic microenvironmental features, including the presence of tumor budding and CD8-positive tumor infiltrating lymphocytes (TILs), were assessed. RESULTS Increased PD-L1 expression in tumor cells was associated with increased TILs (P = .013), high histologic grade (P = .011), advanced pathologic T stage (P = .007), lymph node metastasis (P = .002), distant metastasis (P < .001), perineural invasion (P = .009), high bud score (P = .023), EMT (P < .001), and shorter disease-free survival (P = .029). CONCLUSIONS Overall, PD-L1 expression in colorectal carcinoma tumor cells is a marker of poor prognosis, and the positive correlation detected between EMT status and PD-L1 expression suggests that patients with the mesenchymal phenotype may be more likely to benefit from programmed cell death 1 protein/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Ilke Evrim Secinti
- Department of Pathology, School of Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Tumay Ozgur
- Department of Pathology, School of Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| | - Isa Dede
- Department of Medical Oncology, School of Medicine, Hatay Mustafa Kemal University, Hatay, Turkey
| |
Collapse
|
23
|
To N, Evans RPT, Pearce H, Kamarajah SK, Moss P, Griffiths EA. Current and Future Immunotherapy-Based Treatments for Oesophageal Cancers. Cancers (Basel) 2022; 14:3104. [PMID: 35804876 PMCID: PMC9265112 DOI: 10.3390/cancers14133104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/18/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
Oesophageal cancer is a disease that causes significant morbidity and mortality worldwide, and the prognosis of this condition has hardly improved in the past few years. Standard treatment includes a combination of chemotherapy, radiotherapy and surgery; however, only a proportion of patients go on to treatment intended to cure the disease due to the late presentation of this disease. New treatment options are of utmost importance, and immunotherapy is a new option that has the potential to transform the landscape of this disease. This treatment is developed to act on the changes within the immune system caused by cancer, including checkpoint inhibitors, which have recently shown great promise in the treatment of this disease and have recently been included in the adjuvant treatment of oesophageal cancer in many countries worldwide. This review will outline the mechanisms by which cancer evades the immune system in those diagnosed with oesophageal cancer and will summarize current and ongoing trials that focus on the use of our own immune system to combat disease.
Collapse
Affiliation(s)
- Natalie To
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Richard P. T. Evans
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Sivesh K. Kamarajah
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2SY, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.P.); (P.M.)
| | - Ewen A. Griffiths
- Department of Upper Gastrointestinal Surgery, Queen Elizabeth Hospital Birmingham, University Hospitals Birmingham NHS Trust, Birmingham B15 2GW, UK; (N.T.); (R.P.T.E.); (S.K.K.)
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2SY, UK
| |
Collapse
|
24
|
Hassen G, Kasar A, Jain N, Berry S, Dave J, Zouetr M, Priyanka Ganapathiraju VLN, Kurapati T, Oshai S, Saad M, Pathan J, Kamat S, Tirupathi R, Patel UK, Rana RK. Programmed Death-Ligand 1 (PD-L1) Positivity and Factors Associated with Poor Prognosis in Patients with Gastric Cancer: An Umbrella Meta-Analysis. Cureus 2022; 14:e23845. [PMID: 35530821 PMCID: PMC9076041 DOI: 10.7759/cureus.23845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignancies throughout the world with late diagnosis and poor prognosis. The expression of programmed death-ligand 1 (PD-L1) in GC is attributed to immune evasion and tumor progression. PD-L1 positivity has both predictive and prognostic biomarker potential. Aiming to summarize a large amount of research and to provide a definitive conclusion to the conflicting results on the prognostic significance of PD-L1 expression in GC, we performed an umbrella review based on existing meta-analyses which were published recently (2016-2021) and indexed in the PubMed database. Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline was used in August 2021 to screen articles, and data extraction with quality assessment was performed on the selected meta-analyses. Review Manager (RevMan) 5.3 software was used to analyze the HR and OR with a 95% confidence interval (CI) among PD-L1 positive GC patients. We also assessed the between-study heterogeneity (I 2). Forest and Funnel plots were obtained, and a P-value of <0.05 was considered statistically significant. A total of 567 articles were screened, and we selected three meta-analyses with a total of 40 studies conducted over a period of 14 years. In our umbrella review, a total of 8,419 GC patients with an average PD-L1 positivity of 39% were analyzed. We found that PD-L1 positivity in GC patients is associated with poor prognosis (pooled HR =1.44, 95% CI: 1.24-1.68, P<0.00001) having higher mortality reducing the chances of overall survival (OS). However, there are no significant differences in PD-L1 expression among different lymph node (LN) metastases (OR=1.31, 95% CI: 0.98-1.74, P=0.07) and tumor, node, and metastasis (TNM) stages (OR=1.13, 95% CI: 0.80-1.58, P=0.50). Early identification of PD-L1 expression may help tailor cost-effective and targeted immunotherapy among GC patients. More research is needed to further understand how PD-L1 affects LN metastasis and tumor invasion.
Collapse
Affiliation(s)
- Gashaw Hassen
- Progressive Care, Mercy Medical Center, Baltimore, USA.,Medicine and Surgery, Parma University, Parma, ITA.,Medicine, Addis Ababa University, Addis Ababa, ETH
| | - Amita Kasar
- Internal Medicine, Krishna Institute of Medical Sciences, Secunderabad, IND
| | - Nidhi Jain
- Medicine and Surgery, Himalayan Institute of Medical Sciences, Dehradun, IND.,Hematology and Oncology, Brooklyn Cancer Care, Brooklyn, USA.,Internal Medicine, Sir Ganga Ram Hospital, New Delhi, IND
| | - Shivankshi Berry
- Internal Medicine, Dayanand Medical College and Hospital, Ludhiana, IND.,Internal Medicine, Nassau University Medical Center, New York, USA
| | - Jhanvi Dave
- Internal Medicine, B.J. Medical College, Ahmedabad, IND
| | - Michlene Zouetr
- Family Medicine, American Institute of Antigua College of Medicine, St John's, ATG
| | | | | | - Stephanie Oshai
- Medicine and Surgery, College of Medicine, University of Lagos, Lagos, NGA
| | - Mohamed Saad
- Gastroenterology, Theodor Bilharz Research Institute, Giza, EGY
| | | | | | | | - Urvish K Patel
- Public Health and Neurology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Rishabh K Rana
- Preventive and Social Medicine/Community Medicine, Shahid Nirmal Mahto Medical College, Dhanbad, Dhanbad, IND
| |
Collapse
|
25
|
Ge Y, Liu H, Zhang Y, Liu J, Yan R, Xiao Z, Fan X, Huang X, An G. Inhibition of DCLK1 kinase reverses epithelial-mesenchymal transition and restores T-cell activity in pancreatic ductal adenocarcinoma. Transl Oncol 2022; 17:101317. [PMID: 34998236 PMCID: PMC8739467 DOI: 10.1016/j.tranon.2021.101317] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/20/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy has recently become a promising cancer therapy with extensive applications of immune checkpoint inhibitors (ICIs). However, pancreatic ductal adenocarcinoma (PDAC) appears to be unresponsive to immunotherapy due to the immunosuppressive microenvironment. Recent studies showed that cancer stem cell marker DCLK1 promoted the initiation and development of PDAC. Nevertheless, the mechanism driving this process remains unclear. Here, by performing gain-of-function investigations in PDAC cell lines, we demonstrate that both DCLK1 long (DCLK1-iso1, DCLK1-AS) and short (DCLK1-iso4, DCLK1-BL) isoforms can efficiently activate EMT leading to tumor migration and invasion. Consistent with experiments in vitro, bioinformatic analysis demonstrates that DCLK1 may act as a driver of EMT activation in PDAC. Further analysis showed that EMT was associated with an immunosuppressive microenvironment, which includes more immunosuppressive cells and chemokines, and patients with a higher EMT score were less sensitive to immune checkpoint inhibitors according to the TIDE (Tumor Immune Dysfunction and Exclusion) algorithm. Multiplexed immunofluorescence results demonstrated the close correlation between DCLK1, EMT and immunosuppression in PDAC patients. The findings were further confirmed in vivo reflected by decreased CD4+, CD8+ T cells and increased M2 macrophages as well as E-cad loss in DCLK1-overexpressing subcutaneous tumors. Importantly, the highly-specific DCLK1 inhibitor (DCLK1-IN-1) was able to effectively block EMT process and restore T-cell activity. Altogether, our data demonstrate that DCLK1 is strongly associated with tumor immune escape in PDAC and inhibiting DCLK1 kinase activity may be a promising therapeutic modality.
Collapse
Affiliation(s)
- Yang Ge
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Heshu Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuanyuan Zhang
- Department of Oncology, Beijing Huai-Rou Hospital, Beijing 101400, China
| | - Jian Liu
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Rui Yan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zeru Xiao
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiaona Fan
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xuying Huang
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Guangyu An
- Department of Oncology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
26
|
Saleh RR, Scott JL, Meti N, Perlon D, Fazelzad R, Ocana A, Amir E. Prognostic Value of Programmed Death Ligand-1 Expression in Solid Tumors Irrespective of Immunotherapy Exposure: A Systematic Review and Meta-Analysis. Mol Diagn Ther 2022; 26:153-168. [PMID: 35106739 DOI: 10.1007/s40291-022-00576-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND The programmed cell death-1/programmed cell death ligand-1 (PD-L1) pathway, which plays a crucial role in cancer immune surveillance, is the target of several approved immunotherapeutic agents and is used as a predictive biomarker in some solid tumors. However, its use as a prognostic marker (i.e., regardless of therapy used) is not established clearly with available data demonstrating inconsistent prognostic impact of PD-L1 expression in solid tumors. METHODS We conducted a systematic literature search of electronic databases and identified publications exploring the effect of PD-L1 expression on overall survival and/or disease-free survival. Hazard ratios were pooled in a meta-analysis using generic inverse-variance and random-effects modeling. We used the Deeks method to explore subgroup differences based on disease site, stage of disease, and method of PD-L1 quantification. RESULTS One hundred and eighty-six studies met the inclusion criteria. Programmed cell death ligand-1 expression was associated with worse overall survival (hazard ratio 1.33, 95% confidence interval 1.26-1.39; p < 0.001). There was significant heterogeneity between disease sites (subgroup p = 0.002) with pancreatic, hepatocellular, and genitourinary cancers associated with the highest magnitude of adverse outcomes. Programmed cell death ligand-1 was also associated with worse overall disease-free survival (hazard ratio 1.19, 95% confidence interval 1.09-1.30; p < 0.001). Stage of disease did not significantly affect the results (subgroup p = 0.52), nor did the method of quantification via immunohistochemistry or messenger RNA (subgroup p = 0.70). CONCLUSIONS High expression of PD-L1 is associated with worse survival in solid tumors albeit with significant heterogeneity among tumor types. The effect is consistent in early-stage and metastatic disease and is not sensitive to method of PD-L1 quantification. These data can provide additional information for the counseling of patients with cancer about prognosis.
Collapse
Affiliation(s)
- Ramy R Saleh
- Department of Medical Oncology, McGill University, Montreal, QC, Canada
| | - Jordan L Scott
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Nicholas Meti
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Danielle Perlon
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada
| | - Rouhi Fazelzad
- Information Specialist, Library and Information Services, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Alberto Ocana
- Hospital Clinico San Carlos and Instituto de Investigación Sanitaria San Carlos (IdISSC), and Centro Regional de Investigaciones Biomedicas (CRIB), Centro de Investigación Biomédica en Red Cáncerci (CIBERONC), Universidad Castilla La Mancha (UCLM), Madrid, Spain
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre and the University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
27
|
Wan W, Ao X, Chen Q, Yu Y, Ao L, Xing W, Guo W, Wu X, Pu C, Hu X, Li Z, Yao M, Luo D, Xu X. METTL3/IGF2BP3 axis inhibits tumor immune surveillance by upregulating N 6-methyladenosine modification of PD-L1 mRNA in breast cancer. Mol Cancer 2022; 21:60. [PMID: 35197058 PMCID: PMC8864846 DOI: 10.1186/s12943-021-01447-y] [Citation(s) in RCA: 157] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Continual expression of PD-L1 in tumor cells is critical for tumor immune escape and host T cell exhaustion, however, knowledge on its clinical benefits through inhibition is limited in breast cancer. N6-methyladenosine (m6A) plays a crucial role in multiple biological activities. Our study aimed to investigate the regulatory role of the m6A modification in PD-L1 expression and immune surveillance in breast cancer. METHODS MeRIP-seq and epitranscriptomic microarray identified that PD-L1 is the downstream target of METTL3. MeRIP-qPCR, absolute quantification of m6A modification assay, and RIP-qPCR were used to examine the molecular mechanism underlying METTL3/m6A/IGF2BP3 signaling axis in PD-L1 expression. B-NDG and BALB/c mice were used to construct xenograft tumor models to verify the phenotypes upon METTL3 and IGF2BP3 silencing. In addition, breast cancer tissue microarray was used to analyze the correlation between PD-L1 and METTL3 or IGF2BP3 expression. RESULTS We identified that PD-L1 was a downstream target of METTL3-mediated m6A modification in breast cancer cells. METTL3 knockdown significantly abolished m6A modification and reduced stabilization of PD-L1 mRNA. Additionally, METTL3-mediated PD-L1 mRNA activation was m6A-IGF2BP3-dependent. Moreover, inhibition of METTL3 or IGF2BP3 enhanced anti-tumor immunity through PD-L1-mediated T cell activation, exhaustion, and infiltration both in vitro and in vivo. PD-L1 expression was also positively correlated with METTL3 and IGF2BP3 expression in breast cancer tissues. CONCLUSION Our study suggested that METTL3 could post-transcriptionally upregulate PD-L1 expression in an m6A-IGF2BP3-dependent manner to further promote stabilization of PD-L1 mRNA, which may have important implications for new and efficient therapeutic strategies in the tumor immunotherapy.
Collapse
Affiliation(s)
- Weijun Wan
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.,Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Xiang Ao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Quan Chen
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Yang Yu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Luoquan Ao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Wei Xing
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Wei Guo
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Xiaofeng Wu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Chengxiu Pu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Xueting Hu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Zhan Li
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Mengwei Yao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China
| | - Donglin Luo
- Department of Breast, Thyroid Surgery, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, No. 10, Changjiang Branch Road, Yuzhong District, Chongqing, 400042, China.
| |
Collapse
|
28
|
Takamaru N, Fukuda N, Akita K, Kudoh K, Miyamoto Y. Association of PD‑L1 and ZEB‑1 expression patterns with clinicopathological characteristics and prognosis in oral squamous cell carcinoma. Oncol Lett 2022; 23:75. [DOI: 10.3892/ol.2022.13195] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/13/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Natsumi Takamaru
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8504, Japan
| | - Naoyuki Fukuda
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8504, Japan
| | - Kazuya Akita
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8504, Japan
| | - Keiko Kudoh
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8504, Japan
| | - Youji Miyamoto
- Department of Oral Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima 770‑8504, Japan
| |
Collapse
|
29
|
Sahoo S, Nayak SP, Hari K, Purkait P, Mandal S, Kishore A, Levine H, Jolly MK. Immunosuppressive Traits of the Hybrid Epithelial/Mesenchymal Phenotype. Front Immunol 2022; 12:797261. [PMID: 34975907 PMCID: PMC8714906 DOI: 10.3389/fimmu.2021.797261] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Recent preclinical and clinical data suggests enhanced metastatic fitness of hybrid epithelial/mesenchymal (E/M) phenotypes, but mechanistic details regarding their survival strategies during metastasis remain unclear. Here, we investigate immune-evasive strategies of hybrid E/M states. We construct and simulate the dynamics of a minimalistic regulatory network encompassing the known associations among regulators of EMT (epithelial-mesenchymal transition) and PD-L1, an established immune-suppressor. Our simulations for the network consisting of SLUG, ZEB1, miR-200, CDH1 and PD-L1, integrated with single-cell and bulk RNA-seq data analysis, elucidate that hybrid E/M cells can have high levels of PD-L1, similar to those seen in cells with a full EMT phenotype, thus obviating the need for cancer cells to undergo a full EMT to be immune-evasive. Specifically, in breast cancer, we show the co-existence of hybrid E/M phenotypes, enhanced resistance to anti-estrogen therapy and increased PD-L1 levels. Our results underscore how the emergent dynamics of interconnected regulatory networks can coordinate different axes of cellular fitness during metastasis.
Collapse
Affiliation(s)
- Sarthak Sahoo
- Undergraduate Program, Indian Institute of Science, Bangalore, India.,Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | | | - Kishore Hari
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Prithu Purkait
- Undergraduate Program, Indian Institute of Science, Bangalore, India
| | - Susmita Mandal
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | - Akash Kishore
- Department of Computer Science & Engineering, Sri Sivasubramaniya Nadar (SSN) College of Engineering, Chennai, India
| | - Herbert Levine
- Center for Theoretical Biological Physics, Northeastern University, Boston, MA, United States.,Departments of Physics and Bioengineering, Northeastern University, Boston, MA, United States
| | - Mohit Kumar Jolly
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
30
|
Zhou ZY, Liu SR, Xu LB, Liu C, Zhang R. Clinicopathological and Prognostic Value of Programmed Cell Death 1 Expression in Hepatitis B Virus-related Hepatocellular Carcinoma: A Meta-analysis. J Clin Transl Hepatol 2021; 9:889-897. [PMID: 34966652 PMCID: PMC8666368 DOI: 10.14218/jcth.2021.00056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/07/2021] [Accepted: 04/16/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND AIMS The efficacy of targeted programmed cell death 1/programmed death ligand 1 (PD-1/PD-L1) monoclonal antibodies (mAbs) has been confirmed in many solid malignant tumors. The overexpression of PD-1/PD-L1 serves as a biomarker to predict prognosis and clinical progression. However, the role of PD-1 in patients with hepatitis B virus-related hepatocellular carcinoma (HBV-HCC) remains indeterminate. Given that HBV is the most important cause for HCC, this study aimed to investigate the prognostic and clinicopathological value of PD-1 in HBV-HCC via a meta-analysis. METHODS We searched PubMed, Embase, Scopus, the Cochrane Library, Web of Science and Google Scholar up to January 2021 for studies on the correlation between clinicopathology/prognosis and PD-1 in patients with HBV-HCC. The pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to investigate the prognostic significance of PD-1 expression. The odds ratios (ORs) and 95% CIs were determined to explore the association between PD-1 expression and clinicopathological features. RESULTS Our analysis included seven studies with 658 patients, which showed that high PD-1 expression was statistically correlated with poorer overall survival (HR=2.188, 95% CI: [1.262-3.115], p<0.001) and disease-free survival (HR=2.743, 95% CI: [1.980-3.506], p<0.001). PD-1 overexpression was correlated with multiple tumors (OR=2.268, 95% CI: [1.209-4.257], p=0.011), high level of alpha fetoprotein (AFP; OR=1.495, 95% CI: [1.005-2.223], p=0.047) and advanced Barcelona Clinic Liver Cancer (BCLC) stage (OR=3.738, 95% CI: [2.101-6.651], p<0.001). CONCLUSIONS Our meta-analysis revealed that the high level of PD-1 expression was associated with multiple tumors, high level of AFP and advanced BCLC stage. It significantly predicted a poor prognosis of HBV-HCC, which suggests that anti-PD-1 therapy for HBV-HCC patients is plausible.
Collapse
Affiliation(s)
| | | | | | - Chao Liu
- Correspondence to: Rui Zhang and Chao Liu, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China. ORCID: https://orcid.org/0000-0003-3335-0931 (RZ), https://orcid.org/0000-0001-6659-4372 (CL). Tel: +86-20-34070133, Fax: +86-20-34071091, E-mail: (RZ) or (CL)
| | - Rui Zhang
- Correspondence to: Rui Zhang and Chao Liu, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation and Department of Biliary-Pancreatic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China. ORCID: https://orcid.org/0000-0003-3335-0931 (RZ), https://orcid.org/0000-0001-6659-4372 (CL). Tel: +86-20-34070133, Fax: +86-20-34071091, E-mail: (RZ) or (CL)
| |
Collapse
|
31
|
Xu C, Marelli B, Qi J, Qin G, Yu H, Wang H, Jenkins MH, Lo KM, Lan Y. NHS-IL12 and bintrafusp alfa combination therapy enhances antitumor activity in preclinical cancer models. Transl Oncol 2021; 16:101322. [PMID: 34954456 PMCID: PMC8718653 DOI: 10.1016/j.tranon.2021.101322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/06/2021] [Accepted: 12/17/2021] [Indexed: 01/22/2023] Open
Abstract
Combination therapy induced both adaptive and innate antitumor immunity. Combination therapy significantly enhanced antitumor activity. Combination therapy generated tumor antigen specific immune memory. Bintrafusp alfa mediated the reduction of lung metastases.
Combinatorial immunotherapy approaches are emerging as viable cancer therapeutic strategies for improving patient responses and outcomes. This study investigated whether two such immunotherapies, with complementary mechanisms of action, could enhance antitumor activity in murine tumor models. The immunocytokine NHS-IL12, and surrogate NHS-muIL12, are designed to deliver IL-12 and muIL-12, respectively, to the tumor microenvironment (TME) to activate NK cells and CD8+ T cells and increase their cytotoxic functions. Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domains of the TGF-β receptor II to function as a TGF-β “trap” fused to a human IgG1 antibody blocking PD-L1. With this dual-targeting strategy, BA enhances efficacy over that of monotherapies in preclinical studies. In this study, NHS-muIL12 and BA combination therapy enhanced antitumor activity, prolonged survival, and induced tumor-specific antitumor immunity. This combination therapy increased tumor-specific CD8+ T cells and induced immune profiles, consistent with the activation of both adaptive and innate immune systems. In addition, BA reduced lung metastasis in the 4T1 model. Collectively, these findings could support clinical trials designed to investigate NHS-IL12 and BA combination therapy for patients with advanced solid tumors
Collapse
Affiliation(s)
- Chunxiao Xu
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA.
| | - Bo Marelli
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Jin Qi
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Guozhong Qin
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Huakui Yu
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Hong Wang
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Molly H Jenkins
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Kin-Ming Lo
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA
| | - Yan Lan
- EMD Serono Research and Development Institute, Inc, 45 Middlesex Turnpike, Billerica, MA 01821, USA.
| |
Collapse
|
32
|
Vuletić A, Mirjačić Martinović K, Tišma Miletić N, Zoidakis J, Castellvi-Bel S, Čavić M. Cross-Talk Between Tumor Cells Undergoing Epithelial to Mesenchymal Transition and Natural Killer Cells in Tumor Microenvironment in Colorectal Cancer. Front Cell Dev Biol 2021; 9:750022. [PMID: 34858978 PMCID: PMC8631470 DOI: 10.3389/fcell.2021.750022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/14/2021] [Indexed: 12/19/2022] Open
Abstract
Tumor cells undergoing epithelial to mesenchymal transition (EMT) and immune cells in tumor microenvironment (TME) reciprocally influence each other. Immune cells, by supplying TME with bioactive molecules including cytokines, chemokines, enzymes, metabolites, and by physical interactions with tumor cells via their receptors, represent an important factor that affects EMT. Chronical inflammation in TME favorizes tumor growth and invasiveness and stimulates synthesis of EMT promoting transcription factors. Natural killer (NK) cells, owing to their unique ability to exert cytotoxic function independent of major histocompatibility (MHC)-mediated antigen presentation, play a significant role in the control of metastasis in colorectal cancer (CRC). Although, the cross-talk between immune cells and tumor cells in general favors the induction of EMT and inhibition of antitumor immune responses, there are some changes in the immunogenicity of tumor cells during EMT of CRC cells that increase their susceptibility to NK cell cytotoxic lysis. However, suppressive TME downmodulates the expression of activating NK cell receptors, decreases the expression of activating and increases the expression of inhibitory NK cell ligands on tumor cells, and impairs NK cell metabolism that altogether negatively affects the overall NK cell function. Furthermore, process of EMT is often associated with increased expression of programmed cell death ligand (PD-L) and expression of immune checkpoint molecules PD-1, TIGIT, and TIM3 on functionally exhausted NK cells in TME in CRC. In this review we discuss modalities of cross-talk between tumor cells and NK cells, with regard of EMT-driven changes.
Collapse
Affiliation(s)
- Ana Vuletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | | | - Nevena Tišma Miletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Sergi Castellvi-Bel
- Gastroenterology Department, Hospital Clínic, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomčdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Milena Čavić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Belgrade, Serbia
| |
Collapse
|
33
|
Morelli MB, Amantini C, Rossi de Vermandois JA, Gubbiotti M, Giannantoni A, Mearini E, Maggi F, Nabissi M, Marinelli O, Santoni M, Cimadamore A, Montironi R, Santoni G. Correlation between High PD-L1 and EMT/Invasive Genes Expression and Reduced Recurrence-Free Survival in Blood-Circulating Tumor Cells from Patients with Non-Muscle-Invasive Bladder Cancer. Cancers (Basel) 2021; 13:cancers13235989. [PMID: 34885101 PMCID: PMC8656875 DOI: 10.3390/cancers13235989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND PD-L1 represents a crucial immune checkpoint molecule in the tumor microenvironment, identified as a key target for cancer immunotherapy. A correlation between PD-L1 and EMT-related genes expression in various human cancers has been suggested. METHODS By ScreenCell filtration, digital droplet PCR and confocal microscopy analysis, we aimed to investigate the expression of PD-L1 and EMT/invasive genes (TWIST1, ZEB1, VIMENTIN, TIMP2) in circulating tumor cells (CTCs) collected from the blood of non-muscle-invasive bladder cancer (NMIBC) patients, assessing the prognostic value of these biomarkers in the disease. Welchs' test and Mann-Whitney U test, correlation index, Kaplan-Meier, Univariate and Multivariate Cox hazard proportional analysis were used. RESULTS Higher PD-L1, TIMP2 and VIM mRNA levels were found in pT1 compared to pTa NMIBC. As evaluated by Kaplan-Meier and Univariate and Multivariate Cox analysis, enhancement of PD-L1, TWIST1 and TIMP2 expression reduces the recurrent free survival in NMIBC patients. CONCLUSIONS High PD-L1, TWIST1 and TIMP2 mRNAs mark the recurrent-NMIBC patients and by reducing the RFS represent negative prognostic biomarkers in these patients.
Collapse
Affiliation(s)
- Maria Beatrice Morelli
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy; (F.M.); (M.N.); (O.M.)
- Correspondence: (M.B.M.); (G.S.); Tel.: +39-0737403312 (M.B.M.); +39-0737403319 (G.S.)
| | - Consuelo Amantini
- School of Biosciences and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | | | | | - Antonella Giannantoni
- Department of Medical and Surgical Sciences, University of Siena, 53100 Siena, Italy;
- Neurosciences, Functional and Surgical Urology Unit, Santa Maria alle Scotte Hospital, 53100 Siena, Italy
| | - Ettore Mearini
- Urologic and Andrologic Clinics, University of Perugia, 05100 Perugia, Italy; (J.A.R.d.V.); (E.M.)
| | - Federica Maggi
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy; (F.M.); (M.N.); (O.M.)
- Department of Molecular Medicine, University Sapienza, 00185 Rome, Italy
| | - Massimo Nabissi
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy; (F.M.); (M.N.); (O.M.)
| | - Oliviero Marinelli
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy; (F.M.); (M.N.); (O.M.)
| | - Matteo Santoni
- Oncology Unit, Macerata Hospital, 62100 Macerata, Italy;
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Department of Biomedical Sciences and Public Health School of Medicine, Polytechnic University of Marche Region, Umberto I Hospitals, 60121 Ancona, Italy;
| | - Rodolfo Montironi
- Molecular Medicine and Cell Therapy Foundation, Department of Clinical and Molecular Sciences, Polytechnic University of Marche Region, 60126 Ancona, Italy;
| | - Giorgio Santoni
- School of Pharmacy, Section of Experimental Medicine, University of Camerino, 62032 Camerino, Italy; (F.M.); (M.N.); (O.M.)
- Correspondence: (M.B.M.); (G.S.); Tel.: +39-0737403312 (M.B.M.); +39-0737403319 (G.S.)
| |
Collapse
|
34
|
Yang YL, Li ZQ, Wang QL, Gu JJ, Fang XJ, Huang GH. Efficacy and Safety of Programmed Cell Death 1 Inhibitor Monotherapy Versus Chemotherapy as Second-Line Treatment for Advanced Esophageal Cancer: A Meta-analysis and Systematic Review. Clin Ther 2021; 43:1997-2012. [PMID: 34794831 DOI: 10.1016/j.clinthera.2021.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 08/25/2021] [Accepted: 09/24/2021] [Indexed: 02/09/2023]
Abstract
PURPOSE With programmed cell death 1 (PD-1) inhibitors approved for second-line treatment of advanced esophageal cancer, immunotherapy and chemotherapy have gradually become the main treatments for second-line treatment of patients with advanced esophageal cancer (AEC). This meta-analysis and systematic review were conducted to evaluate the efficacy and safety of PD-1 inhibitors monotherapy versus chemotherapy in second-line treatment of AEC. METHODS Eligible randomized controlled trials were searched in PubMed, Embase, and the Cochrane Library and abstracts presented at the American Society of Clinical Oncology or European Society of Medical Oncology were reviewed to assess the efficacy and tolerability of PD-1/programmed cell death ligand 1 (PD-L1) inhibitors relative to chemotherapy for AEC from January 2016 to October 2020. Patients diagnosed with AEC and progressing after first-line therapy were included in this study. Hazard ratios (HRs) of progression-free survival (PFS) and overall survival (OS), risk ratios (RRs) of objective response rate (ORR), and the odds ratios (ORs) of adverse effects (AEs) were calculated. FINDINGS The study included 4 randomized controlled trials with 1683 patients. The results indicated that PD-1 inhibitors prolonged the OS (HR = 0.79; 95% CI, 0.71-0.88; P < 0.01) and improved the ORR (RR = 3.00; 95% CI, 2.36-3.82; P = 0.01) but did not improve the PFS (HR = 0.96; 95% CI, 0.76-1.20; P = 0.692) compared with chemotherapy in the second-line treatment of AEC. PD-1 inhibitors alone were associated with a lower incidence of all treatment-related AEs (OR = 0.29; 95% CI, 0.09-0.89; P = 0.03) and grade 3 to 5 treatment-related AEs (OR = 0.26; 95% CI, 0.16-0.44; P < 0.01) versus chemotherapy. PD-1 inhibitors prolonged OS mainly in the following patient groups: male, age <65 years, Eastern Cooperative Oncology Group performance status of 1, or PD-L1 tumor proportion score ≥10%. Asian patients had a longer OS than non-Asian patients (P = 0.01). IMPLICATIONS The available evidence indicates that the efficacy and tolerability of PD-1 inhibitors were better than chemotherapy in the second-line treatment of AEC, and the benefiting population of these patients was limited to males, those <65 years of age, those with a Eastern Cooperative Oncology Group performance status of 1, or those with a PD-L1 tumor proportion score ≥10%. Notably, Asian patients receiving immune monotherapy had longer OS than non-Asian patients.
Collapse
Affiliation(s)
- Yong-Liang Yang
- The Second People's Hospital of Lianyungang, The Affiliated Hospital of Bengbu Medical College, Lianyungang, Jiangsu, China
| | - Zhi-Qiang Li
- The Second People's Hospital of Lianyungang, The Affiliated Hospital of Bengbu Medical College, Lianyungang, Jiangsu, China
| | - Qiu-Lu Wang
- The Second People's Hospital of Lianyungang, The Affiliated Hospital of Bengbu Medical College, Lianyungang, Jiangsu, China
| | - Jing-Jing Gu
- The Second People's Hospital of Lianyungang, The Affiliated Hospital of Bengbu Medical College, Lianyungang, Jiangsu, China
| | - Xin-Jian Fang
- The Second People's Hospital of Lianyungang, The Affiliated Hospital of Bengbu Medical College, Lianyungang, Jiangsu, China
| | - Guan-Hong Huang
- The Second People's Hospital of Lianyungang, The Affiliated Hospital of Bengbu Medical College, Lianyungang, Jiangsu, China.
| |
Collapse
|
35
|
He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B 2021; 11:3379-3392. [PMID: 34900524 PMCID: PMC8642427 DOI: 10.1016/j.apsb.2021.03.008] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common cancers with high morbidity and mortality rates. EC includes two histological subtypes, namely esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCC primarily occurs in East Asia, whereas EAC occurs in Western countries. The currently available treatment strategies for EC include surgery, chemotherapy, radiation therapy, molecular targeted therapy, and combinations thereof. However, the prognosis remains poor, and the overall five-year survival rate is very low. Therefore, achieving the goal of effective treatment remains challenging. In this review, we discuss the latest developments in chemotherapy and molecular targeted therapy for EC, and comprehensively analyze the application prospects and existing problems of immunotherapy. Collectively, this review aims to provide a better understanding of the currently available drugs through in-depth analysis, promote the development of new therapeutic agents, and eventually improve the treatment outcomes of patients with EC.
Collapse
Affiliation(s)
- Shiming He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jian Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
36
|
Xia Y, Wang WC, Shen WH, Xu K, Hu YY, Han GH, Liu YB. Thalidomide suppresses angiogenesis and immune evasion via lncRNA FGD5-AS1/miR-454-3p/ZEB1 axis-mediated VEGFA expression and PD-1/PD-L1 checkpoint in NSCLC. Chem Biol Interact 2021; 349:109652. [PMID: 34520751 DOI: 10.1016/j.cbi.2021.109652] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/01/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) accounts for about 80-85% of total lung cancer cases. Identifying the molecular mechanisms of anti-tumor drugs is essential for improving therapeutic effects. Herein, we aim to investigate the role of thalidomide in the tumorigenicity of NSCLC. METHODS The A549 xenograft nude mouse model was established to explore therapeutic effects of thalidomide. The expression of FGD5-AS1 was evaluated in carcinomatous and paracarcinomatous tissues from NSCLC patients as well as NSCLC cell lines. CCK-8 assay was performed to assess cell viability. The invasive capacity was examined using transwell assay. The tube formation assay was applied to determine cell angiogenesis. Flow cytometry was subjected to validate CD8+ T cell activity. The FGD5-AS1/miR-454-3p/ZEB1 regulatory network was analyzed using luciferase reporter, RIP and ChIP assays. RESULTS Thalidomide reduced tumor growth and angiogenesis and increased CD8+ T cell ratio in a mouse model. Enhanced expression of FGD5-AS1 was positively correlated with the poor survival of NSCLC patients. Knockdown of FGD5-AS1 notably suppressed the proliferation, invasion and angiogenesis of cancer cells as well as the apoptosis of CD8+ T cells. Thalidomide targeted FGD5-AS1 to exert its anti-tumor activity in NSCLC. FGD5-AS1 acted as a sponge of miR-454-3p to upregulate ZEB1, thus increasing the expression of PD-L1 and VEGFA. Simultaneous overexpression of FGD5-AS1 and silencing of miR-454-3p reversed thalidomide-mediated anti-tumor effects in NSCLC. CONCLUSION Thalidomide inhibits NSCLC angiogenesis and immune evasion via FGD5-AS1/miR-454-3p/ZEB1 axis-mediated regulation of VEGFA expression and PD-1/PD-L1 checkpoint.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, Taizhou Clinical Medical School of Nanjing Medical University; Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, China; Department of Radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Wei-Cheng Wang
- Department of Oncology, Taizhou Clinical Medical School of Nanjing Medical University; Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, China
| | - Wen-Hao Shen
- Department of Oncology, Taizhou Clinical Medical School of Nanjing Medical University; Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, China
| | - Kun Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yang-Yang Hu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Gao-Hua Han
- Department of Oncology, Taizhou Clinical Medical School of Nanjing Medical University; Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, China.
| | - Yong-Biao Liu
- Department of Radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
37
|
Zhou X, Wang F, Wu H, Chen X, Zhang Y, Lin J, Cai Y, Xiang J, He N, Hu Z, Jin X. Thymoquinone Suppresses the Proliferation, Migration and Invasiveness through Regulating ROS, Autophagic Flux and miR-877-5p in Human Bladder Carcinoma Cells. Int J Biol Sci 2021; 17:3456-3475. [PMID: 34512159 PMCID: PMC8416733 DOI: 10.7150/ijbs.60401] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 08/01/2021] [Indexed: 12/13/2022] Open
Abstract
Bladder carcinoma is among the top 10 most frequently diagnosed cancer types in the world. As a phytochemical active metabolic, thymoquinone (TQ) is extracted from seeds of Nigella sativa, possessing various biological properties in a wide range of diseases. Moreover, the outstanding anti-cancer effect of TQ is attracting increasing attentions. In certain circumstances, moderate autophagy is regarded to facilitate the adaptation of malignant cells to different stressors. Conversely, closely linked with the mitochondrial membrane potential (MMP) loss, the upregulation of intracellular reactive oxygen species (ROS) is reported to activate the cell apoptosis in many cancer types. Furthermore, the vital effects of microRNAs in the pathological processes of cancer cells have also been confirmed by previous studies. The present research confirms that TQ restrains the viability, proliferation, migration and invasion through activating caspase-dependent apoptosis in bladder carcinoma cells, which is mediated by TQ induced ROS increase in bladder carcinoma cells. Furthermore, TQ is proved to block the fusion of autophagosomes and lysosomes, causing the accumulation of autophagosomes and subsequent cell apoptosis. In addition, TQ is also found to initiate the miR-877-5p/PD-L1 axis, which suppresses the epithelial mesenchymal transition (EMT) and invasion of bladder carcinoma cells. Taken together, TQ induces the apoptosis through upregulating ROS level and impairing autophagic flux, and inhibiting the EMT and cell invasion via activating the miR-877-5p/PD-L1 axis in bladder carcinoma cells.
Collapse
Affiliation(s)
- Xuejian Zhou
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Feifan Wang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Hongshen Wu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Xianwu Chen
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Yan Zhang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Juntao Lin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Yueshu Cai
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Jiayong Xiang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Ning He
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Zhenghui Hu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| | - Xiaodong Jin
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, PR China
| |
Collapse
|
38
|
Cui H, Li Y, Li S, Liu G. Prognostic Function of Programmed Cell Death-Ligand 1 in Esophageal Squamous Cell Carcinoma Patients Without Preoperative Therapy: A Systematic Review and Meta-Analysis. Front Oncol 2021; 11:693886. [PMID: 34490091 PMCID: PMC8416500 DOI: 10.3389/fonc.2021.693886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/26/2021] [Indexed: 12/24/2022] Open
Abstract
Background Studies investigating the correlation between the expression of programmed cell death-ligand 1 (PD-L1) and prognosis in patients with esophageal squamous cell carcinoma (ESCC) not receiving preoperative therapy have increased significantly, but conclusions remain inconclusive. Therefore, this study aimed to determine the association between clinical outcomes and expression of PD-L1 in ESCC patients without preoperative therapy. Methods We conducted a comprehensive literature search using four databases up to May 2020. Quality assessment was carried out according to the Newcastle-Ottawa Quality Assessment Scale (NOS). Hazard ratios (HRs) were used to analyze the association between PD-L1 expression with prognosis. Furthermore, we evaluated the correlation between PD-L1 and clinicopathological characteristics using odds ratios (ORs) and 95% confidence intervals (CIs). Results Twenty studies (19 publications) comprising 3,677 patients were included in this meta-analysis. We found that the expression of PD-L1 was not related to overall survival (OS, HR: 1.16, 95% CI: 0.94-1.42, p = 0.16) or disease-free survival (DFS, HR: 0.85, 95% CI: 0.66-1.10, p = 0.21) in ESCC. Furthermore, although PD-L1 expression was not significantly associated with sex, degree of differentiation, TNM stage, T stage, lymph node status, smoking, or alcohol use, the merged OR demonstrated that the expression of PD-L1 was higher in older patients compared to younger patients (OR: 1.40, 95% CI: 1.07-1.83, p = 0.01). No obvious publication bias was observed. Conclusions Our present study illustrated that PD-L1 expression was not related to poor prognosis of ESCC patients not receiving preoperative therapy, albeit the association only showed a tendency for statistical significance. Notably, PD-L1 expression showed a significant association with age. This meta-analysis had several limitations; therefore, our results need to be verified through further large-scale and prospective studies.
Collapse
Affiliation(s)
- Hongxia Cui
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Yarong Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Su Li
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Guangxuan Liu
- Department of Pharmacy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| |
Collapse
|
39
|
Huang TC, Liang CW, Li YI, Guo JC, Lin CC, Chen YJ, Cheng AL, Hsu CH. Prognostic value of PD-L1 expression on immune cells or tumor cells for locally advanced esophageal squamous cell carcinoma in patients treated with neoadjuvant chemoradiotherapy. J Cancer Res Clin Oncol 2021; 148:1803-1811. [PMID: 34432128 DOI: 10.1007/s00432-021-03772-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/17/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE Programmed death-ligand 1 (PD-L1) expression may influence the prognosis of patients with localized esophageal cancer. The current study compared the prognostic value of PD-L1 expression between tumor cells and immune cells. METHODS Archival esophageal tumor tissue samples were collected from patients who received paclitaxel and cisplatin-based neoadjuvant chemoradiotherapy (CRT) for locally advanced esophageal squamous cell carcinoma (ESCC) in three prospective phase II trials. PD-L1 expression on tumor and immune cells was examined immunohistochemically by using the SP142 antibody and scored by two independent pathologists. The association of PD-L1 expression with patient's outcomes was analyzed using a log-rank test and Cox regression multivariate analysis. RESULTS A total of 100 patients were included. PD-L1 expression on tumor cells was positive (≥ 1%, TC-positive) in 55 patients; PD-L1 expression on immune cells was high (≥ 5%, IC-high) in 30 patients. TC-positive status was associated with poor overall survival (OS) (HR: 1.63, P = 0.035), whereas IC-high status was associated with improved OS (HR: 0.44, P = 0.0024). Multivariate analysis revealed that TC-positive, IC-high, and performance status were independent prognostic factors for progression-free survival and that IC-high and performance status were independent factors for OS. Furthermore, the combination of IC-high and TC-negative status was associated with the optimal OS, whereas that of TC-positive and IC-low status was associated with the worst OS. CONCLUSION PD-L1 expression on tumor and immune cells may have different prognostic value for patients with locally advanced ESCC receiving neoadjuvant CRT. A combination of these two indexes may further improve the prognostic prediction.
Collapse
Affiliation(s)
- Ta-Chen Huang
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Cher-Wei Liang
- Department of Pathology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Yu-I Li
- Department of Pathology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Jhe-Cyuan Guo
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chia-Chi Lin
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ya-Jhen Chen
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ann-Lii Cheng
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. .,Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan. .,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Chih-Hung Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan. .,National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
40
|
Gong H, Li B. Guidelines for Radiotherapy of Esophageal Carcinoma (2020 Edition). PRECISION RADIATION ONCOLOGY 2021. [DOI: 10.1002/pro6.1119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
41
|
Liu Z, Sun L, Cai L, Guo M, Xu G, Liu S, Zheng G, Wang Q, Lian X, Feng F, Zhang H. Clinicopathological and prognostic values of PD-L1 expression in oesophageal squamous cell carcinoma: a meta-analysis of 31 studies with 5368 patients. Postgrad Med J 2021; 98:948-957. [PMID: 34253568 DOI: 10.1136/postgradmedj-2021-140029] [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: 02/26/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 11/03/2022]
Abstract
Several immune checkpoint inhibitors targeting programmed death ligand 1 (PD-L1)/programmed death 1 have successfully improved the prognosis of oesophageal squamous cell carcinoma (ESCC) with approval in certain countries. However, whether the expression of PD-L1 is associated with the degree of benefit is unclear yet and a unified standard of antibody and cut-off value of PD-L1 detection is also lacking. The current meta-analysis then aimed to explore the association between PD-L1 expression and clinicopathological features as well as prognosis in ESCC.A systematic search on PubMed, Embase, Cochrane Library and Web of Science databases was performed up to 30 March 2021. The correlation between PD-L1 expression and clinicopathological features, as well as prognosis in ESCC, was estimated with the random-effects model.A total of 5368 patients from 31 retrospective studies were enrolled. The overexpression of PD-L1 was significantly associated with lymph node metastasis (OR 1.342, 95% CI 0.995 to 1.809, p=0.050) and distant metastasis (OR 1.516, 95% CI 1.001 to 2.294, p=0.050). The pooled HR showed that PD-L1 overexpression was significantly correlated with poor overall survival (OS) of patients with ESCC (HR 1.306, 95% CI 1.108 to 1.539, p<0.010) but not disease-free survival (DFS) (HR 1.180, 95% CI 0.937 to 1.487, p=0.160). Heterogeneity decreased significantly in subgroup analyses. The overexpression of PD-L1 was associated with poor DFS at the cut-off point of ≥1% (HR 1.642, 95% CI 1.367 to 1.973, p<0.010; I2=0%) and worse OS at the cut-off point of ≥10% (HR 1.575, 95% CI 1.175 to 2.111, p<0.010; I2=0%).The overexpression of PD-L1 was correlated with lymph node and distant metastasis as well as poor survival of ESCC.
Collapse
Affiliation(s)
- Zhen Liu
- Ddepartment of General Surgery, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China.,Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Li Sun
- Department of Digestive Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Lei Cai
- Department of Digestive Surgery, Xi'an International Medical Center, Xi'an, Shaanxi, People's Republic of China
| | - Man Guo
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Guanghui Xu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Shushang Liu
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Gaozan Zheng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Qiao Wang
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Xiao Lian
- Department of Digestive Surgery, Xi'an International Medical Center, Xi'an, Shaanxi, People's Republic of China
| | - Fan Feng
- Division of Digestive Surgery, Xijing Hospital of Digestive Diseases, The Air Force Medical University, Xi'an, Shaanxi, People's Republic of China
| | - Hongwei Zhang
- Digestive Diseases Center, Wuxi Mingci Hospital, Wuxi, Jiangsu, People's Republic of China
| |
Collapse
|
42
|
Anti-cancer effects of Tranilast: An update. Biomed Pharmacother 2021; 141:111844. [PMID: 34174504 DOI: 10.1016/j.biopha.2021.111844] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/18/2022] Open
Abstract
Tranilast (TRN) or (N-3,4 -dimethoxy cinnamoyl]-anthranilic acid) is an analog of a tryptophan metabolite and is identified mainly as an anti-allergic agent with limited side effects. The anti-cancer effects of tranilast either alone or in combination with chemotherapeutic drugs have been evidenced in several pre-clinical studies. The main mechanism of action of tranilast includes targeting and modulation of various signaling and immune regulatory pathways including Transforming growth factor-beta (TGF-β), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), phosphatidylinositol 3-kinase (PI3K), MAP-Kinase (MAPK), Protein kinase B (Akt/PKB), c-Jun N-terminal kinase, modulation of cancer stem cells, etc. Most of these pathways are involved in tumor proliferation, invasion, and metastasis and it is postulated that tranilast, with its low toxicity profile and high anti-carcinogenic abilities, can serve as a potential anti-tumorigenic agent. The main aim of this review is to provide updated information on the anti-cancer effects of tranilast and its significance as a therapeutic agent.
Collapse
|
43
|
Bornes L, Belthier G, van Rheenen J. Epithelial-to-Mesenchymal Transition in the Light of Plasticity and Hybrid E/M States. J Clin Med 2021; 10:jcm10112403. [PMID: 34072345 PMCID: PMC8197992 DOI: 10.3390/jcm10112403] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/27/2021] [Indexed: 02/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a cellular program which leads to cells losing epithelial features, including cell polarity, cell-cell adhesion and attachment to the basement membrane, while gaining mesenchymal characteristics, such as invasive properties and stemness. This program is involved in embryogenesis, wound healing and cancer progression. Over the years, the role of EMT in cancer progression has been heavily debated, and the requirement of this process in metastasis even has been disputed. In this review, we discuss previous discrepancies in the light of recent findings on EMT, plasticity and hybrid E/M states. Moreover, we highlight various tumor microenvironmental cues and cell intrinsic signaling pathways that induce and sustain EMT programs, plasticity and hybrid E/M states. Lastly, we discuss how recent findings on plasticity, especially on those that enable cells to switch between hybrid E/M states, have changed our understanding on the role of EMT in cancer metastasis, stemness and therapy resistance.
Collapse
|
44
|
Liang Y, Liu Y, Zhang Q, Zhang H, Du J. Tumor-derived extracellular vesicles containing microRNA-1290 promote immune escape of cancer cells through the Grhl2/ZEB1/PD-L1 axis in gastric cancer. Transl Res 2021; 231:102-112. [PMID: 33321257 DOI: 10.1016/j.trsl.2020.12.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) is a highly prevalent malignancy featured by dismal oncological outcomes. Accumulating pieces of evidence have consensus over the therapeutic significance of extracellular vesicles (EVs) and its role in carcinogenesis. Here, we planned to uncover EVs' role in GC by shuttling microRNA-1290 (miR-1290) and to identify the possible molecular mechanism associated with Grhl2, PD-L1, and ZEB1. Grhl2 was under-expressed in GC tissues, exhibiting a negative correlation with PD-L1 expression. In addition, Grhl2 promoted T cell proliferation by down-regulating PD-L1 via inhibiting ZEB1, while miR-1290 was found to negatively regulate Grhl2. EVs were also isolated from GC cells or normal gastric epithelial cells and identified with the presence of EV markers. miR-1290 expression was determined to be enriched in the EVs derived from GC cells and observed to promote the suppressive action of GC cells on T cell activation by up-regulating PD-L1 via the Grhl2/ZEB1 pathway in the co-culture system of GC cells with or without treatment of EVs with T cells. Moreover, we also developed a mouse model of GC and injected the EVs derived from miR-1290-inhibitor-treated GC cells into the tumor-bearing mice for further validation of mechanism in vivo. Intriguingly, the pivotal role of EVs-shuttled miR-1290 as an oncomiR was demonstrated in vivo. Collectively, we found that miR-1290 in EVs secreted from GC cells contributed to immune escape through the Grhl2/ZEB1/PD-L1 axis.
Collapse
Affiliation(s)
- Yuan Liang
- Medical Oncology Department of Thoracic Cancer(2), Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, PR China
| | - Yang Liu
- Department of Pathology, College of Basic Medical Science and The First Affiliated Hospital, China Medical University, Shenyang 110122, PR China
| | - Qingfu Zhang
- Department of Pathology, College of Basic Medical Science and The First Affiliated Hospital, China Medical University, Shenyang 110122, PR China
| | - Heng Zhang
- Department of Pathology, College of Basic Medical Science and The First Affiliated Hospital, China Medical University, Shenyang 110122, PR China
| | - Jiang Du
- Department of Pathology, College of Basic Medical Science and The First Affiliated Hospital, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
45
|
Morisue R, Kojima M, Suzuki T, Nakatsura T, Ojima H, Watanabe R, Sugimoto M, Kobayashi S, Takahashi S, Konishi M, Ishii G, Gotohda N, Fujiwara T, Ochiai A. Sarcomatoid hepatocellular carcinoma is distinct from ordinary hepatocellular carcinoma: Clinicopathologic, transcriptomic and immunologic analyses. Int J Cancer 2021; 149:546-560. [PMID: 33662146 DOI: 10.1002/ijc.33545] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 12/14/2022]
Abstract
Sarcomatoid hepatocellular carcinoma (SHCC), which was a rare histological subtype of hepatocellular carcinoma (HCC), is currently subclassified as poorly differentiated HCC because of insufficient evidence to define SHCC as a subtype of HCC. We aimed to assess the feasibility of classifying SHCC as a histological subtype of HCC by comprehensively identifying novel and distinct characteristics of SHCC compared to ordinary HCC (OHCC). Fifteen SHCCs (1.4%) defined as HCC with at least a 10% sarcomatous component, 15 randomly disease-stage-matched OHCCs and 163 consecutive OHCCs were extracted from 1106 HCCs in the Pathology Database (1997-2019) of our hospital. SHCC patients showed poor prognosis, and the tumors could be histologically subclassified into the pleomorphic, spindle and giant cell types according to the subtype of carcinomas with sarcomatoid or undifferentiated morphology in other organs. The transcriptomic analysis revealed distinct characteristics of SHCC featuring the upregulation of genes associated with epithelial-to-mesenchymal transition and inflammatory responses. The fluorescent multiplex immunohistochemistry results revealed prominent programmed death-ligand 1 (PD-L1) expression on sarcomatoid tumor cells and higher infiltration of CD4+ and CD8+ T cells in SHCCs compared to OHCCs. The density of CD8+ T cells in the nonsarcomatous component of SHCCs was also higher than that in OHCCs. In conclusion, the comprehensive analyses in our study demonstrated that SHCC is distinct from OHCC in terms of clinicopathologic, transcriptomic and immunologic characteristics. Therefore, it is reasonable to consider SHCC as a histological subtype of HCC.
Collapse
Affiliation(s)
- Ryo Morisue
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan.,Department of Gastroenterological Surgery Transplant and Surgical Oncology, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Motohiro Kojima
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Toshihiro Suzuki
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,General Medical Education and Research Center, Teikyo University, Tokyo, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hidenori Ojima
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Reiko Watanabe
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motokazu Sugimoto
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shin Kobayashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shinichiro Takahashi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masaru Konishi
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Division of Pathology, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan.,Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoto Gotohda
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery Transplant and Surgical Oncology, Okayama University Graduate School of Medicine Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsushi Ochiai
- Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
46
|
Wu Y, Sang M, Liu F, Zhang J, Li W, Li Z, Gu L, Zheng Y, Li J, Shan B. Epigenetic modulation combined with PD-1/PD-L1 blockade enhances immunotherapy based on MAGE-A11 antigen-specific CD8+T cells against esophageal carcinoma. Carcinogenesis 2021; 41:894-903. [PMID: 32529260 DOI: 10.1093/carcin/bgaa057] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/15/2020] [Accepted: 06/10/2020] [Indexed: 12/24/2022] Open
Abstract
Cancer testis antigens (CTAs) are promising targets for T cell-based immunotherapy and studies have shown that certain CT genes are epigenetically depressed in cancer cells through DNA demethylation. Melanoma-associated antigen A11 (MAGE-A11) is a CTA that is frequently expressed in esophageal cancer and is correlated with a poor esophageal cancer prognosis. Consequently, MAGE-A11 is a potential immunotherapy target. In this study, we evaluated MAGE-A11 expression in esophageal cancer cells and found that it was downregulated in several tumor cell lines, which restricted the effect of immunotherapy. Additionally, the specific recognition and lytic potential of cytotoxic T lymphocytes (CTLs) derived from the MAGE-A11 was determined. Specific CTLs could kill esophageal cancer cells expressing MAGE-A11 but rarely lysed MAGE-A11-negative tumor cells. Therefore, induction of MAGE-A11 expression is critical for CTLs recognition and lysis of esophageal cancer cells. Treatment with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine increased MAGE-A11 expression in esophageal cancer cells and subsequently enhanced the cytotoxicity of MAGE-A11-specific CD8+T cells against cancer cell lines. Furthermore, we found that PD-L1 expression in esophageal cancer cells affected the antitumor function of CTLs. programmed death-1 (PD-1)/PD-L1 blockade could increase the specific CTL-induced lysis of HLA-A2+/MAGE-A11+ tumor cell lines treated with 5-aza-2'-deoxycytidine. These findings indicate that the treatment of tumor cells with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine augments MAGE-A11 expression in esophageal cancer cells. The combination of epigenetic modulation by 5-aza-2'-deoxycytidine and PD-1/PD-L1 blockade may be useful for T cell-based immunotherapy against esophageal cancer.
Collapse
Affiliation(s)
- Yunyan Wu
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Meixiang Sang
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Institute of Tumor Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Fei Liu
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Jiandong Zhang
- Department of Clinical Laboratory, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Weijing Li
- Department of Anesthesiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Zhenhua Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Lina Gu
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Yang Zheng
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Juan Li
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Baoen Shan
- Department of Research Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.,Institute of Tumor Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
47
|
Harada G, Bonadio RRDCC, de Araújo FCC, Victor CR, Sallum RAA, Junior UR, Cecconello I, Takeda FR, de Castria TB. Induction Chemotherapy for Locally Advanced Esophageal Cancer. J Gastrointest Cancer 2021; 51:498-505. [PMID: 31240598 DOI: 10.1007/s12029-019-00266-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Concurrent chemoradiotherapy followed by surgery is the standard treatment for locally advanced esophageal cancer (EC), and the role of induction chemotherapy (IC) remains unclear. We aimed to study if the addition of IC to standard treatment increases the rate of pathologic complete response (pCR). METHODS We assembled a retrospective analysis of patients (pts) diagnosed with locally advanced EC and treated with preoperative chemoradiotherapy followed by esophagectomy (CRT+S), preceded or not by IC, between 2009 and 2017. Patients' characteristics, tumor variables, and treatment outcomes were evaluated. The Kaplan-Meier method was used to estimate overall survival and the Cox proportional hazard model to evaluate prognostic factors. RESULTS One hundred and three patients were studied, with a median age of 62 years (range 37-84). Seventy-five patients (73%) were male, 67 (65%) had squamous cell carcinoma, and 31 (30%) had adenocarcinoma. Forty-three patients (41.7%) received IC followed by CRT+S (IC+CRT+S). The most frequent IC consisted of paclitaxel and platinum chemotherapy (90%), and the median number of cycles was 2. All patients received CRT+S. Concurrent chemotherapy was a combination of paclitaxel and platinum in 94 patients (91%). There was no statistically significant difference in pCR between the IC group and the standard CRT+S group. The pCR was 41.9% and 46.7% in the IC+CRT+S and CRT+S groups (p = 0.628), respectively. In the multivariate analysis, pCR was an independent prognostic factor for time to treatment failure (TTF) (HR 0.35, p = 0.021), but not for overall survival (OS) (p = 0.863). The factor that significantly affected OS in the multivariate analysis was positive lymph node (HR 5.9, 95%, p = 0.026). CONCLUSIONS Our data suggest that the addition of IC to standard CRT + S does not increase the pCR rate in locally advanced EC. No difference in OS was observed between pts. that received or not IC. Regardless of the treatment received, pts. achieving a pCR presented improved TTF.
Collapse
Affiliation(s)
- Guilherme Harada
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil.
- Centro de Oncologia, Hospital Sírio-Libanês, Sao Paulo, Brazil.
| | | | | | - Carolina Ribeiro Victor
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Rubens Antonio Aissar Sallum
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Ulysses Ribeiro Junior
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Ivan Cecconello
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Flávio Roberto Takeda
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Division of Digestive Surgery - Department of Gastroenterology, São Paulo State Cancer Institute - ICESP-HCFMUSP, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | - Tiago Biachi de Castria
- Instituto do Câncer do Estado de São Paulo, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
- Centro de Oncologia, Hospital Sírio-Libanês, Sao Paulo, Brazil
| |
Collapse
|
48
|
Wang L, Liu Y, Liu H, Tian H, Wang Y, Zhang G, Lei Y, Xue L, Zheng B, Fan T, Zheng Y, Tan F, Xue Q, Gao S, Li C, He J. The therapeutic significance of the novel photodynamic material TPE-IQ-2O in tumors. Aging (Albany NY) 2020; 13:1383-1409. [PMID: 33472175 PMCID: PMC7835032 DOI: 10.18632/aging.202355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
Combination therapies based on photodynamic therapy (PDT) have received much attention in various cancers due to their strong therapeutic effects. Here, we aimed to explore the safety and effectiveness of a new mitochondria-targeting photodynamic material, TPE-IQ-2O, in combination therapies (combined with surgery or immunotherapy). The safety and effectiveness of TPE-IQ-2O PDT were verified with cytotoxicity evaluation in vitro and a zebrafish xenograft model in vivo, respectively. The effectiveness of TPE-IQ-2O PDT combined with surgery or immune checkpoint inhibitors (ICIs) was verified in tumor-bearing mice. Small animal in vivo imaging, immunohistochemistry, and flow cytometry were used to determine the underlying mechanism. TPE-IQ-2O PDT can not only reduce tumor recurrence in surgical treatment but also effectively improve the response to ICIs in immunotherapy without obvious toxicity. It was also found to ameliorate the immunosuppressive tumor microenvironment and promote the antitumor immunity induced by ICIs by increasing CD8+ tumor-infiltrating lymphocyte accumulation. Thus, TPE-IQ-2O PDT is a safe and effective antitumor therapy that can be combined with surgery or immunotherapy.
Collapse
Affiliation(s)
- Liyu Wang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yu Liu
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hengchang Liu
- Department of Colorectal Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yalong Wang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuanyuan Lei
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Liyan Xue
- Department of Pathology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
49
|
Shen H, Liu J, Sun G, Yan L, Li Q, Wang Z, Xie L. The clinicopathological significance and prognostic value of programmed death-ligand 1 in prostate cancer: a meta-analysis of 3133 patients. Aging (Albany NY) 2020; 13:2279-2293. [PMID: 33318295 PMCID: PMC7880326 DOI: 10.18632/aging.202248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022]
Abstract
Background: Programmed death-ligand 1 (PD-L1) is considered an adverse factor predicting poor prognosis in various cancers, but the significance of PD-L1 expression for the prognosis of prostate cancer (PCa) is still unclear. We aimed to investigate the clinicopathological significance and prognostic value of PD-L1 expression in PCa. Methods: Studies were retrieved from PubMed, Web of Science, Cochrane Library and Embase before March 23, 2020. Odds ratios (ORs) and hazard ratios (HRs) with 95% confidence intervals (CIs) were obtained to assess the results. Begg’s test was applied to evaluate publication bias. Results: Fourteen studies involving 3133 cases were analyzed. The pooled data showed that both PD-L1 protein expression and PD-L1 DNA methylation (mPD-L1) were negatively associated with biochemical recurrence-free survival, with HRs of 1.67 (95% CI = 1.38-2.06, p < 0.001) and 2.23 (95% CI = 1.51-3.29, p < 0.001), respectively. In addition, PD-L1 overexpression was significantly related to advanced tumor stage (OR = 1.40, 95% CI= 1.13-1.75, p = 0.003), positive surgical margin (OR = 1.36, 95% CI = 1.03-1.78, p = 0.028), higher Gleason score (OR = 1.81, 95% CI = 1.35-2.42, p < 0.001) and androgen receptor positivity (OR = 2.20, 95% CI = 1.61-3.01, p < 0.001), while no significant correlation with age (p = 0.122), preoperative PSA (p = 0.796) or nodal status (p = 0.113) was observed. Conclusions: The study revealed that high expression of PD-L1 was related to unfavorable prognosis and advanced clinicopathological factors in PCa patients.
Collapse
Affiliation(s)
- Haixiang Shen
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jin Liu
- Department of Surgical Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Guoliang Sun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Libin Yan
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Qinchen Li
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Zhize Wang
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Liping Xie
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| |
Collapse
|
50
|
Liu Y, Zhao L, Xue L, Hou Y. Selected updates in molecular and genomic pathology of esophageal cancer. Ann N Y Acad Sci 2020; 1482:225-235. [PMID: 33215736 DOI: 10.1111/nyas.14527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/18/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022]
Abstract
Recent years have seen rapid advances in the field of molecular and genomic pathology that have not only improved understanding of esophageal carcinogenesis and tumor immune environment in general but also have reshaped pathology practice and clinical management. In this article, we provide updates on three topics (1) human epidermal growth factor receptor 2, the first and most important biomarker in targeted therapy of esophageal cancer; (2) programmed death 1/programmed death ligand 1, recent biomarkers that have shown promise in treating both esophageal adenocarcinoma and esophageal squamous cell carcinoma; and (3) human papillomavirus involvement in esophageal carcinogenesis, one of the most debated topics in the field, discussed here with a renewed understanding from recent genomic and molecular data.
Collapse
Affiliation(s)
- Yueping Liu
- Department of Pathology, Hebei Medical University Fourth Affiliated Hospital and Hebei Provincial Tumor Hospital, Shijiazhuang, China
| | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Liyan Xue
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|