1
|
Uehara Y, Izumi H, Taki T, Sakai T, Udagawa H, Sugiyama E, Umemura S, Zenke Y, Matsumoto S, Yoh K, Kubota S, Aokage K, Sakamoto N, Sakashita S, Kojima M, Nagamine M, Hosomi Y, Tsuboi M, Goto K, Ishii G. Solid Predominant Histology and High Podoplanin Expression in Cancer-Associated Fibroblast Predict Primary Resistance to Osimertinib in EGFR-Mutated Lung Adenocarcinoma. JTO Clin Res Rep 2025; 6:100779. [PMID: 40007550 PMCID: PMC11850751 DOI: 10.1016/j.jtocrr.2024.100779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/29/2024] [Accepted: 12/12/2024] [Indexed: 02/27/2025] Open
Abstract
Introduction Resistance to EGFR tyrosine kinase inhibitors is influenced by tumor-intrinsic and -extrinsic factors. We investigated the impact of tumor cell histology and tumor microenvironment on the efficacy of osimertinib. Methods We evaluated surgically resected adenocarcinoma from patients treated with first-line osimertinib at the National Cancer Center Hospital East (2016-2023), evaluating clinicopathologic characteristics, tumor cell histology, podoplanin expression in cancer-associated fibroblasts (CAFs) identified by immunohistochemistry, and outcomes. We also investigated HGF mRNA expression levels, using The Cancer Genome Atlas Program and Singapore Oncology Data Portal cohorts. Results The study included 93 patients. Solid (n = 19) versus non-solid predominant (n = 74) histology was not associated with worse disease-free survival after surgery (p = 0.12), but was significantly associated with worse progression-free survival (PFS) and overall survival following osimertinib treatment (p = 0.026, p = 0.004). Similarly, high-podoplanin (n = 31) versus low-podoplanin (n = 62) expression in CAFs was not associated with worse disease-free survival after surgery (p = 0.65), but was significantly associated with worse PFS and showed a trend towards worse overall survival following osimertinib treatment (p < 0.001, p = 0.11). In the multivariable analysis, solid predominant histology and high-podoplanin expression in CAFs were independently associated with worse PFS. In the cohorts of The Cancer Genome Atlas Program and Singapore Oncology Data Portal, EGFR-mutated lung adenocarcinoma with solid predominant histology or high-podoplanin expression exhibited significantly higher HGF expression. Conclusions Solid predominant histology and high-podoplanin expression in CAFs predicted osimertinib resistance, potentially guiding the selection of patients for more intensive treatments beyond osimertinib monotherapy.
Collapse
Affiliation(s)
- Yuji Uehara
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hiroki Izumi
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuro Taki
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tetsuya Sakai
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hibiki Udagawa
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Eri Sugiyama
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shigeki Umemura
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshitaka Zenke
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Matsumoto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shoko Kubota
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Keiju Aokage
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Naoya Sakamoto
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shingo Sakashita
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Michiko Nagamine
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Genichiro Ishii
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Innovative Pathology and Laboratory Medicine, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
2
|
Bouchard G, Zhang W, Ilerten I, Li I, Bhattacharya A, Li Y, Trope W, Shrager JB, Kuo C, Ozawa MG, Giaccia AJ, Tian L, Plevritis SK. A quantitative spatial cell-cell colocalizations framework enabling comparisons between in vitro assembloids and pathological specimens. Nat Commun 2025; 16:1392. [PMID: 39915493 PMCID: PMC11802768 DOI: 10.1038/s41467-024-55129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 11/29/2024] [Indexed: 02/09/2025] Open
Abstract
Spatial omics is enabling unprecedented tissue characterization, but the ability to adequately compare spatial features across samples under different conditions is lacking. We propose a quantitative framework that catalogs significant, normalized, colocalizations between pairs of cell subpopulations, enabling comparisons among a variety of biological samples. We perform cell-pair colocalization analysis on multiplexed immunofluorescence images of assembloids constructed with lung adenocarcinoma (LUAD) organoids and cancer-associated fibroblasts derived from human tumors. Our data show that assembloids recapitulate human LUAD tumor-stroma spatial organization, justifying their use as a tool for investigating the spatial biology of human disease. Intriguingly, drug-perturbation studies identify drug-induced spatial rearrangements that also appear in treatment-naïve human tumor samples, suggesting potential directions for characterizing spatial (re)-organization related to drug resistance. Moreover, our work provides an opportunity to quantify spatial data across different samples, with the common goal of building catalogs of spatial features associated with disease processes and drug response.
Collapse
Affiliation(s)
- Gina Bouchard
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Weiruo Zhang
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Ilayda Ilerten
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Irene Li
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Asmita Bhattacharya
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Yuanyuan Li
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Winston Trope
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
| | - Joseph B Shrager
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Calvin Kuo
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Michael G Ozawa
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA, USA
- Department of Oncology, University of Oxford, Oxford, UK
| | - Lu Tian
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Department of Radiology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
3
|
Yu D, Xu H, Zhou J, Fang K, Zhao Z, Xu K. PDPN/CCL2/STAT3 feedback loop alter CAF heterogeneity to promote angiogenesis in colorectal cancer. Angiogenesis 2024; 27:809-825. [PMID: 39115624 DOI: 10.1007/s10456-024-09941-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/31/2024] [Indexed: 11/15/2024]
Abstract
Colorectal cancer (CRC) is one of the common clinical malignancies and the fourth leading cause of cancer-related death in the world. The tumor microenvironment (TME) plays a crucial role in promoting tumor angiogenesis, and cancer-associated fibroblasts (CAFs) are one of the key components of the tumor microenvironment. However, due to the high heterogeneity of CAFs, elucidating the molecular mechanism of CAF-mediated tumor angiogenesis remained elusive. In our study, we found that there is pro-angiogenic functional heterogeneity of CAFs in colorectal cancer and we clarified that Podoplanin (PDPN) can specifically label CAF subpopulations with pro-angiogenic functions. We also revealed that PDPN + CAF could maintain CAF heterogeneity by forming a PDPN/CCL2/STAT3 feedback loop through autocrine CCL2, while activate STAT3 signaling pathway in endothelial cells to promote angiogenesis through paracrine CCL2. We demonstrated WP1066 could inhibit colorectal cancer angiogenesis by blocking both the PDPN/CCL2/STAT3 feedback loop in CAFs and the STAT3 signaling pathway in endothelial cells. Altogether, our study suggests that STAT3 could be a potential therapeutic target for blocking angiogenesis in colorectal cancer. We provide theoretical basis and new therapeutic strategies for the clinical treatment of colorectal cancer.
Collapse
Affiliation(s)
- Die Yu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237, China
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Hanzheng Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237, China
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Jinzhe Zhou
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Kai Fang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, 130 Meilong Rd, Shanghai, 200237, China.
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
| | - Zekun Zhao
- Department of General Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China.
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China.
- Wenzhou Institute of Shanghai University, Wenzhou, China.
| |
Collapse
|
4
|
Zhang D, Liang P, Xia B, Wu J, Hu X. Comprehensive pan-cancer analysis of ZNF337 as a potential diagnostic, immunological, and prognostic biomarker. BMC Cancer 2024; 24:987. [PMID: 39123194 PMCID: PMC11313096 DOI: 10.1186/s12885-024-12703-x] [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: 10/10/2023] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Zinc Finger Protein 337 (ZNF337) is a novel Zinc Finger (ZNF) protein family member. However, the roles of ZNF337 in human cancers have not yet been investigated. METHODS In this study, with the aid of TCGA databases, GTEx databases, and online websites, we determined the expression levels of ZNF337 in pan-cancer and its potential value as a diagnostic and prognostic marker for pan-cancer and analyzed the relationship between ZNF337 expression and immune cell infiltration and immune checkpoint genes. We then focused our research on the potential of ZNF337 as a biomarker for diagnostic and prognostic in KIRC (kidney renal clear cell carcinoma) and validated in the E-MTAB-1980 database. Moreover, the expression of ZNF337 was detected through qRT-PCR and Western blotting (WB). CCK-8 experiment, colony formation experiment, and EDU experiment were performed to evaluate cell proliferation ability. Wound healing assay and transwell assay were used to analyze its migration ability. The qRT-PCR and WB were used to detect the expression of ZNF337 in tumor tissues and paracancerous tissues of KIRC patients. RESULTS The pan-cancer analysis revealed that abnormal ZNF337 expression was found in multiple human cancer types. ZNF337 had a high diagnostic value in pan-cancer and a significant association with the prognosis of certain cancers, indicating that ZNF337 may be a valuable prognostic biomarker for multiple cancers. Further analysis demonstrated that the expression level of ZNF337 displayed significant correlations with cancer-associated fibroblasts, immune cell infiltration, and immune checkpoint genes in many tumors. Additionally, ZNF337 was observed to have a high expression in KIRC. Its expression was significantly associated with poor prognosis [overall survival (OS), disease-specific survival (DSS)], age, TNM stage, histologic grade, and pathologic stage. The high ZNF337 expression was associated with poor prognosis in the E-MTAB-1980 validation cohort. The in vitro experiments suggested that the expression of ZNF337 in KIRC tumor tissues was higher than in adjacent tissues, and ZNF337 knockdown inhibited the proliferation and migration of KIRC cells, whereas overexpression of ZNF337 had the opposite effects. CONCLUSIONS ZNF337 might be an important prognostic and immunotherapeutic biomarker for pan-cancer, especially in KIRC.
Collapse
Affiliation(s)
- Dongxu Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8 Gongti South Road, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Pu Liang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, 100015, China
| | - Bowen Xia
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8 Gongti South Road, Beijing, China
- Institute of Urology, Capital Medical University, Beijing, China
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Xiaopeng Hu
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, NO. 8 Gongti South Road, Beijing, China.
- Institute of Urology, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Tomero-Sanz H, Jiménez-Heffernan JA, Fernández-Chacón MC, Cristóbal-García I, Sainz de la Cuesta R, González-Cortijo L, López-Cabrera M, Sandoval P. Detection of Carcinoma-Associated Fibroblasts Derived from Mesothelial Cells via Mesothelial-to-Mesenchymal Transition in Primary Ovarian Carcinomas. Cancers (Basel) 2024; 16:2697. [PMID: 39123425 PMCID: PMC11311419 DOI: 10.3390/cancers16152697] [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: 05/29/2024] [Revised: 07/23/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Carcinoma-associated fibroblasts (CAFs) are highly accumulated in the tumor-surrounding stroma of primary epithelial ovarian cancer (OC). CAFs exert important functions for the vascularization, growth, and progression of OC cells. However, the origin of CAFs in primary OC had not yet been studied, and they were assumed to arise from the activation of resident fibroblasts. Here, we compared CAFs in the ovary to CAFs found in peritoneal metastases from patients with advanced OC. Our findings show that CAFs from primary tumors and peritoneal metastases share the expression of mesothelial markers. Therefore, similar to peritoneal carcinomatosis, CAFs in primary ovarian carcinomas may originate from mesothelial cells via a mesothelial-to-mesenchymal transition. The detection of mesothelial-derived CAFs in tumors confined to the ovary and identification of biomarkers could be the key to the early detection of OC and peritoneal spread.
Collapse
Affiliation(s)
- Henar Tomero-Sanz
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, 28049 Madrid, Spain;
| | | | | | | | - Ricardo Sainz de la Cuesta
- Hospital Universitario QuirónSalud Madrid, 28223 Madrid, Spain; (R.S.d.l.C.); (L.G.-C.)
- Department of de Medicine, Facultad de Biomédica y Ciencias de la Salud, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Lucía González-Cortijo
- Hospital Universitario QuirónSalud Madrid, 28223 Madrid, Spain; (R.S.d.l.C.); (L.G.-C.)
- Department of de Medicine, Facultad de Biomédica y Ciencias de la Salud, Universidad Europea de Madrid, 28670 Madrid, Spain
| | - Manuel López-Cabrera
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, 28049 Madrid, Spain;
| | - Pilar Sandoval
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa (CBM), CSIC-UAM, 28049 Madrid, Spain;
| |
Collapse
|
6
|
Xu AM, Haro M, Walts AE, Hu Y, John J, Karlan BY, Merchant A, Orsulic S. Spatiotemporal architecture of immune cells and cancer-associated fibroblasts in high-grade serous ovarian carcinoma. SCIENCE ADVANCES 2024; 10:eadk8805. [PMID: 38630822 PMCID: PMC11023532 DOI: 10.1126/sciadv.adk8805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 03/15/2024] [Indexed: 04/19/2024]
Abstract
High-grade serous ovarian carcinoma (HGSOC), the deadliest form of ovarian cancer, is typically diagnosed after it has metastasized and often relapses after standard-of-care platinum-based chemotherapy, likely due to advanced tumor stage, heterogeneity, and immune evasion and tumor-promoting signaling from the tumor microenvironment. To understand how spatial heterogeneity contributes to HGSOC progression and early relapse, we profiled an HGSOC tissue microarray of patient-matched longitudinal samples from 42 patients. We found spatial patterns associated with early relapse, including changes in T cell localization, malformed tertiary lymphoid structure (TLS)-like aggregates, and increased podoplanin-positive cancer-associated fibroblasts (CAFs). Using spatial features to compartmentalize the tissue, we found that plasma cells distribute in two different compartments associated with TLS-like aggregates and CAFs, and these distinct microenvironments may account for the conflicting reports about the role of plasma cells in HGSOC prognosis.
Collapse
Affiliation(s)
- Alexander M. Xu
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marcela Haro
- Department of Obstetrics and Gynecology and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ann E. Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ye Hu
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Joshi John
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Department of Medicine, Division of Geriatrics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Beth Y. Karlan
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Akil Merchant
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Division of Hematology and Cellular Therapy, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Pascual-Antón L, Sandoval P, González-Mateo GT, Kopytina V, Tomero-Sanz H, Arriero-País EM, Jiménez-Heffernan JA, Fabre M, Egaña I, Ferrer C, Simón L, González-Cortijo L, Sainz de la Cuesta R, López-Cabrera M. Targeting carcinoma-associated mesothelial cells with antibody-drug conjugates in ovarian carcinomatosis. J Pathol 2023; 261:238-251. [PMID: 37555348 DOI: 10.1002/path.6170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/14/2023] [Accepted: 06/22/2023] [Indexed: 08/10/2023]
Abstract
Ovarian carcinomatosis is characterized by the accumulation of carcinoma-associated mesothelial cells (CAMs) in the peritoneal stroma and mainly originates through a mesothelial-to-mesenchymal transition (MMT) process. MMT has been proposed as a therapeutic target for peritoneal metastasis. Most ovarian cancer (OC) patients present at diagnosis with peritoneal seeding, which makes tumor progression control difficult by MMT modulation. An alternative approach is to use antibody-drug conjugates (ADCs) targeted directly to attack CAMs. This strategy could represent the cornerstone of precision-based medicine for peritoneal carcinomatosis. Here, we performed complete transcriptome analyses of ascitic fluid-isolated CAMs in advanced OC patients with primary-, high-, and low-grade, serous subtypes and following neoadjuvant chemotherapy. Our findings suggest that both cancer biological aggressiveness and chemotherapy-induced tumor mass reduction reflect the MMT-associated changes that take place in the tumor surrounding microenvironment. Accordingly, MMT-related genes, including fibroblast activation protein (FAP), mannose receptor C type 2 (MRC2), interleukin-11 receptor alpha (IL11RA), myristoylated alanine-rich C-kinase substrate (MARCKS), and sulfatase-1 (SULF1), were identified as specific actionable targets in CAMs of OC patients, which is a crucial step in the de novo design of ADCs. These cell surface target receptors were also validated in peritoneal CAMs of colorectal cancer peritoneal implants, indicating that ADC-based treatment could extend to other abdominal tumors that show peritoneal colonization. As proof of concept, a FAP-targeted ADC reduced tumor growth in an OC xenograft mouse model with peritoneal metastasis-associated fibroblasts. In summary, we propose MMT as a potential source of ADC-based therapeutic targets for peritoneal carcinomatosis. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Lucía Pascual-Antón
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Pilar Sandoval
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Guadalupe T González-Mateo
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Valeria Kopytina
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Henar Tomero-Sanz
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | - Eva María Arriero-País
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| | | | | | | | | | | | | | | | - Manuel López-Cabrera
- Tissue and Organ Homeostasis Program, Centro de Biología Molecular Severo Ochoa - Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid (CBMSO-CSIC-UAM), Madrid, Spain
| |
Collapse
|
8
|
Souza da Silva R, Queiroga EM, de Toledo Osório C, Cunha KS, Neves FP, Andrade JP, Dias EP. Expression Profile of Microenvironmental Factors in the Interface Zone of Colorectal Cancer: Histological-Stromal Biomarkers and Cancer Cell-Cancer-Associated Fibroblast-Related Proteins Combined for the Assessment of Tumor Progression. Pathobiology 2023; 91:99-107. [PMID: 37369175 DOI: 10.1159/000531695] [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: 01/17/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
INTRODUCTION The characterization of tumor microenvironment (TME) related factors and their impact on tumor progression have attracted much interest. We investigated cancer cells and cancer-associated fibroblasts (CAFs) to evaluate biomarkers that are associated with neoplastic progression, observing them in different interface zones of colorectal cancer. METHODS On 357 CRC tissue microarrays, using immunohistochemistry, we examined the associations of podoplanin and α-SMA expressed in cancer cells and CAFs and evaluated them in different areas: tumor core, invasive front, tumor budding, tumor-stroma ratio (TSR) scoring, and desmoplastic stroma. RESULTS CAFs expressing α-SMA were found in more than 90% of the cases. Podoplanin+ was detected in cancer cells and CAFs, with positivities of 38.6% and 70%, respectively. Higher α-SMA+ CAFs and podoplanin+ cancer cells were observed predominantly at the TSR score area: 94.3% and 64.3% of cases, respectively. The status of podoplanin in CAFs+ was higher in the desmoplastic area (71.6%). Stroma-high tumors showed increased expression of α-SMA and podoplanin in comparison with stroma-low tumors. The status of podoplanin in cancer cells was observed in association with lymphatic invasion and distant metastasis. CONCLUSION The substance of the CRC was composed predominantly of the surrounding stroma-α-SMA+ CAFs. Podoplanin expressed in the prognosticator zones was associated with unfavorable pathological features. The combination of histologic and protein-related biomarkers can result in a tool for the stratification of patients with CRC.
Collapse
Affiliation(s)
- Ricella Souza da Silva
- Pathological Anatomy Service, Lauro Wanderley University Hospital of Federal University of Paraíba, João Pessoa, Brazil
| | - Eduardo M Queiroga
- Laboratory of Pathological Anatomy, Alcides Carneiro University Hospital of the Federal University of Campina Grande, Campina Grande, Brazil
| | | | - Karin S Cunha
- Department of Pathology, School of Medicine, Fluminense Federal University, Niterói, Brazil
| | - Fabiana P Neves
- Anatomopathological Diagnostic Center, Napoleão Laureano Hospital, João Pessoa, Brazil
| | - Julieth P Andrade
- Anatomopathological Diagnostic Center, Napoleão Laureano Hospital, João Pessoa, Brazil
| | - Eliane P Dias
- Department of Pathology, School of Medicine, Fluminense Federal University, Niterói, Brazil
| |
Collapse
|
9
|
Prognostic impact of tumor microenvironment-related markers in patients with adenocarcinoma of the lung. Int J Clin Oncol 2023; 28:229-239. [PMID: 36376711 PMCID: PMC9889427 DOI: 10.1007/s10147-022-02271-0] [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: 09/07/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Cancer-associated fibroblasts (CAFs) are a prominent component in the tumor microenvironment (TME), which plays an important role in lung carcinogenesis. Here, we investigated microenvironmental markers expressed by CAFs, including α-smooth muscle actin, CD10, podoplanin, fibroblast-specific protein 1, platelet-derived growth factor α and β, fibroblast-associated protein, tenascin-C, zinc finger E-box binding homeobox 1 (ZEB1), and twist-related protein 1 expression levels. We evaluated samples from 257 patients with lung adenocarcinoma (LAD) to assess the associations of CAF-related protein expression patterns with prognosis. LAD cases were stratified using cluster analysis. To determine the utility of prognostic markers in LAD, univariate and multivariate analyses were performed. LAD cases were classified into subgroups 1 and 2. Subgroup 2 was shown to be significantly correlated with disease-free and overall survival using univariate and multivariate analyses in this group. Upregulation of podoplanin was identified as a single prognostic marker in this study by univariate and multivariate analyses. In addition, ZEB1 overexpression was correlated with disease-free survival. Our current results suggested that the specific CAF phenotype (e.g., the expression pattern of CAF-related proteins) could predict outcomes in patients with LAD. In addition, podoplanin upregulation may predict outcomes in these patients.
Collapse
|
10
|
Ren Z, He Y, Yang Q, Guo J, Huang H, Li B, Wang D, Yang Z, Tian X. A Comprehensive Analysis of the Glutathione Peroxidase 8 (GPX8) in Human Cancer. Front Oncol 2022; 12:812811. [PMID: 35402257 PMCID: PMC8991916 DOI: 10.3389/fonc.2022.812811] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/25/2022] [Indexed: 01/22/2023] Open
Abstract
Objective Nowadays, cancer is still a leading public health problem all over the world. Several studies have reported the GPX8 could be correlated with the poor prognostic of Gastric Cancer and Breast Cancer. However, the prognostic potential of GPX8 in pan-cancer remains unclear. In this work, we aimed to explore the prognostic and immunological role of GPX8 in human cancer and confirm the oncogenic value in GBM. Methods The data of TCGA, CPTAC and GEO databases were adopted for the survival analysis. Based on the RNAseq and Methylation450 data of TCGA, the R language and package “ggplot2” were used to analyze the DNA methylation at the region of the promoter of GPX8 in tumors. The genetic alteration of GPX8 from TCGA cancers was investigated in cBioPortal. The R package “GSVA” and “ssGSEA” were employed to evaluate the correlation of GPX8 expression with the immune infiltration. The KEGG website was used for pathway analysis. The STRING website and GEPIA were performed to predict GPX8-binding proteins. The R package “ggplot2” and “clusterprofile” were used to analyze and visualize the GO and KEGG analysis. A normal human astrocyte cell line and three GBM cell lines were cultured under suitable conditions. The shRNA was transferred to cells by Lipofectamine 3000. The qRT-PCR and WB were adopted to detect the expression of GPX8. The wound-healing assay and transwell assay were taken to analyze the invasive and metastatic abilities. The tumor tissues and paracancerous ones were collected from patients with GBM. WB assay was employed to analyze the expression of GPX8 protein. Results GPX8 was a valuable diagnostic biomarker in multiple cancers, including GBM/LGG (glioblastoma multiforme/Brain lower grade glioma), KIRC (kidney renal clear cell carcinoma), KIRP (kidney renal papillary cell carcinoma) and STAD (stomach adenocarcinoma). Moreover, we observed a correlation between the expression of GPX8 and the reduced DNA methylation at the promoter region in several tumors, such as GBM/LGG. Our results indicated a positive correlation between the GPX8 expression and immune infiltration. In addition, the enrichment analysis demonstrated that antioxidant activity was mainly involved in the functional mechanism of GPX8. In particular, we first confirmed the up-regulated of GPX8 in GBM cells and observed the suppression of migrative and invasive phenotypes by knockdown of GPX8. Furthermore, we confirmed the expression of GPX8 was higher in GBM tumor tissues than paracancerous ones. Conclusion Our study showed a correlation of GPX8 expression with clinical prognosis, DNA methylation and immune infiltrates. Furthermore, we first confirmed GPX8 was highly expressed in GBM cells and contributed to migration and invasion. These results provided a predictive biomarker and an inclusive understanding of the GPX8 expression in multiple tumors types, especially in GBM.
Collapse
Affiliation(s)
- Zhijing Ren
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, China
| | - Yu He
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qinqin Yang
- Medical College, Guizhou University, Guiyang, China
| | - Jiajia Guo
- Medical College, Guizhou University, Guiyang, China
| | - Haifeng Huang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Bo Li
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Dong Wang
- Department of Orthopedics, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | - Zhen Yang
- Department of Orthopedics, Guizhou Provincial People's Hospital, Guiyang, China
| | - Xiaobin Tian
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
11
|
Wu Z, Shi J, Lai C, Li K, Li K, Li Z, Tang Z, Liu C, Xu K. Clinicopathological significance and prognostic value of cancer-associated fibroblasts in prostate cancer patients. Urol Oncol 2021; 39:433.e17-433.e23. [PMID: 34112577 DOI: 10.1016/j.urolonc.2021.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/23/2021] [Accepted: 05/03/2021] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Cancer-associated fibroblasts (CAFs) in the tumor microenvironment were considered to play an essential role in tumor growth and development. However, few studies have assessed the prognostic and clinicopathological significance of CAFs in prostate cancer (PCa) patients. METHODS One hundred thirty pairs of PCa tissues and normal adjacent tissues (NATs) were immunostained with fibroblast activation protein and α-smooth muscle actin to quantify CAFs. Bioinformatics analysis was used to uncover the possible biological functions of CAFs. RESULTS More CAFs were identified in PCa tissues than in NATs. High density of CAFs may be associated with advanced-stage disease, higher Gleason scores, lymphatic metastases, higher PSA, and poor biochemical recurrence-free survival in PCa. Bioinformatics analysis showed that CAFs may regulate tumor progression and recurrence through ECM modification, PI3K-Akt signaling pathway and regulation of cytoskeleton. CONCLUSION In summary, our study uncovered the clinicopathological significance and potential mechanism of CAFs and indicated that CAFs may be a useful prognostic biomarker in PCa.
Collapse
Affiliation(s)
- Zhenyu Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Juanyi Shi
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Cong Lai
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Kaiwen Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Kuiqing Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Zhuohang Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Zhuang Tang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China
| | - Cheng Liu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.
| | - Kewei Xu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P. R. China.
| |
Collapse
|
12
|
Baker AT, Abuwarwar MH, Poly L, Wilkins S, Fletcher AL. Cancer-Associated Fibroblasts and T Cells: From Mechanisms to Outcomes. THE JOURNAL OF IMMUNOLOGY 2021; 206:310-320. [PMID: 33397745 DOI: 10.4049/jimmunol.2001203] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 11/06/2020] [Indexed: 12/20/2022]
Abstract
Over the past decade, T cell immunotherapy has changed the face of cancer treatment, providing robust treatment options for several previously intractable cancers. Unfortunately, many epithelial tumors with high mortality rates respond poorly to immunotherapy, and an understanding of the key impediments is urgently required. Cancer-associated fibroblasts (CAFs) comprise the most frequent nonneoplastic cellular component in most solid tumors. Far from an inert scaffold, CAFs significantly influence tumor neogenesis, persistence, and metastasis and are emerging as a key player in immunotherapy resistance. In this review, we discuss the physical and chemical barriers that CAFs place between effector T cells and their tumor cell targets, and the therapies poised to target them.
Collapse
Affiliation(s)
- Alfie T Baker
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Mohammed H Abuwarwar
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Lylarath Poly
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia
| | - Simon Wilkins
- Cabrini Monash University Department of Surgery, Cabrini Hospital, Malvern 3144, Victoria, Australia.,Department of Epidemiology and Preventive Medicine, School of Public Health and Preventive Medicine, Monash University, Melbourne 3004, Victoria, Australia; and
| | - Anne L Fletcher
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton 3800, Victoria, Australia; .,Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| |
Collapse
|
13
|
Yang C, Ma C, Li Y, Mo P, Yang Y. High Tiam1 expression predicts positive lymphatic metastasis and worse survival in patients with malignant solid tumors: a systematic review and meta-analysis. Onco Targets Ther 2019; 12:5925-5936. [PMID: 31413590 PMCID: PMC6663076 DOI: 10.2147/ott.s191571] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 05/10/2019] [Indexed: 12/16/2022] Open
Abstract
Background Many studies have explored the prognostic value of T-cell lymphoma invasion and metastasis inducing factor 1 (Tiam1) and its association with lymphatic metastasis in malignant solid tumors, but the conclusions remain controversial. Therefore, we performed a meta-analysis to systematically assess the prognostic value of Tiam1 expression and its association with lymphatic metastasis in malignant solid tumors. Methods We searched eligible studies in PubMed, Web of Science and EMBASE databases (from inception up to October 2018). The combined HR with 95% CI was used to estimate the prognostic value of Tiam1 expression. The correlation between Tiam1 expression and lymphatic metastasis was assessed using the combined odds ratio (OR) with 95% CI. Results A total of 17 studies with 2,228 patients with solid tumors were included in this meta-analysis. The overall estimated results showed that high Tiam1 expression was significantly associated with shorter overall survival (HR= 2.08, 95% CI: 1.62-2.68, P<0.01), and disease-free survival (HR = 1.86, 95% CI: 1.49-2.32, P<0.01). Besides, we also found that there was a close relationship between high Tiam1 expression and positive lymphatic metastasis (OR=2.63; 95% CI: 1.79-3.84, P<0.01). Conclusion High Tiam1 expression was significantly associated with shorter survival and positive lymphatic metastasis in patients with malignant solid tumors. Therefore, Tiam1 may be a promising prognostic biomarker and an effective therapeutic target for malignant solid tumors.
Collapse
Affiliation(s)
- Caixia Yang
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Chenlin Ma
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Yingchun Li
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Peng Mo
- Department of Stomatology, Qinghai province people's Hospital, Qinghai, People's Republic of China
| | - Yusheng Yang
- Department of Pathology, Ninbo Yinzhou No. 2 Hospital, Ninbo, People's Republic of China
| |
Collapse
|
14
|
Podoplanin, a Potential Therapeutic Target for Nasopharyngeal Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7457013. [PMID: 31321241 PMCID: PMC6610758 DOI: 10.1155/2019/7457013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/27/2019] [Indexed: 12/11/2022]
Abstract
Introduction The role of podoplanin (PDPN) in nasopharyngeal carcinoma (NPC) is still unknown. The aims of this study were to investigate the expression and role of PDPN in NPC cells. Materials and Methods Immunofluorescence staining and functional tests were used to determine the effects of PDPN knockdown by siRNA in TW01 NPC cells. Microarray analysis was conducted to identify genes regulated by PDPN. The molecular mechanism of PDPN on NPC cells was further determined by Ingenuity Pathways Analysis (IPA). Results PDPN was expressed in most TW01 NPC cells. PDPN knockdown by siRNA decreased NPC cell proliferation, migration, and invasion. The microarray data showed 63 upregulated genes and 12 downregulated genes following PDPN knockdown. The top 5 most upregulated genes analyzed by IPA were IFI27, IFI44L, IFI6, OAS1, and TRIM22, and the most relevant pathway was the interferon signaling pathway. Conclusions To the best of our knowledge, this is the first report to show that knocking down PDPN leads to suppression of NPC cell proliferation, migration, and invasion. Our results suggest that PDPN may serve as a potential chemotherapeutic target for NPC treatment in the future.
Collapse
|
15
|
Blei F. Update December 2018. Lymphat Res Biol 2018. [DOI: 10.1089/lrb.2018.29054.fb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|