1
|
Gorbokon N, Wößner N, Lennartz M, Dwertmann Rico S, Kind S, Reiswich V, Viehweger F, Lutz F, Fraune C, Luebke AM, Hube-Magg C, Menz A, Schlichter R, Krech T, Hinsch A, Burandt E, Sauter G, Simon R, Steurer S, Marx AH, Lebok P, Dum D, Minner S, Jacobsen F, Clauditz TS, Hackert T, Uzunoǧlu FG, Bubendorf L, Bernreuther C, Kluth M. Prevalence of S-methyl-5'-thioadenosine Phosphorylase (MTAP) Deficiency in Human Cancer: A Tissue Microarray Study on 13,067 Tumors From 149 Different Tumor Types. Am J Surg Pathol 2024; 48:1245-1258. [PMID: 39132873 PMCID: PMC11404761 DOI: 10.1097/pas.0000000000002297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Loss of S-methyl-5'-thioadenosine phosphorylase (MTAP) expression is a common event in cancer leading to a critical vulnerability of cancer cells towards anti-cancer drugs. Homozygous MTAP deletions result in a complete expression loss that can be detected by immunohistochemistry (IHC). In this study, a tissue microarray containing 17,078 samples from 149 different tumor entities was analyzed by IHC, and complete MTAP loss was validated by fluorescence in situ hybridization. MTAP loss was observed in 83 of 149 tumor categories, including neuroendocrine neoplasms (up to 80%), Hodgkin lymphoma (50.0%), mesothelioma (32.0% to 36.8%), gastro-intestinal adenocarcinoma (4.0% to 40.5%), urothelial neoplasms (10.5% to 36.7%), squamous cell carcinomas (up to 38%), and various types of sarcomas (up to 20%) and non-Hodgkin lymphomas (up to 14%). Homozygous MTAP deletion was found in 90% to 100% of cases with MTAP expression loss in most tumor categories. However, neuroendocrine tumors, Hodgkin lymphomas, and other lymphomas lacked MTAP deletions. MTAP deficiency was significantly linked to unfavorable tumor phenotype in selected tumor entities and the presence of PD-L1 expression on tumor cells, absence of PD-L1 expression on immune cells, and a low density of CD8 + lymphocytes. In summary, MTAP deficiency can occur in various tumor entities and is linked to unfavorable tumor phenotype and noninflamed tumor microenvironment, but is not always related to deletions. MTAP IHC is of considerable diagnostic value for the detection of neoplastic transformation in multiple different applications.
Collapse
Affiliation(s)
- Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Niklas Wößner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Sebastian Dwertmann Rico
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Simon Kind
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Florian Lutz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Christoph Fraune
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Anne Menz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Ria Schlichter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Till Krech
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Andreas H Marx
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
- Institute of Pathology, Clinical Center Osnabrueck, Am Finkenhügel, Osnabrück, Germany
| | - Patrick Lebok
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - David Dum
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Thilo Hackert
- Department of Pathology, Academic Hospital Fuerth, Jakob-Henle-Straße, Fürth, Germany
| | - Faik G Uzunoǧlu
- Department of Pathology, Academic Hospital Fuerth, Jakob-Henle-Straße, Fürth, Germany
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, Schönbeinstrasse, Basel, Switzerland
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| |
Collapse
|
2
|
Shukrun R, Baron S, Fidel V, Shusterman A, Sher O, Kollender N, Levin D, Peled Y, Gortzak Y, Ben‐Shahar Y, Caspi R, Gordon S, Manisterski M, Elhasid R. Suggested role for neutrophil extracellular trap formation in Ewing sarcoma immune microenvironment. Cancer Sci 2024; 115:36-47. [PMID: 37915266 PMCID: PMC10823276 DOI: 10.1111/cas.15992] [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: 07/06/2023] [Revised: 09/13/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Ewing sarcoma (EWS) is a highly aggressive cancer with a survival rate of 70%-80% for patients with localized disease and under 30% for those with metastatic disease. Tumor-infiltrating neutrophils (TIN) can generate extracellular net-like DNA structures known as neutrophil extracellular traps (NETs). However, little is known about the presence and prognostic significance of tumor-infiltrating NETs in EWS. Herein, we investigated 46 patients diagnosed with EWS and treated in the Tel Aviv Medical Center between 2010 and 2021. TINs and NETs were identified in diagnostic biopsies of EWS by immunofluorescence. In addition, NETs were investigated in neutrophils isolated from peripheral blood samples of EWS patients at diagnosis and following neoadjuvant chemotherapy. The relationships between the presence of TINs and NETs, pathological and clinical features, and outcomes were analyzed. Our results demonstrate that TIN and NETs at diagnosis were higher in EWS patients with metastatic disease compared with those with local disease. High NET formation at diagnosis predicted poor response to neoadjuvant chemotherapy, relapse, and death from disease (p < 0.05). NET formation in peripheral blood samples at diagnosis was significantly elevated among patients with EWS compared with pediatric controls and decreased significantly following neoadjuvant chemotherapy. In conclusion, NET formation seems to have a role in the EWS immune microenvironment. Their presence can refine risk stratification, predict chemotherapy resistance and survival, and serve as a therapeutic target in patients with EWS.
Collapse
Affiliation(s)
- Rachel Shukrun
- Pediatric Hemato‐Oncology Research LaboratoryTel Aviv Medical CenterTel AvivIsrael
- Department of Pediatric Hemato‐OncologyTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Szilvia Baron
- Pediatric Hemato‐Oncology Research LaboratoryTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Victoria Fidel
- Pediatric Hemato‐Oncology Research LaboratoryTel Aviv Medical CenterTel AvivIsrael
| | - Anna Shusterman
- Pediatric Hemato‐Oncology Research LaboratoryTel Aviv Medical CenterTel AvivIsrael
| | - Osnat Sher
- Department of PathologyTel Aviv Medical CenterTel AvivIsrael
| | - Netanya Kollender
- Department of Pediatric Hemato‐OncologyTel Aviv Medical CenterTel AvivIsrael
| | - Dror Levin
- Department of Pediatric Hemato‐OncologyTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Yair Peled
- Department of Pediatric Hemato‐OncologyTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Yair Gortzak
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Pediatric Orthopedic Oncology Unit, National Unit of Orthopedic OncologyTel Aviv Medical CenterTel AvivIsrael
| | - Yoav Ben‐Shahar
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
- Department of Pediatric SurgeryTel Aviv Medical CenterTel AvivIsrael
| | - Revital Caspi
- Pediatric Hemato‐Oncology Research LaboratoryTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Sagi Gordon
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Michal Manisterski
- Department of Pediatric Hemato‐OncologyTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ronit Elhasid
- Pediatric Hemato‐Oncology Research LaboratoryTel Aviv Medical CenterTel AvivIsrael
- Department of Pediatric Hemato‐OncologyTel Aviv Medical CenterTel AvivIsrael
- Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
3
|
Daher M, Zalaquett Z, Chalhoub R, Abi Farraj S, Abdo M, Sebaaly A, Kourie HR, Ghanem I. Molecular and biologic biomarkers of Ewing sarcoma: A systematic review. J Bone Oncol 2023; 40:100482. [PMID: 37180735 PMCID: PMC10173001 DOI: 10.1016/j.jbo.2023.100482] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/23/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
With an annual incidence of less than 1%, Ewing sarcoma mainly occurs in children and young adults. It is not a frequent tumor but is the second most common bone malignancy in children. It has a 5-year survival rate of 65-75%; however, it has a poor prognosis when it relapses in patients. A genomic profile of this tumor can potentially help identify poor prognosis patients earlier and guide their treatment. A systematic review of the articles concerning genetic biomarkers in Ewing sarcoma was conducted using the Google Scholar, Cochrane, and PubMed database. There were 71 articles discovered. Numerous diagnostic, prognostic, and predictive biomarkers were found. However, more research is necessary to confirm the role of some of the mentioned biomarkers. .
Collapse
Affiliation(s)
- Mohammad Daher
- Orthopedic Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
- Corresponding author at: Hotel Dieu de France, Beirut, Lebanon.
| | - Ziad Zalaquett
- Hematology-Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Ralph Chalhoub
- Hematology-Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Sami Abi Farraj
- Orthopedic Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Majd Abdo
- Hematology-Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Amer Sebaaly
- Orthopedic Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Hampig-Raphaël Kourie
- Hematology-Oncology Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| | - Ismat Ghanem
- Orthopedic Department, Faculty of Medicine, Saint Joseph University of Beirut, Lebanon
| |
Collapse
|
4
|
Abrahao-Machado LF, Pinto F, Antunes B, Volc S, Boldrini E, Camargo OPD, Reis RM. Clinical impact of brachyury expression in Ewing sarcoma patients. Adv Med Sci 2021; 66:321-325. [PMID: 34273746 DOI: 10.1016/j.advms.2021.06.002] [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: 09/23/2020] [Revised: 05/21/2021] [Accepted: 06/25/2021] [Indexed: 10/20/2022]
Abstract
PURPOSE The T-box transcription factor brachyury has been demonstrated as a prognostic factor in a variety of cancer types and considered a novel oncotarget in solid tumors. Brachyury acts as a regulator of the epithelial-mesenchymal transition (EMT) process, leading to more aggressive behavior and poorer prognosis. However, recent literature evidence suggests a tumor suppressor role in other neoplasms. In the present study, we aimed to study brachyury expression and its prognostic impact in Ewing sarcoma, an aggressive neoplasm of young individuals. METHODS We analyzed the expression of brachyury by immunohistochemistry in a series of 96 Ewing sarcomas in a tissue microarray and investigated the association of the protein expression with the clinical parameters and overall survival. RESULTS More than half of the cases (51%, n = 49) depicted positive nuclear brachyury expression, while a lack of expression was observed in 49% (n = 47) of cases. Nuclear brachyury staining was significantly associated with non-white ethnicity (p = 0.04) and axial localization (p = 0.025). Importantly, lack of brachyury expression was significantly associated with lower overall survival in multivariate analyses (hazard ratio - HR: 2.227, p = 0.008). CONCLUSIONS Our findings indicate, that brachyury is an independent prognostic biomarker in Ewing sarcoma, which might suggest a tumor suppressor role and which yet to be fully elucidated.
Collapse
|
5
|
Hu Q, Qin Y, Ji S, Shi X, Dai W, Fan G, Li S, Xu W, Liu W, Liu M, Zhang Z, Ye Z, Zhou Z, Yang J, Zhuo Q, Yu X, Li M, Xu X. MTAP deficiency-induced metabolic reprogramming creates a vulnerability to co-targeting de novo purine synthesis and glycolysis in pancreatic cancer. Cancer Res 2021; 81:4964-4980. [PMID: 34385182 DOI: 10.1158/0008-5472.can-20-0414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/18/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022]
Abstract
Methylthioadenosine phosphorylase (MTAP) is a key enzyme associated with the salvage of methionine and adenine that is deficient in 20%-30% of pancreatic cancer. Our previous study revealed that MTAP-deficiency indicates a poor prognosis for pancreatic ductal adenocarcinoma (PDAC) patients. In this study, bioinformatics analysis of The Cancer Genome Atlas (TCGA) data indicated that PDACs with MTAP deficiency display a signature of elevated glycolysis. Metabolomics studies showed that that MTAP deletion-mediated metabolic reprogramming enhanced glycolysis and de novo purine synthesis in pancreatic cancer cells. Western blot analysis revealed that MTAP knockout stabilized hypoxia-inducible factor 1α (HIF-1α) protein via posttranslational phosphorylation. RIO kinase 1 (RIOK1), a downstream kinase upregulated in MTAP-deficient cells, interacted with and phosphorylated HIF-1α to regulate its stability. In vitro experiments demonstrated that the glycolysis inhibitor 2-deoxy-D-glucose (2-DG) and the de novo purine synthesis inhibitor L-alanosine synergized to kill MTAP-deficient pancreatic cancer cells. Collectively, these results reveal that MTAP deficiency drives pancreatic cancer progression by inducing metabolic reprogramming, providing a novel target and therapeutic strategy for treating MTAP-deficient disease.
Collapse
Affiliation(s)
- Qiangsheng Hu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xiuhui Shi
- Department of Medicine, Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Weixing Dai
- Cancer Research Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guixiong Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shuo Li
- Shanghai Pancreatic Cancer Institute, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wensheng Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Mengqi Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Zeng Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Zhijun Zhou
- Department of Medicine, Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Qifeng Zhuo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Min Li
- Department of Medicine, Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
| | - Xiaowu Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Curcumin Treatment Identifies Therapeutic Targets within Biomarkers of Liver Colonization by Highly Invasive Mesothelioma Cells-Potential Links with Sarcomas. Cancers (Basel) 2020; 12:cancers12113384. [PMID: 33207594 PMCID: PMC7696465 DOI: 10.3390/cancers12113384] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Aggressive sarcomatoid tumors designed in inbred strains of immunocompetent rats represent useful tools for both the identification of biomarkers of invasiveness and evaluation of innovative therapies. Our aim was to investigate the molecular determinants of liver colonization and potential common biomarkers of sarcomas and sarcomatoid tumors, using the most invasive (M5-T1) of our four experimental models of peritoneal sarcomatoid malignant mesothelioma in the F344 rat. Using an advanced and robust technique of quantitative proteomics and a bank of paraffin-embedded tumor and tissue samples, we analyzed changes in the proteotype patterns of the liver from normal rats, adjacent non-tumorous liver from untreated tumor-bearing rats, and liver from tumor-bearing rats positively responding to repeated administrations of curcumin given intraperitoneally. The identification of proteome alterations accounting for the antitumor effects of curcumin and changes in the liver microenvironment, which favored the induction of an immune response, could be useful to the research community. Abstract Investigations of liver metastatic colonization suggest that the microenvironment is preordained to be intrinsically hospitable to the invasive cancer cells. To identify molecular determinants of that organotropism and potential therapeutic targets, we conducted proteomic analyses of the liver in an aggressive model of sarcomatoid peritoneal mesothelioma (M5-T1). The quantitative changes between SWATH-MS (sequential window acquisition of all theoretical fragmentation spectra) proteotype patterns of the liver from normal rats (G1), adjacent non-tumorous liver from untreated tumor-bearing rats (G2), and liver from curcumin-treated rats without hepatic metastases (G3) were compared. The results identified 12 biomarkers of raised immune response against M5-T1 cells in G3 and 179 liver biomarker changes in (G2 vs. G1) and (G3 vs. G2) but not in (G3 vs. G1). Cross-comparing these 179 candidates with proteins showing abundance changes related to increasing invasiveness in four different rat mesothelioma tumor models identified seven biomarkers specific to the M5-T1 tumor. Finally, analysis of correlations between these seven biomarkers, purine nucleoside phosphorylase being the main biomarker of immune response, and the 179 previously identified proteins revealed a network orchestrating liver colonization and treatment efficacy. These results highlight the links between potential targets, raising interesting prospects for optimizing therapies against highly invasive cancer cells exhibiting a sarcomatoid phenotype and sarcoma cells.
Collapse
|
7
|
An immune-related gene signature for determining Ewing sarcoma prognosis based on machine learning. J Cancer Res Clin Oncol 2020; 147:153-165. [PMID: 32968877 DOI: 10.1007/s00432-020-03396-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Ewing sarcoma (ES) is one of the most common malignant bone tumors in children and adolescents. The immune microenvironment plays an important role in the development of ES. Here, we developed an optimal signature for determining ES patient prognosis based on immune-related genes (IRGs). METHODS We analyzed the ES gene expression profile dataset, GSE17679, from the GEO database and extracted differential expressed IRGs (DEIRGs). Then, we conducted functional correlation and protein-protein interaction (PPI) analyses of the DEIRGs and used the machine learning algorithm-iterative Lasso Cox regression analysis to build an optimal DEIRG signature. In addition, we applied ES samples from the ICGC database to test the optimal gene signature. We performed univariate and multivariate Cox regressions on clinicopathological characteristics and optimal gene signature to evaluate whether signature is an important prognostic factor. Finally, we calculated the infiltration of 24 immune cells in ES using the ssGSEA algorithm, and analyzed the correlation between the DEIRGs in the optimal gene signature and immune cells. RESULTS A total of 249 DEIRGs were screened and an 11-gene signature with the strongest correlation with patient prognoses was analyzed using a machine learning algorithm. The 11-gene signature also had a high prognostic value in the ES external verification set. Univariate and multivariate Cox regression analyses showed that 11-gene signature is an independent prognostic factor. We found that macrophages and cytotoxic, CD8 T, NK, mast, B, NK CD56bright, TEM, TCM, and Th2 cells were significantly related to patient prognoses; the infiltration of cytotoxic and CD8 T cells in ES was significantly different. By correlating prognostic biomarkers with immune cell infiltration, we found that FABP4 and macrophages, and NDRG1 and Th2 cells had the strongest correlation. CONCLUSION Overall, the IRG-related 11-gene signature can be used as a reliable ES prognostic biomarker and can provide guidance for personalized ES therapy.
Collapse
|
8
|
MTAP-deficiency could predict better treatment response in advanced lung adenocarcinoma patients initially treated with pemetrexed-platinum chemotherapy and bevacizumab. Sci Rep 2020; 10:843. [PMID: 31965001 PMCID: PMC6972892 DOI: 10.1038/s41598-020-57812-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
To investigate the predictive value of methylthioadenosine phosphorylase (MTAP) on treatment response and survival in advanced lung adenocarcinoma. MTAP expression was detected by immunohistochemistry. Treatment response and survival were compared according to MTAP expression level. The results indicated MTAP-low expression was observed in 61.2% (101/165) of all patients. The objective response rate and disease control rate improved in the MTAP-low group (64.4% vs 46.9%, p = 0.035; 92.1% vs. 79.7%, p = 0.03; respectively). The median progression-free survival and survival time in the MTAP-low group were significantly lower than that in the MTAP-high group (8.1 vs. 13.1 months, p = 0.002; 22 vs. 32 months, p = 0.044). Multivariate analysis demonstrated that brain metastasis (HR 1.55, p = 0.046), thoracic radiation (HR 0.52, p = 0.026), and MTAP-low expression (HR 1.36, p = 0.038) were independent factors on survival. It is concluded that MTAP-low expression could predict improved treatment response but worsened survival in advanced lung adenocarcinoma.
Collapse
|