1
|
McLean TD, Duchi S, Di Bella C. Molecular Pathogenesis of Sporadic Desmoid Tumours and Its Implications for Novel Therapies: A Systematised Narrative Review. Target Oncol 2022; 17:223-252. [PMID: 35446005 PMCID: PMC9217905 DOI: 10.1007/s11523-022-00876-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 12/22/2022]
Abstract
Sporadic desmoid-type fibromatosis is a rare, fibroblastic soft-tissue neoplasm with local aggressiveness but no metastatic potential. Aberrant Wnt/β-catenin signalling has been extensively linked to desmoid pathogenesis, although little is known about other molecular drivers and no established treatment approach exists. We aimed to summarise the current literature regarding the molecular pathogenesis of sporadic desmoid-type fibromatosis and to discuss the effects of both current and emerging novel therapies targeting these mechanisms. A literature search was conducted of MEDLINE® ALL and EMBASE databases for published studies (2000–August 2021) using keywords related to ‘fibromatosis aggressive’, ‘immunohistochemistry’, ‘polymerase chain reaction’ and ‘mutation’. Articles were included if they examined the role of proteins in sporadic or extra-abdominal human desmoid-type fibromatosis pathogenesis. Searching identified 1684 articles. Following duplicate removal and eligibility screening, 36 were identified. After a full-text screen, 22 were included in the final review. At least 47% of desmoid-type fibromatosis cases displayed aberrant β-catenin immunoreactivity amongst ten studies. Cyclin D1 overexpression occurred in at least 40% of cases across five studies. Six studies reported oestrogen receptor-β expression with a range of 7.4–90%. Three studies implicated matrix metalloproteinases, with one study demonstrating vascular endothelial growth factor overexpression. One study explored the positive relationship between cyclooxygenase-2 and platelet-derived growth factor receptor-β. Aberrant Wnt/β-catenin signalling is a well-established pathogenic driver that may be targeted via downstream modulation. Growth factor signalling is best appreciated through the clinical trial effects of multi-targeted tyrosine kinase inhibitors, whilst oestrogen receptor expression data may only offer a superficial insight into oestrogen signalling. Finally, the tumour microenvironment presents multiple potential novel therapeutic targets. Sporadic desmoid tumours are rare soft-tissue neoplasms that arise from connective tissues in the chest wall, head, neck and limbs. Whilst lacking metastatic potential, uncertainty surrounding their locally aggressive growth and unpredictable recurrence complicates treatment approaches. At the molecular level, alterations in the Wnt/β-catenin signalling pathway, a fundamental coordinator of cell growth and development, have been strongly linked to desmoid tumour development. Beyond this, however, little is known about other molecular drivers. In the case of progressive or life-threatening disease, complex treatment decisions are made regarding the use of surgery, radiotherapy or systemic treatment modalities. Of the targeted systemic therapies, a lack of comparative clinical studies further complicates medical treatment decision making as no definitive treatment approach exists. Therefore, this review aimed to summarise the literature regarding the molecular drivers of desmoid tumour pathogenesis and to discuss the current and emerging novel therapies targeting such mechanisms. Utilising findings from human desmoid tissue samples, we present the rationale for targeting downstream mediators of the central Wnt/β-catenin pathway and outline potential treatment targets in the tumour microenvironment. We also highlight the knowledge gained from clinical drug trials targeting desmoid growth factor signalling and present the potentially superficial insight provided by oestrogen receptor expression profiles on the role of oestrogen signalling in desmoid pathogenesis. In doing so, this work may assist in the eventual development of an evidence-based treatment approach for sporadic desmoid tumours.
Collapse
Affiliation(s)
- Thomas D McLean
- Melbourne Medical School, The University of Melbourne, Melbourne, VIC, Australia.
| | - Serena Duchi
- Department of Surgery, The University of Melbourne, Melbourne, VIC, Australia.,Biofab 3D, Aikenhead Centre for Medical Discovery, Melbourne, VIC, Australia
| | - Claudia Di Bella
- Department of Surgery, The University of Melbourne, Melbourne, VIC, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, VIC, Australia
| |
Collapse
|
2
|
CRISPR-SID: Identifying EZH2 as a druggable target for desmoid tumors via in vivo dependency mapping. Proc Natl Acad Sci U S A 2021; 118:2115116118. [PMID: 34789568 DOI: 10.1073/pnas.2115116118] [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] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer precision medicine implies identification of tumor-specific vulnerabilities associated with defined oncogenic pathways. Desmoid tumors are soft-tissue neoplasms strictly driven by Wnt signaling network hyperactivation. Despite this clearly defined genetic etiology and the strict and unique implication of the Wnt/β-catenin pathway, no specific molecular targets for these tumors have been identified. To address this caveat, we developed fast, efficient, and penetrant genetic Xenopus tropicalis desmoid tumor models to identify and characterize drug targets. We used multiplexed CRISPR/Cas9 genome editing in these models to simultaneously target a tumor suppressor gene (apc) and candidate dependency genes. Our methodology CRISPR/Cas9 selection-mediated identification of dependencies (CRISPR-SID) uses calculated deviations between experimentally observed gene editing outcomes and deep-learning-predicted double-strand break repair patterns to identify genes under negative selection during tumorigenesis. This revealed EZH2 and SUZ12, both encoding polycomb repressive complex 2 components, and the transcription factor CREB3L1 as genetic dependencies for desmoid tumors. In vivo EZH2 inhibition by Tazemetostat induced partial regression of established autochthonous tumors. In vitro models of patient desmoid tumor cells revealed a direct effect of Tazemetostat on Wnt pathway activity. CRISPR-SID represents a potent approach for in vivo mapping of tumor vulnerabilities and drug target identification.
Collapse
|
3
|
The Prognostic Role of β-Catenin Mutations in Desmoid-type Fibromatosis Undergoing Resection Only: A Meta-analysis of Individual Patient Data. Ann Surg 2021; 273:1094-1101. [PMID: 31804402 DOI: 10.1097/sla.0000000000003698] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVE This meta-analysis (PROSPERO CRD42018100653) uses individual patient data (IPD) to assess the association between recurrence and CTNNB1 mutation status in surgically treated adult desmoid-type fibromatosis (DTF) patients. SUMMARY OF BACKGROUND DATA The majority of sporadic DTF tumors harbor a CTNNB1 (ß-catenin) mutation: T41A, S45F, and S45P or are wild-type (WT). Results are conflicting regarding the recurrence risk after surgery for these mutation types. METHODS A systematic literature search was performed on June 6th, 2018. IPD from eligible studies was used to analyze differences in recurrence according to CTNNB1 mutation status using Cox proportional hazards analysis. Predictive factors included: sex, age, mutation type, tumor site, tumor size, resection margin status, and cohort. The PRISMA-IPD guideline was used. RESULTS Seven studies, describing retrospective cohorts were included and the IPD of 329 patients were used of whom 154 (46.8%) had a T41A mutation, 66 (20.1%) a S45F mutation, and 24 (7.3%) a S45P mutation, whereas 85 (25.8%) patients had a WT CTNNB1. Eighty-three patients (25.2%) experienced recurrence. Multivariable analysis, adjusting for sex, age, and tumor site yielded a P-value of 0.011 for CTNNB1 mutation. Additional adjustment for tumor size yielded a P-value of 0.082 with hazard ratio's of 0.83 [95% confidence interval (CI) 0.48-1.42), 0.37 (95% CI 0.12-1.14), and 0.44 (95% CI 0.21-0.92) for T41A, S45P and WT DTF tumors compared to S45F DTF tumors. The effect modification between tumor size and mutation type suggests that tumor size is an important mediator for recurrence. CONCLUSIONS Primary sporadic DTFs harboring a CTNNB1 S45F mutation have a higher risk of recurrence after surgery compared to T41A, S45P, and WT DTF, but this association seems to be mediated by tumor size.
Collapse
|
4
|
Lee JM, Kim HG, Shin SY, Lee SH. Clinical application of next-generation sequencing for the management of desmoid tumors: A case report and literature review. Medicine (Baltimore) 2021; 100:e24238. [PMID: 33429825 PMCID: PMC7793316 DOI: 10.1097/md.0000000000024238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023] Open
Abstract
RATIONALE Desmoid tumors are rare myofibroblastic neoplasms characterized by local invasiveness and high rates of recurrence, and sometimes mimic local recurrence of previously resected malignancies. Previous studies have suggested that molecular profiling may be useful for the diagnosis of the tumors and risk stratification. However, the clinical utility of next-generation sequencing (NGS) for the management of desmoid tumors has not been established. PATIENT CONCERNS A 42-year-old man visited our clinic for routine follow-up 1 year after left upper lobe lingular segmentectomy for lung adenocarcinoma. DIAGNOSES Chest computed tomography showed a pleural mass adherent to the thoracotomy site. Positron emission tomography revealed mildly increased metabolism with a maximal standardized uptake value of 2.7 within the tumor, suggesting local recurrence of the previous neoplasm. Exploratory thoracotomy and en bloc resection of the tumor revealed spindle cells in a massive collagenous tissue consistent with a desmoid tumor. INTERVENTIONS NGS was performed to confirm the diagnosis and to identify any genetic alterations that might be relevant to the prognosis of this tumor. The tumor harbored an S45F mutation in CTNNB1, which has been correlated with a high recurrence rate. Therefore, we performed adjuvant radiotherapy on the resection bed at a dose of 56 Gy. OUTCOMES The patients experienced no postoperative or radiotherapy-related complications. Periodic follow-up examinations using computed tomography were performed every 3 months, and no evidence of recurrence of either tumor was observed during the 38 months after the last surgery. LESSONS To the best of our knowledge, this is the first case reporting the clinical application of NGS and aggressive treatment based on the genotyping results for the management of a desmoid tumor. Our case highlights the need to consider desmoid tumors among the differential diagnoses when a pleural mass is encountered at a previous thoracotomy site. More importantly, molecular profiling using NGS can be useful for the establishment of a treatment strategy for this tumor, although further investigations are required.
Collapse
Affiliation(s)
- Jeong Mi Lee
- Department of Medicine, Graduate School, Kyung Hee University
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University School of Medicine
| | - Han Gyeol Kim
- Department of Medicine, Graduate School, Kyung Hee University
- Department of Pathology
| | - So Youn Shin
- Department of Radiology, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, South Korea
| | - Seung Hyeun Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University School of Medicine
| |
Collapse
|
5
|
Sekimizu M, Yoshida A, Mitani S, Asano N, Hirata M, Kubo T, Yamazaki F, Sakamoto H, Kato M, Makise N, Mori T, Yamazaki N, Sekine S, Oda I, Watanabe S, Hiraga H, Yonemoto T, Kawamoto T, Naka N, Funauchi Y, Nishida Y, Honoki K, Kawano H, Tsuchiya H, Kunisada T, Matsuda K, Inagaki K, Kawai A, Ichikawa H. Frequent mutations of genes encoding vacuolar H
+
‐ATPase components in granular cell tumors. Genes Chromosomes Cancer 2019; 58:373-380. [DOI: 10.1002/gcc.22727] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/25/2018] [Accepted: 12/27/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Masaya Sekimizu
- Department of Clinical GenomicsNational Cancer Center Research Institute Tokyo Japan
- Department of Musculoskeletal OncologyNational Cancer Center Hospital Tokyo Japan
- Department of Orthopaedic SurgeryShowa University School of Medicine Tokyo Japan
| | - Akihiko Yoshida
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital Tokyo Japan
| | - Sachiyo Mitani
- Department of Clinical GenomicsNational Cancer Center Research Institute Tokyo Japan
| | - Naofumi Asano
- Department of Orthopaedic SurgeryKeio University School of Medicine Tokyo Japan
| | - Makoto Hirata
- Laboratory of Genome TechnologyInstitute of Medical Science, University of Tokyo Tokyo Japan
| | - Takashi Kubo
- Department of Clinical GenomicsNational Cancer Center Research Institute Tokyo Japan
- Division of Translational GenomicsNational Cancer Center‐Exploratory Oncology Research & Clinical Trial Center Tokyo Japan
| | - Fumito Yamazaki
- Department of Clinical GenomicsNational Cancer Center Research Institute Tokyo Japan
- Department of PediatricsKeio University School of Medicine Tokyo Japan
| | - Hiromi Sakamoto
- Department of Clinical GenomicsNational Cancer Center Research Institute Tokyo Japan
| | - Mamoru Kato
- Department of BioinformaticsNational Cancer Center Research Institute Tokyo Japan
| | - Naohiro Makise
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital Tokyo Japan
| | - Taisuke Mori
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital Tokyo Japan
| | - Naoya Yamazaki
- Department of Dermatologic OncologyNational Cancer Center Hospital Tokyo Japan
| | - Shigeki Sekine
- Department of Pathology and Clinical LaboratoriesNational Cancer Center Hospital Tokyo Japan
| | - Ichiro Oda
- Endoscopy DivisionNational Cancer Center Hospital Tokyo Japan
| | - Shun‐ichi Watanabe
- Department of Thoracic SurgeryNational Cancer Center Hospital Tokyo Japan
| | - Hiroaki Hiraga
- Department of Orthopaedic SurgeryHokkaido Cancer Center Sapporo Japan
| | | | - Teruya Kawamoto
- Department of Orthopaedic SurgeryKobe University Graduate School of Medicine Kobe Japan
| | - Norifumi Naka
- Musculoskeletal Oncology ServiceOsaka International Cancer Institute Osaka Japan
| | - Yuki Funauchi
- Department of Orthopaedic SurgeryThe Cancer Institute, Japanese Foundation for Cancer Research Tokyo Japan
| | | | - Kanya Honoki
- Department of Orthopedic SurgeryNara Medical University Kashihara Nara Japan
| | - Hirotaka Kawano
- Department of Orthopaedic SurgeryTeikyo University School of Medicine Tokyo Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic SurgeryKanazawa University Graduate School of Medical Sciences Kanazawa Japan
| | - Toshiyuki Kunisada
- Department of Orthopaedic SurgeryOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences Okayama Japan
| | - Koichi Matsuda
- Laboratory of Clinical Genome SequencingGraduate School of Frontier Sciences, University of Tokyo Tokyo Japan
| | - Katsunori Inagaki
- Department of Orthopaedic SurgeryShowa University School of Medicine Tokyo Japan
| | - Akira Kawai
- Department of Musculoskeletal OncologyNational Cancer Center Hospital Tokyo Japan
| | - Hitoshi Ichikawa
- Department of Clinical GenomicsNational Cancer Center Research Institute Tokyo Japan
- Division of Translational GenomicsNational Cancer Center‐Exploratory Oncology Research & Clinical Trial Center Tokyo Japan
| |
Collapse
|
6
|
TGF-β and CTGF are Mitogenic Output Mediators of Wnt/β-Catenin Signaling in Desmoid Fibromatosis. Appl Immunohistochem Mol Morphol 2018; 25:559-565. [PMID: 26894649 DOI: 10.1097/pai.0000000000000340] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Desmoid fibromatosis is a locally aggressive clonal fibroblastic proliferation with high recurrence rates and no metastatic potential. Implicated molecular aberrations occur within the Wnt/β-catenin pathway (APC and β-catenin gene mutations). Transforming growth factor-β (TGF-β) and connective tissue growth factor (CTGF) are profibrotic growth factors, downstream from nuclear translocation of β-catenin, that lead to increased fibrogenesis. CTGF (a downstream effector of TGF-β) is a matricellular protein that modulates the activity of growth factors, adhesion molecules, integrins, and extracellular matrix thus playing a central role in tissue remodeling and fibrosis. Recently there has been growing interest in use of extracellular matrix inhibitors for treatment of various fibrogenic diseases. Desmoid fibromatosis samples (n=15) were evaluated for expression of β-catenin, TGF-β, and CTGF using immunohistochemistry on formalin paraffin-embedded material. A control group comprising scar tissue and adjacent normal skin (n=10) were simultaneously immunostained with above mentioned markers. Real-time polymerase chain reaction was performed on frozen specimens of desmoid fibromatosis (n=6) and normal skin (n=2). All 15 desmoid tumors were positive for β-catenin (surrogate marker of Wnt/β-catenin pathway dysregulation) which was negative in control normal skin and scar samples. TGF-β and CTGF were negative in 9 of 10 normal skin controls. TGF-β and CTGF were positive in all cases of scar tissue. All 15 cases of desmoid tumors were positive for TGF-β and CTGF. The real-time polymerase chain reaction showed higher expression levels of TGF-β and CTGF in desmoid fibromatosis compared with normal skin. The high constitutive expression of β-catenin downstream effectors; TGF-β, CTGF has the potential for enabling targeted therapy.
Collapse
|
7
|
Zhang L, Song X, Shao Y, Wu C, Jiang J. Prognostic value of Midkine expression in patients with solid tumors: a systematic review and meta-analysis. Oncotarget 2018; 9:24821-24829. [PMID: 29872508 PMCID: PMC5973861 DOI: 10.18632/oncotarget.23892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/13/2017] [Indexed: 12/23/2022] Open
Abstract
Background Accumulated studies have shown the important role of Midkine (MDK) protein in various solid tumors and indicated its correlation with patients' survival. This meta-analysis was performed to further explore the prognostic value of MDK expression in solid tumors. Materials and Methods We collected the literatures through searching PubMed, Embase and the Cochrane Library (last up to April 10, 2017) to assess the effect of MDK on survival in solid tumor patients. The STATA 12.0 software was used for the meta-analysis. Fixed-effects models or random-effects models were used to estimate the pooled hazard ratios (HRs) for overall survival (OS). Results A total of 2097 patients from 17 observational studies were summarized. High expression of MDK was notably associated with worse OS in solid tumor patients. (pooled HR = 1.96; 95% CI = 1.67-2.31). The subgroup analysis of tumor type demonstrated negative impact of elevated MDK on OS in most solid tumor patients (P < 0.05), while MDK had no relevance with OS in the patients with OSCC (pooled HR = 1.68; 95% CI = 0.84-3.36; P = 0.145) or HNSCC (pooled HR = 1.56; 95% CI = 0.96-2.51; P = 0.075). Conclusions The present meta-analysis clarifies that MDK is a potential prognostic biomarker in solid tumor patients. Future large-scale prospective clinical trials are needed to determine the prognostic value of MDK in solid tumor patients.
Collapse
Affiliation(s)
- Luo Zhang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Xing Song
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Yingjie Shao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Changping Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| |
Collapse
|
8
|
Colombo C, Belfiore A, Paielli N, De Cecco L, Canevari S, Laurini E, Fermeglia M, Pricl S, Verderio P, Bottelli S, Fiore M, Stacchiotti S, Palassini E, Gronchi A, Pilotti S, Perrone F. β-Catenin in desmoid-type fibromatosis: deep insights into the role of T41A and S45F mutations on protein structure and gene expression. Mol Oncol 2017. [PMID: 28627792 PMCID: PMC5664003 DOI: 10.1002/1878-0261.12101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Desmoid‐type fibromatosis (DF) is a rare mesenchymal lesion with high risk of local recurrence. Specific β‐catenin mutations (S45F) appeared to be related to this higher risk compared to T41A‐mutated or wild‐type (WT). We explored the influence of both mutations and WT on structure stability and affinity of β‐catenin for α‐catenin and the pattern of gene expression that may influence DF behavior. Using 33 surgically resected primary DFs harboring T41A (n = 14), S45F (n = 10), or WT (n = 9), we performed a comparative molecular analysis by protein/protein interaction modeling, gene expression by DASL microarrays, human inflammation gene panel, and assessment of immune system‐based biomarkers by immunohistochemistry. Mutated proteins were more stable than WT and formed a weaker complex with α‐catenin. Consensus unsupervised gene clustering revealed the presence of two DF group‐mutated (T41A + S45F) and WT (P = 0.0047). The gene sets ‘Inflammatory‐Defense‐Humoral Immune Response’ and ‘Antigen Binding’ were significantly enriched in T41A. The deregulation of 16 inflammation‐related genes was confirmed. Low numbers of T cells and tumor‐associated macrophages (TAM) infiltrating the tumors and low/absent PD‐1/PD‐L1 expression were also identified. We demonstrated that mutated DFs (T41A or S45F) and WT are two distinct molecular subgroups with regard to β‐catenin stability, α‐catenin affinity, and gene expression profiling. A different inflammation signature characterized the two mutated groups, suggesting mediation either by T41A or by S45F. Finally, all mutated cases showed a low number of TIL and TAM cells and a low or absent expression of PD‐1 and PD‐L1 consistent with β‐catenin activation insensitive to checkpoint blockade.
Collapse
Affiliation(s)
- Chiara Colombo
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Antonino Belfiore
- Laboratory of Experimental Molecular Pathology, Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Nicholas Paielli
- Laboratory of Experimental Molecular Pathology, Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Loris De Cecco
- Functional Genomics and Bioinformatics, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Canevari
- Functional Genomics and Bioinformatics, Department of Applied Research and Technology Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Erik Laurini
- Molecular Simulation Engineering (MOSE) Laboratory, DEA, University of Trieste, Italy
| | - Maurizio Fermeglia
- Molecular Simulation Engineering (MOSE) Laboratory, DEA, University of Trieste, Italy
| | - Sabrina Pricl
- Molecular Simulation Engineering (MOSE) Laboratory, DEA, University of Trieste, Italy
| | - Paolo Verderio
- Unit of Medical Statistics, Biometry and Bioinformatics, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Stefano Bottelli
- Unit of Medical Statistics, Biometry and Bioinformatics, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Marco Fiore
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Silvia Stacchiotti
- Adult Mesenchymal Tumor Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Elena Palassini
- Adult Mesenchymal Tumor Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Alessandro Gronchi
- Sarcoma Service, Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Experimental Molecular Pathology, Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Federica Perrone
- Laboratory of Experimental Molecular Pathology, Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
9
|
Hayashi K, Takamura M, Yokoyama H, Sato Y, Yamagiwa S, Nogami H, Wakai T, Hasegawa G, Terai S. A Mesenteric Desmoid Tumor with Rapid Progression. Intern Med 2017; 56:505-508. [PMID: 28250295 PMCID: PMC5399200 DOI: 10.2169/internalmedicine.56.7320] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
We herein report the case of a rapidly progressive sporadic mesenteric desmoid tumor (DT). A 62-year-old woman presented with a 4-cm-diameter palpable mass in the left supraumbilical area. The mass showed an ill-defined margin with heterogeneous delayed enhancement on computed tomography and heterogeneous high intensity on T2-weighted magnetic resonance imaging. Sixteen months after the initial observation, the mass had grown in size, reaching 13 cm in diameter. The resected mass was histologically confirmed as a DT of the mesentery. Since DT often has an unpredictable clinical course, clinicians should bear in mind the need for imaging follow-up.
Collapse
Affiliation(s)
- Kazunao Hayashi
- Division of Gastroenterology and Hepatology, Niigata University Graduate School of Medical and Dental Sciences, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Kim HS, Kim J, Nam KH, Kim WH. Clinical significance of midkine expression in sporadic desmoid tumors. Oncol Lett 2016; 11:1677-1684. [PMID: 26998061 PMCID: PMC4774436 DOI: 10.3892/ol.2016.4129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 12/23/2015] [Indexed: 11/17/2022] Open
Abstract
The aim of the present study was to identify the prognostic factors for the propensity for recurrence in sporadic desmoid tumors. The catenin (cadherin-associated protein) β1 (CTNNB1) genotypes and expression of Wnt pathway proteins and midkine (also termed neurite growth-promoting factor 2) were investigated in 159 patients with sporadic desmoid tumors. Formalin-fixed paraffin-embedded tissues of the surgically resected desmoid tumors were examined by direct sequencing of CTNNB1 exon 3, and immunostained for the expression of β-catenin, T-cell factor 4 (TCF-4), phosphorylated protein kinase B (pAkt), midkine and menin using a tissue microarray method. Among the samples, 70% (111/159) exhibited point mutations of the CTNNB1 gene, including T41A (56%), S45F (8%), S45P (2%), S45N (2%) and T42A (1%). In addition, 100, 57, 24, 15 and 92% of the tumors expressed β-catenin, TCF-4, midkine, pAkt and menin, respectively. Positive midkine expression was significantly associated with the recurrence of tumors (P=0.001). The multivariate analysis of recurrence demonstrated that an extra-abdominal tumor site [hazard ratio (HR), 2.625; P=0.001] and midkine expression (HR, 2.077; P<0.009) were independent prognostic factors of tumor recurrence. In conclusion, the present results suggest that the tumor site and midkine expression may be predictive markers for the recurrence of sporadic desmoid tumors.
Collapse
Affiliation(s)
- Hee Sung Kim
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul 156-755, Republic of Korea
| | - Jin Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| | - Kyung Han Nam
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan 612-896, Republic of Korea
| | - Woo Ho Kim
- Cancer Research Institute, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea; Department of Pathology, College of Medicine, Seoul National University, Seoul 110-799, Republic of Korea
| |
Collapse
|
12
|
Shang H, Braggio D, Lee YJ, Al Sannaa GA, Creighton CJ, Bolshakov S, Lazar AJF, Lev D, Pollock RE. Targeting the Notch pathway: A potential therapeutic approach for desmoid tumors. Cancer 2015; 121:4088-96. [PMID: 26349011 PMCID: PMC4635059 DOI: 10.1002/cncr.29564] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 01/23/2015] [Accepted: 02/12/2015] [Indexed: 12/29/2022]
Abstract
BACKGROUND Desmoid tumors (DTs) are rare mesenchymal lesions that can recur repeatedly. When it is feasible, DTs are surgically resected; however, this often results in high recurrence rates. Recently, treatment with PF‐03084014, a potent γ‐secretase inhibitor, has been shown to have antitumor activity in several tumor types by affecting the WNT/β‐catenin pathway. Consequently, Notch pathway inhibition by PF‐03084014 might be a promising approach for DT treatment. METHODS The expression of Notch pathway components was analyzed in DT tissues and cell strains with immunohistochemistry and Western blotting, respectively. A panel of DT cell strains was exposed to PF‐03084014 and evaluated for cell proliferation. Antitumor effects were assessed via cell cycle, apoptosis, and migration and invasion analysis. Cells treated with PF‐03084014 were characterized with a gene array analysis combined with Ingenuity Pathway Analysis. RESULTS The results showed that Notch pathway components were expressed at different levels in DTs. Hes1 (Hes Family BHLH Transcription Factor 1) was overexpressed in DT tumors versus dermal scar tissue, and PF‐03084014 caused significant decreases in Notch intracellular domain and Hes1 expression in DT cell strains. PF‐03084014 decreased DT cell migration and invasion and also caused cell growth inhibition in DT cell strains, most likely through cell cycle arrest. Gene array analysis combined with Ingenuity Pathway Analysis showed that Wnt1‐inducible signaling pathway protein 2 possibly regulated Notch and WNT pathways after treatment with PF‐03084014 through integrin. CONCLUSION Our findings suggest that the Notch pathway is an important DT therapeutic target. Furthermore, PF‐03084014 has significant antitumor activity against DTs, and it may be an alternative strategy for DT treatment. Cancer 2015;121:4088–4096. © 2015 American Cancer Society. The Notch pathway is an important therapeutic target for desmoid tumors, and the γ‐secretase inhibitor PF‐03084014 has significant antitumor activity against desmoid tumors. The use of this inhibitor may be an alternative strategy for desmoid tumor treatment.
Collapse
Affiliation(s)
- Hui Shang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Orthopedics, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Danielle Braggio
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Surgical Oncology, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio.,Department of Investigative Pathology, International Center for Research, A. C. Camargo Cancer Center, São Paulo, Brazil
| | - Ya-Jung Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ghadah A Al Sannaa
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chad J Creighton
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas
| | - Svetlana Bolshakov
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander J F Lazar
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dina Lev
- Surgery B, Sheba Medical Center, Tel Aviv, Israel
| | - Raphael E Pollock
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Surgical Oncology, Comprehensive Cancer Center, Ohio State University, Columbus, Ohio.,Sarcoma Research Center, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
13
|
Salas S, Brulard C, Terrier P, Ranchere-Vince D, Neuville A, Guillou L, Lae M, Leroux A, Verola O, Jean-Emmanuel K, Bonvalot S, Blay JY, Le Cesne A, Aurias A, Coindre JM, Chibon F. Gene Expression Profiling of Desmoid Tumors by cDNA Microarrays and Correlation with Progression-Free Survival. Clin Cancer Res 2015; 21:4194-200. [DOI: 10.1158/1078-0432.ccr-14-2910] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/28/2015] [Indexed: 11/16/2022]
|
14
|
Enzo MV, Rastrelli M, Rossi CR, Hladnik U, Segat D. The Wnt/β-catenin pathway in human fibrotic-like diseases and its eligibility as a therapeutic target. MOLECULAR AND CELLULAR THERAPIES 2015; 3:1. [PMID: 26056602 PMCID: PMC4452070 DOI: 10.1186/s40591-015-0038-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/04/2015] [Indexed: 02/06/2023]
Abstract
The canonical Wnt signaling pathway is involved in a variety of biological processes like cell proliferation, cell polarity, and cell fate determination. This pathway has been extensively investigated as its deregulation is linked to different diseases, including various types of cancer, skeletal defects, birth defect disorders (including neural tube defects), metabolic diseases, neurodegenerative disorders and several fibrotic diseases like desmoid tumors. In the "on state", beta-catenin, the key effector of Wnt signaling, enters the nucleus where it binds to the members of the TCF-LEF family of transcription factors and exerts its effect on gene transcription. Disease development can be caused by direct or indirect alterations of the Wnt/β-catenin signaling. In the first case germline or somatic mutations of the Wnt components are associated to several diseases such as the familial adenomatous polyposis (FAP) - caused by germline mutations of the tumor suppressor adenomatous polyposis coli gene (APC) - and the desmoid-like fibromatosis, a sporadic tumor associated with somatic mutations of the β-catenin gene (CTNNB1). In the second case, epigenetic modifications and microenvironmental factors have been demonstrated to play a key role in Wnt pathway activation. The natural autocrine Wnt signaling acts through agonists and antagonists competing for the Wnt receptors. Anomalies in this regulation, whichever is their etiology, are an important part in the pathogenesis of Wnt pathway linked diseases. An example is promoter hypermethylation of Wnt antagonists, such as SFRPs, that causes gene silencing preventing their function and consequently leading to the activation of the Wnt pathway. Microenvironmental factors, such as the extracellular matrix, growth factors and inflammatory mediators, represent another type of indirect mechanism that influence Wnt pathway activation. A favorable microenvironment can lead to aberrant fibroblasts activation and accumulation of ECM proteins with subsequent tissue fibrosis that can evolve in fibrotic disease or tumor. Since the development and progression of several diseases is the outcome of the Wnt pathway cross-talk with other signaling pathways and inflammatory factors, it is important to consider not only direct inhibitors of the Wnt signaling pathway but also inhibitors of microenvironmental factors as promising therapeutic approaches for several tumors of fibrotic origin.
Collapse
Affiliation(s)
- Maria Vittoria Enzo
- Genetics Unit, "Mauro Baschirotto" Institute for Rare Diseases, Via B. Bizio, 1- 36023 Vicenza, Italy
| | - Marco Rastrelli
- Melanoma and Sarcoma Unit, Veneto Institute of Oncology, IOV-IRCSS, Via Gattamelata, 64-35128 Padua, Italy
| | - Carlo Riccardo Rossi
- Melanoma and Sarcoma Unit, Veneto Institute of Oncology, IOV-IRCSS, Via Gattamelata, 64-35128 Padua, Italy ; Department of Surgical Oncological and Gastroenterological Science, University of Padua, Via Giustiniani, 2- 35124 Padua, Italy
| | - Uros Hladnik
- Genetics Unit, "Mauro Baschirotto" Institute for Rare Diseases, Via B. Bizio, 1- 36023 Vicenza, Italy
| | - Daniela Segat
- Genetics Unit, "Mauro Baschirotto" Institute for Rare Diseases, Via B. Bizio, 1- 36023 Vicenza, Italy
| |
Collapse
|
15
|
Enzo MV, Rastrelli M, Rossi CR, Hladnik U, Segat D. The Wnt/β-catenin pathway in human fibrotic-like diseases and its eligibility as a therapeutic target. MOLECULAR AND CELLULAR THERAPIES 2015; 3:1. [PMID: 26056602 PMCID: PMC4452070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 01/04/2015] [Indexed: 11/21/2023]
Abstract
The canonical Wnt signaling pathway is involved in a variety of biological processes like cell proliferation, cell polarity, and cell fate determination. This pathway has been extensively investigated as its deregulation is linked to different diseases, including various types of cancer, skeletal defects, birth defect disorders (including neural tube defects), metabolic diseases, neurodegenerative disorders and several fibrotic diseases like desmoid tumors. In the "on state", beta-catenin, the key effector of Wnt signaling, enters the nucleus where it binds to the members of the TCF-LEF family of transcription factors and exerts its effect on gene transcription. Disease development can be caused by direct or indirect alterations of the Wnt/β-catenin signaling. In the first case germline or somatic mutations of the Wnt components are associated to several diseases such as the familial adenomatous polyposis (FAP) - caused by germline mutations of the tumor suppressor adenomatous polyposis coli gene (APC) - and the desmoid-like fibromatosis, a sporadic tumor associated with somatic mutations of the β-catenin gene (CTNNB1). In the second case, epigenetic modifications and microenvironmental factors have been demonstrated to play a key role in Wnt pathway activation. The natural autocrine Wnt signaling acts through agonists and antagonists competing for the Wnt receptors. Anomalies in this regulation, whichever is their etiology, are an important part in the pathogenesis of Wnt pathway linked diseases. An example is promoter hypermethylation of Wnt antagonists, such as SFRPs, that causes gene silencing preventing their function and consequently leading to the activation of the Wnt pathway. Microenvironmental factors, such as the extracellular matrix, growth factors and inflammatory mediators, represent another type of indirect mechanism that influence Wnt pathway activation. A favorable microenvironment can lead to aberrant fibroblasts activation and accumulation of ECM proteins with subsequent tissue fibrosis that can evolve in fibrotic disease or tumor. Since the development and progression of several diseases is the outcome of the Wnt pathway cross-talk with other signaling pathways and inflammatory factors, it is important to consider not only direct inhibitors of the Wnt signaling pathway but also inhibitors of microenvironmental factors as promising therapeutic approaches for several tumors of fibrotic origin.
Collapse
Affiliation(s)
- Maria Vittoria Enzo
- />Genetics Unit, “Mauro Baschirotto” Institute for Rare Diseases, Via B. Bizio, 1- 36023 Vicenza, Italy
| | - Marco Rastrelli
- />Melanoma and Sarcoma Unit, Veneto Institute of Oncology, IOV-IRCSS, Via Gattamelata, 64-35128 Padua, Italy
| | - Carlo Riccardo Rossi
- />Melanoma and Sarcoma Unit, Veneto Institute of Oncology, IOV-IRCSS, Via Gattamelata, 64-35128 Padua, Italy
- />Department of Surgical Oncological and Gastroenterological Science, University of Padua, Via Giustiniani, 2- 35124 Padua, Italy
| | - Uros Hladnik
- />Genetics Unit, “Mauro Baschirotto” Institute for Rare Diseases, Via B. Bizio, 1- 36023 Vicenza, Italy
| | - Daniela Segat
- />Genetics Unit, “Mauro Baschirotto” Institute for Rare Diseases, Via B. Bizio, 1- 36023 Vicenza, Italy
| |
Collapse
|
16
|
Barderas R, Mendes M, Torres S, Bartolomé RA, López-Lucendo M, Villar-Vázquez R, Peláez-García A, Fuente E, Bonilla F, Casal JI. In-depth characterization of the secretome of colorectal cancer metastatic cells identifies key proteins in cell adhesion, migration, and invasion. Mol Cell Proteomics 2013; 12:1602-20. [PMID: 23443137 DOI: 10.1074/mcp.m112.022848] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Liver metastasis in colorectal cancer is the major cause of cancer-related deaths. To identify and characterize proteins associated with colon cancer metastasis, we have compared the conditioned serum-free medium of highly metastatic KM12SM colorectal cancer cells with the parental, poorly metastatic KM12C cells using quantitative stable isotope labeling by amino acids in cell culture (SILAC) analyses on a linear ion trap-Orbitrap Velos mass spectrometer. In total, 1337 proteins were simultaneously identified in SILAC forward and reverse experiments. For quantification, 1098 proteins were selected in both experiments, with 155 proteins showing >1.5-fold change. About 52% of these proteins were secreted directly or using alternative secretion pathways. GDF15, S100A8/A9, and SERPINI1 showed capacity to discriminate cancer serum samples from healthy controls using ELISAs. In silico analyses of deregulated proteins in the secretome of metastatic cells showed a major abundance of proteins involved in cell adhesion, migration, and invasion. To characterize the tumorigenic and metastatic properties of some top up- and down-regulated proteins, we used siRNA silencing and antibody blocking. Knockdown expression of NEO1, SERPINI1, and PODXL showed a significant effect on cellular adhesion. Silencing or blocking experiments with SOSTDC1, CTSS, EFNA3, CD137L/TNFSF9, ZG16B, and Midkine caused a significant decrease in migration and invasion of highly metastatic cells. In addition, silencing of SOSTDC1, EFNA3, and CD137L/TNFSF9 reduced liver colonization capacity of KM12SM cells. Finally, the panel of six proteins involved in invasion showed association with poor prognosis and overall survival after dataset analysis of gene alterations. In summary, we have defined a collection of proteins that are relevant for understanding the mechanisms underlying adhesion, migration, invasion, and metastasis in colorectal cancer.
Collapse
Affiliation(s)
- Rodrigo Barderas
- Functional Proteomics Laboratory, Centro de Investigaciones Biológicas, 28040 Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sri-Ram K, Haddo O, Dannawi Z, Tirabosco R, Cannon S, Briggs T, Sinisi M. The outcome of extra-abdominal fibromatosis treated at a tertiary referral centre. Eur J Surg Oncol 2012; 38:700-5. [DOI: 10.1016/j.ejso.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 02/26/2012] [Accepted: 03/05/2012] [Indexed: 11/29/2022] Open
|
18
|
Krzystek-Korpacka M, Diakowska D, Grabowski K, Gamian A. Tumor location determines midkine level and its association with the disease progression in colorectal cancer patients: a pilot study. Int J Colorectal Dis 2012; 27:1319-24. [PMID: 22562257 PMCID: PMC3449055 DOI: 10.1007/s00384-012-1476-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2012] [Indexed: 02/04/2023]
Abstract
PURPOSE The purpose of this study was to evaluate midkine, multipotential cytokine, and growth factor in colorectal cancer (CRC) stratified by tumor location. METHODS Midkine was assessed immunoenzymatically in paired cancerous and noncancerous tissues from 53 CRCs and referred to CRC stage, tumor location, and size, and circulating cytokine levels. RESULTS Midkine was higher in cancerous versus noncancerous tissue in 98 % cases (424.2 vs. 31.1 pg/mg, p < 0.0001). Mean fold increase was 30.1; in 72.5 %, the relative increase was over fivefold. Midkine upregulation was more pronounced in colon than in rectum (fold increase: 36.6 vs. 12.7, p = 0.005) due to higher midkine level in noncancerous rectal than colonic tissue (45.5 vs. 26.2 pg/mg, p = 0.074). Tumor location affected midkine association with CRC stage. Midkine fold change was higher in advanced stages of rectal cancers (16.8 vs. 5.3, respectively in III/IV vs. I/II, p = 0.013), while it tended to be lower in colonic ones (25.3 vs. 47.8, p = 0.134). In addition, fold change in midkine level was higher in rectal N1 than N0 cancers (17.3 vs. 16.5, p = 0.032), while it tended to be lower in colonic cancers (23.6 vs. 50.1, p = 0.085). Midkine negatively correlated with tumor size (r = 0.40, p = 0.017), while it tended to positively correlate with its serum levels (r = 0.45, p = 0.081). CONCLUSIONS Midkine is differently expressed in tumors arising from colonic and rectal mucosa, where it may play diverse roles in carcinogenesis. High midkine expression in noncancerous rectal mucosa might contribute to, a characteristic for rectal cancers, higher incidence of local recurrence. Divergent expression of midkine and its association pattern ought to be taken into account while designing midkine-directed therapies for CRC.
Collapse
Affiliation(s)
| | - Dorota Diakowska
- Department of Gastrointestinal and General Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Krzysztof Grabowski
- Department of Gastrointestinal and General Surgery, Wroclaw Medical University, Wroclaw, Poland
| | - Andrzej Gamian
- Department of Medical Biochemistry, Wroclaw Medical University, ul. Chalubinskiego 10, 50-368 Wroclaw, Poland ,Wroclaw Research Center EIT+, Wroclaw, Poland
| |
Collapse
|