1
|
Gorbokon N, Teljuk K, Reiswich V, Lennartz M, Minner S, Simon R, Sauter G, Wilczak W, Clauditz TS, Schraps N, Hackert T, Uzunoglu FG, Kluth M, Bubendorf L, Matter M, Viehweger F, Freytag M, Jacobsen F, Möller K, Steurer S. Deficiency of MTAP Is Frequent and Mostly Homogeneous in Pancreatic Ductal Adenocarcinomas. Cancers (Basel) 2025; 17:1205. [PMID: 40227771 PMCID: PMC11987894 DOI: 10.3390/cancers17071205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND The complete loss of S-methyl-5'-thioadenosine phosphorylase (MTAP) expression, often due to homozygous 9p21 deletion, creates a druggable vulnerability in cancer cells. METHODS A total of 769 primary pancreatic ductal adenocarcinomas were analyzed on tissue microarrays with MTAP immunohistochemistry (IHC) and 9p21 fluorescence in situ hybridization (FISH). Intratumoral heterogeneity was assessed on a "heterogeneity" TMA containing up to nine samples from different areas of 236 primary tumor and nodal metastases, and whole sections of all tumor blocks from 19 cancers. RESULTS MTAP expression loss was found in 181 (37.9%) of 478 interpretable primary tumors and was unrelated to pT, pN, grade, and tumor size. MTAP expression loss was homogenous in 37.6% and heterogeneous in 1.1% of the 181 tumors, with at least three evaluable samples on the heterogeneity TMA. On whole sections, 1 of 19 tumors showed heterogeneous MTAP loss. The correlation between IHC and FISH was nearly perfect, with 98.8% of MTAP-deficient samples showing a 9p21 deletion. CONCLUSIONS MTAP expression loss is frequent, caused by homozygous deletion, and mostly homogeneous in pancreatic ductal adenocarcinomas. Considering also their aggressive clinical behavior, pancreatic adenocarcinomas may represent an ideal cancer type for studying new drugs targeting MTAP-deficient cancer cells in clinical trials.
Collapse
Affiliation(s)
- Natalia Gorbokon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Katharina Teljuk
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Viktor Reiswich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Maximilian Lennartz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Till Sebastian Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Nina Schraps
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.S.); (T.H.); (F.G.U.)
| | - Thilo Hackert
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.S.); (T.H.); (F.G.U.)
| | - Faik G. Uzunoglu
- General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.S.); (T.H.); (F.G.U.)
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Lukas Bubendorf
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.B.); (M.M.)
| | - Matthias Matter
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (L.B.); (M.M.)
| | - Florian Viehweger
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Morton Freytag
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Frank Jacobsen
- Pathologie-Hamburg, Labor Lademannbogen MVZ GmbH, 22419 Hamburg, Germany;
| | - Katharina Möller
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (N.G.); (K.T.); (V.R.); (M.L.); (S.M.); (G.S.); (W.W.); (T.S.C.); (M.K.); (F.V.); (K.M.); (S.S.)
| |
Collapse
|
2
|
Taboada RG, Arruda Almeida MF, Santiago KM, Carraro DM, Nunes WA, Diniz AL, Felismino TC, de Jesus VHF. Pancreatic Ductal Adenocarcinoma with Medullary Features and a Complete Pathological Response After Neoadjuvant FOLFIRINOX: A Case Report and Literature Review. J Gastrointest Cancer 2025; 56:42. [PMID: 39804435 DOI: 10.1007/s12029-024-01140-5] [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] [Accepted: 11/07/2024] [Indexed: 05/02/2025]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with poor response to chemotherapy. High-frequency microsatellite instability (MSI-H) is a rare biological phenomenon in conventional PDAC, being more frequently described in tumors with medullary or mucinous features. METHODS AND RESULTS In this manuscript, we report the case of a patient with an MSI-H pancreatic carcinoma with medullary features (medullary carcinoma of the pancreas-MCP) that achieved a complete pathological response after neoadjuvant modified FOLFIRINOX. Additionally, we summarize the available evidence on the clinical, pathological, and molecular features of patients with MCP, along with survival outcomes. CONCLUSIONS MCPs present significant sensitivity not only to immune checkpoint inhibitors, but also to systemic chemotherapy and that the latter treatment modality should not be overlooked. They also present different pathological and molecular features compared with conventional PDAC, meaning they should be considered a separate pathological entity.
Collapse
Affiliation(s)
| | | | - Karina Miranda Santiago
- Department of Clinical and Functional Genomics, International Research Center, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | - Dirce Maria Carraro
- Department of Clinical and Functional Genomics, International Research Center, AC Camargo Cancer Center, São Paulo, SP, Brazil
- National Institute of Science and Technology in Oncogenomics and Therapeutic Innovation (INCITO), São Paulo, SP, Brazil
| | - Warley Abreu Nunes
- Department of Pathology, AC Camargo Cancer Center, São Paulo, SP, Brazil
| | | | | | - Victor Hugo Fonseca de Jesus
- Department of Gastrointestinal Medical Oncology, Oncoclínicas, Florianópolis, SC, Brazil.
- Department of Medical Oncology, Centro de Pesquisas Oncológicas (CEPON), Admar Gonzaga Road, Florianópolis, SC, 65588034-000, Brazil.
| |
Collapse
|
3
|
Wang Y, Zhang C, Zhang J, Huang H, Guo J. Construction and Validation of a Novel T/NK-Cell Prognostic Signature for Pancreatic Cancer Based on Single-Cell RNA Sequencing. Cancer Invest 2024; 42:876-892. [PMID: 39523741 DOI: 10.1080/07357907.2024.2424328] [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: 07/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Evidence with regards to the distinction between primary and metastatic tumors in pancreatic cancer and driving factors for metastases remains limited. METHODS Single-cell RNA sequencing (scRNA-seq) was conducted on metastatic pancreatic cancer. Bioinformatics analysis on relevant sequencing data was used to construct a risk model to predict patient prognosis. Furthermore, immune infiltration and metabolic differences were assessed. The biological function of key differential genes was evaluated. RESULTS Paired primary and metastatic tumor tissues from 3 pancreatic cancer patients were collected and conducted scRNA-seq. Subsequently, the T/NK cell subgroup was the most different cell type between primary tumors and liver metastases and was selected for further analysis. Eventually, 6 specifically expressed genes of T/NK cells (B2M, ZFP36L2, ANXA1, ARL4C, TSPYL2, FYN) were used constructing the prognostic model. The stability of this model was validated by an external cohort. Meanwhile, different immune infiltration abundances occurred between high and low risk groups stratified by the model. The high-risk group had a stronger metabolic capability. CONCLUSIONS A novel prognostic T/NK-cell signature for pancreatic cancer was constructed based on scRNA-seq data and externally validated. The involved key genes may play a role in multiple metabolic pathways of metastasis and affect the tumor immune microenvironment.
Collapse
Affiliation(s)
- Yu Wang
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Jianlu Zhang
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haoran Huang
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junchao Guo
- Department of General Surgery, Key Laboratory of Research in Pancreatic Tumor, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Masetto F, Mafficini A, Saka B, Armutlu A, Chatterjee D, Jang KT, Zen Y, Navale P, Fassan M, Bacchi CE, Mattiolo P, Simbolo M, Ruzzenente A, Lawlor RT, Reid M, Basturk O, Adsay V, Scarpa A, Luchini C. Tubulocystic Carcinoma of Bile Ducts: A Distinct Type of Cholangiocarcinoma Associated With Adenofibroma-type Lesions. Am J Surg Pathol 2024; 48:1082-1092. [PMID: 38946053 DOI: 10.1097/pas.0000000000002278] [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: 07/02/2024]
Abstract
A type of cholangiocarcinoma (CCA) characterized by peculiar histologic patterns and underlying adenofibromatous lesions has been reported in the literature mostly as individual case reports. This study aims to further clarify the defining characteristics of this spectrum of lesions. Clinicopathologic analysis of 8 biliary tumors with tubulocystic architecture arising in the background of adenofibroma-type lesions was performed. Three of these were also investigated with next-generation sequencing with a 174 genes panel. The patients were 5 males and 3 females, with a mean age of 64.6. All tumors were intrahepatic except for one perihilar that protruded into soft tissues. The mean size was 4.4 cm. At histology, all cases showed a peculiar and cytologically bland tubulocystic pattern that closely resembled tubulocystic-type kidney cancers, including back-to-back microcystic units that formed relatively demarcated nodules, and occurring in the background of adenofibromatous lesions. One case showed perineural invasion by otherwise deceptively benign-appearing microcystic structures, one had areas transitioning to intraductal tubulopapillary neoplasm, and 3 cases harbored more conventional small-duct CCA foci. In those 3 cases, both the tubulocystic and conventional CCA components were investigated by next-generation sequencing separately, and they shared the molecular alterations, including recurrent mutations in chromatin remodeling genes, such as ARID1A , BAP1 , and PBRM1 , and the actionable FGFR2-MCU fusion gene. In the limited follow-up, all but one were alive and free of disease after surgical resection. In conclusion, we described a distinct entity of CCA with specific histo-molecular features, for which we propose the designation of tubulocystic carcinoma of bile ducts.
Collapse
Affiliation(s)
- Francesca Masetto
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona
- ARC-Net Research Center, University and Hospital Trust of Verona
| | - Burcu Saka
- Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Ayse Armutlu
- Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Deyali Chatterjee
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kee-Taek Jang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoh Zen
- Institute of Liver Studies, King's College Hospital, London, UK
| | - Pooja Navale
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, and Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | | | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona
| | - Andrea Ruzzenente
- Department of Surgery, Dentistry, Gynecology and Pediatrics, Division of General and Hepatobiliary Surgery, University and Hospital Trust of Verona
| | - Rita T Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona
- Department of Engineering for Innovative Medicine (DIMI), University of Verona, Verona
| | - Michelle Reid
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| | - Olca Basturk
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Volkan Adsay
- Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona
- ARC-Net Research Center, University and Hospital Trust of Verona
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona
- ARC-Net Research Center, University and Hospital Trust of Verona
| |
Collapse
|
5
|
Ciulla C, Luchini C. Genomic determinants of biological aggressiveness and poor prognosis of pancreatic cancers: KRAS and beyond. Expert Rev Mol Diagn 2024; 24:355-362. [PMID: 38708441 DOI: 10.1080/14737159.2024.2348676] [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: 02/14/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
INTRODUCTION A marked histomolecular heterogeneity characterizes pancreatic cancer. Thus, different tumor histologies with divergent genomic profiles exist within the same category. AREAS COVERED Using data from PubMed, SCOPUS, and Embase (last search date: 04/04/2024), this expert-based, narrative review presents and discusses the essential molecular determinants of biological aggressiveness and poor prognosis in pancreatic cancer. First, KRAS mutation still represents one of the most critical difficulties in treating pancreatic cancers. In this district, it is mutated in > 90% of malignant tumors. Notably, actionable alterations for molecular-based therapies are typically lacking in KRAS-mutated pancreatic cancer. Furthermore, transcriptome-based studies clarified that the squamous phenotype is characterized by poorer prognosis and response to standard chemotherapy. We also discuss molecular biomarkers related to dismal prognosis in specific subsets of pancreatic cancer, such as SMAD4 in signet-ring cell carcinoma and TP53 in invasive cancers derived from intraductal tubulopapillary neoplasms. EXPERT OPINION The identification of the subgroups of pancreatic cancer with particularly unfavorable prognoses is a critical step for addressing specific research efforts. In addition to implementing and strengthening current precision oncology strategies, the decisive step for improving the survival of patients affected by pancreatic cancer must pass through targeting the KRAS gene.
Collapse
Affiliation(s)
- Calogero Ciulla
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Center, University of Verona, Verona, Italy
| |
Collapse
|
6
|
Omori Y, Furukawa T, Scarpa A, Luchini C. Co-occurring IPMN and pancreatic cancer: the same or different? An overview from histology to molecular pathology. J Clin Pathol 2023; 76:734-739. [PMID: 37500498 DOI: 10.1136/jcp-2023-209012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023]
Abstract
Intraductal papillary mucinous neoplasm (IPMN) of the pancreas is one of the most well-established precursors of pancreatic cancer. Its progression to acquire invasiveness is a complex process, based on the accumulation of morphological and genetic alterations. Recent advances in DNA sequencing also showed that co-occurring IPMNs and pancreatic cancers could be totally independent, further complicating our understanding of this complex scenario. The distinction between IPMN and related pancreatic cancer vs IPMN and co-occurring-but not related-pancreatic cancer is a challenging task in routine diagnostic activity, but may have important implications for precision oncology. Of note, recent multiregional sequencing-based studies focused not only on IPMN multi-step tumourigenesis, but also on the divergent intratumoural heterogeneity of this neoplasm. Globally considered, there are three different situations in which co-occurring IPMNs and invasive carcinomas can be found in the same pancreata, indicated with different terminologies: (1) IPMN-associated carcinoma: this definition indicates a carcinoma arising from an IPMN and can be also defined as IPMN-derived carcinoma, sequential or likely related; (2) independent IPMN and invasive carcinoma: the two lesions are not related, and this situation is defined as concomitant, de novo or likely independent; (3) branch-off pathway, where an invasive carcinoma and an adjacent IPMN develop divergently in a forked fashion from a common ancestral clone. In this review, we aim at clarifying the most important nomenclature/definitions of these different situations, also providing an overview of the molecular state-of-the-art and of the clinical implications of this complex landscape.
Collapse
Affiliation(s)
- Yuko Omori
- Department of Investigative Patholgy, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Toru Furukawa
- Department of Investigative Patholgy, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
- ARC-Net Research Center, University and Hospital Trust, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, Verona, Italy
- ARC-Net Research Center, University and Hospital Trust, Verona, Italy
| |
Collapse
|
7
|
Zhang Y, Khalilitousi M(S, Park YP. Unraveling dynamically encoded latent transcriptomic patterns in pancreatic cancer cells by topic modeling. CELL GENOMICS 2023; 3:100388. [PMID: 37719139 PMCID: PMC10504675 DOI: 10.1016/j.xgen.2023.100388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/27/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023]
Abstract
Building a comprehensive topic model has become an important research tool in single-cell genomics. With a topic model, we can decompose and ascertain distinctive cell topics shared across multiple cells, and the gene programs implicated by each topic can later serve as a predictive model in translational studies. Here, we present a Bayesian topic model that can uncover short-term RNA velocity patterns from a plethora of spliced and unspliced single-cell RNA-sequencing (RNA-seq) counts. We showed that modeling both types of RNA counts can improve robustness in statistical estimation and can reveal new aspects of dynamic changes that can be missed in static analysis. We showcase that our modeling framework can be used to identify statistically significant dynamic gene programs in pancreatic cancer data. Our results discovered that seven dynamic gene programs (topics) are highly correlated with cancer prognosis and generally enrich immune cell types and pathways.
Collapse
Affiliation(s)
- Yichen Zhang
- Department of Statistics, The University of British Columbia, Vancouver, BC, Canada
| | | | - Yongjin P. Park
- Department of Statistics, The University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer Research, Part of Provincial Health Care Authority, Vancouver, BC, Canada
| |
Collapse
|
8
|
Luo Y. The characteristic of stem-related genes with pancreatic carcinoma cell after irradiation. Heliyon 2023; 9:e17074. [PMID: 37484310 PMCID: PMC10361223 DOI: 10.1016/j.heliyon.2023.e17074] [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: 10/11/2022] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
Purpose To investigate stem-related differentially expressed genes (DEGs) and their potential mechanism in pancreatic cancer cells (MIAPaCa-2) exposed to x-ray and proton radiation, as well as how these factors affected the prognosis of patients with pancreatic adenocarcinoma (PADC). Methods The stem-related DEGs were screened using the online tool Stemchecker after protons and x-rays were used to irradiate MIAPaCa-2 cells. Analysis was done on the probable processes and prognostic significance of the DEGs in PAC patients. Results Four datasets containing 401 DEGs were filtered, and the stem-related DEGs for each irradiation type indicated a variety of radiobiological characteristics. In pancreatic cancer cells, a number of stem-related DEGs may serve as biomarkers of radiation effects. Patients with pancreatic cancer demonstrated predictive significance for GRB7, B2M, and PMAIP1. Conclusions MIAPaCa-2 cells exposed to x-rays and protons repeatedly displayed heterogeneous expression of stem-related DEGs involved in complex radiosensitivity, radio-resistance, and radio-induced mortality pathways. GRB7 and B2M were considered potential radiation sensitivity indicators for pancreatic cancer.
Collapse
|
9
|
Luchini C, Mattiolo P, Basturk O, Mafficini A, Ozcan K, Lawlor RT, Hong SM, Brosens LA, Marchegiani G, Pea A, Manfrin E, Sciacca G, Zampieri F, Polati R, De Robertis R, Milella M, D'Onofrio M, Malleo G, Salvia R, Adsay V, Scarpa A. Acinar Cystic Transformation of the Pancreas: Histomorphology and Molecular Analysis to Unravel its Heterogeneous Nature. Am J Surg Pathol 2023; 47:379-386. [PMID: 36649476 DOI: 10.1097/pas.0000000000002017] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Acinar cystic transformation (ACT) of the pancreas, previously called acinar cell cystadenoma, is a poorly understood and rare entity among pancreatic cystic lesions. This study aims to clarify its real nature. This research cohort included 25 patients with pancreatic ACT, representing the largest series in the literature. We describe their clinicopathological features and molecular profile using next-generation sequencing. ACT arose more often in women (F/M≃2:1), in the body-tail region, with a mean size of ~4 cm. At the latest follow-up, all patients were alive and disease free. Histologically, a typical acinar epithelium lined all cysts, intermingled with ductal-like epithelium in 11/25 (44%) cases. All the cases lacked any evidence of malignancy. Three ACT showed peculiar features: 1 showed an extensive and diffuse microcystic pattern, and the other 2 harbored foci of low-grade pancreatic intraepithelial neoplasia (PanIN) in the ductal-like epithelium. Next-generation sequencing revealed the presence of 2 pathogenic/likely pathogenic mutations in 2 different cases, 1 with ductal-like epithelium and 1 with PanIN, and affecting KRAS (c.34G>C, p.G12R) and SMO (c.1685G>A, p.R562Q) genes, respectively. The other case with PanIN was not available for sequencing. Overall, our findings support that ACT is a benign entity, potentially arising from heterogeneous conditions/background, including: (1) acinar microcysts, (2) malformations, (3) obstructive/inflammatory setting, (4) genetic predisposition, (5) possible neoplastic origin. Although all indications are that ACT is benign, the potential occurrence of driver mutations suggests discussing a potential role of long-term surveillance for these patients.
Collapse
Affiliation(s)
- Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology
- ARC-Net Research Center for Applied Research on Cancer, University of Verona, Verona, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology
| | - Olca Basturk
- Department of Pathology, Memorial Sloan Kettering Cancer Center, NY
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, Section of Pathology
- ARC-Net Research Center for Applied Research on Cancer, University of Verona, Verona, Italy
| | - Kerem Ozcan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, NY
| | - Rita T Lawlor
- Department of Diagnostics and Public Health, Section of Pathology
- ARC-Net Research Center for Applied Research on Cancer, University of Verona, Verona, Italy
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Lodewijk A Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht, and Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Antonio Pea
- Department of Surgery, The Pancreas Institute
| | - Erminia Manfrin
- Department of Diagnostics and Public Health, Section of Pathology
| | - Giuseppe Sciacca
- Department of Diagnostics and Public Health, Section of Pathology
| | | | - Rita Polati
- Department of Diagnostics and Public Health, Section of Pathology
| | | | - Michele Milella
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Mirko D'Onofrio
- Department of Diagnostics and Public Health, Section of Radiology
| | | | | | - Volkan Adsay
- Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology
- ARC-Net Research Center for Applied Research on Cancer, University of Verona, Verona, Italy
| |
Collapse
|
10
|
Gkountakos A, Martelli FM, Silvestris N, Bevere M, De Bellis M, Alaimo L, Sapuppo E, Masetto F, Mombello A, Simbolo M, Bariani E, Milella M, Fassan M, Scarpa A, Luchini C. Extrahepatic Distal Cholangiocarcinoma vs. Pancreatic Ductal Adenocarcinoma: Histology and Molecular Profiling for Differential Diagnosis and Treatment. Cancers (Basel) 2023; 15:1454. [PMID: 36900245 PMCID: PMC10001378 DOI: 10.3390/cancers15051454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/20/2023] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) and distal cholangiocarcinoma (dCCA) are very aggressive tumors with a high mortality rate. Pancreas and distal bile ducts share a common embryonic development. Hence, PDAC and dCCA exhibit similar histological features that make a differential diagnosis during routine diagnostic practice challenging. However, there are also significant differences, with potential clinical implications. Even if PDAC and dCCA are generally associated with poor survival, patients with dCCA seem to present a better prognosis. Moreover, although precision oncology-based approaches are still limited in both entities, their most important targets are different and include alterations affecting BRCA1/2 and related genes in PDAC, as well as HER2 amplification in dCCA. Along this line, microsatellite instability represents a potential contact point in terms of tailored treatments, but its prevalence is very low in both tumor types. This review aims at defining the most important similarities and differences in terms of clinicopathological and molecular features between these two entities, also discussing the main theranostic implications derived from this challenging differential diagnosis.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Filippo M. Martelli
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Michele Bevere
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Mario De Bellis
- Department of Surgery, Dentistry, Gynecology, and Pediatrics, Division of General and Hepatobiliary Surgery, University of Verona, 37134 Verona, Italy
| | - Laura Alaimo
- Department of Surgery, Dentistry, Gynecology, and Pediatrics, Division of General and Hepatobiliary Surgery, University of Verona, 37134 Verona, Italy
| | - Elena Sapuppo
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Francesca Masetto
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy
| | - Aldo Mombello
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Michele Simbolo
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Elena Bariani
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Michele Milella
- Section of Medical Oncology, Department of Medicine, University of Verona, 37134 Verona, Italy
| | - Matteo Fassan
- Section of Pathology, Department of Medicine (DIMED), University of Padua, 35122 Padua, Italy
| | - Aldo Scarpa
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| | - Claudio Luchini
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy
| |
Collapse
|
11
|
Taherian M, Wang H, Wang H. Pancreatic Ductal Adenocarcinoma: Molecular Pathology and Predictive Biomarkers. Cells 2022; 11:cells11193068. [PMID: 36231030 PMCID: PMC9563270 DOI: 10.3390/cells11193068] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis due to the lack of methods or biomarkers for early diagnosis and its resistance to conventional treatment modalities, targeted therapies, and immunotherapies. PDACs are a heterogenous group of malignant epithelial neoplasms with various histomorphological patterns and complex, heterogenous genetic/molecular landscapes. The newly proposed molecular classifications of PDAC based on extensive genomic, transcriptomic, proteomic and epigenetic data have provided significant insights into the molecular heterogeneity and aggressive biology of this deadly disease. Recent studies characterizing the tumor microenvironment (TME) have shed light on the dynamic interplays between the tumor cells and the immunosuppressive TME of PDAC, which is essential to disease progression, as well as its resistance to chemotherapy, newly developed targeted therapy and immunotherapy. There is a critical need for the development of predictive markers that can be clinically utilized to select effective personalized therapies for PDAC patients. In this review, we provide an overview of the histological and molecular heterogeneity and subtypes of PDAC, as well as its precursor lesions, immunosuppressive TME, and currently available predictive molecular markers for patients.
Collapse
Affiliation(s)
- Mehran Taherian
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hua Wang
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Huamin Wang
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-563-1846; Fax: +1-713-563-1848
| |
Collapse
|
12
|
Mattiolo P, Mafficini A, Lawlor RT, Marchegiani G, Malleo G, Pea A, Salvia R, Piccoli P, Sciammarella C, Santonicco N, Parisi A, Silvestris N, Milella M, Adsay V, Scarpa A, Luchini C. "Pure" hepatoid tumors of the pancreas harboring CTNNB1 somatic mutations: a new entity among solid pseudopapillary neoplasms. Virchows Arch 2022; 481:41-47. [PMID: 35359182 PMCID: PMC9226109 DOI: 10.1007/s00428-022-03317-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/04/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023]
Abstract
Hepatoid tumors (HTs) represent a rare group of neoplasms that are histologically similar to hepatocellular carcinoma but arise outside the liver. The current World Health Organization classification recognizes the hepatoid morphology of pancreatic tumors only as a possible variant of pancreatic ductal adenocarcinoma (PDAC). Here, we describe two cases of "pure" HT of the pancreas showing common features and characterized by indolent biological behavior. These tumors were roundish nodules with pushing borders, hyaline globules, and pure hepatoid histology; they were diffusely positive for β-catenin and LEF1 on immunohistochemistry. At next-generation sequencing, both neoplasms harbored only one pathogenic somatic mutation that affected the CTNNB1 gene at exon 3 and showed a loss of heterozygosity on chromosomes 18 and 21. By integrating macroscopic and microscopic features, along with their molecular profiles, we advocate that such tumors represent a distinct entity from PDAC and should be considered a new variant of solid pseudopapillary neoplasms. The recognition of this new neoplastic category may have immediate implications not only for tumor taxonomy but also for clinical practice.
Collapse
Affiliation(s)
- Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Research Center for Applied Research On Cancer, University of Verona, 37134, Verona, Italy
| | - Giovanni Marchegiani
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Giuseppe Malleo
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Antonio Pea
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Roberto Salvia
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Paola Piccoli
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
| | - Concetta Sciammarella
- ARC-Net Research Center for Applied Research On Cancer, University of Verona, 37134, Verona, Italy
| | - Nicola Santonicco
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
| | - Alice Parisi
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, 98125, Messina, Italy
| | - Michele Milella
- Department of Medicine, Section of Medical Oncology, University and Hospital Trust of Verona, 37134, Verona, Italy
| | - Volkan Adsay
- Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), 34010, Istanbul, Turkey
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy
- ARC-Net Research Center for Applied Research On Cancer, University of Verona, 37134, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Piazzale Scuro, 10, 37134, Verona, Italy.
- ARC-Net Research Center for Applied Research On Cancer, University of Verona, 37134, Verona, Italy.
| |
Collapse
|
13
|
Deng S, Feng Y, Pauklin S. 3D chromatin architecture and transcription regulation in cancer. J Hematol Oncol 2022; 15:49. [PMID: 35509102 PMCID: PMC9069733 DOI: 10.1186/s13045-022-01271-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/21/2022] [Indexed: 12/18/2022] Open
Abstract
Chromatin has distinct three-dimensional (3D) architectures important in key biological processes, such as cell cycle, replication, differentiation, and transcription regulation. In turn, aberrant 3D structures play a vital role in developing abnormalities and diseases such as cancer. This review discusses key 3D chromatin structures (topologically associating domain, lamina-associated domain, and enhancer-promoter interactions) and corresponding structural protein elements mediating 3D chromatin interactions [CCCTC-binding factor, polycomb group protein, cohesin, and Brother of the Regulator of Imprinted Sites (BORIS) protein] with a highlight of their associations with cancer. We also summarise the recent development of technologies and bioinformatics approaches to study the 3D chromatin interactions in gene expression regulation, including crosslinking and proximity ligation methods in the bulk cell population (ChIA-PET and HiChIP) or single-molecule resolution (ChIA-drop), and methods other than proximity ligation, such as GAM, SPRITE, and super-resolution microscopy techniques.
Collapse
Affiliation(s)
- Siwei Deng
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, UK
| | - Yuliang Feng
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, UK
| | - Siim Pauklin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Old Road, Headington, Oxford, OX3 7LD, UK.
| |
Collapse
|
14
|
Gkountakos A, Simbolo M, Bariani E, Scarpa A, Luchini C. Undifferentiated Sarcomatoid Carcinoma of the Pancreas: From Histology and Molecular Pathology to Precision Oncology. Int J Mol Sci 2022; 23:1283. [PMID: 35163206 PMCID: PMC8835772 DOI: 10.3390/ijms23031283] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/22/2022] [Accepted: 01/22/2022] [Indexed: 12/12/2022] Open
Abstract
Undifferentiated sarcomatoid carcinoma of the pancreas (SCP) is a rare and aggressive subtype of pancreatic cancer. Histologically, SCP is a poorly differentiated tumor characterized by the lack of glandular differentiation and the presence of mesenchymal-like, spindle-shaped tumor cells. Due to its rarity, only sporadic cases have been reported, while its molecular characterization has not been sufficiently described. Surgical resection with curative intent is the gold-standard of SCP management, but this strategy is possible only in a small proportion of cases due to SCP early metastasization. Although SCP is generally associated with a poor prognosis, some clinical cases amenable to surgical resection and followed by adjuvant chemotherapy have demonstrated a remarkably long survival. Preliminary molecular insights on the SCP molecular landscape have demonstrated the recurrent presence of KRAS and TP53 mutations, highlighting genetic similarities with conventional pancreatic ductal adenocarcinoma (PDAC). Although the use of immunotherapy in PDAC remains an unmet challenge, recent insights indicated a potentially significant role of the PD-L1/Notch3 axis in SCP, opening new horizons for immunotherapy in this cancer subtype. In this review, we described the most important clinic-pathologic features of SCP, with a specific focus on their molecular landscape and the potential targets for precision oncology.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| | - Elena Bariani
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| | - Aldo Scarpa
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| | - Claudio Luchini
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| |
Collapse
|
15
|
Mattiolo P, Kryklyva V, Brosens LA, Mafficini A, Lawlor RT, Milella M, Scarpa A, Corbo V, Luchini C. Refining targeted therapeutic approaches in pancreatic cancer: from histology and molecular pathology to the clinic. Expert Opin Ther Targets 2022; 26:1-4. [PMID: 34936525 DOI: 10.1080/14728222.2022.2021397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 12/18/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Valentyna Kryklyva
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lodewijk A Brosens
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht University, The Netherlands
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Rita T Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Michele Milella
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|