1
|
Pavlíčková K, Hojný J, Waldauf P, Švajdler M, Dundr P, Fabian P, Krkavcová E, Dvořák J, Michálková R, Zambo IS, Hájková N, Flídrová M, Laco J, Hornychová H, Delongová P, Škarda J, Hrudka J, Matěj R. Molecular and Immunohistochemical Classification of Extrapulmonary Small Cell Neuroendocrine Carcinomas: A Study of 181 Cases. J Transl Med 2025; 105:104093. [PMID: 39826683 DOI: 10.1016/j.labinv.2025.104093] [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: 08/13/2024] [Revised: 11/28/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Extrapulmonary small cell neuroendocrine carcinoma (EP-SCNC) is a rare malignancy with a poor prognosis. Most patients with EP-SCNC have metastatic disease upon presentation, and their average overall survival (OS) is less than 12 months. Our study aimed to conduct a complex analysis of EP-SCNC. One hundred eighty-one EP-SCNC tissue samples were subjected to a complex analysis. One hundred fifty-five tumors were pure EP-SCNC, whereas 26 were combined tumors. Immunohistochemistry for ASCL1, NEUROD1, YAP1, POU2F3, Rb1, p53, cyclin D1, p16, PTEN, DLL3, PD-L1, CD56, synaptophysin, chromogranin A, and INSM1 was performed, and 128 samples were analyzed molecularly using next-generation sequencing, comprising DNA and RNA analyses. Detailed results on immunohistochemical and molecular analyses were provided for each primary origin of EP-SCNC separately. Median survival for the whole cohort of patients was 8.94 months. Patient age (≥70 years), tumor mutational burden <15, and TP53 and BRCA2 mutations were negative prognostic factors. High expression of ASCL-1 was associated with shorter OS, whereas high expression of YAP1 was associated with longer OS. Patients with genitourinary tumors had significantly better OS than those with gastrointestinal tract EP-SCNC tumors. Rb1 expression loss was detected more often in genitourinary tract SCNCs. In contrast, p16 overexpression was found more often in genitourinary tract SCNCs. POU2F3 expression was detected more often in combined tumors, whereas NEUROD1 was detected more often in pure EP-SCNC. Regarding "druggable markers," DLL3 was expressed in 66% of tumors and PD-L1 in 17.4%. Detailed analyses of different prognostic and predictive markers are needed to better understand EP-SCNC biology and create more personalized therapy to improve patient prognosis.
Collapse
MESH Headings
- Humans
- Male
- Female
- Aged
- Middle Aged
- Carcinoma, Neuroendocrine/genetics
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/classification
- Carcinoma, Neuroendocrine/mortality
- Carcinoma, Neuroendocrine/metabolism
- Immunohistochemistry
- Adult
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/analysis
- Carcinoma, Small Cell/genetics
- Carcinoma, Small Cell/pathology
- Carcinoma, Small Cell/classification
- Carcinoma, Small Cell/metabolism
- Carcinoma, Small Cell/mortality
- Prognosis
Collapse
Affiliation(s)
- Klára Pavlíčková
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Jan Hojný
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Petr Waldauf
- Department of Anesthesia and Intensive Care, Third Faculty of Medicine, Charles University and University Hospital Kralovske Vinohrady, Prague, Czech Republic
| | - Marián Švajdler
- Bioptická laboratoř s.r.o. and Šikl's Department of Pathology, Charles University, Medical Faculty in Pilsen, Pilsen, Czech Republic
| | - Pavel Dundr
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Pavel Fabian
- Department of Oncological Pathology, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Eva Krkavcová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jiří Dvořák
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Romana Michálková
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Iva Staniczková Zambo
- 1st Institute of Pathologic Anatomy, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Nikola Hájková
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Miroslava Flídrová
- Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Jan Laco
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Kralove, Hradec Králové, Czech Republic
| | - Helena Hornychová
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Králové and University Hospital Hradec Kralove, Hradec Králové, Czech Republic
| | - Patricie Delongová
- Department of Clinical and Molecular Pathology and Medical Genetics, University Hospital Ostrava and Faculty of Medicine University of Ostrava, Ostrava, Czech Republic
| | - Jozef Škarda
- Department of Clinical and Molecular Pathology and Medical Genetics, University Hospital Ostrava and Faculty of Medicine University of Ostrava, Ostrava, Czech Republic
| | - Jan Hrudka
- Department of Pathology, University Hospital Kralovske Vinohrady, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Radoslav Matěj
- Department of Pathology and Molecular Medicine, Thomayer University Hospital, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic; Department of Pathology, University Hospital Kralovske Vinohrady, 3rd Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
2
|
Stefàno E, De Castro F, Ciccarese A, Muscella A, Marsigliante S, Benedetti M, Fanizzi FP. An Overview of Altered Pathways Associated with Sensitivity to Platinum-Based Chemotherapy in Neuroendocrine Tumors: Strengths and Prospects. Int J Mol Sci 2024; 25:8568. [PMID: 39201255 PMCID: PMC11354135 DOI: 10.3390/ijms25168568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Neuroendocrine neoplasms (NENs) are a diverse group of malignancies with a shared phenotype but varying prognosis and response to current treatments. Based on their morphological features and rate of proliferation, NENs can be classified into two main groups with a distinct clinical behavior and response to treatment: (i) well-differentiated neuroendocrine tumors (NETs) or carcinoids (with a low proliferation rate), and (ii) poorly differentiated small- or large-cell neuroendocrine carcinomas (NECs) (with a high proliferation rate). For certain NENs (such as pancreatic tumors, higher-grade tumors, and those with DNA damage repair defects), chemotherapy is the main therapeutic approach. Among the different chemotherapic agents, cisplatin and carboplatin, in combination with etoposide, have shown the greatest efficacy in treating NECs compared to NETs. The cytotoxic effects of cisplatin and carboplatin are primarily due to their binding to DNA, which interferes with normal DNA transcription and/or replication. Consistent with this, NECs, which often have mutations in pathways involved in DNA repair (such as Rb, MDM2, BRCA, and PTEN), have a high response to platinum-based chemotherapy. Identifying mutations that affect molecular pathways involved in the initiation and progression of NENs can be crucial in predicting the response to platinum chemotherapy. This review aims to highlight targetable mutations that could serve as predictors of therapeutic response to platinum-based chemotherapy in NENs.
Collapse
Affiliation(s)
| | | | | | | | | | - Michele Benedetti
- Department of Biological and Environmental Sciences and Technologies (DiSTeBA), University of Salento, Via Monteroni, I-73100 Lecce, Italy; (E.S.); (F.D.C.); (A.C.); (A.M.); (S.M.); (F.P.F.)
| | | |
Collapse
|
3
|
Insulinoma-Associated Protein 1 (INSM1): Diagnostic, Prognostic, and Therapeutic Use in Small Cell Lung Cancer. JOURNAL OF MOLECULAR PATHOLOGY 2022. [DOI: 10.3390/jmp3030013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Small cell lung carcinoma (SCLC) is an aggressive and difficult to treat cancer. Although immunohistochemistry is not mandatory for a SCLC diagnosis, it might be required, especially in small samples. Insulinoma-associated protein 1 (INSM1) is expressed in endocrine and nervous tissues during embryogenesis, generally absent in adults and re-expressed in SCLC and other neuroendocrine neoplasms. Its high specificity propelled its use as diagnostic biomarker and an attractive therapeutic target. Herein, we aim to provide a systematic and critical review on the use of INSM1 for diagnosis, prognostication and the treatment of SCLC. An extensive bibliographic search was conducted in PubMed® focusing on articles published since 2015. According to the literature, INSM1 is a highly sensitive (75–100%) and specific (82–100%) neuroendocrine immunohistochemical marker for SCLC diagnosis. It can be used in histological and cytological samples. Although advantageous, its standalone use is currently not recommended. Studies correlating INSM1 expression and prognosis have disclosed contrasting results, although the expression seemed to entail a worse survival. Targeting INSM1 effectively suppressed SCLC growth either as a suicide gene therapy regulator or as an indirect target of molecular-targeted therapy. INSM1 represents a valuable biomarker for a SCLC diagnosis that additionally offers vast opportunities for the development of new prognostic and therapeutic strategies.
Collapse
|
4
|
Berdugo J, Rooper LM, Chiosea SI. RB1, p16, and Human Papillomavirus in Oropharyngeal Squamous Cell Carcinoma. Head Neck Pathol 2021; 15:1109-1118. [PMID: 33830464 PMCID: PMC8633196 DOI: 10.1007/s12105-021-01317-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/16/2021] [Indexed: 11/24/2022]
Abstract
While P16 immunohistochemistry (IHC) is a well-established surrogate marker of Human Papillomavirus (HPV) in oropharyngeal squamous cell carcinoma (OSCC), Retinoblastoma 1 (RB1) loss may lead to p16 overexpression in the absence of HPV. We determined the proportion of p16-positive/HPV-negative OSCC with RB1 loss and other alterations in RB1/p16 pathway, and tested RB1 IHC as a prognostic biomarker for OSCC, along with the 8th edition of AJCC staging manual. P16 and RB1 IHC and HPV DNA in situ hybridization (ISH) were performed on 257 OSCC. High risk HPV RNA ISH, RB1 fluorescence in situ hybridization (FISH), and next generation sequencing (NGS) were done on p16-positive/HPV DNA ISH-negative OSCC. Disease free survival (DFS) was used as an endpoint. In the entire cohort and in p16-positive (n = 184) and p16-negative (n = 73) subgroups, AJCC 8th edition staging correlated with DFS (p < 0.01). RB1 IHC showed RB1 loss in p16-positive OSCC only (79/184, 43%). RB1 loss by IHC is associated with a better DFS, without providing additional prognostic information for patients with p16-positive OSCC. HPV RNA ISH was positive in 12 of 14 HPV DNA ISH-negative cases. RB1 IHC showed loss in 10 of 15 HPV DNA ISH-negative cases and in 1 of 2 HPV RNA ISH-negative cases. Overall, only one case of p16-positive/HPV RNA ISH-negative OSCC showed RB1 loss by IHC (1/184, 0.5%). Of the 10 p16-positive and HPV DNA ISH-negative cases with RB1 loss by IHC, 2 had RB1 hemizygous deletion and 3 showed Chromosome 13 monosomy by FISH. No RB1 mutations were detected by NGS. Other molecular alterations in p16-positive/HPV DNA ISH-negative cases included TP53 and TERT mutations and DDX3X loss. HPV-independent RB1 inactivation rarely results in false positive p16 IHC. RB1 inactivation by high risk HPV E7 oncoprotein may co-exist with RB1 deletion. RB1 loss is a favorable prognosticator and occurs exclusively in p16-positive OSCC. The 8th edition of the AJCC staging manual satisfactorily predicts DFS of OSCC patients.
Collapse
Affiliation(s)
- Jérémie Berdugo
- Department of Pathology, University of Montreal, Montreal, QC Canada
| | - Lisa M. Rooper
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, MD USA
| | - Simion I. Chiosea
- Department of Pathology, Presbyterian University Hospital, University of Pittsburgh Medical Center, A610.3, 200 Lothrop St., Pittsburgh, PA 15213 USA
| |
Collapse
|
5
|
Lacombe C, De Rycke O, Couvelard A, Turpin A, Cazes A, Hentic O, Gounant V, Zalcman G, Ruszniewski P, Cros J, de Mestier L. Biomarkers of Response to Etoposide-Platinum Chemotherapy in Patients with Grade 3 Neuroendocrine Neoplasms. Cancers (Basel) 2021; 13:643. [PMID: 33562726 PMCID: PMC7915900 DOI: 10.3390/cancers13040643] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 12/18/2022] Open
Abstract
Etoposide-platinum (EP) chemotherapy has long been the reference treatment for grade 3 neuroendocrine neoplasms (G3 NEN). However, G3 NEN are heterogeneous, including well-differentiated tumors (NET) and poorly differentiated large (LCNEC) or small (SCNEC) cell carcinomas, whose response to EP chemotherapy varies considerably. Our aim was to evaluate predictive biomarkers for the response to EP chemotherapy in G3 NEN. We retrospectively studied 89 patients with lung (42%) and digestive (58%) G3 NEN treated by EP chemotherapy between 2006 and 2020. All cases were centrally reviewed for cytomorphology/Ki-67 and immunohistochemistry of retinoblastoma protein (Rb)/p53/p16, analyzed using a semi-quantitative score. The absence of Rb staining (Rbinap) or the absence of very intense p53 staining (p53inap) were considered inappropriate. Rb staining was also studied as a quantitative marker, the best threshold being determined by ROC curve. Intense p16 staining (p16high) also suggested cell cycle dysregulation. Our primary endpoint was the objective response rate (ORR). We included 10 G3 NET, 31 LCNEC and 48 SCNEC, which showed ORR of 20%, 32% and 75%, respectively (NET vs. NEC, p = 0.040; LCNEC vs. SCNEC, p < 0.001). The ORR was significantly higher in NEN presenting with Rbinap (63% vs. 42%, p = 0.025) and p16high (66% vs. 35%, p = 0.006). Rb < 150 optimally identified responders (AUC = 0.657, p < 0.001). The ORR was 67% in Rb < 150 (vs. 25%, p = 0.005). On multivariate analysis, only Rb < 150 was independently associated with ORR (OR 4.16, 95% CI 1.11-15.53, p = 0.034). We confirm the heterogeneity of the response to EP treatment in G3 NEN. Rb < 150 was the best predictive biomarker for the response to EP, and p53 immunostaining had no additional value.
Collapse
Affiliation(s)
- Caroline Lacombe
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| | - Ophélie De Rycke
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| | - Anne Couvelard
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
- Université de Paris, Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat University Hospital (APHP), 75018 Paris, France;
| | - Anthony Turpin
- Department of Medical Oncology, Claude Huriez University Hospital, 59000 Lille, France;
| | - Aurélie Cazes
- Université de Paris, Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat University Hospital (APHP), 75018 Paris, France;
| | - Olivia Hentic
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
| | - Valérie Gounant
- Université de Paris, Department of Thoracic Oncology, CIC INSERM 1425, Bichat University Hospital, 75018 Paris, France; (V.G.); (G.Z.)
| | - Gérard Zalcman
- Université de Paris, Department of Thoracic Oncology, CIC INSERM 1425, Bichat University Hospital, 75018 Paris, France; (V.G.); (G.Z.)
| | - Philippe Ruszniewski
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| | - Jérôme Cros
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
- Université de Paris, Department of Pathology, ENETS Centre of Excellence, Beaujon/Bichat University Hospital (APHP), 75018 Paris, France;
| | - Louis de Mestier
- Université de Paris, Department of Gastroenterology-Pancreatology, ENETS Centre of Excellence, Beaujon University Hospital (APHP), 92110 Clichy, France; (C.L.); (O.D.R.); (O.H.); (P.R.)
- Université de Paris, Centre of Research on Inflammation, INSERM U1149, 75018 Paris, France; (A.C.); (J.C.)
| |
Collapse
|
6
|
Petersen EV, Chudakova DA, Skorova EY, Anikin V, Reshetov IV, Mynbaev OA. The Extracellular Matrix-Derived Biomarkers for Diagnosis, Prognosis, and Personalized Therapy of Malignant Tumors. Front Oncol 2020; 10:575569. [PMID: 33425730 PMCID: PMC7793707 DOI: 10.3389/fonc.2020.575569] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 11/10/2020] [Indexed: 01/18/2023] Open
Abstract
The tumor biomarkers already have proven clinical value and have become an integral part in cancer management and modern translational oncology. The tumor tissue microenvironment (TME), which includes extracellular matrix (ECM), signaling molecules, immune and stromal cells, and adjacent non-tumorous tissue, contributes to cancer pathogenesis. Thus, TME-derived biomarkers have many clinical applications. This review is predominately based on the most recent publications (manuscripts published in a last 5 years, or seminal publications published earlier) and fills a gap in the current literature on the cancer biomarkers derived from the TME, with particular attention given to the ECM and products of its processing and degradation, ECM-associated extracellular vesicles (EVs), biomechanical characteristics of ECM, and ECM-derived biomarkers predicting response to the immunotherapy. We discuss the clinical utility of the TME-incorporating three-dimensional in vitro and ex vivo cell culture models for personalized therapy. We conclude that ECM is a critical driver of malignancies and ECM-derived biomarkers should be included in diagnostics and prognostics panels of markers in the clinic.
Collapse
Affiliation(s)
- Elena V. Petersen
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Daria A. Chudakova
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Ekaterina Yu. Skorova
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Vladimir Anikin
- Harefield Hospital, The Royal Brompton and Harefield Hospitals NHS Foundation Trust, Harefield, United Kingdom
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Igor V. Reshetov
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
- Department of Oncology and Reconstructive Surgery, Sechenov Medical University, Moscow, Russia
| | - Ospan A. Mynbaev
- Department of Molecular and Bio Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| |
Collapse
|
7
|
Inzani F, Santoro A, Angelico G, Feraco A, Spadola S, Arciuolo D, Valente M, Carlino A, Piermattei A, Scaglione G, Scambia G, Rindi G, Zannoni GF. Neuroendocrine Carcinoma of the Uterine Cervix: A Clinicopathologic and Immunohistochemical Study with Focus on Novel Markers (Sst2-Sst5). Cancers (Basel) 2020; 12:cancers12051211. [PMID: 32408525 PMCID: PMC7281076 DOI: 10.3390/cancers12051211] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Gynecological neuroendocrine neoplasms (NENs) are extremely rare, accounting for 1.2-2.4% of the NENs. The aim of this study was to test cervical NENs for novel markers of potential utility for differential diagnosis and target therapy. METHODS All cases of our center (n = 16) were retrieved and tested by immunohistochemistry (IHC) for 12 markers including markers of neuroendocrine differentiation (chromogranin A, synaptophysin, CD56), transcription factors (CDX2 and TTF1), proteins p40, p63, p16INK4a, and p53, somatostatin receptors subtypes (SST2-SST5) and the proliferation marker Ki67 (MIB1). RESULTS All cases were poorly differentiated neuroendocrine carcinomas (NECs), 10 small cell types (small cell-neuroendocrine carcinomas, SCNECs) and 6 large cell types (large cell-neuroendocrine carcinomas, LCNECs); in 3 cases a predominant associated adenocarcinoma component was observed. Neuroendocrine cancer cells expressed at least 2 of the 3 tested neuroendocrine markers; p16 was intensely expressed in 14 (87.5%) cases; SST5 in 11 (56.25%, score 2-3, in 9 cases); SST2 in 8 (50%, score 2-3 in 8), CDX2 in 8 (50%), TTF1 in 5 (31.25%), and p53 in 1 case (0.06%). P63 and p40 expressions were negative, with the exception of one case that showed moderate expression for p63. CONCLUSIONS P40 is a more useful marker for the differential diagnosis compared to squamous cell carcinoma. Neither CDX2 nor TTF1 expression may help the differential diagnosis versus potential cervical metastasis. P16 expression may suggest a cervical origin of NEC; however, it must be always integrated by clinical and instrumental data. The expression of SST2 and SST5 could support a role for SSAs (Somatostatin Analogues) in the diagnosis and therapy of patients with cervical NECs.
Collapse
Affiliation(s)
- Frediano Inzani
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
- ENETS Center of Excellence, Neuroendocrine Tumour (NET) Center, 00168 Rome, Italy
| | - Angela Santoro
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Giuseppe Angelico
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Angela Feraco
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Saveria Spadola
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Damiano Arciuolo
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Michele Valente
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Angela Carlino
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Alessia Piermattei
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
| | - Giulia Scaglione
- Department of Surgical and Diagnostic Sciences, IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy;
| | - Giovanni Scambia
- Oncological Gynaecology Unit, Department of Woman, Child and Public Health Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Obstetric and Gynecologic Clinic Institute, Catholic University of Sacred Hearth, 00168 Rome, Italy
| | - Guido Rindi
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
- ENETS Center of Excellence, Neuroendocrine Tumour (NET) Center, 00168 Rome, Italy
- Pathological Anatomy Institute, Catholic University of Sacred Hearth, 00168 Rome, Italy
| | - Gian Franco Zannoni
- Department of Woman, Child and Public Health Sciences, Gynecopathology and Breast Pathology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (F.I.); (A.S.); (G.A.); (A.F.); (S.S.); (D.A.); (M.V.); (A.C.); (A.P.); (G.R.)
- Pathological Anatomy Institute, Catholic University of Sacred Hearth, 00168 Rome, Italy
- Correspondence:
| |
Collapse
|
8
|
P16 as a marker of carcinoma in effusions and peritoneal washing. BMC Cancer 2020; 20:225. [PMID: 32178642 PMCID: PMC7076945 DOI: 10.1186/s12885-020-6670-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/21/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Considering the potential of p16 as a marker for diagnosis, prognosis and therapeutic response, the aim of this study was to assess its presence, via immunocytochemistry, in metastatic carcinoma of different primary sites and histological types obtained from effusions and peritoneal washings. A total of 118 samples including 85 of metastatic carcinoma and 33 samples of benign effusion/peritoneal washing were prepared by the plasma/thromboplastin method. Immunocytochemistry reactions were performed on cell block sections using antibodies against p16, claudin-4, MOC-31, calretinin, HBME and CD68. RESULTS P16 overexpression was observed in 88.23% of all carcinoma samples. All cervix adenocarcinoma samples showed p16 overexpression. Overexpression in adenocarcinomas of ovary, lung and breast was observed in 93.75, 93.10 and 75% of the samples, respectively. Overexpression was observed in all different histological types analyzed: small cell carcinoma (lung), squamous cell carcinoma (cervical) and urothelial carcinoma (bladder). The specificity of p16 for carcinoma detection was of 96.96%. CONCLUSION Overexpression of p16 was observed in most metastatic carcinoma, from different primary sites and histological types, obtained from effusions and peritoneal washings. Due to its high frequency of overexpression in metastatic carcinoma, p16 may play a possible role in tumor progression and it may be considered as a complementary diagnostic marker depending on histological type and primary site of carcinoma.
Collapse
|
9
|
Rammal G, Fahs A, Kobeissy F, Mechref Y, Zhao J, Zhu R, Diab-Assaf M, Saab R, Ghayad SE. Proteomic Profiling of Rhabdomyosarcoma-Derived Exosomes Yield Insights into Their Functional Role in Paracrine Signaling. J Proteome Res 2019; 18:3567-3579. [PMID: 31448612 DOI: 10.1021/acs.jproteome.9b00157] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Exosomes are important intercellular communication vehicles, secreted into body fluids by multiple cell types, including tumor cells. They have been demonstrated to contribute to the metastatic progression of tumor cells through paracrine signaling. Tumor exosomes contain intact and functional proteins, mRNA and miRNA that may alter the cellular environment to favor tumor growth. We evaluated the protein cargo of exosomes derived from the childhood tumor rhabdomyosarcoma (RMS) and the molecular pathways they are implicated in to decipher their role in the progression of this aggressive disease. We conducted a mass spectrometry analysis of exosome content isolated from five RMS cell lines: three of embryonal RMS (ERMS) and two of alveolar RMS (ARMS) histology and verified results by multiple reaction monitoring and western blot analyses. Results revealed 161 common proteins in ERMS-derived exosomes and 122 common proteins in ARMS-derived exosomes, of which 81 proteins were common to both subtypes. Using both PANTHER gene classification and Pathway Studio software, we assessed the perturbed biological processes and altered pathways in which the exosomal proteins are involved. The 81 commonly expressed proteins included those involved in "cell-signaling," "cell-movement," and "cancer." Pathways engaging the identified proteins revealed 37 common pathways including "integrin signaling pathway," "inflammation mediated by chemokine and cytokine signaling pathway," and "angiogenesis." Finally, a comparison of exosomal proteins of RMS cells with publicly available datasets from other cancer cells revealed that 36 proteins are specific and endogenous to the RMS-exosomes. Taken together, our results reveal that RMS-derived exosomes carry a protein cargo that contributes to conserved cellular signaling networks across multiple cell lines, and we also identify RMS exosome-specific proteins that should be further evaluated as possible novel biomarkers for this tumor.
Collapse
Affiliation(s)
| | | | | | - Yehia Mechref
- Department of Chemistry & Biochemistry , Texas Tech University , Lubbock 79409 , United States
| | - Jingfu Zhao
- Department of Chemistry & Biochemistry , Texas Tech University , Lubbock 79409 , United States
| | - Rui Zhu
- Department of Chemistry & Biochemistry , Texas Tech University , Lubbock 79409 , United States
| | | | | | | |
Collapse
|
10
|
Benzerdjeb N, Traverse-Glehen A, Philouze P, Bishop J, Devouassoux-Shisheboran M. Poorly differentiated neuroendocrine carcinoma of the head and neck: human papillomavirus tumour status/p16 status and impact on overall survival. Histopathology 2019; 76:581-591. [PMID: 31463946 DOI: 10.1111/his.13982] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/06/2019] [Accepted: 08/22/2019] [Indexed: 01/06/2023]
Abstract
AIMS Poorly differentiated neuroendocrine carcinoma (PDNEC) of the head and neck is a rare high-grade neuroendocrine neoplasm. Human papillomavirus (HPV) status and p16 status are as yet unclear among PDNECs, owing to a lack of statistical analysis. The objective of the present study was therefore to evaluate their potential clinicopathological associations, and their prognostic impact on overall survival in PDNECs of the head and neck, regardless to HPV genotype. METHODS AND RESULTS All cases of PDNEC of the head and neck between 1998 and 2019 were identified from the database of the Lyon university hospital pathology department (n = 21); for these cases, p16 immunohistochemistry and HPV in-situ hybridisation were performed. Published cases of PDNEC of the head and neck with assessment of HPV status and p16 status were identified in PubMed (n = 57). Local and published cases were pooled for analysis. HPV positive (HPV+) tumour status was found to be significantly associated with oropharyngeal localisation (P < 0.001) and overexpression of p16 (P < 0.001). Multivariate analysis, adjusted on tumour site, histological subtype, p16 status, HPV status, and source of the case, showed that oropharyngeal localisation [hazard ratio (HR) 3.031, 95% confidence interval (CI) 1.257-7.310] and being a small-cell variant (HR 2.859, 95% CI 1.150-7.109) were significant predictors of worse overall survival; HPV+ tumour status was associated with better overall survival (HR 0.388, 95% CI 0.146-0.995). CONCLUSIONS HPV+ tumour status was associated with oropharyngeal PDNECs and with a better prognosis.
Collapse
Affiliation(s)
- Nazim Benzerdjeb
- Department of Pathology, Institut de Pathologie Multisite, Groupement Hospitalier Sud, Hospices Civils de Lyon, Pierre-Bénite, France.,Université Lyon 1, Villeurbanne, France
| | - Alexandra Traverse-Glehen
- Department of Pathology, Institut de Pathologie Multisite, Groupement Hospitalier Sud, Hospices Civils de Lyon, Pierre-Bénite, France.,Université Lyon 1, Villeurbanne, France
| | - Pierre Philouze
- Service d'Oto-Rhino-Laryngologie et Chirurgie Cervico-Faciale, Hôpital La Croix Rousse, Hospices Civils de Lyon, Lyon, France
| | - Justin Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mojgan Devouassoux-Shisheboran
- Department of Pathology, Institut de Pathologie Multisite, Groupement Hospitalier Sud, Hospices Civils de Lyon, Pierre-Bénite, France.,Université Lyon 1, Villeurbanne, France
| |
Collapse
|
11
|
Ascierto PA, Bifulco C, Palmieri G, Peters S, Sidiropoulos N. Preanalytic Variables and Tissue Stewardship for Reliable Next-Generation Sequencing (NGS) Clinical Analysis. J Mol Diagn 2019; 21:756-767. [PMID: 31251989 DOI: 10.1016/j.jmoldx.2019.05.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/23/2019] [Accepted: 05/24/2019] [Indexed: 12/25/2022] Open
Abstract
An enduring goal of personalized medicine in cancer is the ability to identify patients who are likely to respond to specific therapies. Our growing understanding of the biology and molecular signatures of individual tumor types has facilitated the identification of predictive biomarkers and has led to an increasing number of diagnostic tests to be performed, often as serial and distinct assays on limited tumor specimens. The biomarker diagnostics field has been revolutionized by next-generation sequencing (NGS), which provides a comprehensive overview of the genomic profile of a tumor. Many preanalytic variables can influence the accuracy and reliability of NGS results. Standardization of preanalytic variables is, however, complicated by the plethora of specimen acquisition and processing methods. Variables across the tissue journey, including specimen acquisition, specimen fixation, and sectioning, as well as postfixation processing, such as nucleic acid extraction, library preparation, and choice of sequencing methods, are critical for the reliability of NGS analysis; thus, standardization would be beneficial. In this article, each step in the tissue journey is outlined, with specific focus on preanalytic variables that can influence NGS results. Practical considerations for standardization of these variables are provided to facilitate accurate, reliable, and reproducible NGS-based molecular characterization of tumors, ultimately informing diagnosis and guiding treatment.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", Naples, Italy.
| | - Carlo Bifulco
- Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, Oregon
| | - Giuseppe Palmieri
- Institute of Biomolecular Chemistry - National Research Council, Sassari, Italy
| | - Solange Peters
- Department of Oncology, Lausanne University, Lausanne, Switzerland
| | - Nikoletta Sidiropoulos
- University of Vermont Health Network, Larner College of Medicine at the University of Vermont, Burlington, Vermont
| |
Collapse
|
12
|
Triple marker composed of p16, CD56, and TTF1 shows higher sensitivity than INSM1 for diagnosis of pulmonary small cell carcinoma: proposal for a rational immunohistochemical algorithm for diagnosis of small cell carcinoma in small biopsy and cytology specimens. Hum Pathol 2019; 85:58-64. [DOI: 10.1016/j.humpath.2018.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/12/2018] [Accepted: 10/18/2018] [Indexed: 11/18/2022]
|