1
|
Stuebs FA, Knöll A, Hartmann A, Matek C, John N, Häberle L, Beckmann MW, Erber R, Bercebal C, Geppert CI. Trop2-expression in primary vulvar squamous cell carcinoma. Gynecol Oncol 2025; 196:78-84. [PMID: 40184712 DOI: 10.1016/j.ygyno.2025.03.049] [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: 11/24/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
OBJECTIVES Vulvar squamous cell carcinoma (VSCC) is a rare malignant disease but with increasing incidence. Therapeutic options are generally limited to surgery and radio-(chemo)therapy. New strategies for therapy are lacking. Trophoblast cell surface antigen 2 (Trop2) is a new target being used in different tumor entities. The purpose of the current research is to assess the expression of Trop2 in VSCC. METHODS We constructed a next generation tissue micro array (ngTMA) including women with primary VSCC treated between 2000 and 2021 at the Department of Gynecology and Obstetrics, Erlangen University Hospital analyzing the expression of Trop2. Further clinicopathologic characteristics were analyzed: tumor stage, regional lymph node-status, venous-, lymphatic- and perineural sheath invasion, depth of invasion, grading, p16/p53 subtype and tumor infiltration lymphocytes. RESULTS A total of 217 women were included. Trop2 was expressed in 100 % of VSCC. Women were split in two groups, the "low expression" and the "high expression" groups, according to the median of Trop2 (H-score: 279). p16+/p53- is the most common p16/p53 subtype in patients with high Trop2 expression. Trop2 favored a better prognosis in women with VSCC, hence not clinically impactful for the most common p16-/p53+ subtype. CONCLUSION The ubiquitous expression of Trop2 makes it an interesting potential target for the Trop2 directed antibody drug conjugate (ADC) sacituzumab govitecan (SG). Clinical trials evaluating the safety and effectiveness of SG in VSCC are needed.
Collapse
Affiliation(s)
- Frederik A Stuebs
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany.
| | - Antje Knöll
- Institute of Clinical and Molecular Virology, Erlangen University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - Christian Matek
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - Nelson John
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany; Biostatistics Unit, Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany; Biostatistics Unit, Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - Ramona Erber
- Institute of Pathology, Regensburg University Hospital, Germany
| | - Carlos Bercebal
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| |
Collapse
|
2
|
Stuebs FA, Knöll A, Hartmann A, Leikauf LS, Matek C, John N, Häberle L, Beckmann MW, Geppert CI. PD-L1-Expression in primary and recurrent vulvar squamous cell cancer. Gynecol Oncol 2025; 193:98-104. [PMID: 39798194 DOI: 10.1016/j.ygyno.2025.01.001] [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: 08/20/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Squamous cell vulvar carcinoma is a rare malignant disease of women. In higher tumor stages survival rates are poor. Therapy options are limited. Immunoncology plays an increasing role in the treatment of gynecology cancers. Data on the expression of PD-L1 in vulvar cancer are rare and contradictory. We sought to describe the expression of PD-L1 in VSCC in respect to the clinicopathologic characteristics of the tumor. STUDY DESIGN We conducted a retrospective analysis including women with primary and recurrent vulvar cancer between 2000 and 2021. A next generation tissue micro array (ngTMA) was constructed for the analysis of PD-L1 expression. RESULTS In total 238 women with primary VSCC and 66 cases of local or distant recurrent vulvar cancer were included. 80 women with primary VSCC (33.6 %) had tumors with common positive score (CPS) <1 and 63 women (26.5 %) had tumors with CPS 1- < 10 and 95 women with CPS ≥10 (39.9 %). In the PD-L1 positive group the rates of p53+, groin metastasis, lymphatic invasion and tumor infiltration lymphocytes were higher as compared to PD-L1 negative (CPS <1). There was no significant influence of CPS in overall survival in addition to other prognostic factors (P = 0.13, likelihood ratio test). CONCLUSION PD-L1 expression in primary vulvar cancer is associated with poorer prognosis. Hence, PD-L1 is a possible target for immune checkpoint inhibitors and women might benefit from special treatment options.
Collapse
Affiliation(s)
- Frederik A Stuebs
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany.
| | - Antje Knöll
- Institute of Clinical and Molecular Virology, Erlangen University Hospital, Fried-rich-Alexander-Universität Erlangen-Nürnberg, Schlossgarten 4, 91054 Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - Leah-Sophie Leikauf
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - Christian Matek
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - Nelson John
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany; Biostatistics Unit, Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Lothar Häberle
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany; Biostatistics Unit, Erlangen University Hospital, Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Matthias W Beckmann
- Department of Gynecology and Obstetrics, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Bavarian Cancer Research Center (BZKF), Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, Erlangen University Hospital, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Friedrich-Alexander-Universität Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| |
Collapse
|
3
|
Renne SL, Sama' L, Kumar S, Mintemur O, Ruspi L, Santori I, Sicoli F, Bertuzzi A, Laffi A, Bonometti A, Colombo P, D'amato V, Bressan A, Scorsetti M, Terracciano L, Navarria P, D'incalci M, Quagliuolo V, Pasqualini F, Grizzi F, Cananzi FCM. Disruptions in antigen processing and presentation machinery on sarcoma. Cancer Immunol Immunother 2024; 73:228. [PMID: 39249578 PMCID: PMC11383888 DOI: 10.1007/s00262-024-03822-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND The antigen processing machinery (APM) plays a critical role in generating tumor-specific antigens that can be recognized and targeted by the immune system. Proper functioning of APM components is essential for presenting these antigens on the surface of tumor cells, enabling immune detection and destruction. In many cancers, defects in APM can lead to immune evasion, contributing to tumor progression and poor clinical outcomes. However, the status of the APM in sarcomas is not well characterized, limiting the development of effective immunotherapeutic strategies for these patients. METHODS We investigated 126 patients with 8 types of bone and soft tissue sarcoma operated between 2001-2021. Tissue microarrays mapped 11 specific areas in each case. The presence/absence of APM protein was determined through immunohistochemistry. Bayesian networks were used. RESULTS All investigated sarcomas had some defects in APM. The least damaged component was HLA Class I subunit β2-microglobulin and HLA Class II. The proteasome LMP10 subunit was defective in leiomyosarcoma (LMS), myxoid liposarcoma (MLPS), and dedifferentiated liposarcoma (DDLPS), while MHC I transporting unit TAP2 was altered in undifferentiated pleomorphic sarcoma (UPS), gastrointestinal stromal tumor (GIST), and chordoma (CH). Among different neoplastic areas, high-grade areas showed different patterns of expression compared to high lymphocytic infiltrate areas. Heterogeneity at the patient level was also observed. Loss of any APM component was prognostic of distant metastasis (DM) for LMS and DDLPS and of overall survival (OS) for LMS. CONCLUSION Sarcomas exhibit a high degree of defects in APM components, with differences among histotypes and tumoral areas. The most commonly altered APM components were HLA Class I subunit β2-microglobulin, HLA Class I subunit α (HC10), and MHC I transporting unit TAP2. The loss of APM components was prognostic of DM and OS and clinically relevant for LMS and DDLPS. This study explores sarcoma molecular mechanisms, enriching personalized therapeutic approaches.
Collapse
Affiliation(s)
- Salvatore Lorenzo Renne
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy.
- Pathology Department, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy.
| | - Laura Sama'
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Sonia Kumar
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Omer Mintemur
- Pathology Department, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Laura Ruspi
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Ilaria Santori
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
| | - Federico Sicoli
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Alexia Bertuzzi
- Oncology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Alice Laffi
- Oncology Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Arturo Bonometti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Pathology Department, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Piergiuseppe Colombo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Pathology Department, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Vittoria D'amato
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Alessandra Bressan
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Pathology Department, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Marta Scorsetti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Luigi Terracciano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Pathology Department, IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Pierina Navarria
- Radiotherapy and Radiosurgery Department, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Maurizio D'incalci
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Cancer Pharmacology Laboratory, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Vittorio Quagliuolo
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Fabio Pasqualini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Fabio Grizzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| | - Ferdinando Carlo Maria Cananzi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
- Sarcoma, Melanoma and Rare Tumors Surgery Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089, Milan, Italy
| |
Collapse
|
4
|
Issac R, Masih D, Ranjan M, Pulimood AB. Primary adenocarcinoma of colon: A clinicopathological study with the prevalence and correlation of CDX2 biomarker expression - A tertiary care center experience. Indian J Cancer 2024; 61:238-243. [PMID: 38155448 DOI: 10.4103/ijc.ijc_760_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/14/2021] [Indexed: 12/30/2023]
Abstract
BACKGROUND Colorectal cancer is one of the alarming health problems worldwide. Prognostic biomarkers are the key for risk stratification in patients with colon cancer and the decision to recommend adjuvant chemotherapy. It has been difficult to identify a single prognostic biomarker for colon cancer. Currently, tumor stage, tumor grade, and microsatellite instability remain the most important prognostic variables that aid in the treatment of patients with colon cancer. Some studies highlighted that CDX2 immunohistochemistry negativity is an independent prognostic factor and indicates a worse survival rate. Our aim was to study the prevalence of CDX2 biomarker expression in patients diagnosed with primary adenocarcinoma and to correlate this with the clinical profile and pathological features. METHODS Endoscopic mucosal biopsies and resection specimens of 148 patients diagnosed with colonic adenocarcinoma were analyzed. CDX2 immunohistochemistry was performed, and the result was correlated with clinicopathological features. The results were presented as mean, frequencies, and percentages. Pearson's Chi-square test was used to assess the associations between clinicopathological parameters and CDX2 immunohistochemistry negativity. RESULTS The prevalence of CDX2 expression by immunohistochemistry in colon cancer was found to be 92%. CDX2 biomarker negativity was found to be higher in left-sided colon cancers, poorly differentiated adenocarcinoma, mucinous carcinoma, and higher TNM stages. CONCLUSION CDX2-negative tumors are often associated with several adverse prognostic variables (e.g., advanced stage, poor differentiation, and metastasis). Thus, sub-classification of colon cancer based on the CDX2 biomarker aids to separate them into prognostically relevant categories.
Collapse
Affiliation(s)
- Rijo Issac
- Department of General Pathology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Dipti Masih
- Department of General Pathology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Mark Ranjan
- Department of General Surgery, Christian Medical College, Vellore, Tamil Nadu, India
| | - Anna B Pulimood
- Department of General Pathology, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
5
|
Caldas ÁMC, Nunes WA, Taboada R, Cesca MG, Germano JN, Riechelmann RP. Loss of CDX2 and high COX2 ( PTGS2) expression in metastatic colorectal cancer. Ecancermedicalscience 2024; 18:1666. [PMID: 38439814 PMCID: PMC10911677 DOI: 10.3332/ecancer.2024.1666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Indexed: 03/06/2024] Open
Abstract
Lack of expression of the tumour suppressor gene caudal-type homeobox 2 (CDX2) associates with poor outcomes in early stage colorectal cancer (CRC). Yet its prognostic value in the context of other prognostic biomarkers in metastatic CRC (mCRC) is unknown. Overexpressed cyclooxygenase-2 (COX2) has been reported in advanced CRC. However, CDX2 and COX2 relationship in mCRC remains undetermined. We aimed to assess their expression in mCRC tumours from a clinically characterised cohort and their influence on overall survival (OS) and progression-free survival (PFS) in first line. Among 720 consecutive mCRC patients, 346 had tumour samples appropriate for tissue microarray assembly and immunohistochemistry analyses. Clinical and survival data were retrospectively assessed. Loss of CDX2 expression was detected in 27 (7.8%) samples, enriched in poorly differentiated tumours (20%; p < 0.01) and in those with the BRAF p.V600E variant (40%; p < 0.01). Most tumours (93.4%) expressed COX2. COX2-negative samples were enriched in poorly differentiated mCRC. In unadjusted analyses, median OS (p < 0.001) and median PFS (p < 0.05) were inferior for patients with CDX2-negative versus CDX2-positive tumours. In conclusion, loss of CDX2 was significantly associated with poorly differentiated mCRC and BRAF p.V600E allele and a prognostic marker of worse OS.
Collapse
Affiliation(s)
- Álvaro M C Caldas
- Department of Clinical Oncology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Warley A Nunes
- Department of Pathology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Rodrigo Taboada
- Department of Clinical Oncology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Marcelle G Cesca
- Department of Clinical Oncology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Janaína N Germano
- Statistic Group at the International Research Center (CIPE), AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| | - Rachel P Riechelmann
- Department of Clinical Oncology, AC Camargo Cancer Center, São Paulo 01509-900, Brazil
| |
Collapse
|
6
|
Sinha S, Alcantara J, Perry K, Castillo V, Espinoza CR, Taheri S, Vidales E, Tindle C, Adel A, Amirfakhri S, Sawires JR, Yang J, Bouvet M, Sahoo D, Ghosh P. Machine-Learning Identifies a Strategy for Differentiation Therapy in Solid Tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.13.557628. [PMID: 37745574 PMCID: PMC10515918 DOI: 10.1101/2023.09.13.557628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
BACKGROUND Although differentiation therapy can cure some hematologic malignancies, its curative potential remains unrealized in solid tumors. This is because conventional computational approaches succumb to the thunderous noise of inter-/intratumoral heterogeneity. Using colorectal cancers (CRCs) as an example, here we outline a machine learning(ML)-based approach to track, differentiate, and selectively target cancer stem cells (CSCs). METHODS A transcriptomic network was built and validated using healthy colon and CRC tissues in diverse gene expression datasets (~5,000 human and >300 mouse samples). Therapeutic targets and perturbation strategies were prioritized using ML, with the goal of reinstating the expression of a transcriptional identifier of the differentiated colonocyte, CDX2, whose loss in poorly differentiated (CSC-enriched) CRCs doubles the risk of relapse/death. The top candidate target was then engaged with a clinical-grade drug and tested on 3 models: CRC lines in vitro, xenografts in mice, and in a prospective cohort of healthy (n = 3) and CRC (n = 23) patient-derived organoids (PDOs). RESULTS The drug shifts the network predictably, induces CDX2 and crypt differentiation, and shows cytotoxicity in all 3 models, with a high degree of selectivity towards all CDX2-negative cell lines, xenotransplants, and PDOs. The potential for effective pairing of therapeutic efficacy (IC50) and biomarker (CDX2-low state) is confirmed in PDOs using multivariate analyses. A 50-gene signature of therapeutic response is derived and tested on 9 independent cohorts (~1700 CRCs), revealing the impact of CDX2-reinstatement therapy could translate into a ~50% reduction in the risk of mortality/recurrence. CONCLUSIONS Findings not only validate the precision of the ML approach in targeting CSCs, and objectively assess its impact on clinical outcome, but also exemplify the use of ML in yielding clinical directive information for enhancing personalized medicine.
Collapse
|
7
|
Frei AL, McGuigan A, Sinha RRAK, Glaire MA, Jabbar F, Gneo L, Tomasevic T, Harkin A, Iveson TJ, Saunders M, Oein K, Maka N, Pezella F, Campo L, Hay J, Edwards J, Sansom OJ, Kelly C, Tomlinson I, Kildal W, Kerr RS, Kerr DJ, Danielsen HE, Domingo E, Church DN, Koelzer VH. Accounting for intensity variation in image analysis of large-scale multiplexed clinical trial datasets. J Pathol Clin Res 2023; 9:449-463. [PMID: 37697694 PMCID: PMC10556275 DOI: 10.1002/cjp2.342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/14/2023] [Accepted: 08/20/2023] [Indexed: 09/13/2023]
Abstract
Multiplex immunofluorescence (mIF) imaging can provide comprehensive quantitative and spatial information for multiple immune markers for tumour immunoprofiling. However, application at scale to clinical trial samples sourced from multiple institutions is challenging due to pre-analytical heterogeneity. This study reports an analytical approach to the largest multi-parameter immunoprofiling study of clinical trial samples to date. We analysed 12,592 tissue microarray (TMA) spots from 3,545 colorectal cancers sourced from more than 240 institutions in two clinical trials (QUASAR 2 and SCOT) stained for CD4, CD8, CD20, CD68, FoxP3, pan-cytokeratin, and DAPI by mIF. TMA slides were multi-spectrally imaged and analysed by cell-based and pixel-based marker analysis. We developed an adaptive thresholding method to account for inter- and intra-slide intensity variation in TMA analysis. Applying this method effectively ameliorated inter- and intra-slide intensity variation improving the image analysis results compared with methods using a single global threshold. Correlation of CD8 data derived by our mIF analysis approach with single-plex chromogenic immunohistochemistry CD8 data derived from subsequent sections indicates the validity of our method (Spearman's rank correlation coefficients ρ between 0.63 and 0.66, p ≪ 0.01) as compared with the current gold standard analysis approach. Evaluation of correlation between cell-based and pixel-based analysis results confirms equivalency (ρ > 0.8, p ≪ 0.01, except for CD20 in the epithelial region) of both analytical approaches. These data suggest that our adaptive thresholding approach can enable analysis of mIF-stained clinical trial TMA datasets by digital pathology at scale for precision immunoprofiling.
Collapse
Affiliation(s)
- Anja L Frei
- Department of Pathology and Molecular PathologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
- Life Science Zurich Graduate School, PhD Program in BiomedicineUniversity of ZurichZurichSwitzerland
| | | | | | - Mark A Glaire
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Faiz Jabbar
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | - Luciana Gneo
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
| | | | - Andrea Harkin
- Cancer Research UK Glasgow Clinical Trials UnitUniversity of GlasgowGlasgowUK
| | - Tim J Iveson
- Southampton University Hospital NHS Foundation TrustSouthamptonUK
| | | | - Karin Oein
- Glasgow Tissue Research FacilityUniversity of Glasgow, Queen Elizabeth University HospitalGlasgowUK
| | - Noori Maka
- Glasgow Tissue Research FacilityUniversity of Glasgow, Queen Elizabeth University HospitalGlasgowUK
| | - Francesco Pezella
- Nuffield Division of Clinical Laboratory SciencesUniversity of OxfordOxfordUK
| | | | - Jennifer Hay
- Glasgow Tissue Research FacilityUniversity of Glasgow, Queen Elizabeth University HospitalGlasgowUK
| | | | - Owen J Sansom
- School of Cancer SciencesUniversity of GlasgowGlasgowUK
- Cancer Research UK Beatson InstituteGlasgowUK
- Cancer Research UK Scotland CentreEdinburgh and GlasgowUK
| | - Caroline Kelly
- Cancer Research UK Glasgow Clinical Trials UnitUniversity of GlasgowGlasgowUK
| | | | - Wanja Kildal
- Institute for Cancer Genetics and InformaticsOslo University HospitalOsloNorway
| | | | - David J Kerr
- Nuffield Division of Clinical Laboratory SciencesUniversity of OxfordOxfordUK
| | - Håvard E Danielsen
- Nuffield Division of Clinical Laboratory SciencesUniversity of OxfordOxfordUK
- Institute for Cancer Genetics and InformaticsOslo University HospitalOsloNorway
- Department of InformaticsUniversity of OsloOsloNorway
| | - Enric Domingo
- Department of OncologyUniversity of OxfordOxfordUK
- Cancer Research UK Scotland CentreEdinburgh and GlasgowUK
| | | | - David N Church
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Oxford NIHR Comprehensive Biomedical Research CentreOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Viktor H Koelzer
- Department of Pathology and Molecular PathologyUniversity Hospital Zurich, University of ZurichZurichSwitzerland
- Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Department of OncologyUniversity of OxfordOxfordUK
| |
Collapse
|
8
|
Jungen SH, Noti L, Christe L, Galvan JA, Zlobec I, Müller MD, Imboden S, Siegenthaler F, Carlson JW, Pellinen T, Heredia-Soto V, Ruz-Caracuel I, Hardisson D, Redondo A, Mendiola M, Rau TT. Spatial distribution of CD3- and CD8-positive lymphocytes as pretest for POLE wild-type in molecular subgroups of endometrial carcinoma. Front Med (Lausanne) 2023; 10:1110529. [PMID: 37035329 PMCID: PMC10076655 DOI: 10.3389/fmed.2023.1110529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction Over the years, the molecular classification of endometrial carcinoma has evolved significantly. Both POLEmut and MMRdef cases share tumor biological similarities like high tumor mutational burden and induce strong lymphatic reactions. While therefore use case scenarios for pretesting with tumor-infiltrating lymphocytes to replace molecular analysis did not show promising results, such testing may be warranted in cases where an inverse prediction, such as that of POLEwt, is being considered. For that reason we used a spatial digital pathology method to quantitatively examine CD3+ and CD8+ immune infiltrates in comparison to conventional histopathological parameters, prognostics and as potential pretest before molecular analysis. Methods We applied a four-color multiplex immunofluorescence assay for pan-cytokeratin, CD3, CD8, and DAPI on 252 endometrial carcinomas as testing and compared it to further 213 cases as validation cohort from a similar multiplexing assay. We quantitatively assessed immune infiltrates in microscopic distances within the carcinoma, in a close distance of 50 microns, and in more distant areas. Results Regarding prognostics, high CD3+ and CD8+ densities in intra-tumoral and close subregions pointed toward a favorable outcome. However, TCGA subtyping outperforms prognostication of CD3 and CD8 based parameters. Different CD3+ and CD8+ densities were significantly associated with the TCGA subgroups, but not consistently for histopathological parameter. In the testing cohort, intra-tumoral densities of less than 50 intra-tumoral CD8+ cells/mm2 were the most suitable parameter to assume a POLEwt, irrespective of an MMRdef, NSMP or p53abn background. An application to the validation cohort corroborates these findings with an overall sensitivity of 95.5%. Discussion Molecular confirmation of POLEmut cases remains the gold standard. Even if CD3+ and CD8+ cell densities appeared less prognostic than TCGA, low intra-tumoral CD8+ values predict a POLE wild-type at substantial percentage rates, but not vice versa. This inverse correlation might be useful to increase pretest probabilities in consecutive POLE testing. Molecular subtyping is currently not conducted in one-third of cases deemed low-risk based on conventional clinical and histopathological parameters. However, this percentage could potentially be increased to two-thirds by excluding sequencing of predicted POLE wild-type cases, which could be determined through precise quantification of intra-tumoral CD8+ cells.
Collapse
Affiliation(s)
| | - Luca Noti
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Lucine Christe
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Jose A. Galvan
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Michael D. Müller
- Department of Obstetrics and Gynecology, University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Sara Imboden
- Department of Obstetrics and Gynecology, University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Franziska Siegenthaler
- Department of Obstetrics and Gynecology, University Hospital of Bern, University of Bern, Bern, Switzerland
| | - Joseph W. Carlson
- Karolinska Institutet, Klinisk Patologi KS, Solna, Sweden
- Keck School of Medicine of USC, Pathology, Health Sciences Campus, Los Angeles, CA, United States
| | - Teijo Pellinen
- Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Victoria Heredia-Soto
- Instituto de Investigación Biomédica del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - David Hardisson
- Instituto de Investigación Biomédica del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pathology, Hospital Universitario La Paz, Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Andres Redondo
- Department of Medical Oncology, Hospital Universitario La Paz, Madrid, Spain
| | - Marta Mendiola
- Instituto de Investigación Biomédica del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Tilman T. Rau
- Institute of Pathology, University of Bern, Bern, Switzerland
- Institute of Pathology, Universitätsklinikum Düsseldorf, Düsseldorf, Germany
- *Correspondence: Tilman T. Rau,
| |
Collapse
|
9
|
ATF2 loss promotes tumor invasion in colorectal cancer cells via upregulation of cancer driver TROP2. Cell Mol Life Sci 2022; 79:423. [PMID: 35838828 PMCID: PMC9287261 DOI: 10.1007/s00018-022-04445-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/21/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
In cancer, the activating transcription factor 2 (ATF2) has pleiotropic functions in cellular responses to growth stimuli, damage, or inflammation. Due to only limited studies, the significance of ATF2 in colorectal cancer (CRC) is not well understood. We report that low ATF2 levels correlated with worse prognosis and tumor aggressiveness in CRC patients. NanoString gene expression and ChIP analysis confirmed trophoblast cell surface antigen 2 (TROP2) as a novel inhibitory ATF2 target gene. This inverse correlation was further observed in primary human tumor tissues. Immunostainings revealed that high intratumoral heterogeneity for ATF2 and TROP2 expression was sustained also in liver metastasis. Mechanistically, our in vitro data of CRISPR/Cas9-generated ATF2 knockout (KO) clones revealed that high TROP2 levels were critical for cell de-adhesion and increased cell migration without triggering EMT. TROP2 was enriched in filopodia and displaced Paxillin from adherens junctions. In vivo imaging, micro-computer tomography, and immunostainings verified that an ATF2KO/TROP2high status triggered tumor invasiveness in in vivo mouse and chicken xenograft models. In silico analysis provided direct support that ATF2low/TROP2high expression status defined high-risk CRC patients. Finally, our data demonstrate that ATF2 acts as a tumor suppressor by inhibiting the cancer driver TROP2. Therapeutic TROP2 targeting might prevent particularly the first steps in metastasis, i.e., the de-adhesion and invasion of colon cancer cells.
Collapse
|
10
|
Mucosal Mast Cell Distribution in the Gastrointestinal Tract of Children: A Preliminary Study for Establishing Reference Values. J Pediatr Gastroenterol Nutr 2022; 74:46-53. [PMID: 34694267 DOI: 10.1097/mpg.0000000000003338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVES The physiological number and distribution of mast cells (MCs) in the pediatric gastrointestinal (GI) tract is not well defined and reference values of normality are missing. To define a physiological and disease defining cut-off, a systematic histological exploration of MC distribution from the esophagus to the rectum in healthy as well as in patients with gastrointestinal food allergies (GFA) was performed. METHODS Nine pediatric subjects that exhibited unremarkable histopathological evaluations or underwent endoscopy for surveillance reasons after a previous polypectomy of single colonic juvenile polyps served as reference cohort. In all of these subjects, a chronic inflammatory disease (eg, inflammatory bowel disease, celiac disease) or allergy was excluded. In addition, a group of 15 patients with gastrointestinal complaints suspected to be caused by a GFA were investigated. Immunohistochemistry was performed from all biopsies using CD117 (c-Kit) as a reliable marker to identify MCs in the lamina propria. RESULTS There were distinct differences of MC counts in all parts of the pediatric GI tract. The highest counts of MCs in both symptomatic patients and control cohort, were found in the duodenum, terminal ileum, cecum and ascending colon. The lowest counts were found in the esophagus. Significant disparities between GFA and healthy subjects were found in the gastric corpus (22.1 ± 4.0/ high power field [HPF] vs 32.0 ± 10.1/HPF; P = 0.034) and ascending colon (44.8 ± 10.4/HPF vs 60.4 ± 24.3/HPF; P = 0.047). CONCLUSIONS Mucosal MC counts in the pediatric GI tract are higher than previously reported, with a considerable overlap between healthy and GFA patients. These results provide detailed information on distribution and numbers of MCs in pediatric allergic patients while allowing estimates of physiological values in childhood for the first time. With regard to diagnostic procedures in GFA further laboratory parameters have to be integrated.
Collapse
|
11
|
Pour Farid P, Eckstein M, Merkel S, Grützmann R, Hartmann A, Bruns V, Benz M, Schneider-Stock R, Geppert CI. Novel Criteria for Intratumoral Budding with Prognostic Relevance for Colon Cancer and Its Histological Subtypes. Int J Mol Sci 2021; 22:ijms222313108. [PMID: 34884913 PMCID: PMC8658236 DOI: 10.3390/ijms222313108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
Peritumoral budding and intratumoral budding (ITB) are important prognostic factors for colorectal cancer patients. Scientists worldwide have investigated the role of budding in tumor progression and its prognosis, but guidelines for reliably identifying tumor buds based on morphology are lacking. In this study, next-generation tissue microarray (ngTMA®) construction was used for tumor bud evaluation, and highly detailed rule-out annotation was used for tumor definition in pancytokeratin-stained tissue sections. Initially, tissues of 245 colon cancer patients were evaluated with high interobserver reliability, and a concordance of 96% was achieved. It was shown that high ITB scores were associated with poor distant metastasis-free survival (p = 0.006 with a cut-off of ≥10 buds). This cut-off was defined as the best maximum value from one of two/three ngTMA® cores (0.6 mm diameter). ITB in 30 cases of mucinous, medullary, and signet ring cell carcinoma was analyzed for the subsequent determination of differences in tumor bud analyses between those subtypes. In conclusion, blinded randomized punched cores in the tumor center can be useful for ITB detection. It can be assumed that this method is suitable for its adoption in clinical routines.
Collapse
Affiliation(s)
- Pantea Pour Farid
- Experimental Tumorpathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (P.P.F.); (R.S.-S.)
- Institute of Pathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (M.E.); (A.H.)
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
| | - Markus Eckstein
- Institute of Pathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (M.E.); (A.H.)
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
| | - Susanne Merkel
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
- Department of Surgery, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany
| | - Robert Grützmann
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
- Department of Surgery, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (M.E.); (A.H.)
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
| | - Volker Bruns
- Fraunhofer Institute for Integrated Circuits IIS, Am Wolfsmantel 33, 91058 Erlangen, Germany; (V.B.); (M.B.)
| | - Michaela Benz
- Fraunhofer Institute for Integrated Circuits IIS, Am Wolfsmantel 33, 91058 Erlangen, Germany; (V.B.); (M.B.)
| | - Regine Schneider-Stock
- Experimental Tumorpathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (P.P.F.); (R.S.-S.)
- Institute of Pathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (M.E.); (A.H.)
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
| | - Carol I. Geppert
- Institute of Pathology, University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (M.E.); (A.H.)
- Comprehensive Cancer Center-EMN (CCC), University Hospital, Friedrich-Alexander-University of Erlangen-Nüremberg (FAU), 91054 Erlangen, Germany; (S.M.); (R.G.)
- Correspondence: ; Tel.: +49-9131-85-43649
| |
Collapse
|
12
|
Wang HC, Chan LP, Wu CC, Hsiao HH, Liu YC, Cho SF, Du JS, Liu TC, Yang CH, Pan MR, Moi SH. Progression Risk Score Estimation Based on Immunostaining Data in Oral Cancer Using Unsupervised Hierarchical Clustering Analysis: A Retrospective Study in Taiwan. J Pers Med 2021; 11:jpm11090908. [PMID: 34575686 PMCID: PMC8466609 DOI: 10.3390/jpm11090908] [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: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022] Open
Abstract
This study aimed to investigate whether the progression risk score (PRS) developed from cytoplasmic immunohistochemistry (IHC) biomarkers is available and applicable for assessing risk and prognosis in oral cancer patients. Participants in this retrospective case-control study were diagnosed between 2012 and 2014 and subsequently underwent surgical intervention. The specimens from surgery were stained by IHC for 16 cytoplasmic target markers. We evaluated the results of IHC staining, clinical and pathological features, progression-free survival (PFS), and overall survival (OS) of 102 oral cancer patients using a novel estimation approach with unsupervised hierarchical clustering analysis. Patients were stratified into high-risk (52) and low-risk (50) groups, according to their PRS; a metric consisting of cytoplasmic PLK1, PhosphoMet, SGK2, and SHC1 expression. Moreover, PRS could be extended for use in the Cox proportional hazard regression model to estimate survival outcomes with associated clinical parameters. Our study findings revealed that the high-risk patients had a significantly increased risk in cancer progression compared with low-risk patients (hazard ratio (HR) = 2.20, 95% confidence interval (CI) = 1.10-2.42, p = 0.026). After considering the influences of demographics, risk behaviors, and tumor characteristics, risk estimation with PRS provided distinct PFS groups for patients with oral cancer (p = 0.017, p = 0.019, and p = 0.020). Our findings support that PRS could serve as an ideal biomarker for clinical use in risk stratification and progression assessment in oral cancer.
Collapse
Affiliation(s)
- Hui-Ching Wang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (J.-S.D.); (M.-R.P.)
- Department of Internal Medicine, Division of Hematology and Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-H.H.); (Y.-C.L.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Leong-Perng Chan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Otolaryngology-Head and Neck Surgery, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Kaohsiung Municipal Ta-Tung Hospital and Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Chun-Chieh Wu
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Hui-Hua Hsiao
- Department of Internal Medicine, Division of Hematology and Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-H.H.); (Y.-C.L.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yi-Chang Liu
- Department of Internal Medicine, Division of Hematology and Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-H.H.); (Y.-C.L.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Shih-Feng Cho
- Department of Internal Medicine, Division of Hematology and Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-H.H.); (Y.-C.L.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jeng-Shiun Du
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (J.-S.D.); (M.-R.P.)
- Department of Internal Medicine, Division of Hematology and Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-H.H.); (Y.-C.L.); (S.-F.C.)
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Ta-Chih Liu
- Department of Hematology-Oncology, Chang Bing Show Chwan Memorial Hospital, Changhua 505, Taiwan;
| | - Cheng-Hong Yang
- Department of Electronic Engineering, National Kaohsiung University of Science and Technology, Kaohsiung 807, Taiwan;
- Ph.D. Program in Biomedical Engineering, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (H.-C.W.); (J.-S.D.); (M.-R.P.)
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Sin-Hua Moi
- Center of Cancer Program Development, E-Da Cancer Hospital, I-Shou University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-6150022 (ext. 6135); Fax: +886-7-6150940
| |
Collapse
|
13
|
Combination of GP88 Expression in Tumor Cells and Tumor-Infiltrating Immune Cells Is an Independent Prognostic Factor for Bladder Cancer Patients. Cells 2021; 10:cells10071796. [PMID: 34359965 PMCID: PMC8306318 DOI: 10.3390/cells10071796] [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: 06/08/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/24/2022] Open
Abstract
Urothelial bladder cancer (BCa) is the ninth most commonly diagnosed cancer worldwide and accounts for approximately 3% of global cancer diagnoses. We are interested in prognostic markers that may characterize tumor cells (TCs) and immune cells (ICs) and their relationship in BCa. A potential candidate marker that meets these criteria is progranulin (GP88), which is expressed separately in TCs and ICs. We analyzed GP88 expression by immunohistochemistry (IHC) in 196 muscle-invasive BCa samples using a tissue microarray. The immunoreactive score for GP88 staining in TCs and the percentage of GP88-positive ICs was determined. An easy cutoff for the staining status of TCs (positive vs. negative) and ICs (0% vs. >0%) and, more generally, negative vs. positive GP88 staining could be applied. We detected 93 patients (47.4%) and 92 patients (46.9%) with GP88-positive TCs or ICs, respectively. The IHC results were correlated with clinicopathological and survival data. Positive GP88 staining in TCs appeared to be an independent poor prognostic factor for disease-specific survival (DSS) (RR (relative risk) = 1.74; p = 0.009) and recurrence-free survival (RFS) (RR = 1.92; p = 0.002). In contrast, negative GP88 staining in ICs was an independent negative predictor for overall survival (OS) (RR = 2.18; p < 0.001), DSS (RR = 2.84; p < 0.001) and RFS (RR = 2.91; p < 0.001) in multivariate Cox’s regression analysis. When combining GP88 staining in TCs and ICs, a specific combination of GP88-positive TCs and GP88-negative ICs was associated with a 2.54-fold increased risk of death, a 4.21-fold increased risk of disease-specific death and a 4.81-fold increased risk of recurrence compared to GP88-negative TCs and GP88-positive ICs. In summary, GP88 positivity in TCs is a negative prognostic factor for DSS and RFS. In addition, GP88 positivity can mark ICs that are associated with a good prognosis (OS, DSS and RFS). The combination of GP88 staining in TCs and ICs appears to be a significant independent prognostic biomarker in muscle-invasive BCa.
Collapse
|
14
|
Ji YX, Wang CZ, Li X, Li L. KLF5 promotes HpSlyD induced gastric intestinal metaplasia by activating Wnt/β-catenin pathway. Shijie Huaren Xiaohua Zazhi 2021; 29:274-281. [DOI: 10.11569/wcjd.v29.i6.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Krüppel like factor 5 (KLF5) and the Wnt/β-catenin pathway are hot topics in the research of tumorigenesis. Some studies have found that KLF5 is related to the differentiation and proliferation of intestinal epithelial cells, suggesting that KLF5 may be involved in the occurrence of intestinal metaplasia. However, there are few reports on the effect of KLF5 on intestinal metaplasia and the underlying molecular mechanism.
AIM To explore the effect of KLF5 on gastric intestinal metaplasia (GIM) induced by Helicobacter pylori (H. pylori) and the underlying mechanism.
METHODS The mRNA and protein expression of KLF5 and Wnt3a was detected by RT-PCR and Western blot, respectively. Gastric epithelial cell line GES1 was cultured in vitro and divided into a blank control group, HpSlyD group (200 ng/mL HpSlyD + negative sequence), KLF5 interference group (200 ng/mL HpSlyD + KLF5 siRNA), Wnt agonist group (200 ng/mL HpSlyD + KLF5 siRNA + lithium chloride), and Wnt agonist + KLF5 interference group. GES1 cell proliferation and apoptosis were determined by MTT assay and flow cytometry, respectively. RT-PCR was used to detect KLF5, Wnt3a, beta-catenin, wool protein 1 (VIL1), trefoil factor 2 (TFF2), and caudal homeobox factor 2 (CDX2) mRNA expression. The expression of VIL1, TFF2, and CDX2 proteins was detected by Western blot.
RESULTS The expression of KLF5 and Wnt3a mRNA and protein in intestinal metaplasia was significantly higher than that in the normal gastric mucosa (P < 0.01). The cell proliferation rates in the HpSlyD group and interference control group were significantly higher than that of the blank control group, and the apoptosis rates were significantly lower than that of the blank control group (P < 0.05). The cell proliferation rate of the KLF5 interference group was significantly lower than that of the HpSlyD group, and the apoptosis rate was significantly higher than that of the HpSlyD group (P < 0.05). The mRNA and protein expression of VIL1, TFF2, and CDX2 in the HpSlyD group and interference control group was significantly higher than that of the blank control group (P < 0.05), while the mRNA and protein expression of VIL1, TFF2, and CDX2 in the KLF5 interference group were significantly lower than those of the HpSlyD group (P < 0.05). The expression of Wnt3a, β-catenin, and CDX2 mRNA in the KLF5 interference group was significantly lower than that of the HpSlyD group (P < 0.05). The expression of Wnt3a, β-catenin, and CDX2 mRNA in the Wnt agonist + KLF5 interference group was significantly higher than that of the KLF5 interference group (P < 0.05).
CONCLUSION Interference of KLF5 expression can significantly inhibit HpSlyD induced gastric metaplasia. KLF5 may promote HpSlyD induced gastric metaplasia by activating the Wnt/β-catenin pathway, which provides a new target for clinical prevention of gastric cancer.
Collapse
Affiliation(s)
- Yong-Xin Ji
- Department of Emergency Medicine, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| | - Chun-Zi Wang
- Department of Emergency Medicine, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| | - Xue Li
- Department of Emergency Medicine, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| | - Li Li
- Department of Gastroenterology, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| |
Collapse
|
15
|
Wilkinson K, Ng W, Roberts TL, Becker TM, Lim SHS, Chua W, Lee CS. Tumour immune microenvironment biomarkers predicting cytotoxic chemotherapy efficacy in colorectal cancer. J Clin Pathol 2021; 74:625-634. [PMID: 33753562 PMCID: PMC8461409 DOI: 10.1136/jclinpath-2020-207309] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/22/2020] [Indexed: 12/22/2022]
Abstract
The role of the local tumour and stromal immune landscape is increasingly recognised to be important in cancer development, progression and response to therapy. The composition, function, spatial orientation and gene expression profile of the infiltrate of the innate and adaptive immune system at the tumour and surrounding tissue has an established prognostic role in colorectal cancer (CRC). Multiple studies have confirmed that a tumour immune microenvironment (TIME) reflective of a type 1 adaptive immune response is associated with improved prognosis. There have been significant efforts to evolve these observations into validated, histopathology-based prognostic biomarkers, such as the Immunoscore. However, the clinical need lies much more in the development of predictive, not prognostic, biomarkers which have the potential to improve patient outcomes. This is particularly pertinent to help guide cytotoxic chemotherapy use in CRC, which remains the standard of care. Cytotoxic chemotherapy has recognised immunomodulatory activity distinct from its antimitotic effects, including mechanisms such as immunogenic cell death (ICD) and induction/inhibition of key immune players. Response to chemotherapy may differ with regard to molecular subtype of CRC, which are strongly associated with immune phenotypes. Thus, immune markers are potentially useful, though under-reported, predictive biomarkers. In this review, we discuss the impact of the TIME on response to cytotoxic chemotherapy in CRC, with a focus on baseline immune markers, and associated genomic and transcriptomic signatures.
Collapse
Affiliation(s)
- Kate Wilkinson
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia .,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Weng Ng
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Therese M Becker
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Stephanie Hui-Su Lim
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Macarthur Cancer Therapy Centre, Campbelltown Hospital, Campbelltown, New South Wales, Australia
| | - Wei Chua
- Liverpool Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia.,School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Cheok Soon Lee
- School of Medicine, Western Sydney University, Liverpool, New South Wales, Australia.,Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia.,Department of Anatomical Pathology, Liverpool Hospital, Liverpool, New South Wales, Australia
| |
Collapse
|
16
|
Nguyen HG, Blank A, Dawson HE, Lugli A, Zlobec I. Classification of colorectal tissue images from high throughput tissue microarrays by ensemble deep learning methods. Sci Rep 2021; 11:2371. [PMID: 33504830 PMCID: PMC7840737 DOI: 10.1038/s41598-021-81352-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
Tissue microarray (TMA) core images are a treasure trove for artificial intelligence applications. However, a common problem of TMAs is multiple sectioning, which can change the content of the intended tissue core and requires re-labelling. Here, we investigate different ensemble methods for colorectal tissue classification using high-throughput TMAs. Hematoxylin and Eosin (H&E) core images of 0.6 mm or 1.0 mm diameter from three international cohorts were extracted from 54 digital slides (n = 15,150 cores). After TMA core extraction and color enhancement, five different flows of independent and ensemble deep learning were applied. Training and testing data with 2144 and 13,006 cores included three classes: tumor, normal or "other" tissue. Ground-truth data were collected from 30 ngTMA slides (n = 8689 cores). A test augmentation is applied to reduce the uncertain prediction. Predictive accuracy of the best method, namely Soft Voting Ensemble of one VGG and one CapsNet models was 0.982, 0.947 and 0.939 for normal, "other" and tumor, which outperformed to independent or ensemble learning with one base-estimator. Our high-accuracy algorithm for colorectal tissue classification in high-throughput TMAs is amenable to images from different institutions, core sizes and stain intensity. It helps to reduce error in TMA core evaluations with previously given labels.
Collapse
Affiliation(s)
- Huu-Giao Nguyen
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Annika Blank
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
- Institute of Pathology, Triemli City Hospital, Birmensdorferstrasse 497, 8063, Zurich, Switzerland
| | - Heather E Dawson
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Alessandro Lugli
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland.
| |
Collapse
|
17
|
Immune Cell-Associated Protein Expression Helps to Predict Survival in Muscle-Invasive Urothelial Bladder Cancer Patients after Radical Cystectomy and Optional Adjuvant Chemotherapy. Cells 2021; 10:cells10010159. [PMID: 33467469 PMCID: PMC7829767 DOI: 10.3390/cells10010159] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/12/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Adjuvant chemotherapy following radical cystectomy is a common therapy for muscle invasive bladder cancer (MIBC) patients. No applicable biomarkers exist to predict which patients will benefit from chemotherapy. Three immune cell markers, the chemokine CC motif ligand 2 (CCL2), the pan macrophage marker CD68, and the M2 macrophage marker CD163, were examined using immunohistochemistry to determine their predictive value for chemotherapy responses in different nodal stage and tumor stage subgroups. The presence of tumor-infiltrating immune cells, characterized by the markers CD68, CD163, and CCL2, was associated with a superior prognosis, and chemotherapy may not add an advantage for prognosis. However, a depleted immune microenvironment, here represented as a reduction or loss of macrophages, helped to predict the benefit of chemotherapy in N1 + 2 stage patients. Altogether, it is meaningful to consider the abundance of immune cells, such as macrophages, to better predict the response to chemotherapy for bladder cancer (BCa) patients after radical treatment. Abstract Bladder cancer (BCa) is the tenth most commonly diagnosed malignant cancer worldwide. Although adjuvant chemotherapy following radical cystectomy is a common therapy for muscle invasive bladder cancer patients, no applicable biomarkers exist to predict which patients will benefit from chemotherapy. In this study, we examined three immune cell markers, the chemokine CC motif ligand 2 (CCL2), the pan macrophage marker cluster of differentiation 68 (CD68) and the M2 macrophage marker cluster of differentiation 163 (CD163), using immunohistochemistry to determine their predictive value for the chemotherapy response in different nodal stage (pN0 vs. pN1 + 2) and tumor stage subgroups (pT2 vs. pT3 + 4). The prognosis was studied in terms of the overall survival (OS), disease-specific survival (DSS), and recurrence-free-survival (RFS) in 168 muscle invasive BCa patients. Chemotherapy was associated with a poorer prognosis in patients with a higher expression of the immune markers CCL2 (RFS), CD68 (DSS and RFS), and CD163 (DSS and RFS) in the N0 group and with poorer survival in patients with a higher expression of the immune markers CCL2 (OS, DSS, and RFS), CD68 (OS, DSS, and RFS), and CD163 (OS, DSS, and RFS) in the pT2 group when compared with treatments without chemotherapy. In contrast, chemotherapy was associated with a better prognosis in patients with a low expression of the immune markers CCL2 (DSS and RFS), CD68 (OS, DSS, and RFS), and CD163 (OS) in the N1 + 2 group. In addition, chemotherapy was associated with improved survival in patients with a low expression of the immune marker CD68 (OS and DSS) and there was a trend for a better prognosis in patients with a low expression of CD163 (OS) in the pT3 + 4 group compared to patients not treated with chemotherapy. Interestingly, CD68 appeared to be the most applicable immune marker to stratify patients by the outcome of chemotherapy in the nodal stage and tumor stage groups. Overall, we suggest that, in addition to the clinical factors of tumor stage and nodal stage, it is also meaningful to consider the abundance of immune cells, such as macrophages, to better predict the response to chemotherapy for BCa patients after radical treatment.
Collapse
|
18
|
Bremer FP, Czeczko NG, CollaÇo LM, Rutz LEAC, Gionedis G, Yamakawa CK. ARE CDX2, BETA-CATENIN AND WNT IMMUNOMARCHERS USEFUL FOR EVALUATING THE CHANCE OF DISEASE PROGRESSION OR EVOLUTION TO DEATH IN PATIENTS WITH COLORECTAL CANCER? ACTA ACUST UNITED AC 2020; 33:e1534. [PMID: 33331430 PMCID: PMC7747481 DOI: 10.1590/0102-672020200003e1534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 07/03/2020] [Indexed: 01/05/2023]
Abstract
Background:
Colorectal cancer (CRC) is one of the most common types of cancer in the
world. Over time, intestinal epithelial cells undergo mutations that may
lead to proliferative advantage and the emergence of cancer. Mutations in
the beta-catenin pathway are amongst those described in the development of
CRC.
Aim:
To verify the existence of a relation between the presence of Wnt3,
beta-catenin and CDX2 in colorectal cancer samples and clinical outcomes
such as disease progression or death.
Method:
Wnt3a, beta-catenin and CDX2 immunohistochemistry was performed on CRC tissue
microarray samples (n=122), and analysis regarding the relation between
biomarker expression and disease progression or death was performed.
Results:
No significant difference was found between the presence or absence of CDX2,
beta-catenin or Wnt3a expression and clinical stage, tumor grade, disease
progression or death.
Conclusion:
CDX2, beta-catenin and Wnt3a are not useful to predict prognosis in patients
with CRC.
Collapse
Affiliation(s)
- Fabiola Pabst Bremer
- Mackenzie Evangelical Faculty of Paraná, Curitiba, PR, Brazil.,University Evangelical Mackenzie Hospital, Curitiba, PR, Brazil
| | | | | | | | | | | |
Collapse
|
19
|
Remes SM, Leijon H, Vesterinen T, Louhimo J, Pulkkinen V, Ezer S, Kere J, Haglund C, Arola J. PCSK2 expression in neuroendocrine tumors points to a midgut, pulmonary, or pheochromocytoma-paraganglioma origin. APMIS 2020; 128:563-572. [PMID: 32794589 PMCID: PMC7702075 DOI: 10.1111/apm.13071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Neuroendocrine tumors (NETs) are often diagnosed from the metastases of an unknown primary tumor. Specific immunohistochemical (IHC) markers indicating the location of a primary tumor are needed. The proprotein convertase subtilisin/kexin type 2 (PCSK2) is found in normal neural and neuroendocrine cells, and known to express in NETs. We investigated the tissue microarray (TMA) of 86 primary tumors from 13 different organs and 9 metastatic NETs, including primary tumor‐metastasis pairs, for PCSK2 expression with polymer‐based IHC. PCSK2 was strongly positive in all small intestine and appendiceal NETs, the so‐called midgut NETs, in most pheochromocytomas and paragangliomas, and in some of the typical and atypical pulmonary carcinoid tumors. NETs showing strong positivity were re‐evaluated in larger tumor cohorts confirming the primary observation. In the metastases, the expression of PCSK2 mirrored that of the corresponding primary tumors. We found negative or weak staining in NETs from the thymus, gastric mucosa, pancreas, rectum, thyroid, and parathyroid. PCSK2 expression did not correlate with Ki‐67 in well‐differentiated NETs. Our data suggest that PCSK2 positivity can indicate the location of the primary tumor. Thus, PCSK2 could function in the IHC panel determined from screening metastatic NET biopsies of unknown primary origins.
Collapse
Affiliation(s)
- Satu Maria Remes
- Department of Pathology, Helsinki University Hospital and HUSLAB, University of Helsinki, Helsinki, Finland
| | - Helena Leijon
- Department of Pathology, Helsinki University Hospital and HUSLAB, University of Helsinki, Helsinki, Finland
| | - Tiina Vesterinen
- Department of Pathology, Helsinki University Hospital and HUSLAB, University of Helsinki, Helsinki, Finland.,Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Johanna Louhimo
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Ville Pulkkinen
- Heart and Lung Center, Division of Pulmonary Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Sini Ezer
- Research Programs Unit, Program for Molecular Neurology, University of Helsinki, Helsinki, Finland
| | - Juha Kere
- Research Programs Unit, Program for Molecular Neurology, University of Helsinki, Helsinki, Finland.,Folkhälsan Institute of Genetics, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Novum, Stockholm, Sweden
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Translational Cancer Medicine, Research Program Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Arola
- Department of Pathology, Helsinki University Hospital and HUSLAB, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
CCL2 Expression in Tumor Cells and Tumor-Infiltrating Immune Cells Shows Divergent Prognostic Potential for Bladder Cancer Patients Depending on Lymph Node Stage. Cancers (Basel) 2020; 12:cancers12051253. [PMID: 32429318 PMCID: PMC7281525 DOI: 10.3390/cancers12051253] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/29/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
Bladder cancer (BCa) is the ninth most commonly diagnosed cancer worldwide. Although there are several well-established molecular and immunological classifications, markers for tumor cells and immune cells that are associated with prognosis are still needed. The chemokine CC motif ligand 2 (CCL2) could be such a marker. We analyzed the expression of CCL2 by immunohistochemistry (IHC) in 168 muscle invasive BCa samples using a tissue microarray. Application of a single cut-off for the staining status of tumor cells (TCs; positive vs. negative) and immune cells (ICs; ≤6% of ICs vs. >6% of ICs) revealed 57 cases (33.9%) and 70 cases (41.7%) with CCL2-positive TCs or ICs, respectively. IHC results were correlated with clinicopathological and survival data. Positive CCL2 staining in TCs was associated with shorter overall survival (OS), disease-specific survival (DSS), and relapse-free survival (RFS) (p = 0.004, p = 0.036, and p = 0.047; log rank test) and appeared to be an independent prognostic factor for OS (RR = 1.70; p = 0.007; multivariate Cox’s regression analysis). In contrast, positive CCL2 staining in the ICs was associated with longer OS, DSS, and RFS (p = 0.032, p = 0.001, and p = 0.001; log rank test) and appeared to be an independent prognostic factor for DSS (RR = 1.77; p = 0.031; multivariate Cox’s regression analysis). Most interestingly, after separating the patients according to their lymph node status (N0 vs. N1+2), CCL2 staining in the ICs was differentially associated with prognosis. In the N0 group, CCL2 positivity in the ICs was a positive independent prognostic factor for OS (RR = 1.99; p = 0.014), DSS (RR = 3.17; p = 0.002), and RFS (RR = 3.10; p = 0.002), whereas in the N1+2 group, CCL2 positivity was a negative independent factor for OS (RR = 3.44; p = 0.019)) and RFS (RR = 4.47; p = 0.010; all multivariate Cox’s regression analyses). In summary, CCL2 positivity in TCs is a negative prognostic factor for OS, and CCL2 can mark ICs that are differentially associated with prognosis depending on the nodal stage of BCa patients. Therefore, CCL2 staining of TCs and ICs is suggested as a prognostic biomarker for BCa patients.
Collapse
|
21
|
The Determination of Immunomodulation and Its Impact on Survival of Rectal Cancer Patients Depends on the Area Comprising a Tissue Microarray. Cancers (Basel) 2020; 12:cancers12030563. [PMID: 32121328 PMCID: PMC7139832 DOI: 10.3390/cancers12030563] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/02/2020] [Accepted: 02/17/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND T cell density in colorectal cancer (CRC) has proven to be of high prognostic importance. Here, we evaluated the influence of a hyperfractionated preoperative short-term radiation protocol (25 Gy) on immune cell density in tumor samples of rectal cancer (RC) patients and on patient survival. In addition, we assessed spatial tumor heterogeneity by comparison of analogue T cell quantification on full tissue sections with digital T cell quantification on a virtually established tissue microarray (TMA). METHODS A total of 75 RC patients (60 irradiated, 15 treatment-naïve) were defined for retrospective analysis. RC samples were processed for immunohistochemistry (CD3, CD8, PD-1, PD-L1). Analogue (score 0-3) as well as digital quantification (TMA: 2 cores vs. 6 cores, mean T cell count) of marker expression in 2 areas (central tumor, CT; invasive margin, IM) was performed. Survival was estimated on the basis of analogue as well as digital marker densities calculated from 2 cores (Immunoscore: CD3/CD8 ratio) and 6 cores per tumor area. RESULTS Irradiated RC samples showed a significant decrease in CD3 and CD8 positive T cells, independent of quantification mode. T cell densities of 6 virtual cores approximated to T cell densities of full tissue sections, independent of individual core density or location. Survival analysis based on full tissue section quantification demonstrated that CD3 and CD8 positive T cells as well as PD-1 positive tumor infiltrating leucocytes (TILs) in the CT and the IM had a significant impact on disease-free survival (DFS) as well as overall survival (OS). In addition, CD3 and CD8 positive T cells as well as PD-1 positive TILs in the IM proved as independent prognostic factors for DFS and OS; in the CT, PD-1 positive TILs predicted DFS and CD3 and CD8 positive T cells as well as PD-1 positive TILs predicted OS. Survival analysis based on virtual TMA showed no impact on DFS or OS. CONCLUSION Spatial tumor heterogeneity might result in inadequate quantification of immune marker expression; however, if using a TMA, 6 cores per tumor area and patient sample represent comparable amounts of T cell densities to those quantified on full tissue sections. Consistently, the tissue area used for immune marker quantification represents a crucial factor for the evaluation of prognostic and predictive biomarker potential.
Collapse
|
22
|
Lopes N, Bergsland CH, Bjørnslett M, Pellinen T, Svindland A, Nesbakken A, Almeida R, Lothe RA, David L, Bruun J. Digital image analysis of multiplex fluorescence IHC in colorectal cancer recognizes the prognostic value of CDX2 and its negative correlation with SOX2. J Transl Med 2020; 100:120-134. [PMID: 31641225 PMCID: PMC6917572 DOI: 10.1038/s41374-019-0336-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/18/2019] [Accepted: 09/18/2019] [Indexed: 01/10/2023] Open
Abstract
Flourescence-based multiplex immunohistochemistry (mIHC) combined with multispectral imaging and digital image analysis (DIA) is a quantitative high-resolution method for determination of protein expression in tissue. We applied this method for five biomarkers (CDX2, SOX2, SOX9, E-cadherin, and β-catenin) using tissue microarrays of a Norwegian unselected series of primary colorectal cancer. The data were compared with previously obtained chromogenic IHC data of the same tissue cores, visually assessed by the Allred method. We found comparable results between the methods, although confirmed that DIA offered improved resolution to differentiate cases with high and low protein expression. However, we experienced inherent challenges with digital image analysis of membrane staining, which was better assessed visually. DIA and mIHC enabled quantitative analysis of biomarker coexpression on the same tissue section at the single-cell level, revealing a strong negative correlation between the differentiation markers CDX2 and SOX2. Both methods confirmed known prognostic associations for CDX2, but DIA improved data visualization and detection of clinicopathological and biological associations. In summary, mIHC combined with DIA is an efficient and reliable method to evaluate protein expression in tissue, here shown to recapitulate and improve detection of known clinicopathological and survival associations for the emerging biomarker CDX2, and is therefore a candidate approach to standardize CDX2 detection in pathology laboratories.
Collapse
Affiliation(s)
- Nair Lopes
- 0000 0004 0389 8485grid.55325.34Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway ,0000 0001 1503 7226grid.5808.5i3S—Institute for Research and Innovation in Health, University of Porto, Porto, Portugal ,0000 0001 1503 7226grid.5808.5IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal ,0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Christian Holst Bergsland
- 0000 0004 0389 8485grid.55325.34Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway ,0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway ,0000 0004 1936 8921grid.5510.1Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Merete Bjørnslett
- 0000 0004 0389 8485grid.55325.34Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway ,0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Teijo Pellinen
- 0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway ,0000 0004 0410 2071grid.7737.4Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Aud Svindland
- 0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway ,0000 0004 1936 8921grid.5510.1Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Arild Nesbakken
- 0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway ,0000 0004 1936 8921grid.5510.1Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Raquel Almeida
- 0000 0001 1503 7226grid.5808.5i3S—Institute for Research and Innovation in Health, University of Porto, Porto, Portugal ,0000 0001 1503 7226grid.5808.5Faculty of Medicine, University of Porto, Porto, Portugal ,0000 0001 1503 7226grid.5808.5Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Ragnhild A. Lothe
- 0000 0004 0389 8485grid.55325.34Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway ,0000 0004 0389 8485grid.55325.34K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway ,0000 0004 1936 8921grid.5510.1Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Leonor David
- 0000 0001 1503 7226grid.5808.5i3S—Institute for Research and Innovation in Health, University of Porto, Porto, Portugal ,0000 0001 1503 7226grid.5808.5IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal ,0000 0001 1503 7226grid.5808.5Faculty of Medicine, University of Porto, Porto, Portugal
| | - Jarle Bruun
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway. .,K.G. Jebsen Colorectal Cancer Research Centre, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
23
|
Zahnd S, Braga-Lagache S, Buchs N, Lugli A, Dawson H, Heller M, Zlobec I. A Digital Pathology-Based Shotgun-Proteomics Approach to Biomarker Discovery in Colorectal Cancer. J Pathol Inform 2019; 10:40. [PMID: 31921488 PMCID: PMC6939342 DOI: 10.4103/jpi.jpi_65_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 02/21/2019] [Indexed: 12/26/2022] Open
Abstract
Background Biomarkers in colorectal cancer are scarce, especially for patients with Stage 2 disease. The aim of our study was to identify potential prognostic biomarkers from colorectal cancers using a novel combination of approaches, whereby digital pathology is coupled to shotgun proteomics followed by validation of candidates by immunohistochemistry (IHC) using digital image analysis (DIA). Methods and Results Tissue cores were punched from formalin-fixed paraffin-embedded colorectal cancers from patients with Stage 2 and 3 disease (n = 26, each). Protein extraction and liquid chromatography-mass spectrometry (MS) followed by analysis using three different methods were performed. Fold changes were evaluated. The candidate biomarker was validated by IHC on a series of 413 colorectal cancers from surgically treated patients using a next-generation tissue microarray. DIA was performed by using a pan-cytokeratin serial alignment and quantifying staining within the tumor and normal tissue epithelium. Analysis was done in QuPath and Brightness_Max scores were used for statistical analysis and clinicopathological associations. MS identified 1947 proteins with at least two unique peptides. To reinforce the validity of the biomarker candidates, only proteins showing a significant (P < 0.05) fold-change using all three analysis methods were considered. Eight were identified, and of these, cathepsin B was selected for further validation. DIA revealed strong associations between higher cathepsin B expression and less aggressive tumor features, including tumor node metastasis stage and lymphatic vessel and venous vessel invasion (P < 0.001, all). Cathepsin B was associated with more favorable survival in univariate analysis only. Conclusions Our results present a novel approach to biomarker discovery that includes MS and digital pathology. Cathepsin B expression analyzed by DIA within the tumor epithelial compartment was identified as a strong feature of less aggressive tumor behavior and favorable outcome, a finding that should be further investigated on a more functional level.
Collapse
Affiliation(s)
- Stefan Zahnd
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Sophie Braga-Lagache
- Department for BioMedical Research, Proteomics and Mass Spectrometry Core Facility, University of Bern, Bern, Switzerland
| | - Natasha Buchs
- Department for BioMedical Research, Proteomics and Mass Spectrometry Core Facility, University of Bern, Bern, Switzerland
| | | | - Heather Dawson
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Department for BioMedical Research, Proteomics and Mass Spectrometry Core Facility, University of Bern, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
24
|
Böhm J, Muenzner JK, Caliskan A, Ndreshkjana B, Erlenbach-Wünsch K, Merkel S, Croner R, Rau TT, Geppert CI, Hartmann A, Roehe AV, Schneider-Stock R. Loss of enhancer of zeste homologue 2 (EZH2) at tumor invasion front is correlated with higher aggressiveness in colorectal cancer cells. J Cancer Res Clin Oncol 2019; 145:2227-2240. [PMID: 31317325 PMCID: PMC6708512 DOI: 10.1007/s00432-019-02977-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/06/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Enhancer of zeste homolog 2 (EZH2) is associated with epigenetic gene silencing and aggressiveness in many tumor types. However, the prognostic impact of high EZH2 expression is controversially discussed for colorectal cancer. For this reason, we immunohistochemically analyzed EZH2 expression in 105 specimens from colon cancer patients separately for tumor center and invasion front. METHODS All sections from tissue microarrays were evaluated manually and digitally using Definiens Tissue Studio software (TSS). To mirror-image the EZH2 status at the tumor invasion front, we treated HCT116 colon cancer cells with the EZH2 inhibitor 3-Deazaneplanocin A (DZNep) and studied the growth of in ovo xenografts in the chorioallantoic membrane (CAM) assay. RESULTS We showed a significant decrease in EZH2 expression and the repressive H3K27me3 code at the tumor invasion front as supported by the TSS-constructed heatmaps. Loss of EZH2 at tumor invasion front, but not in tumor center was correlated with unfavorable prognosis and more advanced tumor stages. The observed cell cycle arrest in vitro and in vivo was associated with higher tumor aggressiveness. Xenografts formed by DZNep-treated HCT116 cells showed loosely packed tumor masses, infiltrative growth into the CAM, and high vessel density. CONCLUSION The differences in EZH2 expression between tumor center and invasion front as well as different scoring and cutoff values can most likely explain controversial literature data concerning the prognostic value of EZH2. Epigenetic therapies using EZH2 inhibitors have to be carefully evaluated for each specific tumor type, since alterations in cell differentiation might lead to unfavorable results.
Collapse
Affiliation(s)
- Julian Böhm
- Experimental Tumorpathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Julienne Kathrin Muenzner
- Experimental Tumorpathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Aylin Caliskan
- Experimental Tumorpathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Benardina Ndreshkjana
- Experimental Tumorpathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany.,Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Katharina Erlenbach-Wünsch
- Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Susanne Merkel
- Department of Surgery, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 12, 91054, Erlangen, Germany
| | - Roland Croner
- Department of Surgery, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 12, 91054, Erlangen, Germany.,Department of Surgery, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Tilman T Rau
- Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany.,Institute of Pathology, University Bern, Murtenstr. 31, 3008, Bern, Switzerland
| | - Carol Immanuel Geppert
- Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Arndt Hartmann
- Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Adriana Vial Roehe
- Department of Pathology, Federal University of Health Sciences of Porto Alegre (UFCSPA), R. Sarmento Leite, 245-Centro Histórico, Porto Alegre, RS, 90050-170, Brazil
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Universitätsstr. 22, 91054, Erlangen, Germany. .,Institute of Pathology, Friedrich-Alexander-University of Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany.
| |
Collapse
|
25
|
Vesterinen T, Leijon H, Mustonen H, Remes S, Knuuttila A, Salmenkivi K, Vainio P, Arola J, Haglund C. Somatostatin Receptor Expression Is Associated With Metastasis and Patient Outcome in Pulmonary Carcinoid Tumors. J Clin Endocrinol Metab 2019; 104:2083-2093. [PMID: 30657933 DOI: 10.1210/jc.2018-01931] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 01/11/2019] [Indexed: 02/07/2023]
Abstract
CONTEXT Pulmonary carcinoids (PCs) belong to neuroendocrine tumors that often overexpress somatostatin receptors (SSTRs). This overexpression provides a molecular basis for tumor imaging and treatment with somatostatin analogs. OBJECTIVE To evaluate SSTR1 to SSTR5 distribution in a large set of PC tumors and to investigate whether the expression is associated with clinicopathological and outcome data. DESIGN, SETTING, AND PATIENTS This retrospective study was conducted at Helsinki University Hospital and University of Helsinki. It included 178 PC tumors coupled with patients' clinical data retrieved through Finnish biobanks. After histological reclassification, tissue specimens were processed into next-generation tissue microarray format and stained immunohistochemically with monoclonal SSTR1 to SSTR5 antibodies. MAIN OUTCOME MEASURE SSTR1 to SSTR5 expression in PC tumors. RESULTS Expression of SSTR1 to SSTR5 was detected in 52%, 75%, 56%, 16%, and 32% of the tumors, respectively. Membrane-bound staining was observed for all receptors. SSTR2 negativity and SSTR4 positivity was associated with lymph node involvement at the time of surgery (P = 0.014 and P = 0.017, respectively) and with distant metastasis (P = 0.027 and P = 0.015, respectively). SSTR3 and SSTR4 expression was associated with increased risk of shorter survival [P = 0.046, hazard ratio (HR) 4.703, 95% CI 1.027 to 21.533; and P = 0.013, HR 6.64, 95% CI 1.48 to 29.64, respectively], whereas expression of SSTR1 and SSTR2 was associated with improved outcome (P = 0.021, HR 0.167, 95% CI 0.037 to 0.765; and P = 0.022, HR 0.08, 95% CI 0.01 to 0.70, respectively). CONCLUSION SSTR1 to SSTR5 expression is observed in PCs. As SSTR expression is associated with the tumor's metastatic potential and patient outcome, these receptors may offer the possibility for individualized prognosis estimation.
Collapse
Affiliation(s)
- Tiina Vesterinen
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Helena Leijon
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Mustonen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Satu Remes
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aija Knuuttila
- Department of Pulmonary Medicine, Heart and Lung Center and Cancer Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kaisa Salmenkivi
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Paula Vainio
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Johanna Arola
- HUSLAB, Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
26
|
CDX2 and Muc2 immunohistochemistry as prognostic markers in stage II colon cancer. Hum Pathol 2019; 90:70-79. [PMID: 31121192 DOI: 10.1016/j.humpath.2019.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 12/31/2022]
Abstract
The treatment for colorectal cancer is largely surgical followed by adjuvant chemotherapy in high-risk cases. In patients with stage II cancer, there is no clear benefit for chemotherapy, and the current tools for assessment of risk are inadequate. A recent study identified that colorectal cancer with a gene signature similar to undifferentiated colonic stem cells was associated with a worse outcome. It was later shown that loss of CDX2 detected by immunohistochemistry (IHC) alone resulted in a worse prognosis and that this could be used to predict patients who would benefit from chemotherapy. Having observed that CDX2 expression can be patchy, we elected to validate these prior results for clinical practice using whole-slide IHC. The pathology of all cases was reviewed, and 3 blocks were selected for CDX2 IHC. We also expanded the panel beyond CDX2 to assess whether other markers in the gene signature including CDX1, Muc2, GPX2, and villin could better predict outcome. Among 210 cases, CDX2 expression was diffusely lost in 11% and focally lost in 23% of cases. There was no difference in survival based on CDX2 expression, but Muc2 loss was associated with reduced survival (hazard ratio, 3.32; 95% confidence interval, 1.20 to 9.20). No significant differences in outcome were identified based on CDX1, GPX2, or villin expression. In keeping with this, assessment of The Cancer Genome Atlas gene expression data demonstrated that decreased Muc2 expression was associated with reduced overall survival. Our results with whole-slide IHC are different from the previous studies and caution against the use of CDX2 in isolation as a prognostic marker in clinical practice. We have identified that loss of Muc2 is associated with reduced survival. This supports the use of the colonic differentiation gene expression signature to identify high-risk patients but cautions against the use of any one IHC-based marker in isolation.
Collapse
|
27
|
Challenging the prognostic impact of the new WHO and TNM classifications with special emphasis on HPV status in penile carcinoma. Virchows Arch 2019; 475:211-221. [DOI: 10.1007/s00428-019-02566-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/12/2019] [Accepted: 03/25/2019] [Indexed: 12/21/2022]
|
28
|
Sandberg TP, Sweere I, van Pelt GW, Putter H, Vermeulen L, Kuppen PJ, Tollenaar RAEM, Mesker WE. Prognostic value of low CDX2 expression in colorectal cancers with a high stromal content - a short report. Cell Oncol (Dordr) 2019; 42:397-403. [PMID: 30847807 DOI: 10.1007/s13402-019-00436-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2019] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Lack of expression of the intestinal transcription factor CDX2 in colorectal cancer (CRC) identifies patients with a poor prognosis. This biomarker has previously been suggested to be prognostic in CRCs with a high stromal content based on mRNA expression data. We investigated the prognostic value of CDX2 expression in microsatellite stable CRC stratified by stromal content using microscopy-based techniques. METHODS AND RESULTS The study included a cohort of 236 patients with stage I-IV CRC. We assessed by microscopy the tumour-stroma ratio (TSR) and the immunohistochemical CDX2 intensity. We found that patients of the stroma-high group had a worse prognosis compared to those of the stroma-low group [disease-free survival in a multivariate analysis (DFSmultivariate) HR 1.52 (95% CI 1.05-2.21)]. In our cohort, low CDX2 expression (14.6%) showed prognostic value for DFSmultivariate [HR 1.93 (95% CI 1.16-3.23)]. Interestingly, when stratifying the cohort by TSR, no prognostic difference was observed related to CDX2 expression in stroma-low tumours. However, CDX2 expression was found to be prognostic within the stroma-high group [DFSmultivariate HR 3.02 (95% CI 1.49-6.13)]. The p value for interaction between TSR and CDX2 status was borderline significant in DFS (p = 0.071). CONCLUSIONS The present study confirms a poor outcome of patients with stroma-high tumours. Low CDX2 expression in tumours with a high stromal content identified patients with a particularly poor prognosis. The present study did not reveal a clear difference in TSR associated with CDX2 status and survival. This method, solely based on microscopy, identifies patients who have a high risk of relapse and a poor outcome, and who may benefit from targeted therapy.
Collapse
Affiliation(s)
- Tessa P Sandberg
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands. .,Department of Pathology, Leiden University Medical Centrum, Leiden, The Netherlands.
| | - Iris Sweere
- Department of Pathology, Leiden University Medical Centrum, Leiden, The Netherlands
| | - Gabi W van Pelt
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Hein Putter
- Department of Medical Statistics, Leiden University Medical Centrum, Leiden, The Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Academic Medical Center & Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Peter J Kuppen
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Rob A E M Tollenaar
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| | - Wilma E Mesker
- Department of Surgery, Leiden University Medical Centrum, Albinusdreef 2, 2333ZA, Leiden, The Netherlands
| |
Collapse
|
29
|
Eckstein M, Jung R, Weigelt K, Sikic D, Stöhr R, Geppert C, Agaimy A, Lieb V, Hartmann A, Wullich B, Wach S, Taubert H. Piwi-like 1 and -2 protein expression levels are prognostic factors for muscle invasive urothelial bladder cancer patients. Sci Rep 2018; 8:17693. [PMID: 30523270 PMCID: PMC6283838 DOI: 10.1038/s41598-018-35637-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 11/06/2018] [Indexed: 01/03/2023] Open
Abstract
Piwi-like proteins are essential for stem-cell maintenance and self-renewal in multicellular organisms. We analyzed the expression of Piwi-like 1 and Piwi-like 2 by immunohistochemistry (IHC) in 95 muscle invasive bladder cancer (MIBC) samples using tissue microarray. Application of an immunoreactive score (IRS) revealed 37 and 45 patients who were Piwi-like 1 and -2 positive (IRS > 2). IHC results were correlated with clinico-pathological and survival data. The expression of both proteins was positively correlated with each other, lymph node metastasis and expression of CK20 and GATA 3. A negative correlation for both proteins was detected for disease-specific survival (DSS), recurrence, Ki67/MIB1 proliferation index, and CK5 expression. Detection of Piwi-like 1 protein positivity was associated with poor DSS (P = 0.019; log rank test, Kaplan-Meier analysis), and in multivariate Cox’s analysis (adjusted to tumor stage and tumor grade), it was an independent prognostic factor for DSS (RR = 2.16; P = 0.011). Piwi-like 2 positivity was associated with DSS (P = 0.008) and recurrence-free survival (RFS; P = 0.040), and in multivariate Cox’s analysis, Piwi-like 2 positivity was an independent prognostic factor for DSS (RR = 2.46; P = 0.004) and RFS (RR = 3.0; P = 0.003). Most interestingly, in the basal type patient subgroup (CK5+/GATA3−), Piwi-like 2 positivity was associated with poorer DSS, OS and RFS (P < 0.001, P = 0.004 and P = 0.05; log rank test). In multivariate analysis, Piwi-like 2 positivity was an independent prognostic factor for DSS (RR = 12.70; P = 0.001), OS (RR = 6.62; = 0.008) and RFS (RR=13.0; P = 0.040). In summary, Piwi-like 1 and -2 positivity are associated with clinico-pathological factors and survival. Both Piwi-like proteins are suggested as biomarkers for MIBC patients.
Collapse
Affiliation(s)
- Markus Eckstein
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Rudolf Jung
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Katrin Weigelt
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Danijel Sikic
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Robert Stöhr
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Carol Geppert
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Abbas Agaimy
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Verena Lieb
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Arndt Hartmann
- Institute of Pathology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Bernd Wullich
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Sven Wach
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany
| | - Helge Taubert
- Department of Urology and Pediatric Urology, University Hospital Erlangen, FAU Erlangen-Nürnberg, Germany.
| |
Collapse
|
30
|
Graule J, Uth K, Fischer E, Centeno I, Galván JA, Eichmann M, Rau TT, Langer R, Dawson H, Nitsche U, Traeger P, Berger MD, Schnüriger B, Hädrich M, Studer P, Inderbitzin D, Lugli A, Tschan MP, Zlobec I. CDX2 in colorectal cancer is an independent prognostic factor and regulated by promoter methylation and histone deacetylation in tumors of the serrated pathway. Clin Epigenetics 2018; 10:120. [PMID: 30257705 PMCID: PMC6158822 DOI: 10.1186/s13148-018-0548-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/27/2018] [Indexed: 01/01/2023] Open
Abstract
Background In colorectal cancer, CDX2 expression is lost in approximately 20% of cases and associated with poor outcome. Here, we aim to validate the clinical impact of CDX2 and investigate the role of promoter methylation and histone deacetylation in CDX2 repression and restoration. Methods CDX2 immunohistochemistry was performed on multi-punch tissue microarrays (n = 637 patients). Promoter methylation and protein expression investigated on 11 colorectal cancer cell lines identified two CDX2 low expressors (SW620, COLO205) for treatment with decitabine (DNA methyltransferase inhibitor), trichostatin A (TSA) (general HDAC inhibitor), and LMK-235 (specific HDAC4 and HDAC5 inhibitor). RNA and protein levels were assessed. HDAC5 recruitment to the CDX2 gene promoter region was tested by chromatin immunoprecipitation. Results Sixty percent of tumors showed focal CDX2 loss; 5% were negative. Reduced CDX2 was associated with lymph node metastasis (p = 0.0167), distant metastasis (p = 0.0123), and unfavorable survival (multivariate analysis: p = 0.0008; HR (95%CI) 0.922 (0.988–0.997)) as well as BRAFV600E, mismatch repair deficiency, and CpG island methylator phenotype. Decitabine treatment alone induced CDX2 RNA and protein with values from 2- to 25-fold. TSA treatment ± decitabine also led to successful restoration of RNA and/or protein. Treatment with LMK-235 alone had marked effects on RNA and protein levels, mainly in COLO205 cells that responded less to decitabine. Lastly, decitabine co-treatment was more effective than LMK-235 alone at restoring CDX2. Conclusion CDX2 loss is an adverse prognostic factor and linked to molecular features of the serrated pathway. RNA/protein expression is restored in CDX2 low-expressing CRC cell lines by demethylation and HDAC inhibition. Importantly, our data underline HDAC4 and HDAC5 as new epigenetic CDX2 regulators that warrant further investigation. Electronic supplementary material The online version of this article (10.1186/s13148-018-0548-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Janina Graule
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Kristin Uth
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Elia Fischer
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Irene Centeno
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - José A Galván
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Micha Eichmann
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Tilman T Rau
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Rupert Langer
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Heather Dawson
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Ulrich Nitsche
- Department of Surgery, Klinikum rechts der Isar, Technische Universität München, Ismaninger Strasse 22, Munich, 81675, Germany
| | - Peter Traeger
- Careanesth AG, Nelkenstrasse 15, Zürich, 8006, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, University Hospital of Bern, 3010, Bern, Switzerland.,Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, 90033, CA, USA
| | - Beat Schnüriger
- Department of Visceral and Internal Medicine, University Hospital of Bern, 3008, Bern, Switzerland
| | - Marion Hädrich
- Department of Visceral and Internal Medicine, University Hospital of Bern, 3008, Bern, Switzerland
| | - Peter Studer
- Department of Visceral and Internal Medicine, University Hospital of Bern, 3008, Bern, Switzerland
| | - Daniel Inderbitzin
- University of Bern and Bürgerspital Solothurn, Schöngrünstrasse 42, 4500, Solothurn, Switzerland
| | - Alessandro Lugli
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland
| | - Mario P Tschan
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Freiestrasse 1, 3012, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Murtenstrasse 31, Room L310, 3008, Bern, Switzerland.
| |
Collapse
|
31
|
Association of CDX2 Expression With Survival in Early Colorectal Cancer: A Systematic Review and Meta-analysis. Clin Colorectal Cancer 2018. [DOI: 10.1016/j.clcc.2018.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
32
|
Eckstein M, Wirtz RM, Pfannstil C, Wach S, Stoehr R, Breyer J, Erlmeier F, Günes C, Nitschke K, Weichert W, Otto W, Keck B, Eidt S, Burger M, Taubert H, Wullich B, Bolenz C, Hartmann A, Erben P. A multicenter round robin test of PD-L1 expression assessment in urothelial bladder cancer by immunohistochemistry and RT-qPCR with emphasis on prognosis prediction after radical cystectomy. Oncotarget 2018; 9:15001-15014. [PMID: 29599921 PMCID: PMC5871092 DOI: 10.18632/oncotarget.24531] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 02/10/2018] [Indexed: 01/05/2023] Open
Abstract
Background Immunohistochemical PD-L1 assessment is currently used to identify responders towards checkpoint inhibitors although it is limited by inter-observer effects. Here, we conducted a multi-center round robin test to prove the possibility of assessing the PD-L1 status by gene expression to avoid inter-observer effects. Patients and methods Gene expression of PD-L1 was analyzed in a total of 294 samples (14 cases non-muscle invasive and muscle-invasive bladder cancer; MIBC) in seven centers by a RT-qPCR kit and compared with immunohistochemical scoring of three pathologists (DAKO, 22c3). Both assays were compared towards prognosis prediction in a cohort of 88 patients with MIBC. Results PD-L1 gene expression revealed very high inter center correlation (centrally extracted RNA: r = 0.68–0.98, p ≤ 0.0076; locally extracted RNA: r = 0.81–0.98, p ≤ 0.0014). IHC Inter-observer concordance was moderate to substantial for immune cells (IC), fair for combined IC/ tumor cell (TC) (IC: κ = 0.50–0.61; IC + TC: κ = 0.50), and fair for TC scoring (κ = 0.26–0.35). Gene expression assessment resulted in more positive cases (9/14 cases positive vs. 6/14 cases [IHC]) which could be validated in the independent cohort. Positive mRNA status was associated with significantly better overall and disease-specific survival (5-year OS: 50% vs. 26%, p = 0.0042, HR = 0.48; 5 year DSS: 65% vs. 40%, p = 0.012, HR = 0.49). The 1% IHC IC cut-off also revealed significant better OS (5 year OS: 58% vs. 31%, p = 0.036, HR = 0.62). Conclusion Gene expression showed very high inter-center agreement. Gene expression assessment also resulted in more positive cases and revealed better prognosis prediction. PD-L1 mRNA expression seems to be a reproducible and robust tool for PD-L1 assessment.
Collapse
Affiliation(s)
- Markus Eckstein
- Institute of Pathology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ralph M Wirtz
- STRATIFYER Molecular Pathology GmbH, Cologne, Germany.,Institute of Pathology at The St. Elisabeth Hospital Köln-Hohenlind, Cologne, Germany
| | - Carolin Pfannstil
- Institute of Pathology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sven Wach
- Department of Urology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Robert Stoehr
- Institute of Pathology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Breyer
- Department of Urology, University of Regensburg, Regensburg, Germany
| | | | - Cagatay Günes
- Department of Urology, University of Ulm, Ulm, Germany
| | - Katja Nitschke
- Department of Urology Mannheim, University of Heidelberg, Mannheim, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, Munich, Germany
| | - Wolfgang Otto
- Department of Urology, University of Regensburg, Regensburg, Germany
| | - Bastian Keck
- Department of Urology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Sebastian Eidt
- Institute of Pathology at The St. Elisabeth Hospital Köln-Hohenlind, Cologne, Germany
| | - Maximilian Burger
- Department of Urology, University of Regensburg, Regensburg, Germany
| | - Helge Taubert
- Department of Urology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Bernd Wullich
- Department of Urology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | - Arndt Hartmann
- Institute of Pathology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Philipp Erben
- Department of Urology Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
33
|
Tse JWT, Jenkins LJ, Chionh F, Mariadason JM. Aberrant DNA Methylation in Colorectal Cancer: What Should We Target? Trends Cancer 2017; 3:698-712. [PMID: 28958388 DOI: 10.1016/j.trecan.2017.08.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 12/16/2022]
Abstract
Colorectal cancers (CRCs) are characterized by global hypomethylation and promoter-specific DNA methylation. A subset of CRCs with extensive and co-ordinate patterns of promoter methylation has also been identified, termed the CpG-island methylator phenotype. Some genes methylated in CRC are established tumor suppressors; however, for the majority, direct roles in disease initiation or progression have not been established. Herein, we examine functional evidence of specific methylated genes contributing to CRC pathogenesis, focusing on components of commonly deregulated signaling pathways. We also review current knowledge of the mechanisms underpinning promoter methylation in CRC, including genetic events, altered transcription factor binding, and DNA damage. Finally, we summarize clinical trials of DNA methyltransferase inhibitors in CRC, and propose strategies for enhancing their efficacy.
Collapse
Affiliation(s)
- Janson W T Tse
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia; These authors contributed equally
| | - Laura J Jenkins
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia; School of Cancer Medicine, La Trobe University, Melbourne, Australia; These authors contributed equally
| | - Fiona Chionh
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia
| | - John M Mariadason
- Olivia Newton-John Cancer Research Institute, Melbourne, Australia; School of Cancer Medicine, La Trobe University, Melbourne, Australia.
| |
Collapse
|
34
|
Homeobox protein CDX2 as a prognostic biomarker in solid malignancies: a meta-analysis. Oncotarget 2017; 8:89160-89172. [PMID: 29179508 PMCID: PMC5687678 DOI: 10.18632/oncotarget.20808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023] Open
Abstract
Background CDX2 is a caudal-homeobox gene and its expression is abnormal in numerous tumour cell types. Nevertheless, its prognostic value for solid tumours requires further investigation. Hence, we conducted a meta-analysis to determine the significance of CDX2 as a prognostic biomarker in solid malignancies systematically. Materials and Methods We performed a systematic literature search in PUBMED and EMBASE up to May 2017. Retrospective studies comparing the prognostic value of different CDX2 levels in human malignancies were included. Data extractions and methodological assessments were performed separately by two investigators using a standard procedure. The statistical procedures were performed using Review Manager 5.3 and STATA/MP 14.0. Results A total of 26 retrospective studies met the inclusion criteria and comprised 5008 participants. Patients with CDX2 overexpression had significantly better 3-year, 5-year, 10-year and disease-free survival outcomes in solid malignancies, regardless of the cancer type, mean age, and source region. Nevertheless, there was no significant difference in the patients from Europe. The expression level of CDX2 was not statistically associated with cancer relapse. Moreover, our analysis showed that CDX2 overexpression is correlated to better responses to chemotherapy in patients with TNM IV stage cancers. The stability of the pooled outcomes was verified by sensitivity analysis. The funnel plots, Egger's test and Begg's test jointly confirmed that there was no publication bias. Conclusions Overexpression of CDX2 is a reliable biomarker of a better prognosis in solid malignancies.
Collapse
|
35
|
Reggiani Bonetti L, Lionti S, Vitarelli E, Barresi V. Prognostic significance of CDX2 immunoexpression in poorly differentiated clusters of colorectal carcinoma. Virchows Arch 2017; 471:731-741. [PMID: 28819729 DOI: 10.1007/s00428-017-2219-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/01/2017] [Accepted: 08/09/2017] [Indexed: 12/26/2022]
Abstract
CDX2 is a transcription factor that acts as a tumor suppressor in colorectal cancer (CRC). Its loss triggers metastatic process and tumor progression; however, its prognostic role in patients with CRC is still controversial. Poorly differentiated clusters (PDCs) are aggregates of neoplastic cells which likely have high metastatic potential in CRC. In this study, we analyzed and compared CDX2 expression in PDC (CDX2-PDC) and corresponding main tumor (CDX2 main tumor) in 42 CRCs showing at least 10 PDC (PDC G3). Five of 42 CRCs (12%) were classified as CDX2 main tumor negative (4/5 were also PDC-CDX2 negative); all had tumor recurrence and died of CRC. Twenty nine of 42 cases were CDX2-PDC negative. Among CRC CDX2 main tumor positive, 15 had recurrences and 13 died from CRC; 13 and 11 of them, respectively, were CDX2-PDC negative. By assigning one point to CDX2 main tumor or CDX2-PDC positivity, we assessed CDX2-staining score for each case. Twelve cases had CDX2-staining score 2 (CDX2 positive in main tumor and PDC); 26 had score 1 (CDX2 positive in main tumor or PDC), and 4 had CDX2 score 0 (CDX2 negative in main tumor and PDC). In our patients, CDX2-staining score had higher prognostic value compared to CDX2 main tumor or CDX2-PDC alone. In addition, it represented a significant and independent prognostic variable for disease-free survival (DFS) and cancer-specific survival (CSS). Our findings suggest that, although loss of CDX2 in the main tumor identifies high-risk patients with high specificity, CDX2-PDC should also be considered in CDX2 main tumor positive cases to predict prognosis.
Collapse
Affiliation(s)
- Luca Reggiani Bonetti
- Department of Laboratory Integrated Activities, Anatomic Pathology and Legal Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Lionti
- Department of Pathology in Evolutive Age and Adulthood "Gaetano Barresi", University of Messina, Polyclinic G. Martino, Pad D, Via Consolare Valeria, 98125, Messina, Italy
| | - Enrica Vitarelli
- Department of Pathology in Evolutive Age and Adulthood "Gaetano Barresi", University of Messina, Polyclinic G. Martino, Pad D, Via Consolare Valeria, 98125, Messina, Italy
| | - Valeria Barresi
- Department of Pathology in Evolutive Age and Adulthood "Gaetano Barresi", University of Messina, Polyclinic G. Martino, Pad D, Via Consolare Valeria, 98125, Messina, Italy.
| |
Collapse
|