1
|
Rajput SK, Minhas K, Azam I, Habib S, Shaikh U, Lalani EN. Prognostic implications of MUC1 and XBP1 concordant expression in multiple myeloma: A retrospective study. PLoS One 2025; 20:e0320934. [PMID: 40179083 PMCID: PMC11967961 DOI: 10.1371/journal.pone.0320934] [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: 11/20/2023] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Multiple myeloma (MM) is a disease of malignant plasma cells (PC) with poor survival. Disease progression and treatment relapse are attributed to MM cancer stem cells (CSCs) and signaling molecules such as MUC1 and XBP1. The study aimed to determine the prognostic value of expression of CSC-associated biomarkers, MUC1 and XBP1 in MM, which has not been explored previously. In this study, we determined the immunohistochemical expression of CSC markers (ALDH1, CD117, and CD34), MUC1, and XBP1 in 128 MM formalin-fixed paraffin-embedded bone marrow archival blocks. The expression of biomarkers was assessed for association with clinicopathological variables and patient survival. Descriptive analysis, survival plots and crude association between outcome and independent variables were assessed using Kaplan Meier and Log rank test. Univariate and multivariable analyses were performed using simple and multiple Cox regression models. The results are reported as crude and adjusted hazard ratios with 95% confidence intervals. Expression of ALDH1 and CD117 was found in 51% and 48% of the tumors, respectively. ALDH1 expression was associated with 1.83 years of reduced survival for patients with CD56-negative tumors. MUC1 expression was observed in 62%, whereas XBP1 was expressed in 48% of tumors. Combinatorial group analysis of XBP1 and MUC1 stratified patients into two prognostic groups. Cases with tumors negative for expression of MUC1 and XBP1 (XBP1-/ MUC1-) were categorized as a good prognostic group with increased survival of 3.42 years compared to cases with tumors expressing both (Worst prognosis, XBP1 + /MUC1+). Concordant expression of MUC1 and XBP1 in MM defines a subset of patients with adverse outcomes. The adjusted hazard ratio showed a four-fold increased risk of mortality associated with the concordant expression of MUC1 and XBP1 in patients > 65 years of age.
Collapse
Affiliation(s)
- Sheerien Kareem Rajput
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Khurram Minhas
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - Iqbal Azam
- Department of Community Health Sciences, The Aga Khan University, Karachi, Pakistan
| | - Sadia Habib
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| | - Usman Shaikh
- Department of Pathology and Laboratory Medicine, The Aga Khan University, Karachi, Pakistan
| | - El-Nasir Lalani
- Centre for Regenerative Medicine and Stem Cell Research, The Aga Khan University, Karachi, Pakistan
| |
Collapse
|
2
|
Xu C, Mannucci A, Esposito F, Oliveres H, Alonso-Orduña V, Yubero A, Fernández-Martos C, Salud A, Gallego J, Martín-Richard M, Fernández-Plana J, Guillot M, Aparicio J, Fakih M, Kopetz S, Feliu J, Maurel J, Goel A. An Exosome-Based Liquid Biopsy Predicts Depth of Response and Survival Outcomes to Cetuximab and Panitumumab in Metastatic Colorectal Cancer: The EXONERATE Study. Clin Cancer Res 2025; 31:1002-1015. [PMID: 39820673 PMCID: PMC11913580 DOI: 10.1158/1078-0432.ccr-24-1934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 01/15/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE The EXOsome and cell-free miRNAs of anti-EGFR ResistAnce (EXONERATE) study was an open-label, biomarker interventional study designed to develop, test, and validate a liquid biopsy predictive of progression-free survival (PFS), overall survival (OS), and objective response rate (ORR) for first-line EGFR inhibitors in metastatic colorectal cancer (mCRC). PATIENTS AND METHODS Patients with newly diagnosed RAS wild-type, chemotherapy-naïve mCRC, both right- and left-sided, were enrolled in two nationwide trials to receive cetuximab or panitumumab along with chemotherapy. The primary endpoint was 12-month PFS, which was hierarchically tested in left- and right-sided mCRCs to predict PFS, OS, and ORR. RESULTS Genome-wide small RNA sequencing identified 12 cell-free and 14 exosomal candidates that were differentially expressed in both plasma and tumor tissue of good versus poor responders (based on PFS <12 months). The 8 and 9 best performing candidates, respectively, were used to generate the EXONERATE assay. In left-sided mCRC, 65% were EXONERATE-high, correlating with shorter median PFS (9.5 vs. 18.5 months; P < 0.001). In the independent right-sided mCRC cohort, 80.8% were EXONERATE-high and experienced a similarly shorter median PFS (8.6 vs. 41.2 months; P = 0.0004). In the right-sided group, EXONERATE predicted PFS ≥12 months with 100% sensitivity. A linear relationship existed between EXONERATE values and response depth. Multivariate analysis revealed that EXONERATE predicts PFS and OS independently of tumor sidedness. CONCLUSIONS The EXONERATE assay robustly predicted PFS and OS outcomes in patients with mCRC, both right- and left-sided, before they received either panitumumab or cetuximab. It stratified PFS, OS, and ORR better than a right versus left approach.
Collapse
Affiliation(s)
- Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Alessandro Mannucci
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Francis Esposito
- Department of Medical Oncology, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Helena Oliveres
- Department of Medical Oncology, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - Alfonso Yubero
- Medical Oncology Service, Hospital Universitario Lozano Blesa, Zaragoza, Spain
| | | | - Antonieta Salud
- Medical Oncology Service, Hospital Universitari Arnau de Vilanova, Lleida, Spain
| | - Javier Gallego
- Medical Oncology Service, Hospital General Universitario of Elche, Elche, Spain
| | - Marta Martín-Richard
- Medical Oncology Service, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Mónica Guillot
- Medical Oncology Service, Hospital Son Espases, Palma, Spain
| | - Jorge Aparicio
- Medical Oncology Department, Hospital La Fe de Valencia, Valencia, Spain
| | | | - Scott Kopetz
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jaime Feliu
- Medical Oncology Service, Hospital Universitario La Paz, CIBERONC, Madrid, Spain
| | - Joan Maurel
- Department of Medical Oncology, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, California
- City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
3
|
Ruzzo A, Graziano F, Palladino S, Fischer NW, Catalano V, Giordani P, Malkin D, Tamburrano T, Patriti A, Petrelli F, Sarti D, Chiari R. Clinical impact of TP53 functional mutations in patients with metastatic colorectal cancer treated with bevacizumab and chemotherapy. Oncologist 2025; 30:oyae277. [PMID: 39436921 PMCID: PMC11954512 DOI: 10.1093/oncolo/oyae277] [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: 04/06/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Clinical and experimental studies indicate that the tumor protein p53 (TP53) gene loss of function due to missense mutations (MMs) may confer sensitivity to anti-angiogenics. This effect seems to be linked to cross-talk mechanisms among TP53, vascular endothelial growth factor (VEGF), and VEGF receptors. We investigated whether specific TP53 MMs are associated with clinical outcomes of patients with metastatic colorectal cancer (mCRC) treated with first-line chemotherapy plus Bevacizumab. The study focused on KRAS-mutated, liver-only mCRC cases as a homogeneous subgroup that may represent a relevant setting for exploring this association. MATERIALS AND METHODS MMs were identified on primary tumors. MMs were classified by mutant-specific residual transcriptional activity scores (TP53RTAS) as transcriptionally inactive (TP53inactive = TP53RTAS 0%) or active (TP53active = TP53RTAS ≥ 1%) and used for stratifying patients in progression-free survival (PFS), response rate, and overall survival (OS) analyses. RESULTS The study population consisted of 62 patients. MMs were found in 39 cases (62%) with 16 having TP53inactive and 23 TP53active MMs. Patients with TP53inactive MMs showed better PFS in comparison with the remaining groups (wild-type and TP53active). This effect was retained in the multivariate model. A similar clinical impact was observed in the OS analysis. There was a significant difference in the overall response rate and rate of post-treatment resection of liver metastases between the TP53inactive and the wild-type or TP53active MMs cases. CONCLUSIONS Specific TP53 MMs may identify sub-groups of patients who benefit from Bevacizumab-based systemic therapy and these findings could lead to novel tailored treatment strategies in this setting.
Collapse
Affiliation(s)
- Annamaria Ruzzo
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy
| | | | - Silvia Palladino
- Department of Biomolecular Sciences, University of Urbino, 61029 Urbino, Italy
| | - Nicholas W Fischer
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | | | | | - David Malkin
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, M5G 0A4, Canada
- Division of Hematology-Oncology, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
- Department of Pediatrics, The Hospital for Sick Children, Toronto, M5G 0A4, Canada
| | | | | | | | | | - Rita Chiari
- Medical Oncology Unit, AST1, 61121 Pesaro, Italy
| |
Collapse
|
4
|
Stevão CG, Dominguete MHL, Moraes PDC, Montalli VAM, de Freitas ALS, Sperandio M. Comparison of two fluorescence detectors in flow cytometry to predict malignant transformation of oral leukoplakia from cell cycle fractions. Oral Surg Oral Med Oral Pathol Oral Radiol 2025:S2212-4403(25)00757-6. [PMID: 39988513 DOI: 10.1016/j.oooo.2025.01.726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVE The objective of this paper was to compare the accuracy of predicting malignant transformation of oral leukoplakia using the 585-620 nm detector when compared with data obtained with the 661-683 nm detector. STUDY DESIGN This was a single-factor retrospective longitudinal experimental study, whose factor will be DNA content (DNA ploidy) at 4 levels, G1 phase, S phase, mitosis phase (G2), and DNA excess (4cER). The sample is a cohort of individuals diagnosed with oral leukoplakia (N = 97) where 18 cases evolved into cancer and 79 did not. RESULTS The DNA content parameters 4cER, G2, and G1/G2 ratios were associated with malignant transformation (P < .05). Two analog algorithms of these findings generated positive predictive values (PPVs) of 38% (67% sensitivity and 76% specificity) and 40% (61% sensitivity and 80% specificity). DNA content by flow cytometry can achieve a PPV of 40% in high-risk lesions and negative predictive value (NPV) of 90% in low-risk lesions. CONCLUSIONS There was no significant difference among the percentages of nuclei in the G1, S, G2, and excess G2 (4cER) phases comparing the 661-683 nm and 585-620 nm detectors. The 585-620 nm detector achieved PPV and NPV equivalent to those observed with the 661-683 nm detector.
Collapse
Affiliation(s)
- Caroline Gennari Stevão
- Department of Oral Medicine and Pathology, Faculdade São Leopoldo Mandic, Research Institute, Campinas, São Paulo, Brazil
| | | | - Paulo de Camargo Moraes
- Department of Oral Medicine and Pathology, Faculdade São Leopoldo Mandic, Research Institute, Campinas, São Paulo, Brazil
| | - Victor Angelo Martins Montalli
- Department of Oral Medicine and Pathology, Faculdade São Leopoldo Mandic, Research Institute, Campinas, São Paulo, Brazil
| | | | - Marcelo Sperandio
- Department of Oral Medicine and Pathology, Faculdade São Leopoldo Mandic, Research Institute, Campinas, São Paulo, Brazil.
| |
Collapse
|
5
|
Heinrichs L, Fluegen G, Loosen SH, Loberg C, Wittig L, Quaas A, Plum PS, Große Hokamp N, Minko P, Krieg A, Antoch G, Knoefel WT, Luedde T, Roderburg C, Jördens MS. Bone mineral density as a prognostic marker in patients with biliary tract cancer undergoing surgery. BJC REPORTS 2024; 2:72. [PMID: 39323978 PMCID: PMC11420066 DOI: 10.1038/s44276-024-00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024]
Abstract
Background Biliary tract cancer (BTC) is one of the most aggressive malignancies and surgery represents the only curative treatment approach. However, even in patients with complete tumor resection 5-year survival rates are below 30%. So far, prognostic markers to assess the outcome of these patients are lacking. We therefore evaluated bone mineral density (BMD) as a prognostic tool in patients receiving surgery for BTC. methods 76 BTC patients undergoing tumor resection in our clinic (Duesseldorf cohort) as well as an external validation cohort of 34 BTC patients (Cologne cohort) were included. BMD was analyzed at the first lumbar vertebra, using routine CT scans which has been proven comparable to DXA. Results Median overall survival (OS) of the Duesseldorf cohort after surgery was 527 days, one- and five-year survival probabilities were 62 and 18%. Patients with BMD above 156.5 HU had significantly improved OS (1435 days vs. 459 days; p = 0.002). The prognostic value for BMD was confirmed using Cox-regression analysis, as well as an external validation cohort. In subgroup analysis the prognostic effect of BMD was only present in female patients, suggesting sex specific differences. Conclusion BMD is a valuable, easily accessible and independent prognostic marker in patients receiving liver surgery for BTC.
Collapse
Affiliation(s)
- Lisa Heinrichs
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Georg Fluegen
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Sven H. Loosen
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Christina Loberg
- Department of Diagnostic and Interventional Radiology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Linda Wittig
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital Cologne and Medical Faculty, University of Cologne, 50937 Cologne, Germany
| | - Patrick S. Plum
- Department of General, Visceral, Cancer and Transplantation Surgery, University Hospital Cologne, 50937 Cologne, Germany
| | - Nils Große Hokamp
- Institute for Diagnostic and Interventional Radiology, University Hospital Cologne, 50937 Cologne, Germany
| | - Peter Minko
- Department of Diagnostic and Interventional Radiology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Andreas Krieg
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Gerald Antoch
- Department of Diagnostic and Interventional Radiology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Wolfram T. Knoefel
- Department of Surgery (A), Heinrich-Heine-University and University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Christoph Roderburg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus S. Jördens
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
6
|
Abdullah AR, Gamal El-Din AM, Ismail Y, El-Husseiny AA. The FSCN1 gene rs2966447 variant is associated with increased serum fascin-1 levels and breast cancer susceptibility. Gene 2024; 927:148743. [PMID: 38964493 DOI: 10.1016/j.gene.2024.148743] [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: 01/01/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Fascin-1 (FSCN1) is recognized as an actin-binding protein, commonly exhibits up-regulation in breast cancer (BC) and is crucial for tumor invasion and metastasis. The existence of FSCN1 gene polymorphisms may raise the potential for developing BC, and there are still no studies focusing on the relationship between the FSCN1 rs2966447 variant and BC risk in Egyptian females. Thus, we investigated the serum fascin-1 levels in BC patients and the association between the FSCN1 rs2966447 variant with its serum levels and BC susceptibility. Genotyping was conducted in 153 treatment-naïve BC females with different stages and 144 apparent healthy females by TaqMan® allelic discrimination assay, whereas serum fascin-1 level quantification was employed by ELISA. The FSCN1 rs2966447 variant demonstrated a significant association with BC susceptibility under all utilized genetic models, cancer stages and estrogen receptor negativity. Also, BC females with AT and TT genotypes had higher serum fascin-1 levels and tumor size than those with the AA genotype. Moreover, serum fascin-1 levels were significantly elevated in the BC females, notably in those with advanced-stages. Furthermore, serum fascin-1 levels were markedly positively correlated with number of positive lymph nodes as well as tumor size. Collectively, these findings revealed that the FSCN1 rs2966447 variant may be regarded as a strong candidate for BC susceptibility. Also, this intronic variant is associated with increased serum fascin-1 levels and tumor size.
Collapse
Affiliation(s)
- Ahmed R Abdullah
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ayman M Gamal El-Din
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Yahia Ismail
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt; Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City 11829, Cairo, Egypt.
| |
Collapse
|
7
|
Seliem MA, Mohamadin AM, El-Sayed MIK, Ismail Y, El-Husseiny AA. The clinical signature of genetic variants and serum levels of macrophage migration inhibitory factor in Egyptian breast cancer patients. Breast Cancer Res Treat 2024; 208:57-66. [PMID: 38916819 PMCID: PMC11452551 DOI: 10.1007/s10549-024-07393-9] [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: 11/23/2023] [Accepted: 05/30/2024] [Indexed: 06/26/2024]
Abstract
PURPOSE Macrophage migration inhibitory factor (MIF) is an integral cytokine for the modulation of both innate and adaptive immunity and is involved in the pathogenesis of various cancers. However, conflicting findings on the relationship between MIF polymorphisms and breast cancer (BC) have been reported in earlier research. We investigated the clinical value of serum MIF levels and the association between MIF rs1049829 and rs755622 variants with their serum levels and propensity to develop BC. METHODS A total of 133 treatment-naïve Egyptian BC females and 126 apparently healthy controls were matriculated in this case-control study. The serum MIF protein levels were quantified by ELISA, whereas the genotyping was executed utilizing the TaqMan® allelic discrimination assay. RESULTS A significant increase in the serum MIF level in BC cases was observed in comparison to control subjects (P < 0.0001), with a diagnostic potential to discriminate BC with 92.5% sensitivity and 73.7% specificity at a cut-off value > 9.47 ng/mL. Besides, a significant difference in serum MIF level was observed in BC cases with progesterone receptor (PR) negativity compared to those with PR positivity (P = 0.046). Moreover, a significant association was depicted between the rs1049829 variant of MIF gene and the protective effect against BC meanwhile the rs755622 variant demonstrated no significant link with BC risk. CONCLUSIONS This study revealed that serum MIF levels may be regarded as a promising serum tumor marker for BC. Also, the rs1049829 variant of the MIF gene is considered a protective candidate against BC.
Collapse
Affiliation(s)
- Mahmoud A Seliem
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11231, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, Egypt
| | - Ahmed M Mohamadin
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11231, Egypt
| | - Mohamed I Kotb El-Sayed
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, Helwan, Cairo, 11790, Egypt
| | - Yahia Ismail
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Ahmed A El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City, Cairo, 11231, Egypt.
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, 11829, Egypt.
| |
Collapse
|
8
|
Klümper N, Tran NK, Zschäbitz S, Hahn O, Büttner T, Roghmann F, Bolenz C, Zengerling F, Schwab C, Nagy D, Toma M, Kristiansen G, Heers H, Ivanyi P, Niegisch G, Grunewald CM, Darr C, Farid A, Schlack K, Abbas M, Aydogdu C, Casuscelli J, Mokry T, Mayr M, Niedersüß-Beke D, Rausch S, Dietrich D, Saal J, Ellinger J, Ritter M, Alajati A, Kuppe C, Meeks J, Vera Badillo FE, Nakauma-González JA, Boormans J, Junker K, Hartmann A, Grünwald V, Hölzel M, Eckstein M. NECTIN4 Amplification Is Frequent in Solid Tumors and Predicts Enfortumab Vedotin Response in Metastatic Urothelial Cancer. J Clin Oncol 2024; 42:2446-2455. [PMID: 38657187 PMCID: PMC11227306 DOI: 10.1200/jco.23.01983] [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: 09/12/2023] [Revised: 01/31/2024] [Accepted: 02/26/2024] [Indexed: 04/26/2024] Open
Abstract
PURPOSE The anti-NECTIN4 antibody-drug conjugate enfortumab vedotin (EV) is approved for patients with metastatic urothelial cancer (mUC). However, durable benefit is only achieved in a small, yet uncharacterized patient subset. NECTIN4 is located on chromosome 1q23.3, and 1q23.3 gains represent frequent copy number variations (CNVs) in urothelial cancer. Here, we aimed to evaluate NECTIN4 amplifications as a genomic biomarker to predict EV response in patients with mUC. MATERIALS AND METHODS We established a NECTIN4-specific fluorescence in situ hybridization (FISH) assay to assess the predictive value of NECTIN4 CNVs in a multicenter EV-treated mUC patient cohort (mUC-EV, n = 108). CNVs were correlated with membranous NECTIN4 protein expression, EV treatment responses, and outcomes. We also assessed the prognostic value of NECTIN4 CNVs measured in metastatic biopsies of non-EV-treated mUC (mUC-non-EV, n = 103). Furthermore, we queried The Cancer Genome Atlas (TCGA) data sets (10,712 patients across 32 cancer types) for NECTIN4 CNVs. RESULTS NECTIN4 amplifications are frequent genomic events in muscle-invasive bladder cancer (TCGA bladder cancer data set: approximately 17%) and mUC (approximately 26% in our mUC cohorts). In mUC-EV, NECTIN4 amplification represents a stable genomic alteration during metastatic progression and associates with enhanced membranous NECTIN4 protein expression. Ninety-six percent (27 of 28) of patients with NECTIN4 amplifications demonstrated objective responses to EV compared with 32% (24 of 74) in the nonamplified subgroup (P < .001). In multivariable Cox analysis adjusted for age, sex, and Bellmunt risk factors, NECTIN4 amplifications led to a 92% risk reduction for death (hazard ratio, 0.08 [95% CI, 0.02 to 0.34]; P < .001). In the mUC-non-EV, NECTIN4 amplifications were not associated with outcomes. TCGA Pan-Cancer analysis demonstrated that NECTIN4 amplifications occur frequently in other cancers, for example, in 5%-10% of breast and lung cancers. CONCLUSION NECTIN4 amplifications are genomic predictors of EV responses and long-term survival in patients with mUC.
Collapse
Affiliation(s)
- Niklas Klümper
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
| | - Ngoc Khanh Tran
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Stefanie Zschäbitz
- Department of Medical Oncology, National Center for Tumor Disease (NCT), University Hospital, Heidelberg, Germany
| | - Oliver Hahn
- Department of Urology and Pediatric Urology, Julius Maximilians University Medical Center of Würzburg, Würzburg, Germany
| | - Thomas Büttner
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Florian Roghmann
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology, Marien Hospital, Ruhr-University Bochum, Herne, Germany
| | - Christian Bolenz
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology and Pediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Friedemann Zengerling
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Department of Urology and Pediatric Urology, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Constantin Schwab
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
| | - Dora Nagy
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Marieta Toma
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Glen Kristiansen
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Hendrik Heers
- Department of Urology, University Hospital Marburg, Marburg, Germany
| | - Philipp Ivanyi
- Department of Hemostaseology, Oncology and Stem Cell Transplantation, Medical University Hannover, Hannover, Germany
| | - Günter Niegisch
- Department of Urology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Christopher Darr
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Arian Farid
- Department of Urology, University Medical Center Göttingen, Göttingen, Germany
| | - Katrin Schlack
- Department of Urology, University Hospital Münster, Münster, Germany
| | - Mahmoud Abbas
- Department of Pathology, University Hospital Münster, Münster, Germany
| | - Can Aydogdu
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Jozefina Casuscelli
- Department of Urology, University Hospital, Ludwig Maximilian University of Munich, Munich, Germany
| | - Theresa Mokry
- Department of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Mayr
- Clinic Ottakring, Institute of Pathology and Microbiology, Wien, Austria
| | | | - Steffen Rausch
- Department of Urology, Eberhard Karls University, Tübingen, Germany
| | - Dimo Dietrich
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Jonas Saal
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- Medical Clinic III for Oncology, Hematology, Immune-Oncology and Rheumatology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Jörg Ellinger
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Manuel Ritter
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
| | - Abdullah Alajati
- Department of Urology and Pediatric Urology, University Hospital Bonn, Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Christoph Kuppe
- Institute of Experimental Medicine and Systems Biology and Division of Nephrology, RWTH Aachen University, Aachen, Germany
| | - Joshua Meeks
- Department of Urology, Feinberg School of Medicine, Chicago, IL
| | | | - J. Alberto Nakauma-González
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Joost Boormans
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Kerstin Junker
- Department of Urology and Pediatric Urology, Saarland University, Homburg, Germany
| | - Arndt Hartmann
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
| | - Viktor Grünwald
- Clinic for Internal Medicine (Tumor Research) and Clinic for Urology, Interdisciplinary Genitourinary Oncology at the West-German Cancer Center, Essen University Hospital, Essen, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, University Medical Center Bonn (UKB), Bonn, Germany
- Center for Integrated Oncology Aachen/Bonn/Cologne/Düsseldorf (CIO-ABCD), Bonn, Germany
| | - Markus Eckstein
- BRIDGE-Consortium Germany e.V., Mannheim, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- Comprehensive Cancer Center EMN, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Bavarian Center for Cancer Research (Bayerisches Zentrum für Krebsforschung, BZKF), Erlangen, Germany
| |
Collapse
|
9
|
Okuda Y, Shimura T, Abe Y, Iwasaki H, Nishigaki R, Fukusada S, Sugimura N, Kitagawa M, Yamada T, Taguchi A, Kataoka H. Urinary dipeptidase 1 and trefoil factor 1 are promising biomarkers for early diagnosis of colorectal cancer. J Gastroenterol 2024; 59:572-585. [PMID: 38836911 DOI: 10.1007/s00535-024-02110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/24/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Currently utilized serum tumor markers and fecal immunochemical tests do not have sufficient diagnostic power for colorectal cancer (CRC) due to their low sensitivities. To establish non-invasive urinary protein biomarkers for early CRC diagnosis, we performed stepwise analyses employing urine samples from CRCs and healthy controls (HCs). METHODS Among 474 urine samples, 363 age- and sex-matched participants (188 HCs, 175 stage 0-III CRCs) were randomly divided into discovery (16 HCs, 16 CRCs), training (110 HCs, 110 CRCs), and validation (62 HCs, 49 CRCs) cohorts. RESULTS Of the 23 urinary protein candidates comprehensively identified from mass spectrometry in the discovery cohort, urinary levels of dipeptidase 1 (uDPEP1) and Trefoil factor1 (uTFF1) were the two most significant diagnostic biomarkers for CRC in both training and validation cohorts using enzyme-linked immunosorbent assays. A urinary biomarker panel comprising uDPEP1 and uTFF1 significantly distinguished CRCs from HCs, showing area under the curves of 0.825-0.956 for stage 0-III CRC and 0.792-0.852 for stage 0/I CRC. uDPEP1 and uTFF1 also significantly distinguished colorectal adenoma (CRA) patients from HCs, with uDPEP1 and uTFF1 increasing significantly in the order of HCs, CRA patients, and CRC patients. Moreover, expression levels of DPEP1 and TFF1 were also significantly higher in the serum and tumor tissues of CRC, compared to HCs and normal tissues, respectively. CONCLUSIONS This study established a promising and non-invasive urinary protein biomarker panel, which enables the early detection of CRC with high sensitivity.
Collapse
Affiliation(s)
- Yusuke Okuda
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan.
| | - Yuichi Abe
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-Ku, Nagoya, Aichi, 464-8681, Japan
| | - Hiroyasu Iwasaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Ruriko Nishigaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Shigeki Fukusada
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Naomi Sugimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Mika Kitagawa
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| | - Tamaki Yamada
- Okazaki Public Health Center, 1-3 Harusaki, Harisaki-Cho, Okazaki, 444-0827, Japan
| | - Ayumu Taguchi
- Division of Molecular Diagnostics, Aichi Cancer Center, 1-1 Kanokoden, Chikusa-Ku, Nagoya, Aichi, 464-8681, Japan
- Division of Advanced Cancer Diagnostics, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, Aichi, 466-8550, Japan
| | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, 1-Kawasumi, Mizuho-Cho, Mizuho-Ku, Nagoya, 467-8601, Japan
| |
Collapse
|
10
|
Su DG, Djureinovic D, Schoenfeld D, Marquez-Nostra B, Olino K, Jilaveanu L, Kluger H. Melanocortin-1 Receptor Expression as a Marker of Progression in Melanoma. JCO Precis Oncol 2024; 8:e2300702. [PMID: 38662983 PMCID: PMC11513442 DOI: 10.1200/po.23.00702] [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: 12/17/2023] [Revised: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 04/30/2024] Open
Abstract
PURPOSE Melanocortin-1 receptor (MC1R) plays a critical role in human pigmentation and DNA repair mechanisms. MC1R-targeting agents are being investigated in clinical trials in patients with melanoma, yet large studies investigating the rate and degree of MC1R expression in primary and metastatic human melanoma tissue are lacking. METHODS Using tissue microarrays containing three large cohorts of 225 cases of benign nevi, 189 with primary melanoma, and 271 with metastatic melanoma, we applied quantitative immunofluorescence and immunohistochemistry to comprehensively study MC1R protein expression. RESULTS We show a stepwise elevation of MC1R expression in different stages of melanoma progression (nevi, primary, metastasis). Higher MC1R expression was seen in deeper (>1 mm) primary lesions and ulcerated lesions and was associated with shorter survival in primary and metastatic tumors. On multivariable analysis, Breslow thickness, male sex, and chronic sun exposure were independent predictors of worse overall survival in the primary melanoma cohort. CONCLUSION Our data suggest that MC1R might be a valuable drug target in aggressive melanoma. Additional studies are warranted to determine its functional significance in melanoma progression and its utility as a predictive biomarker in patients receiving MC1R-directed therapies.
Collapse
Affiliation(s)
- David G. Su
- Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT
| | - Dijana Djureinovic
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - David Schoenfeld
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Bernadette Marquez-Nostra
- Department of Radiology, Division of Advanced Medical Imaging Research, University of Alabama at Birmingham, Birmingham, AL
| | - Kelly Olino
- Division of Surgical Oncology, Yale University School of Medicine, New Haven, CT
| | - Lucia Jilaveanu
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| | - Harriet Kluger
- Division of Medical Oncology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
11
|
Elkahwagy DM, Kiriacos CJ, Sobeih ME, Khorshid OMR, Mansour M. The lncRNAs Gas5, MALAT1 and SNHG8 as diagnostic biomarkers for epithelial malignant pleural mesothelioma in Egyptian patients. Sci Rep 2024; 14:4823. [PMID: 38413635 PMCID: PMC10899637 DOI: 10.1038/s41598-024-55083-9] [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: 01/24/2024] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
Long noncoding RNAs have been shown to be involved in a myriad of physiological and pathological pathways. To date, malignant pleural mesothelioma (MPM) is considered an extremely aggressive cancer. One reason for this is the late diagnosis of the disease, which can occur within 30-40 years of asbestos exposure. There is an immense need for the development of new, sensitive, inexpensive and easy methods for the early detection of this disease other than invasive methods such as biopsy. The aim of this study was to determine the expression of circulating lncRNAs in mesothelioma patient plasma to identify potential biomarkers. Ten previously identified lncRNAs that were shown to be aberrantly expressed in mesothelioma tissues were selected as candidates for subsequent validation. The expression of the ten selected candidate lncRNAs was verified via quantitative PCR (qPCR) in human plasma samples from mesothelioma patients versus healthy controls. The expression levels of circulating GAS5, SNHG8 and MALAT1 were significantly greater in plasma samples from patients than in those from controls. The ROC analysis of both MALAT1 and SNHG8 revealed 88.89% sensitivity and 66.67% specificity. The sensitivity of these markers was greater than that of GAS5 (sensitivity 72.22% and specificity 66.67%). The regression model for GAS5 was statistically significant, while that for SNHG8 and MALAT1 was not significant due to the small sample size. The area under the curve (AUC) of the three ROC curves was acceptable and significant: 0.7519 for GAS5, 0.7352 for SNHG8 and 0.7185 for MALAT1. This finding confirmed their ability to be used as markers. The three lncRNAs were not affected by age, sex or smoking status. The three lncRNAs showed great potential as independent predictive diagnostic biomarkers. Although the prediction model for MALAT1 did not significantly differ, MALAT1 was significantly expressed in patients more than in controls (p = 0.0266), and the recorded sensitivity and specificity were greater than those of GAS5.
Collapse
Affiliation(s)
- Dina Mohamed Elkahwagy
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Caroline Joseph Kiriacos
- Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt
| | - Mohamed Emam Sobeih
- Department of Medical Oncology, National Cancer Institute, NCI, Cairo University, Cairo, Egypt
| | - Ola M Reda Khorshid
- Department of Medical Oncology, National Cancer Institute, NCI, Cairo University, Cairo, Egypt
| | - Manar Mansour
- Pharmaceutical Biology and Microbiology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt.
- Department of Pharmaceutical Biology, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, 11835, Egypt.
| |
Collapse
|
12
|
Nishio S, Murotani K, Yamagami W, Suzuki S, Nakai H, Kato K, Tokunaga H, Nomura H, Yokoyama Y, Takehara K, Okamoto A. Pretreatment systemic inflammatory markers predict survival in endometrial cancer: A Japanese Gynecologic Oncology Group 2043 exploratory data analysis. Gynecol Oncol 2024; 181:46-53. [PMID: 38113633 DOI: 10.1016/j.ygyno.2023.12.007] [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: 08/22/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVE We investigated whether pretreatment systemic inflammatory markers are associated with survival outcomes in patients with endometrial cancer (EC). METHODS Data from the Japanese Gynecologic Oncology Group 2043 were analyzed. Patients who did not receive chemotherapy or were lost to follow-up were excluded. Associations of pretreatment systemic inflammatory markers, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and hemoglobin, albumin, lymphocyte, and platelet (HALP) score, with progression-free survival (PFS) and overall survival (OS) were analyzed. The optimal NLR, PLR, and HALP score cutoff values for PFS and OS were determined. Survival estimates were calculated and compared using the Kaplan-Meier method and log-rank test. RESULTS We included 712 patients (median age: 55 [range, 28-74] years; body mass index [BMI]: 21.1 [15.2-38.6] kg/m2). For PFS, optimal NLR, PLR, and HALP score cutoff values were 1.48, 0.017, and 35.52, respectively, and for OS, the values were 1.88, 0.026, and 19.87, respectively. At optimal PFS-related cutoff values, NLR was associated with BMI; PLR with age, BMI, and clinical stage; and HALP score with BMI, clinical stage, and lymph node metastasis. At optimal OS-related cutoff values, NLR was associated with BMI, PLR, and BMI; the HALP score was associated with age and BMI. The HALP score was a prognostic factor for PFS (p = 0.025), while PLR and HALP scores were prognostic factors for OS (both p = 0.028). CONCLUSIONS Pretreatment systemic inflammatory markers are associated with survival outcomes in patients with EC, with the HALP score being a prognostic factor for PFS and OS.
Collapse
Affiliation(s)
- Shin Nishio
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Japan.
| | - Kenta Murotani
- Biostatistics Center, Graduate School of Medicine, Kurume University, Kurume, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Shiro Suzuki
- Department of Gynecologic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hidekatsu Nakai
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Kazuyoshi Kato
- Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Hideki Tokunaga
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Nomura
- Department of Obstetrics and Gynecology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Yoshihito Yokoyama
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kazuhiro Takehara
- Department of Gynecologic Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Thannickal HH, Eltoum N, Henderson NL, Wallner LP, Wagner LI, Wolff AC, Rocque GB. Physicians' Hierarchy of Tumor Biomarkers for Optimizing Chemotherapy in Breast Cancer Care. Oncologist 2024; 29:e38-e46. [PMID: 37405703 PMCID: PMC10769784 DOI: 10.1093/oncolo/oyad198] [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: 03/08/2023] [Accepted: 06/13/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Tumor biomarkers are regularly used to guide breast cancer treatment and clinical trial enrollment. However, there remains a lack of knowledge regarding physicians' perspectives towards biomarkers and their role in treatment optimization, where treatment intensity is reduced to minimize toxicity. METHODS Thirty-nine academic and community oncologists participated in semi-structured qualitative interviews, providing perspectives on optimization approaches to chemotherapy treatment. Interviews were audio-recorded, transcribed, and analyzed by 2 independent coders utilizing a constant comparative method in NVivo. Major themes and exemplary quotes were extracted. A framework outlining physicians' conception of biomarkers, and their comfortability with their use in treatment optimization, was developed. RESULTS In the hierarchal model of biomarkers, level 1 is comprised of standard-of-care (SoC) biomarkers, defined by a strong level of evidence, alignment with national guidelines, and widespread utilization. Level 2 includes SoC biomarkers used in alternative contexts, in which physicians expressed confidence, yet less certainty, due to a lack of data in certain subgroups. Level 3, or experimental, biomarkers created the most diverse concerns related to quality and quantity of evidence, with several additional modulators. CONCLUSION This study demonstrates that physicians conceptualize the use of biomarkers for treatment optimization in successive levels. This hierarchy can be used to guide trialists in the development of novel biomarkers and design of future trials.
Collapse
Affiliation(s)
- Halle H Thannickal
- University of Alabama at Birmingham Heersink School of Medicine, Birmingham, AL, USA
| | - Noon Eltoum
- University of Alabama at Birmingham, Department of Medicine, Division of Hematology and Oncology; Birmingham, AL, USA
| | - Nicole L Henderson
- University of Alabama at Birmingham, Department of Medicine, Division of Hematology and Oncology; Birmingham, AL, USA
| | - Lauren P Wallner
- University of Michigan, Departments of Internal Medicine and Epidemiology, Rogel Cancer Center, Ann Arbor, MI, USA
| | | | - Antonio C Wolff
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Gabrielle B Rocque
- University of Alabama at Birmingham, Department of Medicine, Division of Hematology and Oncology; Birmingham, AL, USA
- University of Alabama at Birmingham, Department of Medicine, Division of Gerontology, Geriatrics, and Palliative CareBirmingham, AL, USA
- O’Neal Comprehensive Cancer Center; Birmingham, AL, USA
| |
Collapse
|
14
|
Mai Z, Xie J, Leng C, Xie X, Wen J, Yang H, Liu Q, Fu J. An optimized postsurgery follow-up strategy for patients with esophageal cancer: a cohort study. Int J Surg 2024; 110:332-341. [PMID: 37916933 PMCID: PMC10793741 DOI: 10.1097/js9.0000000000000827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/24/2023] [Indexed: 11/03/2023]
Abstract
BACKGROUND After radical surgery, patients with esophageal cancer should undergo long-term surveillance of disease relapse. However, the optimal follow-up strategy remains to be explored. METHOD A total of 4688 patients were recruited. Recursive partition analysis was applied to develop recurrence risk stratification for patients. The follow-up strategies of each stratification were developed based on monthly recurrence probability and validated by bootstrap validation and an external dataset. A Markov decision-analytic model was constructed to evaluate the cost-effectiveness of the follow-up strategies. RESULTS Patients were stratified into four groups according to four pathological features. The authors applied a random survival forest to calculate the monthly recurrence probability of each group. Based on the temporal distribution of recurrences, the authors further established surveillance strategies for four groups. The strategies were validated as optimal protocols by bootstrap resampling and another dataset. Markov cost-effective analysis indicated that our recommended strategies outperformed the mainstream protocols from guidelines. Using less than 12 visits across the first 5 years on average, our follow-up strategies were more efficient than the NCCN recommended strategies (14 visits average). Our results also supported the computerized tomography from the neck to the upper abdomen as a routine examination and PETCT of distant metastasis for some groups with high risks. CONCLUSION Our study provided data-driven evidence of personalized and economic follow-up strategies for esophageal cancer patients and shed light on follow-up optimization for other cancer types.
Collapse
Affiliation(s)
- Zihang Mai
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Guangdong Esophageal Cancer Institute, Guangzhou
| | - Jiaxin Xie
- National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Changsen Leng
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Guangdong Esophageal Cancer Institute, Guangzhou
| | - Xiuying Xie
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Guangdong Esophageal Cancer Institute, Guangzhou
| | - Jing Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
| | - Hong Yang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Guangdong Esophageal Cancer Institute, Guangzhou
| | - Qianwen Liu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Guangdong Esophageal Cancer Institute, Guangzhou
| | - Jianhua Fu
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine
- Guangdong Esophageal Cancer Institute, Guangzhou
| |
Collapse
|
15
|
Edland KH, Tjensvoll K, Oltedal S, Dalen I, Lapin M, Garresori H, Glenjen N, Gilje B, Nordgård O. Monitoring of circulating tumour DNA in advanced pancreatic ductal adenocarcinoma predicts clinical outcome and reveals disease progression earlier than radiological imaging. Mol Oncol 2023; 17:1857-1870. [PMID: 37341038 PMCID: PMC10483602 DOI: 10.1002/1878-0261.13472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with a need for better tools to guide treatment selection and follow-up. The aim of this prospective study was to investigate the prognostic value and treatment monitoring potential of longitudinal circulating tumour DNA (ctDNA) measurements in patients with advanced PDAC undergoing palliative chemotherapy. Using KRAS peptide nucleic acid clamp-PCR, we measured ctDNA levels in plasma samples obtained at baseline and every 4 weeks during chemotherapy from 81 patients with locally advanced and metastatic PDAC. Cox proportional hazard regression showed that ctDNA detection at baseline was an independent predictor of progression-free and overall survival. Joint modelling demonstrated that the dynamic ctDNA level was a strong predictor of time to first disease progression. Longitudinal ctDNA measurements during chemotherapy successfully revealed disease progression in 20 (67%) of 30 patients with ctDNA detected at baseline, with a median lead time of 23 days (P = 0.01) over radiological imaging. Here, we confirmed the clinical relevance of ctDNA in advanced PDAC with regard to both the prediction of clinical outcome and disease monitoring during treatment.
Collapse
Affiliation(s)
| | - Kjersti Tjensvoll
- Department of Hematology and OncologyStavanger University HospitalNorway
| | - Satu Oltedal
- Department of Hematology and OncologyStavanger University HospitalNorway
| | - Ingvild Dalen
- Section of Biostatistics, Department of ResearchStavanger University HospitalNorway
| | - Morten Lapin
- Department of Hematology and OncologyStavanger University HospitalNorway
| | - Herish Garresori
- Department of Hematology and OncologyStavanger University HospitalNorway
| | - Nils Glenjen
- Department of OncologyHaukeland University HospitalBergenNorway
| | - Bjørnar Gilje
- Department of Hematology and OncologyStavanger University HospitalNorway
| | - Oddmund Nordgård
- Department of Hematology and OncologyStavanger University HospitalNorway
- Department of Chemistry, Bioscience and Environmental Technology, Faculty of Science and TechnologyUniversity of StavangerNorway
| |
Collapse
|
16
|
Ye SB, Cheng YK, Li PS, Zhang L, Zhang LH, Huang Y, Chen P, Wang Y, Wang C, Peng JH, Shi LS, Ling L, Wu XJ, Qin J, Yang ZH, Lan P. High-throughput proteomics profiling-derived signature associated with chemotherapy response and survival for stage II/III colorectal cancer. NPJ Precis Oncol 2023; 7:50. [PMID: 37258779 DOI: 10.1038/s41698-023-00400-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 05/12/2023] [Indexed: 06/02/2023] Open
Abstract
Adjuvant chemotherapy (ACT) is usually used to reduce the risk of disease relapse and improve survival for stage II/III colorectal cancer (CRC). However, only a subset of patients could benefit from ACT. Thus, there is an urgent need to identify improved biomarkers to predict survival and stratify patients to refine the selection of ACT. We used high-throughput proteomics to analyze tumor and adjacent normal tissues of stage II/III CRC patients with /without relapse to identify potential markers for predicting prognosis and benefit from ACT. The machine learning approach was applied to identify relapse-specific markers. Then the artificial intelligence (AI)-assisted multiplex IHC was performed to validate the prognostic value of the relapse-specific markers and construct a proteomic-derived classifier for stage II/III CRC using 3 markers, including FHL3, GGA1, TGFBI. The proteomics profiling-derived signature for stage II/III CRC (PS) not only shows good accuracy to classify patients into high and low risk of relapse and mortality in all three cohorts, but also works independently of clinicopathologic features. ACT was associated with improved disease-free survival (DFS) and overall survival (OS) in stage II (pN0) patients with high PS and pN2 patients with high PS. This study demonstrated the clinical significance of proteomic features, which serve as a valuable source for potential biomarkers. The PS classifier provides prognostic value for identifying patients at high risk of relapse and mortality and optimizes individualized treatment strategy by detecting patients who may benefit from ACT for survival.
Collapse
Affiliation(s)
- Shu-Biao Ye
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Yi-Kan Cheng
- Department of Radiation Oncology; The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Pei-Si Li
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
- Sun Yat-sen University School of Medicine, Sun Yat-sen University, Guangzhou, PR China
| | - Lin Zhang
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Lian-Hai Zhang
- Department of Surgery, Peking University Cancer Hospital and Institute, Beijing, PR China
| | - Yan Huang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Ping Chen
- Department of VIP, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Yi Wang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing, China
| | - Chao Wang
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China
| | - Jian-Hong Peng
- Department of Colorectal Surgery, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, PR China
| | - Li-Shuo Shi
- Department of Probability and Statistics, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, PR China
| | - Li Ling
- Department of Probability and Statistics, School of Public Health, Sun Yat-sen University, Guangzhou, PR China
| | - Xiao-Jian Wu
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| | - Jun Qin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing Proteome Research Center, Beijing, China.
| | - Zi-Huan Yang
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| | - Ping Lan
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
- Department of Colorectal Surgery, Department of General Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
17
|
Kumar N, Gann PH, McGregor SM, Sethi A. Quantification of subtype purity in Luminal A breast cancer predicts clinical characteristics and survival. Breast Cancer Res Treat 2023:10.1007/s10549-023-06961-9. [PMID: 37209182 DOI: 10.1007/s10549-023-06961-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/26/2023] [Indexed: 05/22/2023]
Abstract
PURPOSE PAM50 profiling assigns each breast cancer to a single intrinsic subtype based on a bulk tissue sample. However, individual cancers may show evidence of admixture with an alternate subtype that could affect prognosis and treatment response. We developed a method to model subtype admixture using whole transcriptome data and associated it with tumor, molecular, and survival characteristics for Luminal A (LumA) samples. METHODS We combined TCGA and METABRIC cohorts and obtained transcriptome, molecular, and clinical data, which yielded 11,379 gene transcripts in common and 1,178 cases assigned to LumA. We used semi-supervised non-negative matrix factorization (ssNMF) to compute the subtype admixture proportions of the four major subtypes-pLumA, pLumB, pHER2, and pBasal-for each case and measured associations with tumor characteristics, molecular features, and survival. RESULTS Luminal A cases in the lowest versus highest quartile for pLumA transcriptomic proportion had a 27% higher prevalence of stage > 1, nearly a threefold higher prevalence of TP53 mutation, and a hazard ratio of 2.08 for overall mortality. We found positive associations between pHER2 and HER2 positivity by IHC or FISH; between pLumB and PR negativity; and between pBasal and younger age, node positivity, TP53 mutation, and EGFR expression. Predominant basal admixture, in contrast to predominant LumB or HER2 admixture, was not associated with shorter survival. CONCLUSION Bulk sampling for genomic analyses provides an opportunity to expose intratumor heterogeneity, as reflected by subtype admixture. Our results elucidate the striking extent of diversity among LumA cancers and suggest that determining the extent and type of admixture holds promise for refining individualized therapy. LumA cancers with a high degree of basal admixture appear to have distinct biological characteristics that warrant further study.
Collapse
Affiliation(s)
- Neeraj Kumar
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Peter H Gann
- Department of Pathology, College of Medicine, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Stephanie M McGregor
- Department of Pathology and Laboratory Medicine, University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Amit Sethi
- Department of Pathology, College of Medicine, University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
- Department of Electrical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
18
|
REMARK guidelines for tumour biomarker study reporting: a remarkable history. Br J Cancer 2023; 128:443-445. [PMID: 36476656 PMCID: PMC9938190 DOI: 10.1038/s41416-022-02046-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/17/2022] [Accepted: 10/21/2022] [Indexed: 12/12/2022] Open
Abstract
In 2005, several experts in tumor biomarker research publishe the REporting recommendations for Tumor MARKer prognostic studies (REMARK) criteria. Coupled with the subsequent Biospecimen Reporting for Improved Study Quality (BRISQ) criteria, these initiatives provide a framework for transparently reporting of the methods of study conduct and analyses.
Collapse
|
19
|
Lin Y, Lin L, Yang Y, Li M, Jiang X, Fu T, Long Y, Guo Q, He H, Chen Z, Du L, Liao G, Liao B, Huang J. DNA Methylation Architecture Provides Insight into the Pathogenesis of Upper Tract Urothelial Carcinoma: A Systematic Review and Meta-Analysis. Clin Genitourin Cancer 2023; 21:32-42. [PMID: 36376170 DOI: 10.1016/j.clgc.2022.10.008] [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: 03/22/2022] [Revised: 10/12/2022] [Accepted: 10/15/2022] [Indexed: 02/01/2023]
Abstract
PURPOSE Numerous studies suggested methylation modifications play an important role in upper tract urothelial carcinoma (UTUC), but few have depicted DNA methylation architecture on the pathological process of UTUC. We aimed to better understand the pathogenesis of UTUC and provide precision medicine references when managing UTUC patients. METHODS PubMed, Cochrane Library, EMBASE, and Scopus were searched for UTUC until December 31, 2020. Methodological quality assessment was conducted according to NIH recommendations. Meta-analysis was conducted to assess the prognostic effect of methylated genes. Kaplan-Meier survival analyses were performed to validate methylated genes and cytosine-phosphate-guanine (CpG) sites. RESULTS Eleven studies (3619 patients) were eligible to investigate 12 methylated genes and 10 CpGs. The quality of all the studies was fair to good. Meta-analysis found the pooled effect of eligible methylated genes had a low risk of tumor recurrence (HR = 0·67; 95% CI: 0·51-0·87; P = ·003), but a high risk of tumor progression (HR = 1·60; 95% CI: 1·17-2·18; P = ·003) and cancer-specific mortality (HR = 1·35; 95% CI: 1·06-1·72; P = ·01). For individual methylation status of GDF15, HSPA2, RASSF1A, TMEFF2, and VIM, the pooled effect of each gene was found pleiotropic on both diagnosis and prognosis. Survival analysis suggested higher methylation of SPARCL1 had a better disease-specific survival (P = ·048). CONCLUSION We combined meta-analysis and Kaplan-Meier survival analysis using the most updated evidence on the methylation of UTUC. Candidate biomarkers with essential diagnosis and prognosis function might provide precision medicine references for personalized therapies.
Collapse
Affiliation(s)
- Yifei Lin
- West China Hospital, Sichuan University, Chengdu, PR China
| | - Ling Lin
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yong Yang
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China
| | - Mei Li
- Medical Equipment Innovation Research Center, West China School of Medicine, Med+X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R.China
| | - Xin Jiang
- Medical Equipment Innovation Research Center, West China School of Medicine, Med+X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R.China
| | - Tingting Fu
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China; Medical Device Regulatory Research and Evaluation Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital/West China School of Medicine, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education,College of Life Sciences, Sichuan University, Chengdu, Sichuan, P.R.China
| | - Youlin Long
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China; Chinese Evidence-Based Medicine Centre, West China Hospital, Sichuan University, Chengdu, PR China
| | - Qiong Guo
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China
| | - He He
- Department of laboratory medicine, West China Hospital, Sichuan University, Chengdu, PR China
| | - Zhenglong Chen
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China
| | - Liang Du
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, PR China
| | - Banghua Liao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, PR China.
| | - Jin Huang
- Medical Device Regulatory Research and Evaluation Centre, West China Hospital, Sichuan University, Chengdu, PR China; Medical Equipment Innovation Research Center, West China School of Medicine, Med+X Center for Manufacturing, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R.China.
| |
Collapse
|
20
|
Valenza C, Taurelli Salimbeni B, Santoro C, Trapani D, Antonarelli G, Curigliano G. Tumor Infiltrating Lymphocytes across Breast Cancer Subtypes: Current Issues for Biomarker Assessment. Cancers (Basel) 2023; 15:cancers15030767. [PMID: 36765724 PMCID: PMC9913599 DOI: 10.3390/cancers15030767] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/17/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) represent a surrogate biomarker of anti-tumor, lymphocyte-mediated immunity. In early, triple-negative breast cancer, TILs have level 1B of evidence to predict clinical outcomes. TILs represent a promising biomarker to select patients who can experience a better prognosis with de-intensified cancer treatments and derive larger benefits from immune checkpoint inhibitors. However, the assessment and the validation of TILs as a biomarker require a prospective and rigorous demonstration of its clinical validity and utility, provided reproducible analytical performance. With pending data about the prospective validation of TILs' clinical validity to modulate treatments in early breast cancer, this review summarizes the most important current issues and future challenges related to the implementation of TILs assessments across all breast cancer subtypes and their potential integration into clinical practice.
Collapse
Affiliation(s)
- Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| | - Celeste Santoro
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Via Festa del Perdono 7, 20122 Milan, Italy
- Correspondence: ; Tel.: +39-02-5748-9599
| |
Collapse
|
21
|
Wang A, Tan Y, Wang S, Chen X. The prognostic value of separate lymphatic invasion and vascular invasion in oesophageal squamous cell carcinoma: a meta-analysis and systematic review. BMC Cancer 2022; 22:1329. [PMID: 36536299 PMCID: PMC9764535 DOI: 10.1186/s12885-022-10441-6] [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: 06/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Lymphovascular invasion (LVI) is a factor correlated with a poor prognosis in oesophageal squamous cell carcinoma (ESCC). Lymphatic invasion (LI) and vascular invasion (VI) should be reported separately because they may indicate a difference in prognosis. The prognostic role of LI and VI in ESCC patients remains controversial. A meta-analysis was conducted to resolve this question. METHODS We searched the PubMed, EMBASE, Web of Science, Scopus and Cochrane Library databases for studies on the association between LI and VI and the prognosis of patients with ESCC. The PICOs (Participant, Intervention, Comparison, Outcome) strategy were selected for the systematic review and meta-analysis. The effect size (ES) was the hazard ratio (HR) or relative ratio (RR) with 95% confidence intervals (CI) for overall survival (OS) and recurrence-free survival (RFS). RESULTS A total of 27 studies with 5740 patients were included. We calculated the pooled results from univariate and multivariate analysis using the Cox proportional hazards method. The heterogeneity was acceptable in OS and RFS. According to the pooled results of multivariate analysis, both LI and VI were correlated with a worse OS. VI was a negative indicator for RFS, while the p value of VI was greater than 0.05. The prognostic role was weakened in subgroup analysis with studies using haematoxylin-eosin staining method. CONCLUSIONS Both LI and VI were indicators of a worse OS outcome. LI was a more significant indicator in predicting a worse RFS. More larger sample studies with immunohistochemical staining and good designs are required to detect the prognostic value of separate LI and VI in ESCC.
Collapse
Affiliation(s)
- An Wang
- grid.8547.e0000 0001 0125 2443Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yulong Tan
- grid.8547.e0000 0001 0125 2443Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Shaohua Wang
- grid.8547.e0000 0001 0125 2443Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaofeng Chen
- grid.8547.e0000 0001 0125 2443Department of Thoracic Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Basyuni S, Nugent G, Ferro A, Barker E, Reddin I, Jones O, Lechner M, O’Leary B, Jones T, Masterson L, Fenton T, Schache A. Value of p53 sequencing in the prognostication of head and neck cancer: a systematic review and meta-analysis. Sci Rep 2022; 12:20776. [PMID: 36456616 PMCID: PMC9715723 DOI: 10.1038/s41598-022-25291-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
This review aimed to examine the relationship between TP53 mutational status, as determined by genomic sequencing, and survival in squamous cell carcinoma of the head and neck. The databases Medline, Embase, Web of Science (core collection), Scopus and Cochrane Library were searched from inception to April 2021 for studies assessing P53 status and survival. Qualitative analysis was carried out using the REMARK criteria. A meta-analyses was performed and statistical analysis was carried out to test the stability and reliability of results. Twenty-five studies met the inclusion criteria, of which fifteen provided enough data for quantitative evaluation. TP53 mutation was associated with worse overall survival (HR 1.75 [95% CI 1.45-2.10], p < 0.001), disease-specific survival (HR 4.23 [95% CI 1.19-15.06], p = 0.03), and disease-free survival (HR 1.80 [95% CI 1.28-2.53], p < 0.001). Qualitative assessment identified room for improvement and the pooled analysis of all anatomical subsites leads to heterogeneity that may erode the validity of the observed overall effect and its subsequent extrapolation and application to individual patients. Our systematic review and meta-analysis supports the utility of TP53 mutational as a prognostic factor for survival in head and neck squamous cell carcinoma. A well designed prospective, multi-centre trial is needed to definitively answer this question.
Collapse
Affiliation(s)
- Shadi Basyuni
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.24029.3d0000 0004 0383 8386Department of Oral and Maxillo-Facial Surgery, Cambridge University Hospitals, Cambridge, CB2 0QQ UK
| | - Gareth Nugent
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.24029.3d0000 0004 0383 8386Department of Oral and Maxillo-Facial Surgery, Cambridge University Hospitals, Cambridge, CB2 0QQ UK
| | - Ashley Ferro
- grid.24029.3d0000 0004 0383 8386Department of Oral and Maxillo-Facial Surgery, Cambridge University Hospitals, Cambridge, CB2 0QQ UK
| | - Eleanor Barker
- grid.5335.00000000121885934Medical Library, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ian Reddin
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.5491.90000 0004 1936 9297Faculty of Medicine, University of Southampton, Southampton, UK
| | - Oliver Jones
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.8250.f0000 0000 8700 0572University of Durham, Durham, UK
| | - Matt Lechner
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.83440.3b0000000121901201Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - Ben O’Leary
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.18886.3fThe Institute of Cancer Research, London, UK
| | - Terry Jones
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.10025.360000 0004 1936 8470Liverpool Head & Neck Centre, University of Liverpool, Liverpool, UK
| | - Liam Masterson
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.24029.3d0000 0004 0383 8386Department of Ear, Nose and Throat Surgery, Cambridge University Hospitals, Cambridge, UK
| | - Tim Fenton
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.5491.90000 0004 1936 9297Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andrew Schache
- grid.498322.6Genomics England Head and Neck Cancer Clinical Interpretation Partnership, England, UK ,grid.10025.360000 0004 1936 8470Liverpool Head & Neck Centre, University of Liverpool, Liverpool, UK
| |
Collapse
|
23
|
Integration of OV6 expression and CD68 + tumor-associated macrophages with clinical features better predicts the prognosis of patients with hepatocellular carcinoma. Transl Oncol 2022; 25:101509. [PMID: 36030750 PMCID: PMC9428913 DOI: 10.1016/j.tranon.2022.101509] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/31/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Reliable prognostic indicators for accurately predicting postoperative outcomes in Hepatocellular carcinoma (HCC) patients are lacking. Although cancer stem-like cells (CSCs) and tumor-associated macrophages (TAMs) in tumor microenvironment are implicated in the occurrence and development of HCC, whether the combination of CSC biomarkers and TAM populations could achieve better performance in predicting the prognosis of patients with HCC has been rarely reported. METHODS A total of 306 HCC patients were randomly divided into the training and validation cohorts at a 1:1 ratio, and the expression of OV6 and CD68 was assessed using immunohistochemistry in HCC samples. The prognostic value of these biomarkers for post-surgical survival and recurrence were evaluated by the curve of receiver operating characteristic and multivariate Cox regression analyses. RESULTS The density of OV6+ CSCs was positively correlated with the infiltration of CD68+ TAMs in HCC. Both high OV6 expression and CD68+ TAM infiltration was closely associated with poor overall survival (OS) and progression-free survival (PFS) of HCC patients. Moreover, overexpression of OV6 and infiltration of CD68+ TAMs were identified as independent prognostic factors for OS and PFS after liver resection. The integration of OV6 and CD68 with tumor size and microvascular invasion exhibited highest C-index value for survival predictivity in HCC patients than any other biomarkers or clinical indicators alone. CONCLUSION Incorporating intratumoral OV6 expression and CD68+ TAMs infiltration with established clinical indicators may serve as a promising prognostic signature for HCC, and could more accurately predict the clinical outcomes for HCC patients after liver resection.
Collapse
|
24
|
Fu T, Lin Y, Lin L, Yang Y, Guo Q, Long Y, He H, Bao Y, Lin T, Chen J, Chen Z, Du L, Liao G, Liao B, Huang J. Network architecture of non-coding RNAs provides insights into the pathogenesis of upper tract urothelial carcinoma. Urol Oncol 2022; 40:383.e11-383.e21. [DOI: 10.1016/j.urolonc.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/18/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
25
|
Nordgård O, Lapin M, Tjensvoll K, Oltedal S, Edland KH, Neverdahl NB, Fostenes D, Garresori H, Glenjen N, Smaaland R, Gilje B. Prognostic value of disseminated tumor cells in unresectable pancreatic ductal adenocarcinoma: a prospective observational study. BMC Cancer 2022; 22:609. [PMID: 35659265 PMCID: PMC9166481 DOI: 10.1186/s12885-022-09714-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although pancreatic ductal adenocarcinoma (PDAC) rarely metastasizes to the skeleton, disseminated tumor cells have been detected in bone marrow samples from patients with this disease. The prognostic value of such findings is currently unclear. Thus, the current study aimed to clarify the prognostic information associated with disseminated tumor cell detection in samples from patients with PDAC. METHODS Bone marrow aspirates were obtained from 48 patients with locally advanced (n = 11) or metastatic (n = 37) PDAC, before and after 2 months of chemotherapy. Disseminated tumor cells were detected with an mRNA panel and quantitative reverse transcription PCR. We used the highest levels measured in healthy bone marrow (n = 30) as a threshold to define the positive detection of disseminated tumor cells. Progression-free and overall survival were analyzed with Kaplan-Meier and Cox proportional hazards regression analyses. RESULTS Disseminated tumor cells were detected in 15/48 (31%) bone marrow samples obtained before starting chemotherapy and in 8/25 (32%) samples obtained during chemotherapy. Patients with disseminated tumor cells detected before therapy had significantly shorter progression-free (p = 0.03; HR = 2.0) and overall survival (p = 0.03; HR = 2.0), compared to those without disseminated tumor cells in the bone marrow. When restricting disseminated tumor cell detection to keratins KRT7 and KRT8, the prognostic information was substantially stronger (p = 1 × 10-6; HR = 22, and p = 2 × 10-5; HR = 7.7, respectively). The multivariable Cox regression analysis demonstrated that disseminated tumor cell detection prior to treatment had independent prognostic value. In contrast, disseminated tumor cells detected during treatment did not have prognostic value. CONCLUSIONS Disseminated tumor cells detected before commencing chemotherapy had prognostic value in patients with inoperable PDAC.
Collapse
Affiliation(s)
- Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway.
- Department of Chemistry, Bioscience and Environmental Technology, Faculty of Science and Technology, University of Stavanger, Stavanger, Norway.
| | - Morten Lapin
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Kjersti Tjensvoll
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Satu Oltedal
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Karin Hestnes Edland
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Nicolay Bore Neverdahl
- Department of Chemistry, Bioscience and Environmental Technology, Faculty of Science and Technology, University of Stavanger, Stavanger, Norway
| | - Dmitrij Fostenes
- Department of Chemistry, Bioscience and Environmental Technology, Faculty of Science and Technology, University of Stavanger, Stavanger, Norway
| | - Herish Garresori
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| | - Nils Glenjen
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Rune Smaaland
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
- Present Address: Mosaic Oncology AS, Sandnes, Norway
| | - Bjørnar Gilje
- Department of Hematology and Oncology, Stavanger University Hospital, Stavanger, Norway
| |
Collapse
|
26
|
Hwang CY, Yu SJ, Won JK, Park SM, Noh H, Lee S, Cho EJ, Lee JH, Lee KB, Kim YJ, Suh KS, Yoon JH, Cho KH. Systems analysis identifies endothelin 1 axis blockade for enhancing the anti-tumor effect of multikinase inhibitor. Cancer Gene Ther 2022; 29:845-858. [PMID: 34363028 DOI: 10.1038/s41417-021-00373-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/14/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023]
Abstract
Multikinase inhibitors, such as sorafenib, are used for the treatment of advanced carcinomas but the response shows limited efficacy or varies a lot with patients. Here we adopted the systems approach combined with high-throughput data analysis to discover key mechanism embedded in the drug response. When analyzing the transcriptomic data from the Cancer Cell Line Encyclopedia (CCLE) database, endothelin 1 (EDN1) was enriched in cancer cells with low responsiveness to sorafenib. We found that the level of EDN1 is higher in the tissue and blood of hepatocellular carcinoma (HCC) patients showing poor response to sorafenib. In vitro experiment showed that EDN1 not only induces activation of angiogenic-promoting pathways in HCC cells but also stimulates proliferation and migration. Moreover, EDN1 is related with poor responsiveness to sorafenib by mitigating unfolded protein response (UPR), which was validated in both transcriptomic data analysis and in silico simulation. Finally, we found that endothelin receptor B (EDNRB) antagonists can enhance the efficacy of sorafenib in both HCC cells and xenograft mouse models. Our findings provide that EDN1 is a novel diagnostic marker for sorafenib responsiveness in HCC and a basis for testing macitentan, which is currently used for pulmonary artery hypertension, in combination with sorafenib in advanced HCC patients.
Collapse
Affiliation(s)
- Chae Young Hwang
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Aventi, Inc., Daejeon, Korea
| | - Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jae-Kyung Won
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Min Park
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,KM Data Division, Korea Institute of Oriental Medicine, Daejeon, Korea
| | - Hyojin Noh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Soobeom Lee
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Eun Ju Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong-Hoon Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung Bun Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yoon Jun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Suk Suh
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jung-Hwan Yoon
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Kwang-Hyun Cho
- Laboratory for Systems Biology and Bio-inspired Engineering, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
| |
Collapse
|
27
|
KLK3 germline mutation I179T complements DNA repair genes for predicting prostate cancer progression. Prostate Cancer Prostatic Dis 2022; 25:749-754. [PMID: 35149774 DOI: 10.1038/s41391-021-00466-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND Germline mutations in DNA repair genes and KLK3 have been associated with adverse prostate cancer (PCa) outcomes in separate studies but never jointly. The objective of this study is to simultaneously assess these two types of germline mutations. METHODS Germline rare pathogenic mutations (RPMs) in 9 commonly tested DNA repair genes and KLK3 variants were tested for their associations with PCa progression in two PCa cohorts: (1) hospital-based PCa patients treated with radical surgery at the Johns Hopkins Hospital (JHH, N = 1943), and (2) population-based PCa patients in the UK Biobank (UKB, N = 10,224). Progression was defined as metastasis and/or PCa-specific death (JHH) and PCa-specific death (UKB). RPMs of DNA repair genes were annotated using the American College of Medical Genetics recommendations. Known KLK3 variants were genotyped. Associations were tested using a logistic regression model adjusting for genetic background (top ten principal components). RESULTS In the JHH, 3.2% (59/1,843) of patients had RPMs in 9 DNA repair genes; odds ratio (OR, 95% confidence interval) for progression was 2.99 (1.6-5.34), P < 0.001. In comparison, KLK3 I179T mutation was more common; 9.7% (189/1,943) carried the mutation, OR = 1.6 (1.05-2.37), P = 0.02. Similar results were found in the UKB. Both types of mutations remained statistically significant in multivariable analyses. In the combined cohort, compared to patients without any mutations (RPMs-/KLK3-), RPMs-/KLK3+ patients had modestly increased risk for progression [OR = 1.54 (1.15-2.02), P = 0.003], and RPMs+/KLK3+ patients had greatly increased risk for progression [OR = 5.41 (2.04-12.99), P < 0.001]. Importantly, associations of mutations with PCa progression were found in patients with clinically defined low- or intermediate risk for disease progression. CONCLUSIONS Two different cohorts consistently demonstrate that KLK3 I179T and RPMs of nine commonly tested DNA repair genes are complementary for predicting PCa progression. These results are highly relevant to PCa germline testing and provide critical information for KLK3 I179T to be considered in guidelines.
Collapse
|
28
|
Luque M, Cristóbal I, Sanz-Álvarez M, Santos A, Zazo S, Eroles P, Arpí O, Rovira A, Albanell J, Madoz-Gúrpide J, García-Foncillas J, Rojo F. CIP2A as a Key Regulator for AKT Phosphorylation Has Partial Impact Determining Clinical Outcome in Breast Cancer. J Clin Med 2022; 11:jcm11061610. [PMID: 35329936 PMCID: PMC8955826 DOI: 10.3390/jcm11061610] [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: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/12/2022] [Indexed: 12/07/2022] Open
Abstract
Together with its reported ability to modulate AKT phosphorylation (p-AKT) status in several tumor types, the oncoprotein CIP2A has been described to induce breast cancer progression and drug resistance. However, the clinical and therapeutic relevance of the CIP2A/AKT interplay in breast cancer remains to be fully clarified. Here, we found high p-AKT levels in 80 out of 220 cases (36.4%), which were associated with negative estrogen receptor expression (p = 0.049) and CIP2A overexpression (p < 0.001). Interestingly, p-AKT determined substantially shorter overall (p = 0.002) and progression-free survival (p = 0.003), and multivariate analyses showed its CIP2A-independent prognostic value. Moreover, its clinical relevance was further confirmed in the triple negative and HER2-positive subgroups after stratifying our series by molecular subtype. Functionally, we confirmed in vitro the role of CIP2A as a regulator of p-AKT levels in breast cancer cell lines, and the importance of the CIP2A/AKT axis was also validated in vivo. Finally, p-AKT also showed a higher predictive value of response to doxorubicin than CIP2A in ex vivo analyses. In conclusion, our findings suggest that CIP2A overexpression is a key contributing event to AKT phosphorylation and highlights the CIP2A/AKT axis as a promising therapeutic target in breast cancer. However, our observations highlight the existence of alternative mechanisms that regulate AKT signaling in a subgroup of breast tumors without altered CIP2A expression that determines its independent value as a marker of poor outcome in this disease.
Collapse
Affiliation(s)
- Melani Luque
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| | - Marta Sanz-Álvarez
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, ISS-FJD-UAM, 28040 Madrid, Spain;
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Sandra Zazo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Pilar Eroles
- Institute of Health Research INCLIVA, 46010 Valencia, Spain;
| | - Oriol Arpí
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Ana Rovira
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Joan Albanell
- Medical Oncology Department, Hospital del Mar, 08003 Barcelona, Spain; (O.A.); (A.R.); (J.A.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain;
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Diaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-Á.); (S.Z.); (J.M.-G.)
- Correspondence: (I.C.); (F.R.); Tel.: +34-915-504-800 (I.C. & F.R.)
| |
Collapse
|
29
|
Santos A, Cristóbal I, Rubio J, Caramés C, Luque M, Sanz-Alvarez M, Morales-Gallego M, Madoz-Gúrpide J, Rojo F, García-Foncillas J. MicroRNA-199b Deregulation Shows Oncogenic Properties and Promising Clinical Value as Circulating Marker in Locally Advanced Rectal Cancer Patients. Int J Mol Sci 2022; 23:2203. [PMID: 35216319 PMCID: PMC8875596 DOI: 10.3390/ijms23042203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/14/2022] [Accepted: 02/14/2022] [Indexed: 02/01/2023] Open
Abstract
The identification of robust prognostic markers still represents a need in locally advanced rectal cancer (LARC). MicroRNAs (miRs) have progressively emerged as promising circulating markers, overcoming some limitations that traditional biopsy comprises. Tissue miR-199b deregulation has been reported to predict outcome and response to neoadjuvant chemoradiotherapy (nCRT) in LARC, and was also found to be associated with disease progression in colorectal cancer. However, its biological and clinical relevance remains to be fully clarified. Thus, we observed here that miR-199b regulates cell migration, aggressiveness, and cell growth, and inhibits colonosphere formation and induces caspase-dependent apoptosis. Moreover, miR-199b expression was quantified by real-time PCR in plasma samples from LARC patients and its downregulation was observed in 22.7% of cases. This alteration was found to be associated with higher tumor size (p = 0.002) and pathological stage (p = 0.020) after nCRT. Notably, we observed substantially lower global miR-199b expression associated with patient downstaging (p = 0.009), as well as in non-responders compared to those cases who responded to nCRT in both pre- (p = 0.003) and post-treatment samples (p = 0.038). In concordance, we found that miR-199b served as a predictor marker of response to neoadjuvant therapy in our cohort (p = 0.011). Altogether, our findings here demonstrate the functional relevance of miR-199b in this disease and its potential value as a novel circulating marker in LARC.
Collapse
Affiliation(s)
- Andrea Santos
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (A.S.); (J.R.); (C.C.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
| | - Ion Cristóbal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (A.S.); (J.R.); (C.C.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
| | - Jaime Rubio
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (A.S.); (J.R.); (C.C.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
- Medical Oncology Department, University Hospital “Fundación Jiménez Díaz”, UAM, 28040 Madrid, Spain
| | - Cristina Caramés
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (A.S.); (J.R.); (C.C.)
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
- Medical Oncology Department, University Hospital “Fundación Jiménez Díaz”, UAM, 28040 Madrid, Spain
| | - Melani Luque
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-A.); (M.M.-G.); (J.M.-G.); (F.R.)
| | - Marta Sanz-Alvarez
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-A.); (M.M.-G.); (J.M.-G.); (F.R.)
| | - Miriam Morales-Gallego
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-A.); (M.M.-G.); (J.M.-G.); (F.R.)
| | - Juan Madoz-Gúrpide
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-A.); (M.M.-G.); (J.M.-G.); (F.R.)
| | - Federico Rojo
- Pathology Department, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain; (M.L.); (M.S.-A.); (M.M.-G.); (J.M.-G.); (F.R.)
| | - Jesús García-Foncillas
- Translational Oncology Division, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM, 28040 Madrid, Spain
- Medical Oncology Department, University Hospital “Fundación Jiménez Díaz”, UAM, 28040 Madrid, Spain
| |
Collapse
|
30
|
Nilsen A, Hillestad T, Skingen VE, Aarnes E, Fjeldbo CS, Hompland T, Evensen TS, Stokke T, Kristensen GB, Grallert B, Lyng H. miR-200a/b/-429 downregulation is a candidate biomarker of tumor radioresistance and independent of hypoxia in locally advanced cervical cancer. Mol Oncol 2022; 16:1402-1419. [PMID: 35064630 PMCID: PMC8936520 DOI: 10.1002/1878-0261.13184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/21/2021] [Accepted: 01/19/2022] [Indexed: 11/07/2022] Open
Abstract
Many patients with locally advanced cervical cancer experience recurrence within the radiation field after chemoradiotherapy. Biomarkers of tumor radioresistance are required to identify patients in need of intensified treatment. Here, the biomarker potential of miR-200 family members was investigated in this disease. Also, involvement of tumor hypoxia in the radioresistance mechanism was determined, using a previously defined 6-gene hypoxia classifier. miR-200 expression was measured in pre-treatment tumor biopsies of an explorative cohort (n=90) and validation cohort 1 (n=110) by RNA sequencing. Publicly available miR-200 data of 79 patients were included for validation of prognostic significance. A score based on expression of the miR-200a/b/-429 (miR-200a, miR-200b and miR-429) cluster showed prognostic significance in all cohorts. The score was significant in multivariate analysis of central pelvic recurrence. No association with distant recurrence or hypoxia status was found. Potential miRNA target genes were identified from gene expression profiles and showed enrichment of genes in extracellular matrix organization and cell adhesion. miR-200a/b/-429 overexpression had a pronounced radiosensitizing effect in tumor xenografts, whereas the effect was minor in vitro. In conclusion, miR-200a/b/-429 downregulation is a candidate biomarker of central pelvic recurrence and seems to predict cell-adhesion-mediated tumor radioresistance independent of clinical markers and hypoxia.
Collapse
Affiliation(s)
- Anja Nilsen
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Tiril Hillestad
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Vilde E. Skingen
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Eva‐Katrine Aarnes
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Christina S. Fjeldbo
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Tord Hompland
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Tina Sandø Evensen
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Trond Stokke
- Department of Core FacilitiesNorwegian Radium HospitalOslo University HospitalNorway
| | - Gunnar B. Kristensen
- Department of Gynecological OncologyNorwegian Radium HospitalOslo University HospitalNorway
- Institute of Cancer Genetics and InformaticsOslo University HospitalNorway
| | - Beata Grallert
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
| | - Heidi Lyng
- Department of Radiation BiologyNorwegian Radium HospitalOslo University HospitalNorway
- Department of PhysicsUniversity of OsloNorway
| |
Collapse
|
31
|
A Novel Urinary miRNA Biomarker for Early Detection of Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14020461. [PMID: 35053622 PMCID: PMC8773893 DOI: 10.3390/cancers14020461] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/27/2021] [Accepted: 01/14/2022] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Early diagnosis is critically important to achieve life-saving therapy for colorectal cancer (CRC). Since colonoscopy is not suitable as a screening method for CRC due to its invasiveness and high-cost, reliable and non-invasive diagnostic biomarkers are hopeful for CRC. In this case-control study, we established completely non-invasive, novel urinary microRNA (miRNA) biomarker panel combining miR-129-1-3p and miR-566 for the diagnosis of CRC. In the independent age- and sex-matched three cohorts comprising 415 participants, urinary levels of these miRNAs were consistently elevated in the CRC group compared to the healthy controls. Notably, the panel of combining miR-129-1-3p and miR-566 revealed an AUC of 0.845 for stage 0/I CRC that can be treated with endoscopic resection. Abstract Since noninvasive biomarkers as an alternative to invasive colonoscopy to detect colorectal cancer (CRC) are desired, we conducted this study to determine the urinary biomarker consisting of microRNAs (miRNAs). In total, 415 age- and sex-matched participants, including 206 patients with CRC and 209 healthy controls (HCs), were randomly divided into three groups: (1) the discovery cohort (CRC, n = 3; HC, n = 6); (2) the training cohort (140 pairs); and (3) the validation cohort (63 pairs). Among 11 urinary miRNAs with aberrant expressions between the two groups, miR-129-1-3p and miR-566 were significantly independent biomarkers that detect CRC. The panel consisting of two miRNAs could distinguish patients with CRC from HC participants with an area under the curve (AUC) = 0.811 in the training cohort. This panel showed good efficacy with an AUC = 0.868 in the validation cohort. This urinary biomarker combining miR-129-1-3p and miR-566 could detect even stage 0/I CRC effectively with an AUC = 0.845. Moreover, the expression levels of both miR-129-1-3p and miR-566 were significantly higher in primary tumor tissues than in adjacent normal tissue. Our established novel biomarker consisting of urinary miR-129-1-3p and miR-566 enables noninvasive and early detection of CRC.
Collapse
|
32
|
Shi Z, Lu L, Resurreccion WK, Yang W, Wei J, Wang Q, Engelmann V, Zheng SL, Cooney KA, Isaacs WB, Helfand BT, Lu J, Xu J. Association of germline rare pathogenic mutations in guideline-recommended genes with prostate cancer progression: A meta-analysis. Prostate 2022; 82:107-119. [PMID: 34674288 DOI: 10.1002/pros.24252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Germline mutations in several genes, mainly DNA repair genes, have been associated with prostate cancer (PCa) progression. However, primarily due to the rarity of mutations, statistical evidence for these associations is not consistently established. The objective of this study is to synthesize evidence from multiple studies using a meta-analysis. METHODS Genes analyzed were chosen based on National Comprehensive Cancer Network guidelines recommendations (10 genes) and a commonly reported gene (NBN). PCa progression in this analysis was defined as either having metastases or PCa-specific mortality. We searched PubMed for papers published before April 26, 2021, using selected keywords. Pooled odds ratio (OR) was estimated in all races and Caucasians-only using both fixed- and random-effect models. RESULTS The search identified 1028 papers and an additional five from a manual review of references. After a manual process that excluded noneligible studies, 11 papers remained, including a total of 3944 progressors and 20,054 nonprogressors. Combining results from these eligible studies, mutation carrier rates were significantly higher in progressors than nonprogressors for NBN, BRCA2, ATM (under both fixed- and random-effect models), for CHEK2 (under fixed-effect model only), and for PALB2 (under random-effect model only), p < 0.05. Pooled OR (95% confidence interval) was 6.38 (2.25-18.05), 3.41 (2.31; 5.03), 1.93 (1.17-3.20), and 1.53 (1.00-2.33) for NBN, BRCA2, ATM, and CHEK2, respectively, under fixed-effect model and 2.63 (1.12-6.13) for PALB2 under random-effect model. No significant association was found for the six remaining genes. Certainty of evidence was low for many genes due primarily to the limited number of eligible studies and mutation carriers. CONCLUSIONS Statistical evidence for five genes was obtained in this first meta-analysis of germline mutations and PCa progression. While these results may help urologists and genetic counselors interpret germline testing results for PCa progression, more original studies are needed.
Collapse
Affiliation(s)
- Zhuqing Shi
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Lucy Lu
- GoPath Laboratories LLC, Buffalo Grove, Illinois, USA
| | - William Kyle Resurreccion
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Wancai Yang
- GoPath Laboratories LLC, Buffalo Grove, Illinois, USA
| | - Jun Wei
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Qiang Wang
- GoPath Laboratories LLC, Buffalo Grove, Illinois, USA
| | | | - Siqun Lilly Zheng
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Kathleen A Cooney
- Department of Medicine, Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - William B Isaacs
- Department of Urology, The Johns Hopkins School of Medicine, The Brady Urological Institute, Baltimore, Maryland, USA
| | - Brian T Helfand
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Jim Lu
- GoPath Laboratories LLC, Buffalo Grove, Illinois, USA
| | - Jianfeng Xu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, NorthShore University HealthSystem, Evanston, Illinois, USA
| |
Collapse
|
33
|
Targeted Therapy Modulates the Secretome of Cancer-Associated Fibroblasts to Induce Resistance in HER2-Positive Breast Cancer. Int J Mol Sci 2021; 22:ijms222413297. [PMID: 34948097 PMCID: PMC8706990 DOI: 10.3390/ijms222413297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 12/29/2022] Open
Abstract
The combination of trastuzumab plus pertuzumab plus docetaxel as a first-line therapy in patients with HER2-positive metastatic breast cancer has provided significant clinical benefits compared to trastuzumab plus docetaxel alone. However, despite the therapeutic success of existing therapies targeting HER2, tumours invariably relapse. Therefore, there is an urgent need to improve our understanding of the mechanisms governing resistance, so that specific therapeutic strategies can be developed to provide improved efficacy. It is well known that the tumour microenvironment (TME) has a significant impact on cancer behaviour. Cancer-associated fibroblasts (CAFs) are essential components of the tumour stroma that have been linked to acquired therapeutic resistance and poor prognosis in breast cancer. For this reason, it would be of interest to identify novel biomarkers in the tumour stroma that could emerge as therapeutic targets for the modulation of resistant phenotypes. Conditioned medium experiments carried out in our laboratory with CAFs derived from HER2-positive patients showed a significant capacity to promote resistance to trastuzumab plus pertuzumab therapies in two HER2-positive breast cancer cell lines (BCCLs), even in the presence of docetaxel. In order to elucidate the components of the CAF-conditioned medium that may be relevant in the promotion of BCCL resistance, we implemented a multiomics strategy to identify cytokines, transcription factors, kinases and miRNAs in the secretome that have specific targets in cancer cells. The combination of cytokine arrays, label-free LC-MS/MS quantification and miRNA analysis to explore the secretome of CAFs under treatment conditions revealed several up- and downregulated candidates. We discuss the potential role of some of the most interesting candidates in generating resistance in HER2-positive breast cancer.
Collapse
|
34
|
Dudgeon SN, Wen S, Hanna MG, Gupta R, Amgad M, Sheth M, Marble H, Huang R, Herrmann MD, Szu CH, Tong D, Werness B, Szu E, Larsimont D, Madabhushi A, Hytopoulos E, Chen W, Singh R, Hart SN, Sharma A, Saltz J, Salgado R, Gallas BD. A Pathologist-Annotated Dataset for Validating Artificial Intelligence: A Project Description and Pilot Study. J Pathol Inform 2021; 12:45. [PMID: 34881099 PMCID: PMC8609287 DOI: 10.4103/jpi.jpi_83_20] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/23/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Purpose: Validating artificial intelligence algorithms for clinical use in medical images is a challenging endeavor due to a lack of standard reference data (ground truth). This topic typically occupies a small portion of the discussion in research papers since most of the efforts are focused on developing novel algorithms. In this work, we present a collaboration to create a validation dataset of pathologist annotations for algorithms that process whole slide images. We focus on data collection and evaluation of algorithm performance in the context of estimating the density of stromal tumor-infiltrating lymphocytes (sTILs) in breast cancer. Methods: We digitized 64 glass slides of hematoxylin- and eosin-stained invasive ductal carcinoma core biopsies prepared at a single clinical site. A collaborating pathologist selected 10 regions of interest (ROIs) per slide for evaluation. We created training materials and workflows to crowdsource pathologist image annotations on two modes: an optical microscope and two digital platforms. The microscope platform allows the same ROIs to be evaluated in both modes. The workflows collect the ROI type, a decision on whether the ROI is appropriate for estimating the density of sTILs, and if appropriate, the sTIL density value for that ROI. Results: In total, 19 pathologists made 1645 ROI evaluations during a data collection event and the following 2 weeks. The pilot study yielded an abundant number of cases with nominal sTIL infiltration. Furthermore, we found that the sTIL densities are correlated within a case, and there is notable pathologist variability. Consequently, we outline plans to improve our ROI and case sampling methods. We also outline statistical methods to account for ROI correlations within a case and pathologist variability when validating an algorithm. Conclusion: We have built workflows for efficient data collection and tested them in a pilot study. As we prepare for pivotal studies, we will investigate methods to use the dataset as an external validation tool for algorithms. We will also consider what it will take for the dataset to be fit for a regulatory purpose: study size, patient population, and pathologist training and qualifications. To this end, we will elicit feedback from the Food and Drug Administration via the Medical Device Development Tool program and from the broader digital pathology and AI community. Ultimately, we intend to share the dataset, statistical methods, and lessons learned.
Collapse
Affiliation(s)
- Sarah N Dudgeon
- Division of Imaging Diagnostics and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiologic Health, United States Food and Drug Administration, White Oak, MD, USA
| | - Si Wen
- Division of Imaging Diagnostics and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiologic Health, United States Food and Drug Administration, White Oak, MD, USA
| | | | - Rajarsi Gupta
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | - Mohamed Amgad
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Manasi Sheth
- Division of Biostatistics, Center for Devices and Radiologic Health, United States Food and Drug Administration, White Oak, MD, USA
| | - Hetal Marble
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard Huang
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Markus D Herrmann
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | - Evan Szu
- Arrive Bio, San Francisco, CA, USA
| | - Denis Larsimont
- Department of Pathology, Institute Jules Bordet, Brussels, Belgium
| | - Anant Madabhushi
- Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA
| | | | - Weijie Chen
- Division of Imaging Diagnostics and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiologic Health, United States Food and Drug Administration, White Oak, MD, USA
| | - Rajendra Singh
- Northwell Health and Zucker School of Medicine, New York, NY, USA
| | - Steven N Hart
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ashish Sharma
- Department of Biomedical Informatics, Emory University, Atlanta, GA, USA
| | - Joel Saltz
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | - Roberto Salgado
- Division of Research, Peter Mac Callum Cancer Centre, Melbourne, Australia.,Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
| | - Brandon D Gallas
- Division of Imaging Diagnostics and Software Reliability, Office of Science and Engineering Laboratories, Center for Devices and Radiologic Health, United States Food and Drug Administration, White Oak, MD, USA
| |
Collapse
|
35
|
El Bairi K, Haynes HR, Blackley E, Fineberg S, Shear J, Turner S, de Freitas JR, Sur D, Amendola LC, Gharib M, Kallala A, Arun I, Azmoudeh-Ardalan F, Fujimoto L, Sua LF, Liu SW, Lien HC, Kirtani P, Balancin M, El Attar H, Guleria P, Yang W, Shash E, Chen IC, Bautista V, Do Prado Moura JF, Rapoport BL, Castaneda C, Spengler E, Acosta-Haab G, Frahm I, Sanchez J, Castillo M, Bouchmaa N, Md Zin RR, Shui R, Onyuma T, Yang W, Husain Z, Willard-Gallo K, Coosemans A, Perez EA, Provenzano E, Ericsson PG, Richardet E, Mehrotra R, Sarancone S, Ehinger A, Rimm DL, Bartlett JMS, Viale G, Denkert C, Hida AI, Sotiriou C, Loibl S, Hewitt SM, Badve S, Symmans WF, Kim RS, Pruneri G, Goel S, Francis PA, Inurrigarro G, Yamaguchi R, Garcia-Rivello H, Horlings H, Afqir S, Salgado R, Adams S, Kok M, Dieci MV, Michiels S, Demaria S, Loi S. The tale of TILs in breast cancer: A report from The International Immuno-Oncology Biomarker Working Group. NPJ Breast Cancer 2021; 7:150. [PMID: 34853355 PMCID: PMC8636568 DOI: 10.1038/s41523-021-00346-1] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 09/28/2021] [Indexed: 02/08/2023] Open
Abstract
The advent of immune-checkpoint inhibitors (ICI) in modern oncology has significantly improved survival in several cancer settings. A subgroup of women with breast cancer (BC) has immunogenic infiltration of lymphocytes with expression of programmed death-ligand 1 (PD-L1). These patients may potentially benefit from ICI targeting the programmed death 1 (PD-1)/PD-L1 signaling axis. The use of tumor-infiltrating lymphocytes (TILs) as predictive and prognostic biomarkers has been under intense examination. Emerging data suggest that TILs are associated with response to both cytotoxic treatments and immunotherapy, particularly for patients with triple-negative BC. In this review from The International Immuno-Oncology Biomarker Working Group, we discuss (a) the biological understanding of TILs, (b) their analytical and clinical validity and efforts toward the clinical utility in BC, and (c) the current status of PD-L1 and TIL testing across different continents, including experiences from low-to-middle-income countries, incorporating also the view of a patient advocate. This information will help set the stage for future approaches to optimize the understanding and clinical utilization of TIL analysis in patients with BC.
Collapse
Affiliation(s)
- Khalid El Bairi
- Department of Medical Oncology, Mohammed VI University Hospital, Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco.
| | - Harry R Haynes
- Department of Cellular Pathology, Great Western Hospital, Swindon, UK
- Translational Health Sciences, University of Bristol, Bristol, UK
| | - Elizabeth Blackley
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Susan Fineberg
- Department of Pathology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jeffrey Shear
- Chief Information Officer, WISS & Company, LLP and President J. Shear Consulting, LLC-Ardsley, Ardsley, NY, USA
| | | | - Juliana Ribeiro de Freitas
- Department of Pathology and Legal Medicine, Medical School of the Federal University of Bahia, Salvador, Brazil
| | - Daniel Sur
- Department of Medical Oncology, University of Medicine "I. Hatieganu", Cluj Napoca, Romania
| | | | - Masoumeh Gharib
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Indu Arun
- Department of Histopathology, Tata Medical Center, Kolkata, India
| | - Farid Azmoudeh-Ardalan
- Department of Pathology, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Luciana Fujimoto
- Pathology and Legal Medicine, Amazon Federal University, Belém, Brazil
| | - Luz F Sua
- Department of Pathology and Laboratory Medicine, Fundacion Valle del Lili, and Faculty of Health Sciences, Universidad ICESI, Cali, Colombia
| | | | - Huang-Chun Lien
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Pawan Kirtani
- Department of Histopathology, Manipal Hospitals Dwarka, New Delhi, India
| | - Marcelo Balancin
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | | | - Prerna Guleria
- Army Hospital Research and Referral, Delhi Cantt, New Delhi, India
| | | | - Emad Shash
- Breast Cancer Comprehensive Center, National Cancer Institute, Cairo University, Cairo, Egypt
| | - I-Chun Chen
- Department of Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Veronica Bautista
- Department of Pathology, Breast Cancer Center FUCAM, Mexico City, Mexico
| | | | - Bernardo L Rapoport
- The Medical Oncology Centre of Rosebank, Johannesburg, South Africa
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, corner Doctor Savage Road and Bophelo Road, Pretoria, 0002, South Africa
| | - Carlos Castaneda
- Department of Medical Oncology, Instituto Nacional de Enfermedades Neoplásicas, Lima, 15038, Peru
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru
| | - Eunice Spengler
- Departmento de Patologia, Hospital Universitario Austral, Pilar, Argentina
| | - Gabriela Acosta-Haab
- Department of Pathology, Hospital de Oncología Maria Curie, Buenos Aires, Argentina
| | - Isabel Frahm
- Department of Pathology, Sanatorio Mater Dei, Buenos Aires, Argentina
| | - Joselyn Sanchez
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima, 15038, Peru
| | - Miluska Castillo
- Department of Research, Instituto Nacional de Enfermedades Neoplasicas, Lima, 15038, Peru
| | - Najat Bouchmaa
- Institute of Biological Sciences, Mohammed VI Polytechnic University (UM6P), 43 150, Ben-Guerir, Morocco
| | - Reena R Md Zin
- Department of Pathology, Faculty of Medicine, UKM Medical Centre, Kuala Lumpur, Malaysia
| | - Ruohong Shui
- Department of Pathology, Fudan University Cancer Center, Shanghai, China
| | | | - Wentao Yang
- Department of Pathology, Fudan University Cancer Center, Shanghai, China
| | | | - Karen Willard-Gallo
- Molecular Immunology Unit, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - An Coosemans
- Laboratory of Tumour Immunology and Immunotherapy, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Edith A Perez
- Department of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Elena Provenzano
- Department of Histopathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Paula Gonzalez Ericsson
- Breast Cancer Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eduardo Richardet
- Clinical Oncology Unit, Instituto Oncológico Córdoba, Córdoba, Argentina
| | - Ravi Mehrotra
- India Cancer Research Consortium-ICMR, Department of Health Research, New Delhi, India
| | - Sandra Sarancone
- Department of Pathology, Laboratorio QUANTUM, Rosario, Argentina
| | - Anna Ehinger
- Department of Clinical Genetics and Pathology, Skåne University Hospital, Lund University, Lund, Sweden
| | - David L Rimm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - John M S Bartlett
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, Canada
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Giuseppe Viale
- Department of Pathology, Istituto Europeo di Oncologia IRCCS, and University of Milan, Milan, Italy
| | - Carsten Denkert
- Institute of Pathology, Universitätsklinikum Gießen und Marburg GmbH, Standort Marburg and Philipps-Universität Marburg, Marburg, Germany
| | - Akira I Hida
- Department of Pathology, Matsuyama Shimin Hospital, Matsuyama, Japan
| | - Christos Sotiriou
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Stephen M Hewitt
- Laboratory of Pathology, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Indiana University, Indianapolis, USA
| | - William Fraser Symmans
- Department of Pathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Rim S Kim
- National Surgical Adjuvant Breast and Bowel Project (NSABP)/NRG Oncology, Pittsburgh, PA, USA
| | - Giancarlo Pruneri
- Department of Pathology, RCCS Fondazione Istituto Nazionale Tumori and University of Milan, School of Medicine, Milan, Italy
| | - Shom Goel
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Prudence A Francis
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
- Medical Oncology Department, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Rin Yamaguchi
- Department of Pathology and Laboratory Medicine, Kurume University Medical Center, Kurume, Fukuoka, Japan
| | - Hernan Garcia-Rivello
- Servicio de Anatomía Patológica, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Hugo Horlings
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Said Afqir
- Department of Medical Oncology, Mohammed VI University Hospital, Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
| | - Roberto Salgado
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
| | - Sylvia Adams
- Perlmutter Cancer Center, New York University Medical School, New York, NY, USA
| | - Marleen Kok
- Divisions of Medical Oncology, Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maria Vittoria Dieci
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Stefan Michiels
- Service de Biostatistique et d'Epidémiologie, Gustave Roussy, Oncostat U1018, Inserm, University Paris-Saclay, labeled Ligue Contre le Cancer, Villejuif, France
| | - Sandra Demaria
- Department of Radiation Oncology, Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Sherene Loi
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
36
|
Atakpa EC, Thorat MA, Cuzick J, Brentnall AR. Mammographic density, endocrine therapy and breast cancer risk: a prognostic and predictive biomarker review. Cochrane Database Syst Rev 2021; 10:CD013091. [PMID: 34697802 PMCID: PMC8545623 DOI: 10.1002/14651858.cd013091.pub2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Endocrine therapy is effective at preventing or treating breast cancer. Some forms of endocrine therapy have been shown to reduce mammographic density. Reduced mammographic density for women receiving endocrine therapy could be used to estimate the chance of breast cancer returning or developing breast cancer in the first instance (a prognostic biomarker). In addition, changes in mammographic density might be able to predict how well a woman responds to endocrine therapy (a predictive biomarker). The role of breast density as a prognostic or predictive biomarker could help improve the management of breast cancer. OBJECTIVES To assess the evidence that a reduction in mammographic density following endocrine therapy for breast cancer prevention in women without previous breast cancer, or for treatment in women with early-stage hormone receptor-positive breast cancer, is a prognostic or predictive biomarker. SEARCH METHODS We searched the Cochrane Breast Cancer Group Specialised Register, CENTRAL, MEDLINE, Embase, and two trials registers on 3 August 2020 along with reference checking, bibliographic searching, and contact with study authors to obtain further data. SELECTION CRITERIA We included randomised, cohort and case-control studies of adult women with or without breast cancer receiving endocrine therapy. Endocrine therapy agents included were selective oestrogen receptor modulators and aromatase inhibitors. We required breast density before start of endocrine therapy and at follow-up. We included studies published in English. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. Two review authors independently extracted data and assessed risk of bias using adapted Quality in Prognostic Studies (QUIPS) and Risk Of Bias In Non-randomised Studies - of Interventions (ROBINS-I) tools. We used the GRADE approach to evaluate the certainty of the evidence. We did not perform a quantitative meta-analysis due to substantial heterogeneity across studies. MAIN RESULTS Eight studies met our inclusion criteria, of which seven provided data on outcomes listed in the protocol (5786 women). There was substantial heterogeneity across studies in design, sample size (349 to 1066 women), participant characteristics, follow-up (5 to 14 years), and endocrine therapy agent. There were five breast density measures and six density change definitions. All studies had at least one domain as at moderate or high risk of bias. Common concerns were whether the study sample reflected the review target population, and likely post hoc definitions of breast density change. Most studies on prognosis for women receiving endocrine therapy reported a reduced risk associated with breast density reduction. Across endpoints, settings, and agents, risk ratio point estimates (most likely value) were between 0.1 and 1.5, but with substantial uncertainty. There was greatest consistency in the direction and magnitude of the effect for tamoxifen (across endpoints and settings, risk ratio point estimates were between 0.3 and 0.7). The findings are summarised as follows. Prognostic biomarker findings: Treatment Breast cancer mortality Two studies of 823 women on tamoxifen (172 breast cancer deaths) reported risk ratio point estimates of ~0.4 and ~0.5 associated with a density reduction. The certainty of the evidence was low. Recurrence Two studies of 1956 women on tamoxifen reported risk ratio point estimates of ~0.4 and ~0.7 associated with a density reduction. There was risk of bias in methodology for design and analysis of the studies and considerable uncertainty over the size of the effect. One study of 175 women receiving an aromatase inhibitor reported a risk ratio point estimate of ~0.1 associated with a density reduction. There was considerable uncertainty about the effect size and a moderate or high risk of bias in all domains. One study of 284 women receiving exemestane or tamoxifen as part of a randomised controlled trial reported risk ratio point estimates of ~1.5 (loco-regional recurrence) and ~1.3 (distance recurrence) associated with a density reduction. There was risk of bias in reporting and study confounding, and uncertainty over the size of the effects. The certainty of the evidence for all recurrence endpoints was very low. Incidence of a secondary primary breast cancer Two studies of 451 women on exemestane, tamoxifen, or unknown endocrine therapy reported risk ratio point estimates of ~0.5 and ~0.6 associated with a density reduction. There was risk of bias in reporting and study confounding, and uncertainty over the effect size. The certainty of the evidence was very low. We were unable to find data regarding the remaining nine outcomes prespecified in the review protocol. Prevention Incidence of invasive breast cancer and ductal carcinoma in situ (DCIS) One study of 507 women without breast cancer who were receiving preventive tamoxifen as part of a randomised controlled trial (51 subsequent breast cancers) reported a risk ratio point estimate of ~0.3 associated with a density reduction. The certainty of the evidence was low. Predictive biomarker findings: One study of a subset of 1065 women from a randomised controlled trial assessed how much the effect of endocrine therapy could be explained by breast density declines in those receiving endocrine therapy. This study evaluated the prevention of invasive breast cancer and DCIS. We found some evidence to support the hypothesis, with a risk ratio interaction point estimate ~0.5. However, the 95% confidence interval included unity, and data were based on 51 women with subsequent breast cancer in the tamoxifen group. The certainty of the evidence was low. AUTHORS' CONCLUSIONS There is low-/very low-certainty evidence to support the hypothesis that breast density change following endocrine therapy is a prognostic biomarker for treatment or prevention. Studies suggested a potentially large effect size with tamoxifen, but the evidence was limited. There was less evidence that breast density change following tamoxifen preventive therapy is a predictive biomarker than prognostic biomarker. Evidence for breast density change as a prognostic treatment biomarker was stronger for tamoxifen than aromatase inhibitors. There were no studies reporting mammographic density change following endocrine therapy as a predictive biomarker in the treatment setting, nor aromatase inhibitor therapy as a prognostic or predictive biomarker in the preventive setting. Further research is warranted to assess mammographic density as a biomarker for all classes of endocrine therapy and review endpoints.
Collapse
Affiliation(s)
- Emma C Atakpa
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mangesh A Thorat
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Breast Services, Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jack Cuzick
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Adam R Brentnall
- Wolfson Institute of Population Health, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
37
|
Van Den Bosch T, van Dijk E, Vermeulen L, Miedema DM. Predicting survival of cancer patients by chromosomal copy number heterogeneity. Mol Cell Oncol 2021; 8:1949956. [PMID: 34616875 PMCID: PMC8489936 DOI: 10.1080/23723556.2021.1949956] [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] [Indexed: 10/24/2022]
Abstract
We recently introduced a method to derive intra-tumor heterogeneity (ITH) from a single copy number measurement. This method stratifies patients for survival and could potentially help to identify low and high-risk patients with clinical relevance.
Collapse
Affiliation(s)
- Tom Van Den Bosch
- LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| | - Erik van Dijk
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Louis Vermeulen
- LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| | - Daniël M Miedema
- LEXOR, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology & Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
38
|
Okuda Y, Shimura T, Iwasaki H, Katano T, Kitagawa M, Nishigaki R, Fukusada S, Natsume M, Tanaka M, Nishie H, Ozeki K, Yamada T, Kataoka H. Serum Exosomal Dicer Is a Useful Biomarker for Early Detection of Differentiated Gastric Adenocarcinoma. Digestion 2021; 102:640-649. [PMID: 33049740 DOI: 10.1159/000510993] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/16/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM A recent basic study identified that Dicer is contained in exosomes derived from cancer cells and plays crucial roles in microRNA maturation and cancer development. Based on this novel basic concept, we analyzed the usefulness of serum exosomal Dicer as a diagnostic biomarker for gastrointestinal cancers. METHODS Enrolled participants (691) were categorized into 3 groups: gastric cancer (GC) cohort, 183 patients (90 healthy controls (HCs) and 93 GC patients); esophageal cancer (EC) cohort, 115 patients (90 HCs and 25 EC patients); and colorectal cancer (CRC) cohort, 188 patients (92 HCs and 96 CRC patients) after age- and sex matching using the propensity score. The quality of isolated serum exosomes was validated with an electron microscope, particle size analyzer, and exosome marker, CD63. RESULTS Serum exosomal Dicer was significantly higher in the GC group than in the HC group (p = 0.004), whereas no significant differences were found in both EC and CRC cohorts. Serum exosomal Dicer was significantly higher in only differentiated gastric adenocarcinoma and not in the undifferentiated type. Moreover, serum exosomal Dicer showed no significant differences regardless of Helicobacter pylori (H. pylori) status. The biomarker panel combining serum exosomal Dicer with H. pylori status distinguished between HC and differentiated GC patients with an area under the curve (AUC) of 0.762. As for early-stage diagnosis, this combination distinguished between HC and stage I differentiated GC with an AUC = 0.758. CONCLUSIONS Serum exosomal Dicer is a potential noninvasive diagnostic biomarker for early detection of differentiated gastric adenocarcinoma.
Collapse
Affiliation(s)
- Yusuke Okuda
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan,
| | - Hiroyasu Iwasaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahito Katano
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mika Kitagawa
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ruriko Nishigaki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shigeki Fukusada
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Makoto Natsume
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mamoru Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hirotada Nishie
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Keiji Ozeki
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | | | - Hiromi Kataoka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
39
|
Filipits M, Rudas M, Kainz V, Singer CF, Fitzal F, Bago-Horvath Z, Greil R, Balic M, Regitnig P, Halper S, Hulla W, Egle D, Barron S, Loughman T, O'Leary D, Gallagher WM, Hlauschek D, Gnant M, Dubsky P. The OncoMasTR test predicts distant recurrence in estrogen receptor-positive, HER2-negative early-stage breast cancer: A validation study in ABCSG Trial 8. Clin Cancer Res 2021; 27:5931-5938. [PMID: 34380638 DOI: 10.1158/1078-0432.ccr-21-1023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/17/2021] [Accepted: 08/05/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE To validate the clinical performance of the OncoMasTR Risk Score in the biomarker cohort of ABCSG Trial 8. EXPERIMENTAL DESIGN We evaluated the OncoMasTR test in 1200 formalin-fixed, paraffin-embedded surgical specimens from postmenopausal women with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative primary breast cancer with 0-3 involved lymph nodes in the prospective, randomized ABCSG Trial 8. Time to distant recurrence (DR) was analyzed by Cox models. RESULTS The OncoMasTR Risk Score categorized 850 of 1087 (78.2%) evaluable patients as "low risk". At 10 years, the DR rate for patients in the low-risk group was 5.8% versus 21.1% for patients in the high-risk group (P<0.0001, absolute risk reduction 15.3%). The OncoMasTR Risk Score was highly prognostic for prediction of DR in years 0-10 in all patients (hazard ratio (HR) 1.91, 95% confidence interval (CI) 1.62 to 2.26, P<0.0001; C-index 0.73), in node-negative patients (HR 1.79, 95% CI 1.43 to 2.24, P<0.0001; C-index 0.72), and in patients with 1-3 involved lymph nodes (HR 1.93, 95% CI 1.44 to 2.58, P<0.0001; C-index 0.71). The OncoMasTR Risk Score provided significant additional prognostic information beyond clinical parameters, Ki67, Nottingham Prognostic Index, and Clinical Treatment Score. CONCLUSIONS OncoMasTR Risk Score is highly prognostic for DR in postmenopausal women with ER-positive, HER2-negative primary breast cancer with 0-3 involved lymph nodes. In combination with prior validation studies, this fully independent validation in ABCSG Trial 8 provides level 1B evidence for the prognostic capability of the OncoMasTR Risk Score.
Collapse
Affiliation(s)
- Martin Filipits
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna
| | | | - Verena Kainz
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna
| | - Christian F Singer
- Department of OB/GYN and Comprehensive Cancer Center, Medical University of Vienna
| | | | | | - Richard Greil
- Department of Internal Medicine III, Salzburg Cancer Research Institute - Laboratory for Immunological and Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University Salzburg
| | | | - Peter Regitnig
- Diagnostic- and Researchinstitute of Pathology, Medical University of Graz
| | - Stefan Halper
- Department of Surgery, Breast Health Center, Hospital Wiener Neustadt
| | - Wolfgang Hulla
- Department of Clinical Pathology and Molecular Pathology, Hospital Wiener Neustadt
| | - Daniel Egle
- Department of Obstetrics and Gynecology, Medical University of Innsbruck
| | | | | | | | | | | | - Michael Gnant
- Comprehensive Cancer Center, Medical University of Vienna
| | | |
Collapse
|
40
|
Mismatch Repair Deficiency as a Predictive and Prognostic Biomarker in Molecularly Classified Endometrial Carcinoma. Cancers (Basel) 2021; 13:cancers13133124. [PMID: 34206702 PMCID: PMC8268938 DOI: 10.3390/cancers13133124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary We studied mismatch repair (MMR) deficiency as a predictive and prognostic biomarker in endometrial carcinoma. MMR deficiency was associated with poor outcome only when p53 aberrant and polymerase-ϵ mutant tumors were excluded from the MMR proficient subgroup, in accordance with molecular classification based on The Cancer Genome Atlas. MMR deficiency was associated with an increased risk of death in the absence of various clinicopathologic risk factors, but the outcome was not worsened when such risk factors were present. The proportion of pelvic relapses and lymphatic dissemination, defined as primary lymph node involvement or relapses in regional lymph nodes, were higher in the MMR deficient subgroup. In conclusion, the effect of MMR deficiency on the outcome of endometrial carcinoma depends on how MMR proficiency is defined. MMR deficiency is associated with an increased risk of death in the absence of established risk factors and a unique pattern of disease spread. Abstract The aggressiveness of mismatch repair (MMR) deficient endometrial carcinomas was examined in a single institution retrospective study. Outcomes were similar for MMR proficient (n = 508) and deficient (n = 287) carcinomas, identified by immunohistochemistry. In accordance with molecular classification based on The Cancer Genome Atlas (TCGA), tumors with abnormal p53 staining or polymerase-ϵ exonuclease domain mutation were excluded from the MMR proficient subgroup, termed as “no specific molecular profile” (NSMP). Compared with NSMP (n = 218), MMR deficiency (n = 191) was associated with poor disease-specific survival (p = 0.001). MMR deficiency was associated with an increased risk of cancer-related death when controlling for confounders (hazard ratio 2.0). In the absence of established clinicopathologic risk factors, MMR deficiency was invariably associated with an increased risk of cancer-related death in univariable analyses (hazard ratios ≥ 2.0). In contrast, outcomes for MMR deficient and NSMP subgroups did not differ when risk factors were present. Lymphatic dissemination was more common (p = 0.008) and the proportion of pelvic relapses was higher (p = 0.029) in the MMR deficient subgroup. Our findings emphasize the need for improved triage to adjuvant therapy and new therapeutic approaches in MMR deficient endometrial carcinomas.
Collapse
|
41
|
Thomopoulou K, Papadaki C, Monastirioti A, Koronakis G, Mala A, Kalapanida D, Mavroudis D, Agelaki S. MicroRNAs Regulating Tumor Immune Response in the Prediction of the Outcome in Patients With Breast Cancer. Front Mol Biosci 2021; 8:668534. [PMID: 34179081 PMCID: PMC8220200 DOI: 10.3389/fmolb.2021.668534] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/27/2021] [Indexed: 12/29/2022] Open
Abstract
MicroRNAs (miRNAs) are key regulators in immune surveillance and immune escape as well as modulators in the metastatic process of breast cancer cells. We evaluated the differential expression of plasma miR-10b, miR-19a, miR-20a, miR-126 and miR-155, which regulate immune response in breast cancer progression and we investigated their clinical relevance in the outcomes of breast cancer patients. Plasma samples were obtained from early (eBC; n = 140) and metastatic (mBC; n = 64) breast cancer patients before adjuvant or first-line chemotherapy, respectively. Plasma miRNA expression levels were assessed by qRT-PCR. We revealed a 4-miRNA panel consisted of miR-19a, miR-20a, miR-126, and miR-155 able to discriminate eBC from mBC patients with an AUC of 0.802 (p < 0.001). Survival analysis in eBC patients revealed that low miR-10b and miR-155 expression was associated with shorter disease free survival (disease free survival; p = 0.012 and p = 0.04, respectively) compared to high expression. Furthermore, miR-126 expression was associated with shorter overall survival (overall survival; p = 0.045). In multivariate analysis the number of infiltrated axillary lymph nodes and low miR-10b expression independently predicted for shorter DFS (HR: 2.538; p = 0.002 and HR: 1.943; p = 0.033, respectively) and axillary lymph nodes and low miR-126 for shorter OS (HR: 3.537; p = 0.001 and HR: 2.558; p = 0.018). In the subgroup of triple negative breast cancer (TNBC) patients, low miR-155 expression independently predicted for shorter DFS (HR: 5.056; p = 0.037). Accordingly in mBC, patients with low miR-10b expression had shorter progression free survival and OS compared to patients with high expression (p = 0.0017 and p = 0.042, respectively). In multivariate analysis, recurrent disease and low miR-10b expression independently predicted for shorter PFS (HR: 2.657; p = 0.001 and HR: 1.920; p = 0.017, respectively), whereas performance status two independently predicted for shorter OS (HR: 2.031; p = 0.03). In summary, deregulated expression of circulating miRNAs involved in tumor and immune cell interactions evaluated before adjuvant and 1st-line chemotherapy can distinguish disease status and emerge as independent predictors for outcomes of breast cancer patients.
Collapse
Affiliation(s)
- Konstantina Thomopoulou
- Department of Medical Oncology, University General Hospital, Crete, Heraklion, Greece
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
| | - Chara Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
| | - Alexia Monastirioti
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
| | - George Koronakis
- Department of Medical Oncology, University General Hospital, Crete, Heraklion, Greece
| | - Anastasia Mala
- Department of Medical Oncology, University General Hospital, Crete, Heraklion, Greece
| | - Despoina Kalapanida
- Department of Medical Oncology, University General Hospital, Crete, Heraklion, Greece
| | - Dimitrios Mavroudis
- Department of Medical Oncology, University General Hospital, Crete, Heraklion, Greece
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
| | - Sofia Agelaki
- Department of Medical Oncology, University General Hospital, Crete, Heraklion, Greece
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Greece
| |
Collapse
|
42
|
Ortiz-Romero PL. The time for new biomarkers in mycosis fungoides/Sézary syndrome is here. Br J Dermatol 2021; 185:250-251. [PMID: 34096050 DOI: 10.1111/bjd.20491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 11/29/2022]
Affiliation(s)
- P L Ortiz-Romero
- Service of Dermatology, Hospital 12 de Octubre. Institute i+12, CIBERONC, Medical School, University Complutense, Madrid, Spain
| |
Collapse
|
43
|
Sanz-Álvarez M, Martín-Aparicio E, Luque M, Zazo S, Martínez-Useros J, Eroles P, Rovira A, Albanell J, Madoz-Gúrpide J, Rojo F. The Novel Oral mTORC1/2 Inhibitor TAK-228 Reverses Trastuzumab Resistance in HER2-Positive Breast Cancer Models. Cancers (Basel) 2021; 13:cancers13112778. [PMID: 34204960 PMCID: PMC8199905 DOI: 10.3390/cancers13112778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Hyperactivation of the PI3K/AKT/mTOR cell signalling pathway is an important and well-described mechanism of trastuzumab resistance in HER2-positive breast cancer. In cell-line models of acquired trastuzumab resistance generated in our laboratory, we demonstrate this type of activation, which is independent of HER2-mediated regulation. We investigate whether the use of specific mTOR inhibitors, a PI3K/AKT/mTOR pathway effector, could lead to decreased activity of the pathway, influencing trastuzumab resistance. We demonstrate that TAK-228, a mTORC1 and mTORC2 inhibitor, can reverse resistance and increasing response to trastuzumab in models of primary and acquired resistance. Abstract The use of anti-HER2 therapies has significantly improved clinical outcome in patients with HER2-positive breast cancer, yet a substantial proportion of patients acquire resistance after a period of treatment. The PI3K/AKT/mTOR pathway is a good target for drug development, due to its involvement in HER2-mediated signalling and in the emergence of resistance to anti-HER2 therapies, such as trastuzumab. This study evaluates the activity of three different PI3K/AKT/mTOR inhibitors, i.e., BEZ235, everolimus and TAK-228 in vitro, in a panel of HER2-positive breast cancer cell lines with primary and acquired resistance to trastuzumab. We assess the antiproliferative effect and PI3K/AKT/mTOR inhibitory capability of BEZ235, everolimus and TAK-228 alone, and in combination with trastuzumab. Dual blockade with trastuzumab and TAK-228 was superior in reversing the acquired resistance in all the cell lines. Subsequently, we analyse the effects of TAK-228 in combination with trastuzumab on the cell cycle and found a significant increase in G0/G1 arrest in most cell lines. Likewise, the combination of both drugs induced a significant increase in apoptosis. Collectively, these experiments support the combination of trastuzumab with PI3K/AKT/mTOR inhibitors as a potential strategy for inhibiting the proliferation of HER2-positive breast cancer cell lines that show resistance to trastuzumab.
Collapse
Affiliation(s)
- Marta Sanz-Álvarez
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Ester Martín-Aparicio
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Melani Luque
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Sandra Zazo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
| | - Javier Martínez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-Fundación Jiménez Díaz (IIS-FJD, UAM), 28040 Madrid, Spain;
| | - Pilar Eroles
- Institute of Health Research INCLIVA-CIBERONC, 46010 Valencia, Spain;
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain; (A.R.); (J.A.)
- Medical Oncology Department, Hospital del Mar-CIBERONC, 08003 Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Research Institute), 08003 Barcelona, Spain; (A.R.); (J.A.)
- Medical Oncology Department, Hospital del Mar-CIBERONC, 08003 Barcelona, Spain
- Department of Experimental and Health Sciences, Faculty of Medicine, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Juan Madoz-Gúrpide
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
- Correspondence: (J.M.-G.); (F.R.); Tel.: +34-915-504-800 (J.M.-G.); +34-915-504-800 (F.R.)
| | - Federico Rojo
- Department of Pathology, Fundación Jiménez Díaz University Hospital Health Research Institute (IIS—FJD, UAM)—CIBERONC, 28040 Madrid, Spain; (M.S.-Á.); (E.M.-A.); (M.L.); (S.Z.)
- Correspondence: (J.M.-G.); (F.R.); Tel.: +34-915-504-800 (J.M.-G.); +34-915-504-800 (F.R.)
| |
Collapse
|
44
|
Lee JY, Kim JH, Bang H, Cho J, Ko YH, Kim SJ, Kim WS. EGR1 as a potential marker of prognosis in extranodal NK/T-cell lymphoma. Sci Rep 2021; 11:10342. [PMID: 33990633 PMCID: PMC8121831 DOI: 10.1038/s41598-021-89754-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
Extranodal natural killer T-cell lymphoma (ENKTL) is an aggressive malignancy with a dismal prognosis. In the present study, gene expression profiling was performed to provide more information on ENKTL molecular signature and offer a rationale for further investigation of prognostic markers in ENKTL. NanoString nCounter Analysis encompassing 133 target genes was used to compare gene expression levels of 43 ENKTL tumor samples. The majority of the patients were under 60 years of age (79.1%); 32 (74.4%) patients had nasal type ENKTL and 23 patients (53.5%) had intermediate/high risk ENKTL based on the prognostic index for natural killer cell lymphoma (PINK). The median follow-up was 15.9 months and the median overall survival (OS) was 16.1 months (95% CI 13.0-69.8). EGR1 upregulation was consistently identified in the localized stage with a low risk of prognostic index based on the PINK. Among the six significantly relevant genes for EGR1 expression, high expression levels of genes, including CD59, GAS1, CXCR7, and RAMP3, were associated with a good survival prognosis. The in vitro test showed EGR1 modulated the transcriptional activity of the target genes including CD59, GAS1, CXCR7, and RAMP3. Downregulation of EGR1 and its target genes significantly inhibited apoptosis and decreased chemosensitivity and attenuated radiation-induced apoptosis. The findings showed EGR1 may be a candidate for prognostic markers in ENKTL. Considerable additional characterization may be necessary to fully understand EGR1.
Collapse
Affiliation(s)
- Ji Yun Lee
- Division of Hematology-Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Joo Hyun Kim
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Heejin Bang
- Department of Pathology, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Republic of Korea
| | - Junhun Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Hyeh Ko
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Won Seog Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
45
|
Urinary microRNA biomarkers for detecting the presence of esophageal cancer. Sci Rep 2021; 11:8508. [PMID: 33879806 PMCID: PMC8058072 DOI: 10.1038/s41598-021-87925-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/05/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal cancer (EC) including esophageal squamous cell carcinoma (ESCC) and adenocarcinoma (EAC) generally exhibits poor prognosis; hence, a noninvasive biomarker enabling early detection is necessary. Age- and sex-matched 150 healthy controls (HCs) and 43 patients with ESCC were randomly divided into two groups: 9 individuals in the discovery cohort for microarray analysis and 184 individuals in the training/test cohort with cross-validation for qRT-PCR analysis. Using 152 urine samples (144 HCs and 8 EACs), we validated the urinary miRNA biomarkers for EAC diagnosis. Among eight miRNAs selected in the discovery cohort, urinary levels of five miRNAs (miR-1273f, miR-619-5p, miR-150-3p, miR-4327, and miR-3135b) were significantly higher in the ESCC group than in the HC group, in the training/test cohort. Consistently, these five urinary miRNAs were significantly different between HC and ESCC in both training and test sets. Especially, urinary miR-1273f and miR-619-5p showed excellent values of area under the curve (AUC) ≥ 0.80 for diagnosing stage I ESCC. Similarly, the EAC group had significantly higher urinary levels of these five miRNAs than the HC group, with AUC values of approximately 0.80. The present study established novel urinary miRNA biomarkers that can early detect ESCC and EAC.
Collapse
|
46
|
Effects of Epstein-Barr Virus Infection on the Risk and Prognosis of Primary Laryngeal Squamous Cell Carcinoma: A Hospital-Based Case-Control Study in Taiwan. Cancers (Basel) 2021; 13:cancers13071741. [PMID: 33917480 PMCID: PMC8038767 DOI: 10.3390/cancers13071741] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
Mounting molecular evidence supports Epstein-Barr virus (EBV) involvement in the pathogenesis of laryngeal squamous cell carcinoma (LSCC); however, the epidemiological data are inconsistent. In this retrospective case-control study, we aimed to determine whether EBV infection underlies the risk and prognosis of LSCC. The prevalence of EBV infection, as analyzed using an EBV DNA polymerase chain reaction assay, was significantly higher in 42 Taiwanese patients with newly diagnosed primary LSCC, compared to 39 age- and sex-matched control patients without cancer (48% vs. 19%). Furthermore, most of the EBER signals detected using in situ hybridization were localized to the nuclei of tumor-infiltrating lymphocytes. In multivariate analysis, EBV DNA positivity, age ≥ 55 years, cigarette smoking, and high BCL-2, B2M, and CD161 expression (assessed using immunohistochemistry) were identified as independent risk factors for LSCC. Furthermore, five-year local recurrence and disease-free survival rates were 34% and 58%, respectively, with a high EBER signal and low CD3 expression independently predicting five-year local recurrence and disease-free survival. Our comprehensive profiling data accurately identified patients at risk for LSCC development, local recurrence, or disease-free survival. The information obtained in this study improves our understanding of EBV infection in LSCC, and may guide precision medicine for patients with LSCC.
Collapse
|
47
|
Harada K, Hwang H, Wang X, Abdelhakeem A, Iwatsuki M, Blum Murphy MA, Maru DM, Weston B, Lee JH, Rogers JE, Thomas I, Shanbhag N, Zhao M, Bhutani MS, Nguyen QN, Swisher SG, Ikoma N, Badgwell BD, Hofstetter WL, Ajani JA. Frequency and Implications of Paratracheal Lymph Node Metastases in Resectable Esophageal or Gastroesophageal Junction Adenocarcinoma. Ann Surg 2021; 273:751-757. [PMID: 31188215 DOI: 10.1097/sla.0000000000003383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE We aimed to evaluate the frequency of paratracheal lymph nodes (LN) metastases and their prognostic influence. SUMMARY BACKGROUND DATA Paratracheal LNs are considered regional nodes in the esophageal cancer classification, but their metastatic rate and influence on survival remain unclear. METHODS One thousand one hundred ninety-nine patients with resectable esophageal or gastroesophageal junction adenocarcinoma (EAC) (January 2002 and December 2016) in our Gastrointestinal Medical Oncology Database were analyzed. Paratracheal LNs were defined as1R, 1L, 2R, 2L, 4R, and 4L, according to the 8th American Joint Committee on Cancer classification. RESULTS Of 1199 patients, 73 (6.1%) had positive paratracheal LNs at diagnosis. The median overall survival (OS) of 73 patients with initial paratracheal LN involvement was 2.10 years (range 0.01-10.1, 5-yrs OS 24.2%). Of 1071 patients who were eligible for recurrence evaluation, 70 patients (6.5%) developed paratracheal LN metastases as the first recurrence. The median time to recurrence was 1.28 years (range 0.28-5.96 yrs) and the median OS following recurrence was only 0.95 year (range 0.03-7.88). OS in 35 patients who had only paratracheal LN recurrence was significantly longer than in patients who had other recurrences (median OS 2.26 vs 0.51 yrs, 5-yrs OS; 26.8% vs 0%, P < 0.0001). Higher T stage (T3/T4) was an independently risk factor for paratracheal LN recurrence (odds ratio 5.10, 95% confidence interval 1.46-17.89). We segregated patients in 3 groups based on the distance of tumor's proximal edge to esophagogastric junction (low; ≤2 cm, medium; 2.0-7.0 cm, and high; >7.0 cm). Paratracheal LN metastases were more frequent with the proximal tumors (low, 4.2%; medium, 12.0%; high, 30.3%; Cochran-Armitage Trend test, P < 0.001). CONCLUSION Paratracheal LN metastases were associated with a shorter survival in resectable EAC patients. Alternate approaches to prolong survival of this group of patients are warranted.
Collapse
Affiliation(s)
- Kazuto Harada
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Hyunsoo Hwang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ahmed Abdelhakeem
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Masaaki Iwatsuki
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Mariela A Blum Murphy
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dipen M Maru
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brian Weston
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jeffrey H Lee
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jane E Rogers
- Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Irene Thomas
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Namita Shanbhag
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Meina Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Manoop S Bhutani
- Department of Gastroenterology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
48
|
Wang C, Hong T, Wang Y, Peng G, Yu Y, Zhang J, Zhuo D, Zheng J, Ma X, Cui X. Combining UBR5 and CD163 + tumor-associated macrophages better predicts prognosis of clear cell renal cell carcinoma patients. Cancer Immunol Immunother 2021; 70:2925-2935. [PMID: 33710368 DOI: 10.1007/s00262-021-02885-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/03/2021] [Indexed: 01/23/2023]
Abstract
PURPOSE Identification of reliable postoperative indicators for accurately evaluating prognosis of clear cell renal cell carcinoma (ccRCC) patients remains an important clinical issue. This study determined the prognostic value of UBR5 expression in ccRCC patients by combining with CD163+ tumor-associated macrophages (TAMs) and the established clinical parameters. METHODS The expression of UBR5 was analyzed in ccRCC patients from TCGA databases. A total of 310 ccRCC patients were randomly divided into the training and validation cohorts at a 3:2 or 1:1 ratio, and immunohistochemistry (IHC) and statistical analyses were performed to examine the prognostic value of UBR5 and CD163+ TAMs. RESULTS UBR5 expression was commonly downregulated in human ccRCC specimens, which was associated with TNM stage, SSIGN, WHO/ISUP Grading and poor prognosis of ccRCC patients. In addition, UBR5 expression was negatively correlated with CD163 expression (a TAM marker) in ccRCC tissues, and combining expressions of UBR5 and CD163 better predicted worse overall survival and progression-free survival of ccRCC patients. Even after multivariable adjustment, UBR5, CD163, TNM stage and SSIGN appeared to be independent risk factors. By time-dependent c-index analysis, the integration of intratumoral UBR5 and CD163 achieved higher c-index value than UBR5, CD163, TNM stage or SSIGN alone in predicting ccRCC patients' prognosis. Moreover, the incorporation of both UBR5 and CD163 into the clinical indicators TNM stage or SSIGN exhibited highest c-index value. CONCLUSIONS Integrating intratumoral UBR5 and CD163+ TAMs with the current clinical parameters achieves better accuracy in predicting ccRCC patients' postoperative prognosis.
Collapse
Affiliation(s)
- Chao Wang
- Department of Urinary Surgery, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China
- Shanghai Heath Commission Key Lab of Artificial Intelligence (AI)-Based Management of Inflammation and Chronic Diseases, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China
- Department of Urology, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, 29 Xinglong Road, Changzhou, 213000, Jiangsu, China
| | - TianYu Hong
- Department of Urinary Surgery, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China
| | - Yuning Wang
- Department of Urinary Surgery, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China
| | - Guang Peng
- Department of Urinary Surgery, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), 700 North Moyu Road, Shanghai, 201805, China
- Department of Urology, Joint Logistic Support Force No. 925 Hospital of PLA, 67 Yellow River Road, Guiyang, 550009, Guizhou, China
- Department of Orthopedic, Joint Logistic Support Force No. 925 Hospital of PLA, 67 Yellow River Road, Guiyang, 550009, Guizhou, China
| | - Yongwei Yu
- Department of Pathology, Changhai Hospital, Second Military Medical University (Naval Medical University), 168 Changhai Road, Shanghai, 200438, China
| | - Jing Zhang
- Department of Urinary Surgery, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China
| | - Dong Zhuo
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, 1300 York Avenue, New York, NY, 10065-4805, USA.
| | - Jingcun Zheng
- Department of Urinary Surgery, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China.
| | - Xiaojing Ma
- Department of Urology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), No. 2 West Zheshan RD, Wuhu, Anhui, 241000, China.
| | - Xingang Cui
- Department of Urinary Surgery, Gongli Hospital, The Second Military Medical University (Naval Medical University), 219 Miaopu Road, Shanghai, 200135, China.
- Department of Urinary Surgery, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), 700 North Moyu Road, Shanghai, 201805, China.
| |
Collapse
|
49
|
Expression of Phosphorylated BRD4 Is Markedly Associated with the Activation Status of the PP2A Pathway and Shows a Strong Prognostic Value in Triple Negative Breast Cancer Patients. Cancers (Basel) 2021; 13:cancers13061246. [PMID: 33809005 PMCID: PMC7999847 DOI: 10.3390/cancers13061246] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary The use of BRD4 inhibitors has emerged as a novel therapeutic approach in a wide variety of tumors including the triple negative breast cancer. Moreover, PP2A has been proposed as the phosphatase involved in regulating BRD4 phosphorylation and stabilization. Our aim was to evaluate for the first time the clinical impact of BRD4 phosphorylation in triple negative breast cancer patients and as well as its potential linking with the PP2A activation status in this disease. Our findings are special relevant since they suggest the prognostic value of BRD4 phosphorylation levels, and the potential clinical usefulness of PP2A inhibition markers to anticipate response to BRD4 inhibitors. Abstract The bromodomain-containing protein 4 (BRD4), a member of the bromodomain and extra-terminal domain (BET) protein family, has emerged in the last years as a promising molecular target in many tumors including breast cancer. The triple negative breast cancer (TNBC) represents the molecular subtype with the worst prognosis and a current therapeutic challenge, and TNBC cells have been reported to show a preferential sensitivity to BET inhibitors. Interestingly, BRD4 phosphorylation (pBRD4) was found as an alteration that confers resistance to BET inhibition and PP2A proposed as the phosphatase responsible to regulate pBRD4 levels. However, the potential clinical significance of pBRD4, as well as its potential correlation with the PP2A pathway in TNBC, remains to be investigated. Here, we evaluated the expression levels of pBRD4 in a series of 132 TNBC patients. We found high pBRD4 levels in 34.1% of cases (45/132), and this alteration was found to be associated with the development of patient recurrences (p = 0.007). Interestingly, BRD4 hyperphosphorylation predicted significantly shorter overall (p < 0.001) and event-free survival (p < 0.001). Moreover, multivariate analyses were performed to confirm its independent prognostic impact in our cohort. In conclusion, our findings show that BRD4 hyperphosphorylation is an alteration associated with PP2A inhibition that defines a subgroup of TNBC patients with unfavorable prognosis, suggesting the potential clinical and therapeutic usefulness of the PP2A/BRD4 axis as a novel molecular target to overcome resistance to treatments based on BRD4 inhibition.
Collapse
|
50
|
Luque Paz D, Riou J, Verger E, Cassinat B, Chauveau A, Ianotto JC, Dupriez B, Boyer F, Renard M, Mansier O, Murati A, Rey J, Etienne G, Mansat-De Mas V, Tavitian S, Nibourel O, Girault S, Le Bris Y, Girodon F, Ranta D, Chomel JC, Cony-Makhoul P, Sujobert P, Robles M, Ben Abdelali R, Kosmider O, Cottin L, Roy L, Sloma I, Vacheret F, Wemeau M, Mossuz P, Slama B, Cussac V, Denis G, Walter-Petrich A, Burroni B, Jézéquel N, Giraudier S, Lippert E, Socié G, Kiladjian JJ, Ugo V. Genomic analysis of primary and secondary myelofibrosis redefines the prognostic impact of ASXL1 mutations: a FIM study. Blood Adv 2021; 5:1442-1451. [PMID: 33666653 PMCID: PMC7948260 DOI: 10.1182/bloodadvances.2020003444] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/30/2020] [Indexed: 12/26/2022] Open
Abstract
We aimed to study the prognostic impact of the mutational landscape in primary and secondary myelofibrosis. The study included 479 patients with myelofibrosis recruited from 24 French Intergroup of Myeloproliferative Neoplasms (FIM) centers. The molecular landscape was studied by high-throughput sequencing of 77 genes. A Bayesian network allowed the identification of genomic groups whose prognostic impact was studied in a multistate model considering transitions from the 3 conditions: myelofibrosis, acute leukemia, and death. Results were validated using an independent, previously published cohort (n = 276). Four genomic groups were identified: patients with TP53 mutation; patients with ≥1 mutation in EZH2, CBL, U2AF1, SRSF2, IDH1, IDH2, NRAS, or KRAS (high-risk group); patients with ASXL1-only mutation (ie, no associated mutation in TP53 or high-risk genes); and other patients. A multistate model found that both TP53 and high-risk groups were associated with leukemic transformation (hazard ratios [HRs] [95% confidence interval], 8.68 [3.32-22.73] and 3.24 [1.58-6.64], respectively) and death from myelofibrosis (HRs, 3.03 [1.66-5.56] and 1.77 [1.18-2.67], respectively). ASXL1-only mutations had no prognostic value that was confirmed in the validation cohort. However, ASXL1 mutations conferred a worse prognosis when associated with a mutation in TP53 or high-risk genes. This study provides a new definition of adverse mutations in myelofibrosis with the addition of TP53, CBL, NRAS, KRAS, and U2AF1 to previously described genes. Furthermore, our results argue that ASXL1 mutations alone cannot be considered detrimental.
Collapse
Affiliation(s)
- Damien Luque Paz
- Univ Angers, INSERM, CRCINA, Angers, France
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire (CHU) Angers, Angers, France
- Univ Angers, UFR Santé, Angers, France
| | - Jérémie Riou
- Univ Angers, INSERM, Unit 1066 minT, Angers, France
| | - Emmanuelle Verger
- Laboratoire de Biologie Cellulaire, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
- Université de Paris, U1131 INSERM, IRSL, Paris, France
| | - Bruno Cassinat
- Laboratoire de Biologie Cellulaire, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
- Université de Paris, U1131 INSERM, IRSL, Paris, France
| | | | | | | | | | - Maxime Renard
- Univ Angers, INSERM, CRCINA, Angers, France
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire (CHU) Angers, Angers, France
- Univ Angers, UFR Santé, Angers, France
| | - Olivier Mansier
- Laboratoire d'Hématologie, CHU Bordeaux, Bordeaux, France
- Université de Bordeaux, INSERM U1034, Bordeaux, France
| | - Anne Murati
- Département de Biopathologie et Département d'Oncologie Prédictive and
| | - Jérôme Rey
- Département d'Hématologie, Centre de Recherche en Cancérologie de Marseille, Institut Paoli-Calmettes, INSERM, Marseille, France
| | - Gabriel Etienne
- Département d'Hématologie, Institut Bergonié, Bordeaux, France
| | | | - Suzanne Tavitian
- Service d'Hématologie, CHU Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Toulouse, France
| | - Olivier Nibourel
- Laboratoire d'Hématologie Cellulaire and
- UMR 9020-UMR-S 1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University Lille, CNRS, INSERM, CHU Lille, Lille, France
| | | | - Yannick Le Bris
- Laboratoire d'Hématologie, CHU Nantes, Nantes, France
- Université de Nantes, INSERM, CRCINA, Nantes, France
| | | | - Dana Ranta
- Hématologie Clinique, CHU Nancy, Nancy, France
| | | | | | - Pierre Sujobert
- Service d'Hématologie Biologique, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Margot Robles
- Hématologie Clinique, CH Périgueux, Périgueux, France
| | - Raouf Ben Abdelali
- Pôle Hématologie et Oncologie, Laboratoire Cerba, Saint-Ouen L'Aumône, France
| | - Olivier Kosmider
- Laboratoire d'Hématologie, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | - Laurane Cottin
- Univ Angers, INSERM, CRCINA, Angers, France
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire (CHU) Angers, Angers, France
- Univ Angers, UFR Santé, Angers, France
| | - Lydia Roy
- Service d'Hématologie, Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Créteil, France
- Faculté de Santé, Université Paris Est Créteil (UPEC), Créteil, France
| | - Ivan Sloma
- Département d'Hématologie et Immunologie, Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
| | | | | | - Pascal Mossuz
- Laboratoire d'Hématologie, CHU Grenoble, Grenoble, France
| | - Borhane Slama
- Service d'Onco-Hématologie, CH Avignon, Avignon, France
| | | | | | | | - Barbara Burroni
- Département d'Anatomo-Pathologie, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Paris, France
| | | | - Stéphane Giraudier
- Laboratoire de Biologie Cellulaire, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
- Université de Paris, U1131 INSERM, IRSL, Paris, France
| | | | - Gérard Socié
- Hématologie-Transplantation, Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France; and
| | - Jean-Jacques Kiladjian
- Université de Paris, U1131 INSERM, IRSL, Paris, France
- Centre d'Investigations Cliniques (INSERM CIC1427), Assistance Publique-Hôpitaux de Paris, Hôpital Saint-Louis, Paris, France
| | - Valérie Ugo
- Univ Angers, INSERM, CRCINA, Angers, France
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire (CHU) Angers, Angers, France
- Univ Angers, UFR Santé, Angers, France
| |
Collapse
|