1
|
Coutinho AK, Vazquez YCB, Gifoni MAC, Jansen AM, O’Connor JM, Pérez-Vargas JCS, Rico-Restrepo M, Sanku G, Mendez G. Current landscape of BRAF-V600E metastatic CRC management in Latin America: an expert Latin American panel's recommendations. Ecancermedicalscience 2024; 18:1807. [PMID: 40171464 PMCID: PMC11959131 DOI: 10.3332/ecancer.2024.1807] [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: 06/10/2024] [Indexed: 04/03/2025] Open
Abstract
Colorectal cancer is the second leading cause of cancer death in Latin America (LA) with a projected 65.4% increase by 2040. Up to 10% of metastatic CRC (mCRC) patients in LA had an activating BRAF mutation. In clinical trials, targeted therapies for BRAF-V600E mutated mCRC have improved patient outcomes. However, in LA, BRAF-V600E testing and treatment of positive patients remains variable. To address this need, the Americas Health Foundation convened a meeting of LA experts on BRAF-V600E mCRC to develop treatment recommendations. The expert panel addressed the current landscape of BRAF-V600E mCRC testing, diagnosis and treatment in the region and identified significant limitations. Local guidelines, multidisciplinary boards, and tumor genotyping are among the recommendations. The panel also made first-line, second-line and surgery recommendations for patients after diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guillermo Mendez
- Hospital de Gastroenterologia ‘Carlos Bonorino Udaondo’, Buenos Aires 1264, Argentina
| |
Collapse
|
2
|
Martínez‐Roca A, Cubiella J, García‐Heredia A, Guill‐Berbegal D, Baile‐Maxía S, Mangas‐Sanjuán C, Sala‐Miquel N, Madero‐Velazquez L, Alenda C, Zapater P, González‐Núñez C, Iglesias‐Gómez A, Codesido‐Prado L, Díez‐Martín A, Kaminski MF, Erichsen R, Adami H, Ferlitsch M, Pellisé M, Holme Ø, Dekker E, Bretthauer M, Jover R, For the EPoS Study Group. Prediction of metachronous advanced colorectal neoplasia by KRAS mutation in polyps. United European Gastroenterol J 2024; 12:1179-1189. [PMID: 39400528 PMCID: PMC11578838 DOI: 10.1002/ueg2.12667] [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: 04/29/2024] [Accepted: 07/05/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The potential of molecular markers in the removed polys as reliable predictors of metachronous lesions is still uncertain. AIM Our aim was to evaluate the role of somatic mutations in KRAS in polyps of patients with high-risk adenomas to predict the risk of advanced polyps or colorectal cancer (CRC) within 3 years. METHODS A total of 518 patients were prospectively enrolled. The included patients had adenomas ≥10 mm, high-grade dysplasia, villous component or ≥3 more adenomas at baseline and were scheduled to undergo surveillance colonoscopy at 3 years ± 6 months. Somatic KRAS mutation was performed on 1189 polyps collected from these patients. At surveillance, advanced lesions were defined as adenomas with a size of ≥10 mm. High-grade dysplasia or villous component, serrated polyps ≥10 mm or with dysplasia or CRC. RESULTS At baseline, 81 patients (15.6%) had KRAS mutations in at least one polyp. Patients with KRAS mutated polyps had more frequent villous histological lesions and size ≥20 mm. In the multivariate analysis, adjusted for age and sex, only age (odds ratios [OR], 1.06; 95% confidence interval [CI], 1.02-1.09; p < 0.001), ≥5 adenomas (OR, 3.92; 95% CI, 1.96-7.82), and KRAS mutation (OR, 2.54; 95% CI, 1.48-4.34; p < 0.01) were independently associated with the development of advanced lesions at surveillance. CONCLUSIONS Our results show that, in patients with high-risk adenomas, the presence of somatic mutations in KRAS is an independent risk factor for the development of advanced metachronous polyps.
Collapse
Affiliation(s)
- Alejandro Martínez‐Roca
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - Joaquín Cubiella
- Department of GastroenterologyResearch Group in Gastrointestinal Oncology‐OurenseCIBEREHDHospital Universitario de OurenseOurenseSpain
| | - Anabel García‐Heredia
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - David Guill‐Berbegal
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - Sandra Baile‐Maxía
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - Carolina Mangas‐Sanjuán
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - Noelia Sala‐Miquel
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - Lucía Madero‐Velazquez
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | - Cristina Alenda
- Pathology DepartmentAlicante Institute for Health and Biomedical Research (ISABIAL)Dr. Balmis General University HospitalAlicanteSpain
| | - Pedro Zapater
- Clinical Pharmacology DepartmentAlicante Institute for Health and Biomedical Research (ISABIAL)Dr. Balmis General University HospitalAlicanteSpain
| | | | - Agueda Iglesias‐Gómez
- Department of GastroenterologyResearch Group in Gastrointestinal Oncology‐OurenseCIBEREHDHospital Universitario de OurenseOurenseSpain
| | - Laura Codesido‐Prado
- Department of GastroenterologyResearch Group in Gastrointestinal Oncology‐OurenseCIBEREHDHospital Universitario de OurenseOurenseSpain
| | - Astrid Díez‐Martín
- Department of GastroenterologyResearch Group in Gastrointestinal Oncology‐OurenseCIBEREHDHospital Universitario de OurenseOurenseSpain
| | - Michal F. Kaminski
- Department of Oncological GastroenterologyMaria Sklodowska‐Curie National Research Institute of OncologyWarsawPoland
- Department of Cancer PreventionMaria Sklodowska‐Curie National Research Institute of OncologyWarsawPoland
| | - Rune Erichsen
- Department of Clinical EpidemiologyAarhus University HospitalAarhusDenmark
- Department of SurgeryRanders Regional HospitalRandersDenmark
| | - Hans‐Olov Adami
- Department of Medical Epidemiology and BiostatisticsKarolinska InstitutetStockholmSweden
| | - Monika Ferlitsch
- Department of Internal Medicine IIIMedical University of ViennaWienAustria
| | - María Pellisé
- Department of GastroenterologyHospital Clínic de BarcelonaInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)University of BarcelonaBarcelonaSpain
| | - Øyvind Holme
- Department of GastroenterologySørlandet Sykehus HFKristiansandNorway
| | - Evelien Dekker
- Department of Gastroenterology and Hepatology C2‐115Amsterdam University Medical CentresDuivendrechtThe Netherlands
- Bergman Clinics IZAAmsterdamThe Netherlands
| | | | - Rodrigo Jover
- Servicio de Medicina DigestivaInstituto de Investigación Sanitaria ISABIALHospital General Universitario Dr. BalmisAlicanteSpain
- Departamento de Medicina ClínicaUniversidad Miguel HernándezAlicanteSpain
| | | |
Collapse
|
3
|
Santos FA, Reis RM, Barroti LC, Pereira AAL, Matsushita MM, de Carvalho AC, Datorre JG, Berardinelli GN, Araujo RLC. Overall Survival, BRAF, RAS, and MSI Status in Patients Who Underwent Cetuximab After Refractory Chemotherapy for Metastatic Colorectal Cancer. J Gastrointest Cancer 2024; 55:344-354. [PMID: 37608030 DOI: 10.1007/s12029-023-00964-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2023] [Indexed: 08/24/2023]
Abstract
PURPOSE Evaluate overall survival (OS), RAS, BRAF, and MSI frequencies in patients with metastatic colorectal cancer (mCRC), refractory to chemotherapy, and finally treated with cetuximab. METHODS A retrospective cohort study to evaluate 211 mCRC patients with wild-type KRAS treated with cetuximab. BRAF V600E, KRAS, NRAS gene mutations, and MSI status were identified using PCR techniques in a population of pre-treated patients who were refractory to fluoropyrimidines, oxaliplatin, and irinotecan. In addition, we evaluated the mutation frequency of the BRAF and NRAS genes and the MSI status of this population. Uni- and multivariate analyses were performed for independent prognostic factors of OS. RESULTS The median OS was 10.4 months, 6.6 months for patients with right and 11.5 months for left colon cancers (p = 0.02). The frequencies of mutations were BRAF at 3.9% (median OS of 4.9 months), NRAS at 3.38% (median OS of 6.9 months), and MSI-High status at 3.3% (median OS of 4.6 months). The OS, NRAS, and MSI frequencies were similar to those found in other studies that evaluated cetuximab in poly-treated patients and were associated with lower survival rates in univariate analyses. The frequency of BRAF mutations was lower than that found in previous studies. The only variable that remained significant for OS in the multivariate model was tumour laterality, with patients with right colon cancer presenting a worse prognosis (HR = 2.81). CONCLUSION Although BRAF, NRAS mutations, and MSI-High status were associated with shorter OS in univariate analyses, only tumour laterality remained an independent prognostic factor in the multivariate analysis.
Collapse
Affiliation(s)
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), School Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimaraes, Portugal
| | - Lucas C Barroti
- Department of Dermatology, Universidade Federal de Sao Paulo, Sao Paulo, Brazil
| | - Allan A L Pereira
- Clinical Oncology Department, Hospital Sirio Libanes de Brasilia-DF, Sao Paulo, Brazil
| | | | | | | | | | - Raphael L C Araujo
- Department of Surgery, Digestive Surgery Service, Universidade Federal de Sao Paulo, Sao Paulo, Brazil.
- Instituto de Ensino e Pesquisa, Barretos Cancer Hospital, Sao Paulo, Brazil.
| |
Collapse
|
4
|
Waters KM, Singhi AD, Montgomery EA. Exploring the spectrum of serrated epithelium encountered in inflammatory bowel disease. Hum Pathol 2023; 132:126-134. [PMID: 35753410 PMCID: PMC11186602 DOI: 10.1016/j.humpath.2022.06.018] [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: 06/06/2022] [Accepted: 06/17/2022] [Indexed: 02/07/2023]
Abstract
Patients with inflammatory bowel disease (IBD) are at an increased risk for colorectal cancer. Currently, dysplasia is the best marker of CRC risk. Assessing dysplasia is a challenging task for pathologists as the longstanding inflammation causes marked reactive cytologic changes and architectural distortion. Recent descriptions of nonconventional types of dysplasia in IBD have added to the complexity. In this review, we focus on the clinical, endoscopic, histologic, and molecular findings in lesions with serrated epithelium. Serrated epithelial change (SEC), sessile serrated lesion (SSL)-like, serrated lesion-not otherwise specified (SL-NOS), and traditional serrated adenoma (TSA)-like lesions all typically occur in patients with longstanding IBD with mean ages in the fifth-sixth decade. SEC is often encountered in nontargeted biopsies while the others form visible polyps. While serrated lesions have significant histologic overlap, subtle differences can help pathologists separate them. SEC has markedly distorted architecture with crypts losing perpendicular orientation to the muscularis mucosae. The crypts are goblet cell-rich and have irregular serrations that involve the full length of the crypt. SSL-like lesions are goblet cell poor and have microvesicular cytoplasm. Like their sporadic counterpart in non-IBD patients, these lesions have lateral growth at the crypt bases. TSA-like lesions are characterized by their villous architecture, ectopic crypts, pink cytoplasm, and hyperchromatic elongated nuclei. We also explore molecular findings that help in distinguishing these lesions, current knowledge on the association of each of these lesions with dysplasia and CRC, and future research needed to better characterize these entities.
Collapse
Affiliation(s)
- Kevin M Waters
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, 90048, USA
| | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Elizabeth A Montgomery
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
5
|
Lee HW, Song B, Kim K. Colorectal cancers with a residual adenoma component: Clinicopathologic features and KRAS mutation. PLoS One 2022; 17:e0273723. [PMID: 36083889 PMCID: PMC9462729 DOI: 10.1371/journal.pone.0273723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 07/22/2022] [Indexed: 11/19/2022] Open
Abstract
Background/Aim
Colorectal cancer is well known for its “adenoma-carcinoma” sequential carcinogenesis. Some colorectal cancers demonstrate a residual adenoma component during progression from adenoma to invasive carcinoma. However, the clinicopathological significance of residual adenoma component remains unclear. In this study, we aimed to investigate the clinicopathologic and molecular characteristics including the KRAS mutation in colorectal cancers containing a residual adenoma component.
Materials and methods
In this study, 498 surgically resected colorectal cancer patients were enrolled. Their detailed clinicopathologic features and results of molecular study including KRAS mutation test and microsatellite instability were analyzed.
Results
A residual adenoma component was identified in 42 (8.4%) patients with colorectal cancer. The presence of a residual adenoma component was associated with a high frequency of the KRAS mutation (65%, p = 0.031) as well as indolent clinicopathological features, including polypoid gross type (p < 0.001), well-differentiated histology (p < 0.001), low pT (p < 0.001) and pN stage (p = 0.003), absence of vascular invasion (p = 0.005), and a better progression-free prognosis (p = 0.029). The cases with an adenoma component had a 35.7% discordance rate on the KRAS mutation tests in their adenoma and carcinoma regions.
Conclusion
In conclusion, colorectal cancer with a residual adenoma component showed indolent clinicopathologic features and frequent KRAS mutations. Due to the discordance in the incidence of the KRAS mutation between the adenoma and carcinoma components, the adenoma component should be documented in the pathology report, and care should be taken not to include the adenoma component when collecting samples for molecular testing.
Collapse
Affiliation(s)
- Hyoun Wook Lee
- Department of Pathology, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Republic of Korea
| | - Boram Song
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyungneun Kim
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
6
|
Güttlein L, Luca MR, Esteso F, Fresno C, Mariani J, Otero Pizarro M, Brest E, Starapoli S, Kreimberg K, Teves P, Mendoza Bertelli A, R Girotti M, Salanova R, O'Connor JM. Liquid biopsy for KRAS, NRAS and BRAF mutation testing in advanced colorectal cancer patients: the Argentinean experience. Future Oncol 2022; 18:3277-3287. [PMID: 36004810 DOI: 10.2217/fon-2022-0329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Objective: To analyze the frequency of KRAS, NRAS and BRAF hotspot mutations in circulating tumor DNA (ctDNA) from patients with metastatic colorectal cancer (mCRC). Methods: Observational, descriptive and retrospective study in mCRC patients with available ctDNA-based genotype of KRAS, NRAS and BRAF. Results: The frequencies of plasma mutations for KRAS, NRAS and BRAF were 34% (± 7), 4% (± 3) and 4% (± 3), respectively. Median overall survival of plasma-tested RAS/BRAF-mutated patients was 26.6 months (95% CI: 14.4-not estimable [NE]), while RAS/BRAF wild-type patients did not reach the median survival during follow-up. Median progression-free survival for RAS/BRAF wild-type and RAS/BRAF-mutated patients was 12 (95% CI: 7-NE) and 4 months (95% CI: 4-NE), respectively. Conclusion: Our work supports the utility of KRAS, NRAS and BRAF analysis in liquid biopsy from mCRC patients.
Collapse
Affiliation(s)
- Leandro Güttlein
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - María R Luca
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| | - Federico Esteso
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| | | | - Javier Mariani
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| | - Mercedes Otero Pizarro
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Esteban Brest
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Solange Starapoli
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Kevin Kreimberg
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Paula Teves
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Andrea Mendoza Bertelli
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - María R Girotti
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Ruben Salanova
- Molecular Biology Laboratory. Biomakers S.A. Av. Pueyrredón 1777. 9 Floor. Buenos Aires, C1119ACA, Argentina
| | - Juan M O'Connor
- Clinical Oncology, Instituto Alexander Fleming, Av Cramer 1180, Buenos Aires, C1426ANZ, Argentina
| |
Collapse
|
7
|
Yuan Z, Yu X, Chen W, Chen D, Cai J, Jiang Y, Liu X, Wu Z, Wang L, Grady WM, Wang H. Epigenetic silencing and tumor suppressor gene of HAND2 by targeting ERK signaling in colorectal cancer. Cell Commun Signal 2022; 20:111. [PMID: 35870943 PMCID: PMC9308366 DOI: 10.1186/s12964-022-00878-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The screening biomarkers for early detection of colorectal cancer (CRC) is lacking. The aim is to identify epigenetic silenced genes and clarify its roles and underlying mechanism in CRC. We conducted integrative analyses of epigenome-wide Human Methylation 450 K arrays and transcriptome to screen out candidate epigenetic driver genes with transcription silencing. Methylated silencing HAND2 were identified and verified in large CRC cohort. The mechanism of HAND2 expression by promoter inhibition were clarified both in vitro and vivo assays. Cell biofunctional roles of HAND2 methylation was investigated in CRC cells. HAND2 reconstitution were constructed by lentivirus plasmid and tumor xenograft model of HAND2 were built subcutaneously. Genomic mRNA analysis by RNA-sequencing and subsequent GSEA analysis were performed to identify potential target of HAND2 and qPCR/WB was conducted to identify the results. RESULTS We firstly reported high frequency of HAND2 methylation in promoter in CRC and hypermethylation was negatively correlated with expression silencing and leaded to poor survival in several CRC cohort patients. 5-Aza treatment to demethylated HAND2 could revert its expression in CRC cells. Functionally, HAND2 reconstitution can inhibit cell proliferation, invasion and migration in vitro. In tumor xenograft, HAND2 reconstruction significantly repressed tumor growth when compared to control vector. Thousands of aberrant expressed genes were observed in the heatmap of RNA-sequencing data. HAND2 reconstitution could bind to ERK and reduce its phosphorylation by CoIP assay. These above results showed HAND2 reconstitution perturbed the activation of MAPK/ERK signaling by reduction of ERK phosphorylation. CONCLUSIONS HAND2 is one tumor suppressor by targeting ERK signaling and one potential epigenetic driver gene in CRC. Video Abstract.
Collapse
Affiliation(s)
- Zixu Yuan
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China.
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D4-100, Seattle, WA, 98109, USA.
| | - Xihu Yu
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenle Chen
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Daici Chen
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jian Cai
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yingming Jiang
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Xiaoxia Liu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhijie Wu
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - Lei Wang
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China
| | - William M Grady
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, D4-100, Seattle, WA, 98109, USA.
| | - Hui Wang
- Department of Colorectal Surgery, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangzhou, China.
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
8
|
dos Santos W, dos Reis MB, Porto J, de Carvalho AC, Matsushita M, Oliveira G, Syrjänen K, Reis RM, Guimarães DP. Somatic targeted mutation profiling of colorectal cancer precursor lesions. BMC Med Genomics 2022; 15:143. [PMID: 35761395 PMCID: PMC9238170 DOI: 10.1186/s12920-022-01294-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 06/16/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Most colorectal cancers (CRC) arise from precursor lesions. This study aimed to characterize the mutation profile of colorectal cancer precursor lesions in a Brazilian population.
Methods
In total, 90 formalin-fixed paraffin-embedded colorectal precursor lesions, including 67 adenomas, 7 sessile serrated lesions, and 16 hyperplastic polyps, were analyzed by next-generation sequencing using a panel of 50 oncogenes and tumor suppressor genes. The genetic ancestry of the patients was estimated.
Results
Somatic driver mutations were identified in 66.7% of cases, including alterations in APC (32.2%), TP53 (20.0%), KRAS (18.9%), BRAF (13.3%) and EGFR (7.8%). Adenomas displayed a higher number of mutations, mainly in APC, compared to serrated polyps (73.1% vs. 47.8%, p = 0.026). Advanced adenomas had a significantly higher frequency of mutation in KRAS and a high overall mutation rate than early adenomas (92.9% vs. 59%, p = 0.006). A high degree of ancestry admixture was observed in the population studied, with a predominance of European components (mean of 73%) followed by African (mean of 11.3%). No association between genetic ancestry and type of lesions was found. The mutation profile of Brazilian colorectal precursor lesions exhibits alteration in APC, KRAS, TP53, and BRAF at different frequencies according to lesion type.
Conclusions
These results bestow the knowledge of CRC's biologic history and support the potential of these biomarkers for precursor lesions detection in CRC screening of the Brazilian population.
Collapse
|
9
|
España MS. Treatment of advanced BRAF-mutated colorectal cancer: where we are and where we are going. Clin Colorectal Cancer 2022; 21:71-79. [DOI: 10.1016/j.clcc.2022.01.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 11/03/2022]
|
10
|
Sobanski T, Arantes LMRB, Dos Santos W, Matsushita M, de Oliveira MA, Costa M, de Carvalho AC, Berardinelli GN, Syrjänen K, Reis RM, Guimarães DP. Methylation profile of colon cancer genes in colorectal precursor lesions and tumor tissue: perspectives for screening. Scand J Gastroenterol 2021; 56:920-928. [PMID: 34218733 DOI: 10.1080/00365521.2021.1922744] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AIMS Epigenetic alterations of genes involved in colorectal carcinogenesis are likely to be informative biomarkers for early detection. We assessed the methylation profile of a panel of seven colon cancer-related genes comparing normal colon, colorectal cancer (CRC) precursor lesions and cancer tissues from a Brazilian cohort. METHODS The cohort comprised 114 CRC patients, including 40 matched normal tissue, 47 patients with adenomas, 33 with serrated polyps and 8 with normal colonic biopsy. DNA methylation status of SEPT9, ALX4, NDRG4, BMP3, APC, p16 and MLH1 was determined by pyrosequencing and correlated with clinicopathological features. Sensitivity, specificity, positive predictive value and negative predictive value were calculated for all genes using cancer endpoint. RESULTS The most frequently methylated genes in cancer and in precancer lesions were SEPT9, ALX4, NDRG4, and BMP3, ranging from 55.3 to 95% of the samples. Overall, the frequency of methylation of these four genes in normal colonic tissue was significantly lower as compared to cancer or precursor lesions both in adenoma-carcinoma (p < .001 and p < .050) and serrated (sessile-serrated lesion) (p < .001 and p < .050) pathways. Additionally, sensitivity for the cancer endpoint ranged from 65.6 to 91.8%, and specificity from 17.9 to 62.9% for SEPT9, ALX4, NDRG4, and BMP3 genes. Moreover, the comethylation of ≥4 genes was higher in sessile-serrated lesion (87.5%) and conventional adenomas (78.7%) than in hyperplastic polyps (43.7%) (p = .025) and was significantly associated with proximal cancers (p = .042). CONCLUSIONS Our study suggests the DNA methylation can constitute potential biomarkers in CRC screening of Brazilian population.
Collapse
Affiliation(s)
- Thais Sobanski
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | - Maraisa Costa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | - Kari Syrjänen
- SMW Consultants Ltd, Kaarina, Finland.,Department of Clinical Research, Biohit Oyj, Helsinki, Finland
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal.,3ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Endoscopy, Barretos Cancer Hospital, Barretos, Brazil
| |
Collapse
|
11
|
de Paula AE, Galvão HDCR, Bonatelli M, Sabato C, Fernandes GC, Berardinelli GN, Andrade CEM, Neto MC, Romagnolo LGC, Campacci N, Scapulatempo-Neto C, Reis RM, Palmero EI. Clinicopathological and molecular characterization of Brazilian families at risk for Lynch syndrome. Cancer Genet 2021; 254-255:82-91. [PMID: 33647816 DOI: 10.1016/j.cancergen.2021.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/14/2020] [Accepted: 02/08/2021] [Indexed: 11/25/2022]
Abstract
Lynch syndrome (LS), is the most common hereditary colorectal cancer syndrome. However, it is poorly characterized in Brazil. Therefore, we aimed to determine the spectrum of pathogenic variants in Mismatch Repair (MMR) genes and investigate the MLH1 promotor methylation role as a second hit in LS tumors. Tumor screening through microsatellite instability and immunohistochemistry for MMR proteins was performed in 323 cases who met clinical criteria. BRAF-V600E and MLH1 promoter methylation were analyzed for all MLH1-deficient tumors. Patients with MMR deficient tumor proceeded to germline genetic testing. MMR deficient tumors were detected in 41% of patients recruited. About half of patients carried a pathogenic germline variant. Two recurrent variants in MLH1 and three novel pathogenic variants were identified. Furthermore, pathogenic germline variants with concomitant somatic MLH1 hypermethylation were found in 6% of cases. Predictive genetic testing was offered to 387 relatives. Overall, 127 tumors were diagnosed in 100 LS patients, from 62 unrelated families. Our molecular data provide new information about the spectrum of MMR mutations, which contributes to a better characterization of LS in Brazil. Furthermore, we call attention to the possibility of failure in the diagnosis of germline MLH1 mutation carriers when somatic MLH1 hypermethylation is used to rule out LS.
Collapse
Affiliation(s)
| | | | - Murilo Bonatelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | - Cristina Sabato
- Molecular Diagnosis Laboratory, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | | | | | | | | | | | - Natalia Campacci
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil
| | | | - Rui Manuel Reis
- Molecular Diagnosis Laboratory, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Edenir Inêz Palmero
- Molecular Diagnosis Laboratory, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Pele Pequeno Principe Research Institute, Curitiba, Brazil; Faculdades Pequeno Principe, Curitiba, Brazil.
| |
Collapse
|
12
|
Satorres C, García-Campos M, Bustamante-Balén M. Molecular Features of the Serrated Pathway to Colorectal Cancer: Current Knowledge and Future Directions. Gut Liver 2021; 15:31-43. [PMID: 32340435 PMCID: PMC7817929 DOI: 10.5009/gnl19402] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/18/2020] [Accepted: 03/04/2020] [Indexed: 02/05/2023] Open
Abstract
Serrated lesions are the precursor lesions of a new model of colorectal carcinogenesis. From a molecular standpoint, the serrated pathway is thought to be responsible for up to 30% of all colorectal cancer cases. The three major processes of this molecular mechanism are alterations in the mitogen-activated protein kinase pathway, production of the CpG island methylation phenotype, and generation of microsatellite instability. Other contributing processes are activation of WNT, alterations in the regulation of tumor suppressor genes, and alterations in microRNAs or in MUC5AC hypomethylation. Although alterations in the serrated pathway also contribute, their precise roles remain obscure because of the various methodologies and definitions used by different research groups. This knowledge gap affects clinical assessment of precursor lesions for their carcinogenic risk. The present review describes the current literature reporting the molecular mechanisms underlying each type of serrated lesion and each phenotype of serrated pathway colorectal cancer, identifying those areas that merit additional research. We also propose a unified serrated carcinogenesis pathway combining molecular alterations and types of serrated lesions, which ends in different serrated pathway colorectal cancer phenotypes depending on the route followed. Finally, we describe some key issues that need to be addressed in order to incorporate the newest technologies in serrated pathway research and to improve overall knowledge for developing specific prevention strategies and new therapeutic targets.
Collapse
Affiliation(s)
- Carla Satorres
- Gastrointestinal Endoscopy Research Group, La Fe Health Research Institute, Valencia, Spain
- Gastrointestinal Endoscopy Unit, Digestive Diseases Department, La Fe Polytechnic University Hospital, Valencia, Spain
| | - María García-Campos
- Gastrointestinal Endoscopy Unit, Digestive Diseases Department, La Fe Polytechnic University Hospital, Valencia, Spain
| | - Marco Bustamante-Balén
- Gastrointestinal Endoscopy Research Group, La Fe Health Research Institute, Valencia, Spain
- Gastrointestinal Endoscopy Unit, Digestive Diseases Department, La Fe Polytechnic University Hospital, Valencia, Spain
| |
Collapse
|
13
|
Evangelista AF, de Menezes WP, Berardinelli GN, Dos Santos W, Scapulatempo-Neto C, Guimarães DP, Calin GA, Reis RM. Pyknon-Containing Transcripts Are Downregulated in Colorectal Cancer Tumors, and Loss of PYK44 Is Associated With Worse Patient Outcome. Front Genet 2020; 11:581454. [PMID: 33304384 PMCID: PMC7693444 DOI: 10.3389/fgene.2020.581454] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/14/2020] [Indexed: 01/19/2023] Open
Abstract
Pyknons are specific human/primate-specific DNA motifs at least 16 nucleotides long that are repeated in blocks in intergenic and intronic regions of the genome and can be located in a new class of non-coding RNAs of variable length. Recent studies reported that pyknon deregulation could be involved in the carcinogenesis process, including colorectal cancer. We evaluated the expression profile of a set of 12 pyknons in a set of molecularly characterized colorectal cancer (CRC) patients. The pyknons (PYK10, PYK14, PYK17, PYK26, PYK27, PYK40, PYK41, PYK42, PYK43, PYK44, PYK83, and PYK90) expression was determined by qRT-PCR. A pilot analysis of 20 cases was performed, and consistent results were obtained for PYK10, PYK17, PYK42, PYK44, and PYK83. Further, the expression of the selected pyknons was evaluated in 73 CRC cases. Moreover, in 52 patients, we compared the expression profile in both tumor and normal tissues. All five pyknons analyzed showed significantly lower expression levels in the tumor compared to normal tissue. It was observed an association between expression of PYK10 with TP53 mutations (p = 0.029), PYK17 to histologic grade (p = 0.035), and PYK44 to clinical staging (p = 0.016). Moreover, levels of PYK44 were significantly associated with the patient's poor overall survival (p = 0.04). We reported the significant downregulation of pyknons motifs in tumor tissue compared with the normal counterpart, and the association of lower PYK44 expression with worse patient outcome. Further studies are needed to extend and validate these findings and determine the clinical-pathological impact.
Collapse
Affiliation(s)
| | | | | | | | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Endoscopy, Barretos Cancer Hospital, Barretos, Brazil
| | - George A Calin
- Translational Molecular Pathology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Guimarães, Portugal
| |
Collapse
|
14
|
Guimarães DP, Mantuan LA, de Oliveira MA, Junior RL, Costa AMD, Rossi S, Fava G, Taveira LN, Giardina KM, Talarico T, Costa M, Scapulatempo-Neto C, Matsushita MM, Véo CA, Fregnani JHT, Reis RM, Hawk ET, Mauad EC. The Performance of Colorectal Cancer Screening in Brazil: The First Two Years of the Implementation Program in Barretos Cancer Hospital. Cancer Prev Res (Phila) 2020; 14:241-252. [PMID: 32998941 DOI: 10.1158/1940-6207.capr-20-0179] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/01/2020] [Accepted: 09/22/2020] [Indexed: 11/16/2022]
Abstract
Colorectal cancer is the second most common cancer in Brazil. Yet, a nationally organized colorectal screening program is not implemented. Barretos Cancer Hospital (BCH) is one of the largest Brazilian institution that cares for underserved patients. BCH developed a fecal immunochemical test (FIT)-based organized colorectal cancer screening program to improve colorectal cancer outcomes.This study aims to present the quality/performance measures of the first 2 years of the FIT-based colorectal cancer screening program and its impact on the colorectal cancer disease stage. Between 2015 and 2017, a total of 6,737 individuals attending the Outpatient Department of Prevention or the Mobile Unit of BCH, which visits 18 cities of Barretos county, ages 50 to 65 years, were personally invited by a health agent/nurse practitioner. Exclusion criteria were personal history of colorectal cancer, adenomatous polyps, inflammatory bowel disease, and colonoscopy, or flexible sigmoidoscopy performed in the past 5 years. European Union (EU) guidelines for colorectal cancer screening programs were evaluated. Overall, 92.8% returned the FIT, with an inadequate examination rate of 1.5%. Among the 6,253 adequately tested, 12.5% had a positive result. The colonoscopy compliance and completion rates were 84.6 and 98.2%, respectively. The PPVs were 60.0%, 16.5%, and 5.6% for adenoma, advanced adenoma, and cancer, respectively. Stage distribution of screen-detected cancers shows earlier stages than clinically diagnosed colorectal cancer cancers reported at BCH and Brazilian cancer registries. Our colorectal cancer screening program achieved desirable quality metrics, aligned with the EU guidelines. The observed shift toward earlier colorectal cancer stages suggests an exciting opportunity to improve colorectal cancer-related cancers in Brazil.
Collapse
Affiliation(s)
- Denise Peixoto Guimarães
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, Brazil. .,Department of Prevention, Barretos Cancer Hospital, Barretos, Brazil.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | - Silvana Rossi
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, Brazil
| | - Gilberto Fava
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | - Thais Talarico
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Maraisa Costa
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | - Carlos Augusto Véo
- Department of Lower Digestive Surgery, Barretos Cancer Hospital, Barretos, Brazil
| | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Ernest T Hawk
- Cancer Prevention and Population Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
15
|
Campanella NC, Silva EC, Dix G, de Lima Vazquez F, Escremim de Paula F, Berardinelli GN, Balancin M, Chammas R, Mendoza Lopez RV, Silveira HCS, Capelozzi VL, Reis RM. Mutational Profiling of Driver Tumor Suppressor and Oncogenic Genes in Brazilian Malignant Pleural Mesotheliomas. Pathobiology 2020; 87:208-216. [PMID: 32369821 DOI: 10.1159/000507373] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Malignant pleural mesothelioma (MPM) is a highly lethal disease comprising a heterogeneous group of tumors with challenging to predict biological behavior. The diagnosis is complex, and the histologic classification includes 2 major subtypes of MPM: epithelioid (∼60% of cases) and sarcomatous (∼20%). Its identification depends upon pathological investigation supported by clinical and radiological evidence and more recently ancillary molecular testing. Treatment options are currently limited, with no known targeted therapies available. OBJECTIVES To elucidate the mutation profile of driver tumor suppressor and oncogenic genes in a cohort of Brazilian patients. METHODS We sequenced 16 driver genes in a series of 43 Brazilian malignant mesothelioma (MM) patients from 3 distinct Brazilian centers. Genomic DNA was extracted from formalin-fixed paraffin-embedded tumor tissue blocks, and the TERT promoter region was amplified by PCR followed by direct capillary sequencing. The Illumina TruSight Tumor 15 was used to evaluate 250 amplicons from 15 genes associated with solid tumors (AKT1, GNA11, NRAS, BRAF, GNAQ, PDGFRA, EGFR, KIT, PIK3CA, ERBB2, KRAS, RET, FOXL2, MET,and TP53). Library preparation with the TruSight Tumor 15 was performed before sequencing at the MiSeq platform. Data analysis was performed using Sophia DDM software. RESULTS Out of 43 MPM patients, 38 (88.4%) were epithelioid subtype and 5 (11.6%) were sarcomatoid histotype. Asbestos exposure was present in 15 (39.5%) patients with epithelioid MPM and 3 (60%) patients with sarcomatoid MPM. We found a TERT promoter mutation in 11.6% of MM, and the c.-146C>T mutation was the most common event. The next-generation sequencing was successful in 33 cases. A total of 18 samples showed at least 1 pathogenic, with a median of 1.8 variants, ranging from 1 to 6. The most mutated genes were TP53 and ERBB2 with 7 variants each, followed by NRAS BRAF, PI3KCA, EGFR and PDGFRA with 2 variants each. KIT, AKT1, and FOXL2 genes exhibited 1 variant each. Interestingly, 2 variants observed in the PDGFRA gene are classic imatinib-sensitive therapy. CONCLUSIONS We concluded that Brazilian MPM harbor mutation in classic tumor suppressor and oncogenic genes, which might help in the guidance of personalized treatment of MPM.
Collapse
Affiliation(s)
| | | | - Gustavo Dix
- Department of Surgery, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | - Marcelo Balancin
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil.,Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Rossana V Mendoza Lopez
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | | | - Vera Luiza Capelozzi
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil, .,Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal, .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal,
| |
Collapse
|
16
|
Hernández-Sandoval JA, Gutiérrez-Angulo M, Magaña-Torres MT, Alvizo-Rodríguez CR, Ramírez-Plascencia HHF, Flores-López BA, Valenzuela-Pérez JA, Peregrina-Sandoval J, Moreno-Ortiz JM, Domínguez-Valentín M, Ayala-Madrigal MDLL. Prevalence of the BRAF p.v600e variant in patients with colorectal cancer from Mexico and its estimated frequency in Latin American and Caribbean populations. J Investig Med 2020; 68:985-991. [PMID: 32184228 PMCID: PMC7306871 DOI: 10.1136/jim-2020-001301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2020] [Indexed: 01/08/2023]
Abstract
This study aimed to investigate the frequency of the somatic BRAF p.V600E in patients with colorectal cancer (CRC) in Mexico and compare it with those estimated for Latin American and Caribbean populations. One hundred and one patients with CRC with AJCC stages ranging I–IV from Western Mexico were included, out of which 55% were male and 61% had AJCC stage III–IV, with a mean age of 60 years. PCR-Sanger sequencing was used to identify the BRAF p.V600E variant. In addition, a systematic literature search in PubMed/Medline database and Google of the 42 countries in Latin America and the Caribbean led to the collection of information on the BRAF p.V600E variant frequency of 17 population reports. To compare the BRAF variant prevalence among populations, a statistical analysis was performed using GraphPad Prism V.6.0. We found that 4% of patients with CRC were heterozygous for the p.V600E variant. The χ2 test showed no significant difference (p>0.05) in p.V600E detection when comparing with other Latin American and Caribbean CRC populations, except for Chilean patients (p=0.02). Our observational study provides the first evidence on the frequency of BRAF p.V600E in patients with CRC from Western Mexico, which is 4%, but increases to 7.8% for all of Latin America and the Caribbean. The patient mean age and genetic descent on the observed frequencies of the variant in populations could influence the frequency differences.
Collapse
Affiliation(s)
- Jesús Arturo Hernández-Sandoval
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Melva Gutiérrez-Angulo
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México.,Departamento de Clínicas, CUALTOS, Universidad de Guadalajara, Tepatitlán de Morelos, Jalisco, México
| | - María Teresa Magaña-Torres
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Carlos Rogelio Alvizo-Rodríguez
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Helen Haydee Fernanda Ramírez-Plascencia
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Beatriz Armida Flores-López
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | | | - Jorge Peregrina-Sandoval
- Laboratorio de Inmunología, CUCBA, Universidad de Guadalajara, Zapopan, Jalisco, México.,Laboratorio de Patología Clínica, Hospital Civil "Fray Antonio Alcalde", Guadalajara, Jalisco, México
| | - José Miguel Moreno-Ortiz
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Mev Domínguez-Valentín
- Department of Tumor Biology, Institute for Cancer Research, University of Oslo, Oslo, Norway.,Instituto de Investigación, Universidad Católica Los Angeles de Chimbote, Chimbote, Áncash, Perú
| | - María de la Luz Ayala-Madrigal
- Instituto de Genética Humana "Dr. Enrique Corona Rivera" y Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, México
| |
Collapse
|
17
|
Dabir PD, Bruggeling CE, van der Post RS, Dutilh BE, Hoogerbrugge N, Ligtenberg MJL, Boleij A, Nagtegaal ID. Microsatellite instability screening in colorectal adenomas to detect Lynch syndrome patients? A systematic review and meta-analysis. Eur J Hum Genet 2019; 28:277-286. [PMID: 31695176 DOI: 10.1038/s41431-019-0538-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 10/01/2019] [Accepted: 10/13/2019] [Indexed: 11/09/2022] Open
Abstract
The colorectal cancer spectrum has changed due to population screening programs, with a shift toward adenomas and early cancers. Whether it would be a feasible option to test these adenomas for detection of Lynch syndrome (LS) patients is unclear. Through meta-analysis and systematic review, risk factors for DNA mismatch repair deficiency (dMMR) and microsatellite instability (MSI) in adenomas were identified in LS and unselected patient cohorts. Data were extracted for patient age and MMR variant together with adenoma type, grade, size, and location. A total of 41 studies were included, and contained more than 519 LS patients and 1698 unselected patients with 1142 and 2213 adenomas respectively. dMMR/MSI was present in 69.5% of conventional adenomas in LS patients, compared with 2.8% in unselected patients. In the LS cohort, dMMR/MSI was more frequently present in patients older than 60 years (82% versus 54%). dMMR/MSI was also more common in villous adenomas (84%), adenomas over 1 cm (81%), and adenomas with high grade dysplasia (88%). No significant differences were observed for dMMR/MSI in relation to MMR variants and location of adenomas. In the context of screening, we conclude that detection of dMMR/MSI in conventional adenomas of unselected patients is uncommon and might be considered as indication for LS testing. Within the LS cohort, 69.5% of LS patients could have been detected through dMMR/MSI screening of their conventional adenomas.
Collapse
Affiliation(s)
- Parag D Dabir
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Pathology, Randers Regional Hospital, Randers, Denmark
| | - Carlijn E Bruggeling
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rachel S van der Post
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bas E Dutilh
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, The Netherlands.,Centre for Molecular and Biomolecular Informatics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicoline Hoogerbrugge
- Department of Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjolijn J L Ligtenberg
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Annemarie Boleij
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris D Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
18
|
Moosazadeh M, Sadough A, Afshari M, Barzegari S, Janbabaee G, Tabrizi R, Akbari M, Alizadeh-Navaei R, Hedayatizadeh-Omran A, Rostami-Maskopaee F. Prevalence of BRAF gene mutation in samples of primary and metastatic colorectal cancer: A meta-analysis. Eur J Cancer Care (Engl) 2019; 28:e13160. [PMID: 31482595 DOI: 10.1111/ecc.13160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 06/23/2019] [Accepted: 08/01/2019] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Understanding the prevalence and biology of BRAF gene can improve the treatment methods of cancerous patients. This study aims to estimate the prevalence of BRAF gene mutation in samples of primary and metastatic colorectal cancer using meta-analysis method. METHODS We searched PubMed, Scopus, ScienceDirect, Ovid and Google Scholar motor engine using MeSH terms of relevant keywords. During the screening phase, titles, abstracts and full texts were reviewed and risk of bias was assessed for all selected papers based on Newcastle-Ottawa Scale (NOS) checklist. The results of the primary studies were combined using meta-analysis. RESULTS Of 95 eligible studies entered into the meta-analysis, prevalence of BRAF gene mutation had been assessed among 19,484 primary tumour samples as well as 12,256 metastatic samples. The total prevalence of BRAF gene mutation among primary tumour samples was estimated as of 10.16% (8.09-12.22) in the world, 0.41% (0-1.89) in EMRO region, 10.06% (7.54-12.59) in EURO region, 10.33% (7.24-13.43) in SEARO region and 11.33% (7.29-15.37) in WPRO region. The pooled estimates for BRAF gene mutation in metastatic samples were 6.53% (5.09-7.96), 8.07% (5.57-10.56), 5.38% (3.75-7.02) and 5.55% (1.72-9.38) for all regions, EURO, WPRO and PAHO regions respectively. CONCLUSION Our results showed evidences of BRAF gene mutation in one-tenth of primary colorectal tumour samples in EURO, PAHO, SEARO and WPRO regions which was considerably higher than that of the EMRO region.
Collapse
Affiliation(s)
- Mahmood Moosazadeh
- Health Sciences Research center, Addiction Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Azita Sadough
- Traditional Medicine and History of Medical science Research center, Faculty of Iranian medicine, Babol University of Medical Science, Babol, Iran
| | - Mahdi Afshari
- Department of Community Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Saeed Barzegari
- Department of Health Information Management, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Janbabaee
- Gastrointestinal Cancer Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Reza Tabrizi
- Health Policy Research Center, Student Research Committee, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Akbari
- Health Policy Research Center, Student Research Committee, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Alizadeh-Navaei
- Gastrointestinal cancer research center, Mazandaran University of Medical Sciences, Sari, Iran
| | | | | |
Collapse
|
19
|
Takeda T, Banno K, Kobayashi Y, Adachi M, Yanokura M, Tominaga E, Kosaki K, Aoki D. Mutations of RAS genes in endometrial polyps. Oncol Rep 2019; 42:2303-2308. [PMID: 31638232 PMCID: PMC6826305 DOI: 10.3892/or.2019.7353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/27/2019] [Indexed: 11/05/2022] Open
Abstract
Endometrial polyps are common, yet the molecular mechanisms underlying their formation and progression remain unclear. We examined gene mutations possibly related to the pathogenesis of endometrial polyps, as well as to their clinical features. Four premenopausal patients with endometrial polyps, who were not under drug treatment, were recruited. Whole exomes of endometrial polyps and peripheral blood lymphocytes were analyzed by next‑generation sequencing, and somatic mutations were derived by subtraction. Then, 35 samples of endometrial polyps and 12 samples of atypical polypoid adenomyoma were newly recruited to validate the identified mutations by polymerase chain reaction‑reverse sequence specific oligonucleotide method. The mutations were also analyzed in separate stromal and glandular components of the polyps after laser‑capture microdissection. Whole exome sequencing revealed that KRAS mutations were the only type of mutation detectable in multiple cases (2/4). Targeted mutation analysis revealed that 16 of 35 samples (45.7%) of endometrial polyps harbored RAS mutations. Mutation‑positive cases exhibited a significantly higher number of endometrial polyps (3.25±2.70 vs. 1.74±0.87, P=0.045). Laser‑capture microdissection in NRAS‑mutated endometrial polyps revealed that both stromal and glandular components harbored RAS mutations. There was no RAS mutation in 12 samples of atypical polypoid adenomyoma. This is the first report demonstrating that pathogenic RAS mutations are frequent in non‑treated endometrial polyps. RAS mutations may have an important role in tumorigenesis and in the formation of multiple endometrial polyps.
Collapse
Affiliation(s)
- Takashi Takeda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Masataka Adachi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Megumi Yanokura
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo 160‑8582, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo 160‑8582, Japan
| |
Collapse
|
20
|
Lima J, Teixeira Y, Pimenta C, Felipe AV, Silva TD, Junior EEDL, Saad SS, Deak E, Murray H, Manoukian Forones N. Fecal Genetic Mutations and Human DNA in Colorectal Cancer and Polyps Patients. Asian Pac J Cancer Prev 2019; 20:2929-2934. [PMID: 31653137 PMCID: PMC6982689 DOI: 10.31557/apjcp.2019.20.10.2929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Colorectal cancer (CRC) is one of the most frequent cancers. Genetic mutations in CRC already described can be detected in feces. Microarray methods in feces can represent a new diagnostic tool for CRC and significant improvement at public health. Aim: to analyze stool DNA by human DNA quantify and microarray methods as alternatives to CRC screening. Method: Three methods were analyzed in stool samples: Human DNA Quantify, RanplexCRC and KRAS/BRAF/PIK3CA (KBP) Arrays. Results: KBP array mutations were presented in 60.7% of CRC patients and RanplexCRC Array mutations in 61.1% of CRC patients. Sensitivity and specificity for human DNA quantification was 66% and 82% respectively. Fecal KBP Array had 35% sensitivity and 96% specificity and RanplexCRC Array method had 78% sensitivity and 100% specificity. Conclusion: Microarray methods showed promise as potential biomarkers for CRC screening; however, these methods had to be optimized to improve accuracy and applicability by clinical routine.
Collapse
Affiliation(s)
- Jacqueline Lima
- Oncology Group, Gastroenterology Division, Medicine Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Yolanda Teixeira
- Oncology Group, Gastroenterology Division, Medicine Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Célia Pimenta
- Oncology Group, Gastroenterology Division, Medicine Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Aledson Vitor Felipe
- Oncology Group, Gastroenterology Division, Medicine Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Tiago Donizetti Silva
- Oncology Group, Gastroenterology Division, Medicine Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | | | - Sarhan Sydney Saad
- Surgery Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Elisabeth Deak
- Surgery Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| | - Helena Murray
- Randox Laboratories Ltd., Crumlin, Co. Antrim, United Kingdom
| | - Nora Manoukian Forones
- Oncology Group, Gastroenterology Division, Medicine Department, Universidade Federal de Sao Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Dos Santos W, Sobanski T, de Carvalho AC, Evangelista AF, Matsushita M, Berardinelli GN, de Oliveira MA, Reis RM, Guimarães DP. Mutation profiling of cancer drivers in Brazilian colorectal cancer. Sci Rep 2019; 9:13687. [PMID: 31548566 PMCID: PMC6757044 DOI: 10.1038/s41598-019-49611-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/28/2019] [Indexed: 12/24/2022] Open
Abstract
The molecular basis of colorectal cancer (CRC) can guide patient prognosis and therapy. In Brazil, knowledge on the CRC mutation landscape is limited. Here, we investigated the mutation profile of 150 cancer-related genes by next-generation sequencing and associated with microsatellite instability (MSI) and genetic ancestry in a series of 91 Brazilian CRC patients. Driver mutations were found in the APC (71.4%), TP53 (56.0%), KRAS (52.7%), PIK3CA (15.4%) and FBXW7 (10.9%) genes. Overall, genes in the MAPK/ERK, PIK3/AKT, NOTCH and receptor tyrosine kinase signaling pathways were mutated in 68.0%, 23.1%, 16.5%, and 15.3% of patients, respectively. MSI was found in 13.3% of tumors, most of which were proximal (52.4%, P< 0.001) and had a high mutation burden. European genetic ancestry was predominant (median of 83.1%), followed by Native American (4.1%), Asian (3.4%) and African (3.2%). NF1 and BRAF mutations were associated with African ancestry, while TP53 and PIK3CA mutations were inversely correlated with Native American ancestry. Our study suggests that Brazilian CRC patients exhibit a mutation profile similar to other populations and identify the most frequently mutated genes, which could be useful in future target therapies and molecular cancer screening strategies.
Collapse
Affiliation(s)
| | - Thais Sobanski
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | | | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, 4710-057, Portugal.
- 3ICVS/3B's-PT Government Associate Laboratory, Braga, 4710-057, Portugal.
| | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.
- Department of Endoscopy, Barretos Cancer Hospital, Barretos, Brazil.
| |
Collapse
|
22
|
Mutational profile of Brazilian lung adenocarcinoma unveils association of EGFR mutations with high Asian ancestry and independent prognostic role of KRAS mutations. Sci Rep 2019; 9:3209. [PMID: 30824880 PMCID: PMC6397232 DOI: 10.1038/s41598-019-39965-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/06/2019] [Indexed: 01/05/2023] Open
Abstract
Lung cancer is the deadliest cancer worldwide. The mutational frequency of EGFR and KRAS genes in lung adenocarcinoma varies worldwide per ethnicity and smoking. The impact of EGFR and KRAS mutations in Brazilian lung cancer remains poorly explored. Thus, we investigated the frequency of EGFR and KRAS mutations in a large Brazilian series of lung adenocarcinoma together with patients’ genetic ancestry, clinicopathological and sociodemographic characteristics. The mutational frequency of EGFR was 22.7% and KRAS was 20.4%. The average ancestry proportions were 73.1% for EUR, 13.1% for AFR, 6.5% for AME and 7.3% for ASN. EGFR mutations were independently associated with never-smokers, high-Asian ancestry, and better performance status. KRAS mutations were independently associated with tobacco exposure and non-Asian ancestry. EGFR-exon 20 mutations were associated with worse outcome. The Cox regression model indicated a worse outcome for patients whose were older at diagnosis (>61 y), solid histological subtype, loss of weight (>10%), worse performance status (≥2), and presence of KRAS mutations and EGFR mutational status in TKi non-treated patients. In conclusion, we assessed the clinicopathological and ethnic impact of EGFR and KRAS mutations in the largest series reported of Brazilian lung adenocarcinomas. These findings can support future clinical strategies for Brazilian lung cancer patients.
Collapse
|
23
|
de Carvalho AC, de Mattos Pereira L, Datorre JG, dos Santos W, Berardinelli GN, Matsushita MDM, Oliveira MA, Durães RO, Guimarães DP, Reis RM. Microbiota Profile and Impact of Fusobacterium nucleatum in Colorectal Cancer Patients of Barretos Cancer Hospital. Front Oncol 2019; 9:813. [PMID: 31555583 PMCID: PMC6727361 DOI: 10.3389/fonc.2019.00813] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/08/2019] [Indexed: 12/24/2022] Open
Abstract
Microbial diversity has been pointed out as a major factor in the development and progression of colorectal cancer (CRC). We sought to explore the richness and abundance of the microbial community of a series of colorectal tumor samples treated at Barretos Cancer Hospital, Brazil, through 16S rRNA sequencing. The presence and the impact of Fusobacterium nucleatum (Fn) DNA in CRC prognosis was further evaluated by qPCR in a series of 152 CRC cases. An enrichment for potentially oncogenic bacteria in CRC was observed, with Fusobacterium being the most abundant genus in the tumor tissue. In the validation dataset, Fn was detected in 35/152 (23.0%) of fresh-frozen tumor samples and in 6/57 (10.5%) of paired normal adjacent tissue, with higher levels in the tumor (p = 0.0033). Fn DNA in the tumor tissue was significantly associated with proximal tumors (p = 0.001), higher depth of invasion (p = 0.014), higher clinical stages (p = 0.033), poor differentiation (p = 0.011), MSI-positive status (p < 0.0001), BRAF mutated tumors (p < 0.0001), and the loss of expression of mismatch-repair proteins MLH1 (p < 0.0001), MSH2 (p = 0.003), and PMS2 (p < 0.0001). Moreover, the presence of Fn DNA in CRC tissue was also associated with a worse patient cancer-specific survival (69.9 vs. 82.2% in 5 years; p = 0.028) and overall survival (63.5 vs. 76.5%; p = 0.037). Here we report, for the first time, the association of F. nucleatum presence with important clinical and molecular features in a Brazilian cohort of CRC patients. Tumor detection and classification based on the gut microbiome might provide a promising approach to improve the prediction of patient outcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Denise Peixoto Guimarães
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Department of Prevention, Barretos Cancer Hospital, Barretos, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's–PT Government Associate Laboratory, Braga, Portugal
- *Correspondence: Rui Manuel Reis
| |
Collapse
|
24
|
Proença MA, Biselli JM, Succi M, Severino FE, Berardinelli GN, Caetano A, Reis RM, Hughes DJ, Silva AE. Relationship between Fusobacterium nucleatum, inflammatory mediators and microRNAs in colorectal carcinogenesis. World J Gastroenterol 2018; 24:5351-5365. [PMID: 30598580 PMCID: PMC6305535 DOI: 10.3748/wjg.v24.i47.5351] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/29/2018] [Accepted: 12/13/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To examine the effect of Fusobacterium nucleatum (F. nucleatum) on the microenvironment of colonic neoplasms and the expression of inflammatory mediators and microRNAs (miRNAs).
METHODS Levels of F. nucleatum DNA, cytokine gene mRNA (TLR2, TLR4, NFKB1, TNF, IL1B, IL6 and IL8), and potentially interacting miRNAs (miR-21-3p, miR-22-3p, miR-28-5p, miR-34a-5p, miR-135b-5p) were measured by quantitative polymerase chain reaction (qPCR) TaqMan® assays in DNA and/or RNA extracted from the disease and adjacent normal fresh tissues of 27 colorectal adenoma (CRA) and 43 colorectal cancer (CRC) patients. KRAS mutations were detected by direct sequencing and microsatellite instability (MSI) status by multiplex PCR. Cytoscape v3.1.1 was used to construct the postulated miRNA:mRNA interaction network.
RESULTS Overabundance of F. nucleatum in neoplastic tissue compared to matched normal tissue was detected in CRA (51.8%) and more markedly in CRC (72.1%). We observed significantly greater expression of TLR4, IL1B, IL8, and miR-135b in CRA lesions and TLR2, IL1B, IL6, IL8, miR-34a and miR-135b in CRC tumours compared to their respective normal tissues. Only two transcripts for miR-22 and miR-28 were exclusively downregulated in CRC tumour samples. The mRNA expression of IL1B, IL6, IL8 and miR-22 was positively correlated with F. nucleatum quantification in CRC tumours. The mRNA expression of miR-135b and TNF was inversely correlated. The miRNA:mRNA interaction network suggested that the upregulation of miR-34a in CRC proceeds via a TLR2/TLR4-dependent response to F. nucleatum. Finally, KRAS mutations were more frequently observed in CRC samples infected with F. nucleatum and were associated with greater expression of miR-21 in CRA, while IL8 was upregulated in MSI-high CRC.
CONCLUSION Our findings indicate that F. nucleatum is a risk factor for CRC by increasing the expression of inflammatory mediators through a possible miRNA-mediated activation of TLR2/TLR4.
Collapse
Affiliation(s)
- Marcela Alcântara Proença
- Department of Biology, UNESP, Univ. Estadual Paulista, Campus of São José do Rio Preto, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Joice Matos Biselli
- Department of Biology, UNESP, Univ. Estadual Paulista, Campus of São José do Rio Preto, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Maysa Succi
- Department of Biology, UNESP, Univ. Estadual Paulista, Campus of São José do Rio Preto, São José do Rio Preto, São Paulo 15054-000, Brazil
| | - Fábio Eduardo Severino
- Department of Surgery and Orthopedics, Faculty of Medicine, UNESP, Univ. Estadual Paulista, Campus of Botucatu, Botucatu, São Paulo 18618-687, Brazil
| | | | - Alaor Caetano
- Endoscopy Center of Rio Preto, São José do Rio Preto, São Paulo 15015-700, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil
- Life and Health Sciences Research Institute, University of Minho, Campus Gualtar, Braga 4710-057, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Campus Gualtar, Braga 4710-057, Portugal
| | - David J Hughes
- Cancer Biology and Therapeutics Group, UCD Conway Institute, University College Dublin, Dublin D04 V1W8, Ireland
| | - Ana Elizabete Silva
- Department of Biology, UNESP, Univ. Estadual Paulista, Campus of São José do Rio Preto, São José do Rio Preto, São Paulo 15054-000, Brazil
| |
Collapse
|
25
|
Niu W, Wang G, Feng J, Li Z, Li C, Shan B. Correlation between microsatellite instability and RAS gene mutation and stage III colorectal cancer. Oncol Lett 2018; 17:332-338. [PMID: 30655771 PMCID: PMC6313205 DOI: 10.3892/ol.2018.9611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023] Open
Abstract
Correlation between RAS gene mutation and microsatellite instability (MSI) status in cancer tissues and clinicopathological parameters of patients with stage III colorectal cancer (CRC) were investigated. Tissues were collected from 180 patients diagnosed with stage III CRC in the Department of Gastrointestinal Surgery of the Fourth Hospital of Hebei Medical University from 2012 to 2016. RAS gene mutations in paraffin sections were detected by PCR and Sanger sequencing. Expression of mismatch repair proteins MLH1, MSH2, MSH6 and PMS2 was detected by immunohistochemistry, and MSI status was determined based on the positive and negative expression combinations of the above proteins, and the correlation with clinicopathological parameters of CRC was analyzed. Mutation rates of KRAS and NRAS were 48.33% (87/180) and 2.78% (5/180), respectively. Mutation rate of p.G12D in codon 12 of exon 2 in KRAS gene was the highest (31/87, 35.63%). Mutation rate of p.G12D in codon 12 of exon 2 in NRAS gene was the highest (2/5, 40%). Mutation rate of KRAS gene in right colon was higher than that in left colon and rectum (p<0.05), and mutation rate in N2b phase was higher than that in N2a and N1 phases (p<0.01). In low degree of microsatellite instability (MSI-L) and high degree of microsatellite instability (MSI-H) status, negative MKH1 protein expression was dominant (18/32, 56.25%). MSI-H in CRC patients aged ≥50 years was higher than that of CRC patients <50 years. Rates of MSI-H in N1, N2a, and N2b were 1.75, 12.82, and 1.11% (p<0.05). Mutation rate of KRAS gene in MSI-H status of stage III CRC patients was significantly higher than that in MSI-L/microsatellite stability (MSS) (p<0.05). Mutation of RAS gene and the status of MSI are involved in the occurrence and development of stage III CRC. Detection of RAS gene has important significance for the individual treatment of CRC in clinic.
Collapse
Affiliation(s)
- Wenbo Niu
- Department of Surgery Ⅱ, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Guiying Wang
- Department of Surgery Ⅱ, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jun Feng
- Department of Surgery Ⅱ, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Zheng Li
- Department of Surgery Ⅱ, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Chenhui Li
- Department of Surgery Ⅱ, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Baoen Shan
- Center of Scientific Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
26
|
Corcoran RB, André T, Atreya CE, Schellens JHM, Yoshino T, Bendell JC, Hollebecque A, McRee AJ, Siena S, Middleton G, Muro K, Gordon MS, Tabernero J, Yaeger R, O'Dwyer PJ, Humblet Y, De Vos F, Jung AS, Brase JC, Jaeger S, Bettinger S, Mookerjee B, Rangwala F, Van Cutsem E. Combined BRAF, EGFR, and MEK Inhibition in Patients with BRAFV600E-Mutant Colorectal Cancer. Cancer Discov 2018; 8:428-443. [PMID: 29431699 PMCID: PMC5882509 DOI: 10.1158/2159-8290.cd-17-1226] [Citation(s) in RCA: 436] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/21/2018] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
Abstract
Although BRAF inhibitor monotherapy yields response rates >50% in BRAFV600-mutant melanoma, only approximately 5% of patients with BRAFV600E colorectal cancer respond. Preclinical studies suggest that the lack of efficacy in BRAFV600E colorectal cancer is due to adaptive feedback reactivation of MAPK signaling, often mediated by EGFR. This clinical trial evaluated BRAF and EGFR inhibition with dabrafenib (D) + panitumumab (P) ± MEK inhibition with trametinib (T) to achieve greater MAPK suppression and improved efficacy in 142 patients with BRAFV600E colorectal cancer. Confirmed response rates for D+P, D+T+P, and T+P were 10%, 21%, and 0%, respectively. Pharmacodynamic analysis of paired pretreatment and on-treatment biopsies found that efficacy of D+T+P correlated with increased MAPK suppression. Serial cell-free DNA analysis revealed additional correlates of response and emergence of KRAS and NRAS mutations on disease progression. Thus, targeting adaptive feedback pathways in BRAFV600E colorectal cancer can improve efficacy, but MAPK reactivation remains an important primary and acquired resistance mechanism.Significance: This trial demonstrates that combined BRAF + EGFR + MEK inhibition is tolerable, with promising activity in patients with BRAFV600E colorectal cancer. Our findings highlight the MAPK pathway as a critical target in BRAFV600E colorectal cancer and the need to optimize strategies inhibiting this pathway to overcome both primary and acquired resistance. Cancer Discov; 8(4); 428-43. ©2018 AACR.See related commentary by Janku, p. 389See related article by Hazar-Rethinam et al., p. 417This article is highlighted in the In This Issue feature, p. 371.
Collapse
Affiliation(s)
- Ryan B Corcoran
- Massachusetts General Hospital Cancer Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Thierry André
- Hôpital Saint-Antoine, and Sorbonne Universités, Paris, France
| | | | | | | | - Johanna C Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | | | - Autumn J McRee
- University of North Carolina, Chapel Hill, North Carolina
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Osopedale Metropolitano Niguarda and Department of Oncology and Hemato-Oncollogy, Università degli Studi di Milano, Milan, Italy
| | - Gary Middleton
- University of Birmingham and University Hospital, Birmingham, United Kingdom
| | - Kei Muro
- Aichi Cancer Center Hospital, Nagoya, Japan
| | | | | | - Rona Yaeger
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Peter J O'Dwyer
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Filip De Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | | | - Savina Jaeger
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | - Fatima Rangwala
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey
| | | |
Collapse
|
27
|
Cruvinel-Carloni A, Yamane L, Scapulatempo-Neto C, Guimarães D, Reis RM. Absence of TERT promoter mutations in colorectal precursor lesions and cancer. Genet Mol Biol 2018; 41:82-84. [PMID: 29473934 PMCID: PMC5901499 DOI: 10.1590/1678-4685-gmb-2017-0133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 10/13/2017] [Indexed: 06/08/2025] Open
Abstract
Hotspot mutations (c.-124bp G > A and c.-146bp G > A) in the promoter region of the TERT gene have been recently described in several types of solid tumors, including glioma, bladder, thyroid, liver and skin neoplasms. However, knowledge with respect to colorectal precursor lesions and cancer is scarce. In the present study we aimed to determine the frequency of hotspot TERT promoter mutations in 145 Brazilian patients, including 103 subjects with precursor lesions and 42 with colorectal carcinomas, and we associated the presence of such mutations with the patients clinical-pathological features. The mutation analysis was conclusive in 123 cases, and none of the precursor and colorectal carcinoma cases showed TERT promoter mutations. We conclude that TERT mutations are not a driving factor in colorectal carcinogenesis.
Collapse
Affiliation(s)
| | - Letícia Yamane
- Molecular Oncology Research Center, Hospital de Câncer de Barretos, Barretos, SP, Brazil
| | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Hospital de Câncer de Barretos, Barretos, SP, Brazil.,Department of Pathology, Hospital de Câncer de Barretos, Barretos, São Paulo, Brazil
| | - Denise Guimarães
- Molecular Oncology Research Center, Hospital de Câncer de Barretos, Barretos, SP, Brazil.,Department of Endoscopy, Hospital de Câncer de Barretos, Barretos, São Paulo, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Hospital de Câncer de Barretos, Barretos, SP, Brazil.,Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
28
|
KRAS and BRAF somatic mutations in colonic polyps and the risk of metachronous neoplasia. PLoS One 2017; 12:e0184937. [PMID: 28953955 PMCID: PMC5617162 DOI: 10.1371/journal.pone.0184937] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 09/01/2017] [Indexed: 01/09/2023] Open
Abstract
Background & aims High-risk features of colonic polyps are based on size, number, and pathologic characteristics. Surveillance colonoscopy is often recommended according to these findings. This study aimed to determine whether the molecular characteristics of polyps might provide information about the risk of metachronous advanced neoplasia. Methodology We retrospectively included 308 patients with colonic polyps. A total of 995 polyps were collected and tested for somatic BRAF and KRAS mutations. Patients were classified into 3 subgroups, based on the polyp mutational profile at baseline, as follows: non-mutated polyps (Wild-type), at least one BRAF-mutated polyp, or at least one KRAS-mutated polyp. At surveillance, advanced adenomas were defined as adenomas ≥ 10 mm and/or with high grade dysplasia or a villous component. In contrast, advanced serrated polyps were defined as serrated polyps ≥ 10 mm in any location, located proximal to the splenic flexure with any size or with dysplasia. Results At baseline, 289 patients could be classified as wild-type (62.3%), BRAF mutated (14.9%), or KRAS mutated (22.8%). In the univariate analysis, KRAS mutations were associated with the development of metachronous advanced polyps (OR: 2.36, 95% CI: 1.22–4.58; P = 0.011), and specifically, advanced adenomas (OR: 2.42, 95% CI: 1.13–5.21; P = 0.023). The multivariate analysis, adjusted for age and sex, also showed associations with the development of metachronous advanced polyps (OR: 2.27, 95% CI: 1.15–4.46) and advanced adenomas (OR: 2.23, 95% CI: 1.02–4.85). Conclusions Our results suggested that somatic KRAS mutations in polyps represent a potential molecular marker for the risk of developing advanced neoplasia.
Collapse
|
29
|
Basso G, Bianchi P, Malesci A, Laghi L. Hereditary or sporadic polyposis syndromes. Best Pract Res Clin Gastroenterol 2017; 31:409-417. [PMID: 28842050 DOI: 10.1016/j.bpg.2017.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 05/31/2017] [Indexed: 01/31/2023]
Abstract
Polyposis syndromes are encountered in endoscopy practice, and are considered rare entities, accounting for ≤1% of colorectal cancer. Polyposis can occur within inherited syndromes or as "sporadic" cases of unknown etiology. Their proper characterization is relevant for patient management, and should nowadays drive appropriate genetic tests which have a key role in clinical practice for driving surveillance and colorectal cancer prevention, enlarged to relatives. Polyposis classification is based upon polyp number and histology, familial and personal history. This review will explore the polyposis nosology and their genetic determinants in the emerging scenario of Next Generation Sequencing which allow testing multiples genes in parallel. This capability will likely continue to increase the range of polyposis predisposing genes, contributing to define new clinical entities.
Collapse
Affiliation(s)
- Gianluca Basso
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Paolo Bianchi
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Alberto Malesci
- Department of Biotechnologies and Translational Medicine, University of Milan, Via Vanvitelli 32, 20133 Milan, Italy; Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy; Department of Gastroenterology, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy; Hereditary Cancer Genetics Clinic, Humanitas Cancer Center, Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano (Milan), Italy.
| |
Collapse
|
30
|
AKT can modulate the in vitro response of HNSCC cells to irreversible EGFR inhibitors. Oncotarget 2017; 8:53288-53301. [PMID: 28881811 PMCID: PMC5581110 DOI: 10.18632/oncotarget.18395] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 05/16/2017] [Indexed: 12/14/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is overexpressed in up to 90% of head and neck squamous cell carcinoma (HNSCC) tumors. Cetuximab is the first targeted (anti-EGFR) therapy approved for the treatment of HNSCC patients. However, its efficacy is limited due to primary and secondary resistance, and there is no predict biomarkers of response. New generation of EGFR inhibitors with pan HER targeting and irreversible action, such as afatinib and allitinib, represents a significant therapeutic promise. In this study, we intend to compare the potential cytotoxicity of two anti-EGFR inhibitors (afatinib and allitinib) with cetuximab and to identify potential predictive biomarkers of response in a panel of HNSCC cell lines. The mutational analysis in the eight HNSCC cell lines revealed an EGFR mutation (p.H773Y) and gene amplification in the HN13 cells. According to the growth inhibition score (GI), allitinib was the most cytotoxic drug, followed by afatinib and finally cetuximab. The higher AKT phosphorylation level was associated with resistance to anti-EGFR agents. Therefore, we further performed drug combinations with anti-AKT agent (MK2206) and AKT1 gene editing, which demonstrated afatinib and allitinib sensitivity restored. Additionally, in silico analysis of TCGA database showed that AKT1 overexpression was present in 14.7% (41/279) of HNSCC cases, and was associated with perineural invasion in advanced stage. In conclusion, allitinib presented a greater cytotoxic profile when compared to afatinib and cetuximab. AKT pathway constitutes a predictive marker of allitinib response and combination with AKT inhibitors could restore response and increase treatment success.
Collapse
|
31
|
Feasibility of Large-Scale Identification of Sessile Serrated Polyp Patients Using Electronic Records: A Utah Study. Dig Dis Sci 2017; 62:1455-1463. [PMID: 28315031 DOI: 10.1007/s10620-017-4543-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 03/10/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND/AIMS The serrated pathway accounts for 15-25% of sporadic colorectal cancer (CRC). In our study, we sought to accurately characterize sessile serrated polyps (SSP) in a population by electronically interrogating colonoscopy patients' endoscopy and pathology reports using a rules-based text search of pre-defined SSP-related terms. To this aim, we compared a sample of putative SSP and hyperplastic polyps (HP) using our algorithm to a determination of SSP or HP by pathologist and molecular examination to determine the feasibility of large-scale identification of SSP in electronic medical records. METHODS In 23,990 endoscopy reports from colonoscopies with pathology performed at a University of Utah Healthcare facility in 2000-2012, we identified serrated lesions and categorized each as putative SSP or HP using a text search algorithm. We obtained 93 tissue samples for histologic and molecular analysis. RESULTS Serrated polyps were categorized as putative SSP (N = 920) and putative HP (N = 7159) by text search algorithm. Histologic examination of 93 samples identified 37 SSP, 11 probable SSP, and 45 HP. Of 26 putative SSP, 25 were SSP/probable SSP (96%) by histology. Of 67 putative HP, 44 were HP (66%) by histology. Reducing size criterion from ≥1 to ≥5 mm in the search algorithm caused improved sensitivity (77.1%) without decline in specificity (97.8%). CONCLUSIONS A simple rules-based search to identify SSP provides "proof of principle" that SSP can be identified in a large electronic record set. Pilot data indicate defining large, right-sided polyps as ≥5 mm provides adequate sensitivity to detect SSP from electronic records while maintaining high specificity.
Collapse
|
32
|
Evangelista AF, Zanon MF, Carloni AC, de Paula FE, Morini MA, Ferreira-Neto M, Soares IC, Miziara JE, de Marchi P, Scapulatempo-Neto C, Reis RM. Detection of ALK fusion transcripts in FFPE lung cancer samples by NanoString technology. BMC Pulm Med 2017; 17:86. [PMID: 28549458 PMCID: PMC5446704 DOI: 10.1186/s12890-017-0428-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 05/19/2017] [Indexed: 11/29/2022] Open
Abstract
Background ALK-rearranged lung cancers exhibit specific pathologic and clinical features and are responsive to anti-ALK therapies. Therefore, the detection of ALK-rearrangement is fundamental for personalized lung cancer therapy. Recently, new molecular techniques, such as NanoString nCounter, have been developed to detect ALK fusions with more accuracy and sensitivity. Methods In the present study, we intended to validate a NanoString nCounter ALK-fusion panel in routine biopsies of FFPE lung cancer patients. A total of 43 samples were analyzed, 13 ALK-positive and 30 ALK-negative, as previously detected by FISH and/or immunohistochemistry. Results The NanoString panel detected the presence of the EML4-ALK, KIF5B-ALK and TFG-ALK fusion variants. We observed that all the 13 ALK-positive cases exhibited genetic aberrations by the NanoString methodology. Namely, six cases (46.15%) presented EML-ALK variant 1, two (15.38%) presented EML-ALK variant 2, two (15.38%) presented EML-ALK variant 3a, and three (23.07%) exhibited no variant but presented unbalanced expression between 5’/3’ exons, similar to other positive samples. Importantly, for all these analyses, the initial input of RNA was 100 ng, and some cases displayed poor RNA quality measurements. Conclusions In this study, we reported the great utility of NanoString technology in the assessment of ALK fusions in routine lung biopsies of FFPE specimens. Electronic supplementary material The online version of this article (doi:10.1186/s12890-017-0428-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adriane F Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Maicon F Zanon
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Adriana Cruvinel Carloni
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Flávia E de Paula
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Mariana Andozia Morini
- Department of Pathology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Maressa Ferreira-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Iberê Cauduro Soares
- Department of Pathology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Jose Elias Miziara
- Department of Thoracic Surgery, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Pedro de Marchi
- Department of Clinical Oncology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, Sao Paulo, Brazil
| | - Cristovam Scapulatempo-Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil.,Department of Pathology, Barretos Cancer Hospital, Rua Antenor Duarte Villela 1331, Barretos, CEP 14784-400, São Paulo, Brazil
| | - Rui M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Rua Antenor Duarte Villela, 1331, Barretos, CEP 14784-400, São Paulo, Brazil. .,Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, 4710-057, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, 4710-057, Portugal.
| |
Collapse
|
33
|
Gao C. Molecular pathological epidemiology in diabetes mellitus and risk of hepatocellular carcinoma. World J Hepatol 2016; 8:1119-1127. [PMID: 27721917 PMCID: PMC5037325 DOI: 10.4254/wjh.v8.i27.1119] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/28/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023] Open
Abstract
Molecular pathological epidemiology (MPE) is a multidisciplinary and transdisciplinary study field, which has emerged as an integrated approach of molecular pathology and epidemiology, and investigates the relationship between exogenous and endogenous exposure factors, tumor molecular signatures, and tumor initiation, progression, and response to treatment. Molecular epidemiology broadly encompasses MPE and conventional-type molecular epidemiology. Hepatocellular carcinoma (HCC) is the third most common cause of cancer-associated death worldwide and remains as a major public health challenge. Over the past few decades, a number of epidemiological studies have demonstrated that diabetes mellitus (DM) is an established independent risk factor for HCC. However, how DM affects the occurrence and development of HCC remains as yet unclearly understood. MPE may be a promising approach to investigate the molecular mechanisms of carcinogenesis of DM in HCC, and provide some useful insights for this pathological process, although a few challenges must be overcome. This review highlights the recent advances in this field, including: (1) introduction of MPE; (2) HCC, risk factors, and DM as an established independent risk factor for HCC; (3) molecular pathology, molecular epidemiology, and MPE in DM and HCC; and (4) MPE studies in DM and risk of HCC. More MPE studies are expected to be performed in future and I believe that this field can provide some very important insights on the molecular mechanisms, diagnosis, personalized prevention and treatment for DM and risk of HCC.
Collapse
|
34
|
Bidinotto LT, Véo CAR, Loaiza EA, De França APS, Lorenzi AT, Rosa LAR, De Oliveira CM, Levi JE, Scapulatempo-Neto C, Longatto-Filho A, Reis RM. Low mutation percentage of KRAS and BRAF genes in Brazilian anal tumors. Mol Med Rep 2016; 14:3791-7. [PMID: 27573925 DOI: 10.3892/mmr.2016.5684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/01/2016] [Indexed: 11/05/2022] Open
Abstract
Anal cancer is a rare type of digestive tract disease, which has had a crescent incidence in a number of regions. Carcinomas are most frequently found, with squamous cell carcinoma (SCC) comprising ~95% of all anal tumors. The major risk factor for development of this type of tumor is human papillomavirus (HPV) infection. However, previous studies have identified patients with anal cancer that are HPV‑/p16‑and observed that they have a poorer outcome compared with HPV+/p16+ patients. This suggests that molecular profile may drive anal cancer progression. The aim of the present study was to evaluate the mutational status of two important oncogenes, KRAS and BRAF, in a series of anal cancer lesions. Resected tumors of the anal canal (n=43) were evaluated, nine of these were high‑grade squamous intra‑epithelial lesion cases (HSIL), 11 were adenocarcinomas, and 23 SCCs. Direct sequencing of KRAS proto‑oncogene, GTPase (KRAS; codons 12 and 13) and B‑Raf proto‑oncogene, serine/threonine kinase (BRAF; codon 600) was performed and associated with patient clinicopathological and molecular features. There was a trend of poorer prognosis of adenocarcinoma compared with HSIL and SCC. Analysis indicated one SCC patient (2.3%) exhibited a KRAS p.G13D mutation, and one adenocarcinoma patient (2.3%) exhibited a BRAF p.V600E mutation. It was observed that, these mutations are rare in anal tumors, and certain patients may be at a disadvantage using targeted therapies based on KRAS and BRAF mutational status. As there is a low mutation percentage in SCCs, adenocarcinomas and HSIL, there may exist other underlying molecular alterations that result in anal cancer development, which require further elucidation.
Collapse
Affiliation(s)
- Lucas Tadeu Bidinotto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil
| | - Carlos A R Véo
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil
| | - Edgar Aleman Loaiza
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil
| | | | - Adriana Tarla Lorenzi
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil
| | - Luciana Albina Reis Rosa
- Laboratory of Virology, Institute of Tropical Medicine, University of São Paulo, São Paulo 05403 000, Brazil
| | - Cristina Mendes De Oliveira
- Laboratory of Virology, Institute of Tropical Medicine, University of São Paulo, São Paulo 05403 000, Brazil
| | - José Eduardo Levi
- Laboratory of Virology, Institute of Tropical Medicine, University of São Paulo, São Paulo 05403 000, Brazil
| | | | - Adhemar Longatto-Filho
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784 400, Brazil
| |
Collapse
|
35
|
Brown DG, Rao S, Weir TL, O'Malia J, Bazan M, Brown RJ, Ryan EP. Metabolomics and metabolic pathway networks from human colorectal cancers, adjacent mucosa, and stool. Cancer Metab 2016; 4:11. [PMID: 27275383 PMCID: PMC4893840 DOI: 10.1186/s40170-016-0151-y] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/16/2016] [Indexed: 12/18/2022] Open
Abstract
Background Colorectal cancers (CRC) are associated with perturbations in cellular amino acids, nucleotides, pentose-phosphate pathway carbohydrates, and glycolytic, gluconeogenic, and tricarboxylic acid intermediates. A non-targeted global metabolome approach was utilized for exploring human CRC, adjacent mucosa, and stool. In this pilot study, we identified metabolite profile differences between CRC and adjacent mucosa from patients undergoing colonic resection. Metabolic pathway analyses further revealed relationships between complex networks of metabolites. Methods Seventeen CRC patients participated in this pilot study and provided CRC, adjacent mucosa ~10 cm proximal to the tumor, and stool. Metabolomes were analyzed by gas chromatography-mass spectrometry (GC/MS) and ultra-performance liquid chromatography-mass spectrometry (UPLC-MS/MS). All of the library standard identifications were confirmed and further analyzed via MetaboLyncTM for metabolic network interactions. Results There were a total of 728 distinct metabolites identified from colonic tissue and stool matrices. Nineteen metabolites significantly distinguished CRC from adjacent mucosa in our patient-matched cohort. Glucose-6-phosphate and fructose-6-phosphate demonstrated 0.64-fold and 0.75-fold lower expression in CRC compared to mucosa, respectively, whereas isobar: betaine aldehyde, N-methyldiethanolamine, and adenylosuccinate had 2.68-fold and 1.88-fold higher relative abundance in CRC. Eleven of the 19 metabolites had not previously been reported for CRC relevance. Metabolic pathway analysis revealed significant perturbations of short-chain fatty acid metabolism, fructose, mannose, and galactose metabolism, and glycolytic, gluconeogenic, and pyruvate metabolism. In comparison to the 500 stool metabolites identified from human CRC patients, only 215 of those stool metabolites were also detected in tissue. This CRC and stool metabolome investigation identified novel metabolites that may serve as key small molecules in CRC pathogenesis, confirmed the results from previously reported CRC metabolome studies, and showed networks for metabolic pathway aberrations. In addition, we found differences between the CRC and stool metabolomes. Conclusions Stool metabolite profiles were limited for direct associations with CRC and adjacent mucosa, yet metabolic pathways were conserved across both matrices. Larger patient-matched CRC, adjacent non-cancerous colonic mucosa, and stool cohort studies for metabolite profiling are needed to validate these small molecule differences and metabolic pathway aberrations for clinical application to CRC control, treatment, and prevention. Electronic supplementary material The online version of this article (doi:10.1186/s40170-016-0151-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dustin G Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University, 200 West Lake Street, 1680 Campus Delivery, Fort Collins, CO 80523 USA
| | - Sangeeta Rao
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523 USA
| | - Tiffany L Weir
- Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523 USA
| | - Joanne O'Malia
- University of Colorado Health-North, Fort Collins, CO 80522 USA
| | - Marlon Bazan
- University of Colorado Health-North, Fort Collins, CO 80522 USA
| | - Regina J Brown
- Division of Medical Oncology, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, 200 West Lake Street, 1680 Campus Delivery, Fort Collins, CO 80523 USA ; Department of Clinical Sciences, Colorado State University, Fort Collins, CO 80523 USA
| |
Collapse
|
36
|
Cárcano FM, Lengert AH, Vidal DO, Scapulatempo Neto C, Queiroz L, Marques H, Baltazar F, Berardinelli GN, Martinelli CMS, da Silva ECA, Reis RM, Lopes LF. Absence of microsatellite instability and BRAF (V600E) mutation in testicular germ cell tumors. Andrology 2016; 4:866-72. [PMID: 27153176 DOI: 10.1111/andr.12200] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 03/16/2016] [Accepted: 03/18/2016] [Indexed: 12/27/2022]
Abstract
Testicular germ cell tumors (TGCT) are the most common malignant neoplasm in young men. DNA mismatch repair deficiency can lead to microsatellite instability (MSI), an important mechanism of genetic instability. A mutation of the BRAF gene has been implicated in the pathogenesis of several solid tumors and has recently become an important therapeutic target. The role of MSI and BRAF gene mutation in TGCT, particularly in refractory disease, is poorly understood and reported findings are controversial. In this study, we aimed to determine the frequency and clinical impact of MSI status and BRAF mutations in TGCT. DNA was isolated from formalin-fixed paraffin embedded (FFPE) tissue from 150 TGCT cases. The MSI phenotype was evaluated using multiplex PCR for five quasimonomorphic mononucleotide repeat markers. Exon 15 of the BRAF oncogene (V600E) was analyzed by PCR, followed by direct sequencing. Sixteen percent of cases were considered to have refractory disease. In a small subset of cases (17 for MSI and 18 for BRAF), the quantity and quality of DNA recovery were poor and therefore, were unable to be analyzed. The remaining 133 TGCT cases showed a complete absence of MSI. Of the 132 cases successfully evaluated for BRAF mutations, all were V600E wild-type. In conclusion, despite a distinct response of testicular germ cell tumors to therapy, microsatellite instability, and the BRAF V600E mutation were absent in all testicular germ cell tumors tested in this study.
Collapse
Affiliation(s)
- F M Cárcano
- Department of Medical Oncology, Barretos Cancer Hospital, Barretos, Brazil.,Barretos School of Health Sciences, Dr. Paulo Prata - FACISB, Barretos, Brazil
| | - A H Lengert
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos Children's Cancer Hospital, Barretos, Brazil
| | - D O Vidal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos Children's Cancer Hospital, Barretos, Brazil
| | - C Scapulatempo Neto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - L Queiroz
- Department of Medical Oncology, Hospital de Braga, Braga, Guimarães, Portugal
| | - H Marques
- Department of Medical Oncology, Hospital de Braga, Braga, Guimarães, Portugal
| | - F Baltazar
- Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - G N Berardinelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - C M S Martinelli
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - E C A da Silva
- Department of Pathology, Barretos Cancer Hospital, Barretos, Brazil
| | - R M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - L F Lopes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil.,Barretos Children's Cancer Hospital, Barretos, Brazil
| |
Collapse
|
37
|
Pinheiro C, Miranda-Gonçalves V, Longatto-Filho A, Vicente ALSA, Berardinelli GN, Scapulatempo-Neto C, Costa RFA, Viana CR, Reis RM, Baltazar F, Vazquez VL. The metabolic microenvironment of melanomas: Prognostic value of MCT1 and MCT4. Cell Cycle 2016; 15:1462-70. [PMID: 27105345 DOI: 10.1080/15384101.2016.1175258] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BRAF mutations are known drivers of melanoma development and, recently, were also described as players in the Warburg effect, while this reprogramming of energy metabolism has been identified as a possible strategy for treating melanoma patients. Therefore, the aim of this work was to evaluate the expression and prognostic value of a panel of glycolytic metabolism-related proteins in a series of melanomas. The immunohistochemical expression of MCT1, MCT4, GLUT1, and CAIX was evaluated in 356 patients presenting melanoma and 20 patients presenting benign nevi. Samples included 20 benign nevi, 282 primary melanomas, 117 lymph node and 54 distant metastases samples. BRAF mutation was observed in 29/92 (31.5%) melanoma patients and 17/20 (85%) benign nevi samples. NRAS mutation was observed in 4/36 (11.1%) melanoma patients and 1/19 (5.3%) benign nevi samples. MCT4 and GLUT1 expression was significantly increased in metastatic samples, and MCT1, MCT4 and GLUT1 were significantly associated with poor prognostic variables. Importantly, MCT1 and MCT4 were associated with shorter overall survival. In conclusion, the present study brings new insights on metabolic aspects of melanoma, paving the way for the development of new-targeted therapies.
Collapse
Affiliation(s)
- Céline Pinheiro
- a Barretos School of Health Sciences, Dr. Paulo Prata - FACISB , Barretos , São Paulo , Brazil.,b Molecular Oncology Research Center, Barretos Cancer Hospital , Barretos , São Paulo , Brazil
| | - Vera Miranda-Gonçalves
- c Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga , Portugal.,d ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
| | - Adhemar Longatto-Filho
- b Molecular Oncology Research Center, Barretos Cancer Hospital , Barretos , São Paulo , Brazil.,c Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga , Portugal.,d ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal.,e Laboratory of Medical Investigation (LIM-14), School of Medicine, University of São Paulo , São Paulo , Brazil
| | - Anna L S A Vicente
- b Molecular Oncology Research Center, Barretos Cancer Hospital , Barretos , São Paulo , Brazil
| | - Gustavo N Berardinelli
- b Molecular Oncology Research Center, Barretos Cancer Hospital , Barretos , São Paulo , Brazil
| | | | - Ricardo F A Costa
- a Barretos School of Health Sciences, Dr. Paulo Prata - FACISB , Barretos , São Paulo , Brazil
| | - Cristiano R Viana
- f Pathology Department , Barretos Cancer Hospital , Barretos , São Paulo , Brazil
| | - Rui M Reis
- b Molecular Oncology Research Center, Barretos Cancer Hospital , Barretos , São Paulo , Brazil.,c Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga , Portugal.,d ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
| | - Fátima Baltazar
- c Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga , Portugal.,d ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães , Portugal
| | - Vinicius L Vazquez
- b Molecular Oncology Research Center, Barretos Cancer Hospital , Barretos , São Paulo , Brazil.,g Surgery Department , Melanoma/Sarcoma, Barretos Cancer Hospital , Barretos , São Paulo , Brazil
| |
Collapse
|
38
|
Vazquez VDL, Vicente AL, Carloni A, Berardinelli G, Soares P, Scapulatempo C, Martinho O, Reis RM. Molecular profiling, including TERT promoter mutations, of acral lentiginous melanomas. Melanoma Res 2016; 26:93-9. [PMID: 26709572 DOI: 10.1097/cmr.0000000000000222] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Acral lentiginous melanoma (ALM) is the less common subtype with singular characterization. TERT (human telomerase reverse transcriptase) promoter mutations have being described as recurrent in melanomas and infrequent in ALM, but their real incidence and clinical relevance is unclear. The objectives of this study were to describe the prevalence of TERT promoter mutations in ALM, and correlate with the molecular profile of other drive genes and clinical features. Sixty-one samples from 48 patients with ALM were analyzed. After DNA isolation, the mutation profiles of the hotspot region of BRAF, NRAS, KIT, PDGFRA, and TERT genes were determined by PCR amplification followed by direct Sanger sequencing. KIT, PDGFRA, and VEGFR2 gene amplification was performed by quantitative PCR. Clinical information such as survival, clinical stage, and Breslow tumor classification were obtained from medical records. TERT promoter mutations were found in 9.3% of the cases, BRAF in 10.3%, NRAS in 7.5%, KIT in 20.7%, and PDGFRA in 14.8% of ALM. None of the cases showed KIT, PDGFRA, or VEGFR2 gene amplification. We found an association between KIT mutations and advanced Clark level (IV and V, P=0.043) and TERT promoter mutations with low mitotic index. No other significant associations were observed between mutation profile and patients' clinical features nor survival rates. Oncogenic TERT promoter mutations are present in a fraction of ALMs. No relevant associations were found between TERT mutation status and clinical/molecular features nor survival. Mutations of KIT and PDGFRA are the most common genetic alterations, and they can be therapeutic targets for these patients.
Collapse
Affiliation(s)
- Vinicius de Lima Vazquez
- aMolecular Oncology Research Center bDepartment of Surgery, Melanoma and Sarcoma Unity cDepartment of Pathology, Barretos Cancer Hospital, Barretos, São Paulo, Brazil dInstitute of Pathology and Molecular Immunology of University of Porto, (IPATIMUP), Porto eLife and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho fICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
INTRODUCTION Oncogenic hotspot mutations in the promoter region of the TERT gene have been identified in several cancer types as being associated with a worse outcome. Additionally, a polymorphism (rs2853669) in the TERT promoter region was reported to modify the survival of TERT-mutated patients. Our aim is to determine the frequency of c.-124 C>T and c.-146 C>T TERT mutations and to genotype the rs2853669 polymorphism in a series of 68 soft tissue sarcomas (STS) comprising 22 histological subtypes. METHODS PCR was performed, followed by direct sequencing of a fragment of TERT containing the hotspots and the rs2853669. RESULTS We found TERT mutations in 4/68 (5.9%) STSs including 1 pleomorphic liposarcoma (1/1), 1 dedifferentiated liposarcoma (1/1) and 2 myxoid liposarcomas (2/9). The variant C allele of rs2853669 was found in 54.8% (34/62) of all STSs and in 75% (3/4) of TERT-mutated cases. TERT mutations were associated with younger age, and the C allele of the rs2853669 was associated with high histological grade (2 and 3). No association was found between TERT mutation status or rs2853669 genotype and patient prognosis. CONCLUSIONS We showed that TERT promoter mutation is not a recurrent event in STS and is present in particular histological subtypes.
Collapse
|
40
|
Silva-Oliveira RJ, Silva VAO, Martinho O, Cruvinel-Carloni A, Melendez ME, Rosa MN, de Paula FE, de Souza Viana L, Carvalho AL, Reis RM. Cytotoxicity of allitinib, an irreversible anti-EGFR agent, in a large panel of human cancer-derived cell lines: KRAS mutation status as a predictive biomarker. Cell Oncol (Dordr) 2016; 39:253-63. [DOI: 10.1007/s13402-016-0270-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2016] [Indexed: 12/27/2022] Open
|
41
|
Abstract
The hallmarks of premalignant lesions were first described in the 1970s, a time when relatively little was known about the molecular underpinnings of cancer. Yet it was clear there must be opportunities to intervene early in carcinogenesis. A vast array of molecular information has since been uncovered, with much of this stemming from studies of existing cancer or cancer models. Here, examples of how an understanding of cancer biology has informed cancer prevention studies are highlighted and emerging areas that may have implications for the field of cancer prevention research are described. A note of caution accompanies these examples, in that while there are similarities, there are also fundamental differences between the biology of premalignant lesions or premalignant conditions and invasive cancer. These differences must be kept in mind, and indeed leveraged, when exploring potential cancer prevention measures.
Collapse
Affiliation(s)
- Bríd M Ryan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA..
| | | |
Collapse
|
42
|
Cohen JD, Labenski M, Mastrandrea NJ, Canatsey RD, Monks TJ, Lau SS. Transcriptional and post-translational modifications of B-Raf in quinol-thioether induced tuberous sclerosis renal cell carcinoma. Mol Carcinog 2015; 55:1243-50. [PMID: 26333016 DOI: 10.1002/mc.22366] [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: 10/15/2014] [Revised: 06/05/2015] [Accepted: 06/26/2015] [Indexed: 11/12/2022]
Abstract
Increased activity of B-Raf has been identified in approximately 7% of human cancers. Treatment of Eker rats (Tsc-2(EK/+) ), bearing a mutation in one allele of the tuberous sclerosis-2 (Tsc-2) gene, with the nephrocarcinogen 2,3,5-tris-(glutathion-S-yl) hydroquinone (TGHQ) results in loss of the wild-type allele of Tsc-2 in renal preneoplastic lesions and tumors. These tumors have increased protein expression of B-Raf, C-Raf (Raf-1), and increased expression and activity of ERK kinase. Similar changes are observed in Raf kinases following TGHQ-mediated transformation of primary renal epithelial cells derived from Tsc-2(EK/+) rats (QTRRE cells), cells that are also null for tuberin. Herein, we utilized LC-MS/MS to identify constitutive phosphorylation of S345 and S483 in both 100- and 95-kDa forms of B-Raf in QTRRE cells. Using microRotofor liquid-phase isoelectric focusing, we identified four fractions of B-Raf that contain different post-translational modification profiles in QTRRE cells. Amplification of the kinase domain of B-Raf from QTRRE cells, outer-stripe of the outer medulla of 8-month TGHQ- or vehicle-treated Tsc-2(+/+) and Tsc-2(EK/+) rats, as well as tumors excised from 8-month TGHQ-treated Tsc-2(EK/+) rats revealed three splice variants of B-Raf within the kinase domain. These splice variants differed by approximately 340, 544, and 600 bp; confirmed by sequencing. No point mutations within the kinase domain of B-Raf were identified. In addition, B-Raf/Raf-1/14-3-3 complex formation in the QTRRE cells was decreased by sorafenib, with concomitant selective decreases in p-ERK levels. Transcriptional and post-translational characterization of critical kinases, such as B-Raf, may contribute to the progression of tuberous sclerosis RCC. (246/250) © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Southwest Environmental Health Sciences Center, College of Pharmacy, The University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona
| | - Matthew Labenski
- Southwest Environmental Health Sciences Center, College of Pharmacy, The University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona
| | - Nicholas J Mastrandrea
- Southwest Environmental Health Sciences Center, College of Pharmacy, The University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona
| | - Ryan D Canatsey
- Southwest Environmental Health Sciences Center, College of Pharmacy, The University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona
| | - Terrence J Monks
- Southwest Environmental Health Sciences Center, College of Pharmacy, The University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona
| | - Serrine S Lau
- Southwest Environmental Health Sciences Center, College of Pharmacy, The University of Arizona, Tucson, Arizona.,Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona
| |
Collapse
|
43
|
KIAA1549: BRAF Gene Fusion and FGFR1 Hotspot Mutations Are Prognostic Factors in Pilocytic Astrocytomas. J Neuropathol Exp Neurol 2015; 74:743-54. [PMID: 26083571 PMCID: PMC4470527 DOI: 10.1097/nen.0000000000000213] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Up to 20% of patients with pilocytic astrocytoma (PA) experience a poor outcome. BRAF alterations and Fibroblast growth factor receptor 1 (FGFR1) point mutations are key molecular alterations in Pas, but their clinical implications are not established. We aimed to determine the frequency and prognostic role of these alterations in a cohort of 69 patients with PAs. We assessed KIAA1549:BRAF fusion by fluorescence in situ hybridization and BRAF (exon 15) mutations by capillary sequencing. In addition, FGFR1 expression was analyzed using immunohistochemistry, and this was compared with gene amplification and hotspot mutations (exons 12 and 14) assessed by fluorescence in situ hybridization and capillary sequencing. KIAA1549:BRAF fusion was identified in almost 60% of cases. Two tumors harbored mutated BRAF. Despite high FGFR1 expression overall, no cases had FGFR1 amplifications. Three cases harbored a FGFR1 p.K656E point mutation. No correlation was observed between BRAF and FGFR1 alterations. The cases were predominantly pediatric (87%), and no statistical differences were observed in molecular alterations–related patient ages. In summary, we confirmed the high frequency of KIAA1549:BRAF fusion in PAs and its association with a better outcome. Oncogenic mutations of FGFR1, although rare, occurred in a subset of patients with worse outcome. These molecular alterations may constitute alternative targets for novel clinical approaches, when radical surgical resection is unachievable.
Collapse
|
44
|
Campanella NC, Penna V, Ribeiro G, Abrah�o-Machado LF, Scapulatempo-Neto C, Reis RM. Absence of Microsatellite Instability In Soft Tissue Sarcomas. Pathobiology 2015; 82:36-42. [DOI: 10.1159/000369906] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 11/14/2014] [Indexed: 11/19/2022] Open
|