1
|
Devalle S, Aran V, Bastos Júnior CDS, Pannain VL, Brackmann P, Gregório ML, Ferreira Manso JE, Moura Neto V. A panorama of colon cancer in the era of liquid biopsy. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100148. [PMID: 40027146 PMCID: PMC11863817 DOI: 10.1016/j.jlb.2024.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/05/2025]
Abstract
Colon cancer (CC) is one of the most frequent cancers worldwide being responsible for over 500 thousand deaths in 2022. Its financial and human burden is expected to increase in the next decades accompanying the growing and aging of the global population. Much of this burden could be alleviated considering that the lethality of CC is mostly due to its late diagnosis and failure in the individualized management of patients. Coordinated government actions and implementation of better diagnostic tools capable of detecting CC earlier and of tracking tumoral evolution are mandatory to achieve a reduction in CC's social impact. CtDNA-based liquid biopsy (LB) has great potential to contribute to patients' screening adhesion, CC earlier detection, and to longitudinal tumor follow-up. In this review, we will discuss the latest epidemiological data on CC disease, diagnostic, subtypes, genetics, and treatment management focusing on the advantages and limitations of ctDNA-based LB, including important bottlenecks and solutions necessary for its clinical translation. The latest ctDNA-directed CC clinical trials will also be examined.
Collapse
Affiliation(s)
- Sylvie Devalle
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Veronica Aran
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vera Lucia Pannain
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Brackmann
- Clínica de Coloproctologia do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - Marcelo Leal Gregório
- Instituto de Pesquisas Biomédicas do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - José Eduardo Ferreira Manso
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
2
|
Habashy P, Lea V, Wilkinson K, Wang B, Wu XJ, Roberts TL, Ng W, Rutland T, Po JW, Becker T, Descallar J, Lee M, Mackenzie S, Gupta R, Cooper W, Lim S, Chua W, Lee CS. KRAS and BRAF Mutation Rates and Survival Outcomes in Colorectal Cancer in an Ethnically Diverse Patient Cohort. Int J Mol Sci 2023; 24:17509. [PMID: 38139338 PMCID: PMC10743527 DOI: 10.3390/ijms242417509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
KRAS and BRAF mutation rates in colorectal cancer (CRC) reported from various mono-ethnic studies vary amongst different ethnic groups. However, these differences in mutation rates may not be statistically significant or may be due to differences in environmental and/or laboratory factors across countries rather than racial genetic differences. Here, we compare the KRAS/BRAF mutation rates and survival outcomes in CRC between ethnic groups at a single institution. We also investigate the contributions of genetic, environmental, and laboratory factors to the variations in KRAS/BRAF mutation rates reported from different countries. Clinicopathological data from 453 ethnically diverse patients with CRC were retrospectively analyzed at Liverpool Hospital, NSW Australia (2014-2016). KRAS/BRAF mutations were detected using real-time PCR (Therascreen kits from Qiagen). Mismatch repair (MMR) status was determined using immunohistochemical staining. Four ethnic groups were analyzed: Caucasian, Middle Eastern, Asian, and South American. Overall survival data were available for 406 patients. There was no significant difference in KRAS mutation rates between Caucasians (41.1%), Middle Easterners (47.9%), Asians (44.8%), and South Americans (25%) (p = 0.34). BRAF mutation rates differed significantly between races (p = 0.025), with Caucasians having the highest rates (13.5%) and Middle Easterners the lowest (0%). A secondary analysis in which Caucasians were divided into three subgroups showed that ethnic grouping correlated significantly with KRAS mutation rate (p = 0.009), with central and eastern Europeans having the highest rates (58.3%). There were no significant differences in overall survival (OS) or disease-free survival (DFS) between the four races. The similarity in KRAS mutation rates across races raises the possibility that the differences in KRAS mutation rates reported from various countries may either not be statistically significant or may be due to environmental and/or laboratory factors rather than underlying racial genetic differences. In contrast, we verified that BRAF mutation rates differ significantly between races, suggesting racial genetic differences may be responsible for the discrepant BRAF mutation rates reported from different countries.
Collapse
Affiliation(s)
- Paul Habashy
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia; (P.H.); (T.R.)
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
| | - Vivienne Lea
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia; (P.H.); (T.R.)
- Department of Anatomical Pathology, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Kate Wilkinson
- Department of Medical Oncology, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Bin Wang
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia; (P.H.); (T.R.)
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Xiao-Juan Wu
- Department of Anatomical Pathology, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Tara Laurine Roberts
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2170, Australia
| | - Weng Ng
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Department of Medical Oncology, Liverpool Hospital, Sydney, NSW 2170, Australia
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2170, Australia
| | - Tristan Rutland
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia; (P.H.); (T.R.)
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Department of Anatomical Pathology, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Joseph William Po
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- Surgical Innovations Unit, Department of Surgery, Westmead Hospital, Sydney, NSW 2140, Australia
| | - Therese Becker
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2170, Australia
| | - Joseph Descallar
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2170, Australia
| | - Mark Lee
- Department of Radiation Oncology, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Scott Mackenzie
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Department of Surgery, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Ruta Gupta
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Wendy Cooper
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia; (P.H.); (T.R.)
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW 2050, Australia
| | - Stephanie Lim
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- Department of Medical Oncology, Campbelltown Hospital, Sydney, NSW 2560, Australia
| | - Wei Chua
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Department of Medical Oncology, Liverpool Hospital, Sydney, NSW 2170, Australia
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
| | - Cheok Soon Lee
- Discipline of Pathology, School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia; (P.H.); (T.R.)
- Liverpool Clinical School, Western Sydney University, Sydney, NSW 2170, Australia; (T.L.R.); (T.B.)
- Department of Anatomical Pathology, Liverpool Hospital, Sydney, NSW 2170, Australia
- Ingham Institute for Applied Medical Research, Liverpool Hospital, Sydney, NSW 2170, Australia
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW 2170, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| |
Collapse
|
3
|
Wielandt AM, Hurtado C, Moreno C M, Villarroel C, Castro M, Estay M, Simian D, Martinez M, Vial MT, Kronberg U, López-Köstner F. Characterization of Chilean patients with sporadic colorectal cancer according to the three main carcinogenic pathways: Microsatellite instability, CpG island methylator phenotype and Chromosomal instability. Tumour Biol 2020; 42:1010428320938492. [PMID: 32635826 DOI: 10.1177/1010428320938492] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Molecular classification of colorectal cancer is difficult to implement in clinical settings where hundreds of genes are involved, and resources are limited. This study aims to characterize the molecular subtypes of patients with sporadic colorectal cancer based on the three main carcinogenic pathways microsatellite instability (MSI), CpG island methylator phenotype (CIMP), and chromosomal instability (CIN) in a Chilean population. Although several reports have characterized colorectal cancer, most do not represent Latin-American populations. Our study includes 103 colorectal cancer patients who underwent surgery, without neoadjuvant treatment, in a private hospital between 2008 and 2017. MSI, CIN, and CIMP status were assessed. Frequent mutations in KRAS, BRAF, and PIK3CA genes were analyzed by Sanger sequencing, and statistical analysis was performed by Fisher's exact and/or chi-square test. Survival curves were estimated with Kaplan-Meier and log-rank test. Based on our observations, we can classify the tumors in four subgroups, Group 1: MSI-high tumors (15%) are located in the right colon, occur at older age, and 60% show a BRAF mutation; Group 2: CIN-high tumors (38%) are in the left colon, and 26% have KRAS mutations. Group 3: [MSI/CIN/CIMP]-low/negative tumors (30%) are left-sided, and 39% have KRAS mutations; Group 4: CIMP-high tumors (15%) were more frequent in men and left side colon, with 27% KRAS and 7% presented BRAF mutations. Three percent of patients could not be classified. We found that CIMP-high was associated with a worse prognosis, both in MSI-high and MSI stable patients (p = 0.0452). Group 3 (Low/negative tumors) tend to have better overall survival compared with MSI-high, CIMP-high, and CIN-high tumors. This study contributes to understanding the heterogeneity of tumors in the Chilean population being one of the few characterizations performed in Latin-America. Given the limited resources of these countries, these results allow to improve molecular characterization in Latin-American colorectal cancer populations and confirm the possibility of using the three main carcinogenic pathways to define therapeutic strategies.
Collapse
Affiliation(s)
- Ana María Wielandt
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile.,Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Claudia Hurtado
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile.,Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Mauricio Moreno C
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile.,Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Cynthia Villarroel
- Oncology and Molecular Genetics Laboratory, Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Magdalena Castro
- Academic Department Research Unit, Clínica Las Condes, Santiago, Chile
| | - Marlene Estay
- Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | - Daniela Simian
- Academic Department Research Unit, Clínica Las Condes, Santiago, Chile
| | - Maripaz Martinez
- Academic Department Research Unit, Clínica Las Condes, Santiago, Chile
| | | | - Udo Kronberg
- Coloproctology Unit, Clínica Las Condes, Santiago, Chile
| | | |
Collapse
|
4
|
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
|
5
|
Zhang W, Yang H, Zhu L, Luo Y, Nie L, Li G. Role of EGFR/ErbB2 and PI 3K/AKT/e-NOS in Lycium barbarum polysaccharides Ameliorating Endothelial Dysfunction Induced by Oxidative Stress. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:1523-1539. [PMID: 31645123 DOI: 10.1142/s0192415x19500782] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lycium barbarum polysaccharides (LBP) are the major ingredients of wolfberry. In this study, we investigated the role of LBP in endothelial dysfunction induced by oxidative stress and the underlying mechanisms using thoracic aortic endothelial cells of rat (RAECs) as a model. We found that Ang II inhibits cell viability of RAECs with 10-6mol/L of Ang II treatment for 24h most potential (P<0.05), the level of reactive oxygen species (ROS) is increased by Ang II treatment (P<0.01), and the expression of Occludin and Zonula occludens-1 (ZO-1) is decreased by Ang II treatment (P<0.05). However, preincubation of cells with LBP could inhibit the changes caused by Ang II, LBP increased cell viability (P<0.05), decreased the level of ROS (P<0.01), and up-regulated the expression of Occludin (P<0.05) and ZO-1. In addition, Ang II treatment increased the expression of EGFR and p-EGFR (Try1172) and which can be inhibited by LBP. On the contrary, expression of ErbB2, p-ErbB2 (Try1248), PI3K, p-e-NOS (Ser1177) (P<0.05), and p-AKT (Ser473) (P<0.05) was inhibited by Ang II treatment and which can be increased by LBP. Treatment of the cells with inhibitors showed that the regulation of p-e-NOS and p-AKT expression by Ang II and LBP can be blocked by PI3K inhibitor wortmannin but not EGFR and ErbB2 inhibitor AC480. Taken together, our results suggested that LBP plays a critical role in maintaining the integrality of blood vessel endothelium through reduced production of ROS via regulating the activity of EGFR, ErbB2, PI3K/AKT/e-NOS, and which may offer a novel therapeutic option in the management of endothelial dysfunction.
Collapse
Affiliation(s)
- Wenjuan Zhang
- School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Huifang Yang
- School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Lingqin Zhu
- School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Yan Luo
- School of Basic Medical Science, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Lihong Nie
- School of Basic Medical Science, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| | - Guanghua Li
- School of Basic Medical Science, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China.,School of Public Health and Management, Ningxia Medical University, Shengli Street 1160, Yinchuan 750004, P. R. China
| |
Collapse
|
6
|
Zhang X, Zhao W, Wei W, You Z, Ou X, Sun M, Yin Y, Tang X, Zhao Z, Hu C, Liu F, Deng J, Mao L, Zhou D, Ren Y, Li X, Zhang S, Liu C, Geng J, Yao G, Song B, Liu Y, Li D, Jiang Y, Chen Y, Zhao Y, Yu S, Pang D. Parallel Analyses of Somatic Mutations in Plasma Circulating Tumor DNA (ctDNA) and Matched Tumor Tissues in Early-Stage Breast Cancer. Clin Cancer Res 2019; 25:6546-6553. [PMID: 31350313 DOI: 10.1158/1078-0432.ccr-18-4055] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/08/2019] [Accepted: 07/24/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Early detection and intervention can decrease the mortality of breast cancer significantly. Assessments of genetic/genomic variants in circulating tumor DNA (ctDNA) have generated great enthusiasm for their potential application as clinically actionable biomarkers in the management of early-stage breast cancer.Experimental Design: In this study, 861 serial plasma and matched tissue specimens from 102 patients with early-stage breast cancer who need chemotherapy and 50 individuals with benign breast tumors were deeply sequenced via next-generation sequencing (NGS) techniques using large gene panels. RESULTS Cancer tissues in this cohort of patients showed profound intratumor heterogeneities (ITHGs) that were properly reflected by ctDNA testing. Integrating the ctDNA detection rate of 74.2% in this cohort with the corresponding predictive results based on Breast Imaging Reporting and Data System classification (BI-RADS) could increase the positive predictive value up to 92% and potentially dramatically reduce surgical overtreatment. Patients with positive ctDNA after surgery showed a higher percentage of lymph node metastasis, indicating potential recurrence and remote metastasis. The ctDNA-positive rates were significantly decreased after chemotherapy in basal-like and Her2+ tumor subtypes, but were persistent despite chemotherapy in luminal type. The tumor mutation burden in blood (bTMB) assessed on the basis of ctDNA testing was positively correlated with the TMB in tumor tissues (tTMB), providing a candidate biomarker warranting further study of its potentials used for precise immunotherapy in cancer. CONCLUSIONS These data showed that ctDNA evaluation is a feasible, sensitive, and specific biomarker for diagnosis and differential diagnosis of patients with early-stage breast cancer who need chemotherapy.
Collapse
Affiliation(s)
- Xianyu Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Weiwei Zhao
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Wei Wei
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Zilong You
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaohua Ou
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Mingming Sun
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Yanling Yin
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiaoyan Tang
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Zhen Zhao
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Changming Hu
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Feifei Liu
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Junhao Deng
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Linlin Mao
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Danyan Zhou
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Yuxia Ren
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Xiaoxia Li
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Shangfei Zhang
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Chang Liu
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China
| | - Jingshu Geng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guodong Yao
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Bingbing Song
- Cancer Center of Heilongjiang Province, Harbin, China
| | - Yupeng Liu
- Department of Epidemiology, Harbin Medical University, Harbin, China
| | - Dalin Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yongdong Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanbo Chen
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yashuang Zhao
- Department of Epidemiology, Harbin Medical University, Harbin, China
| | - Shihui Yu
- Clinical Genome Center, KingMed Center for Clinical Laboratory Co., Ltd; KingMed College of Laboratory Medicine, Guangzhou Medical University; Guangzhou KingMed Translational Medicine Institute Co., Ltd; KingMed JianShi Innovation Institute (Guangzhou) Co., Ltd, Guangzhou, China.
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
7
|
Huang D, Sun W, Zhou Y, Li P, Chen F, Chen H, Xia D, Xu E, Lai M, Wu Y, Zhang H. Mutations of key driver genes in colorectal cancer progression and metastasis. Cancer Metastasis Rev 2019; 37:173-187. [PMID: 29322354 DOI: 10.1007/s10555-017-9726-5] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The association between mutations of key driver genes and colorectal cancer (CRC) metastasis has been investigated by many studies. However, the results of these studies have been contradictory. Here, we perform a comprehensive analysis to screen key driver genes from the TCGA database and validate the roles of these mutations in CRC metastasis. Using bioinformatics analysis, we identified six key driver genes, namely APC, KRAS, BRAF, PIK3CA, SMAD4 and p53. Through a systematic search, 120 articles published by November 30, 2017, were included, which all showed roles for these gene mutations in CRC metastasis. A meta-analysis showed that KRAS mutations (combined OR 1.18, 95% CI 1.05-1.33) and p53 mutations (combined OR 1.49, 95% CI 1.23-1.80) were associated with CRC metastasis, including lymphatic and distant metastases. Moreover, CRC patients with a KRAS mutation (combined OR 1.29, 95% CI 1.13-1.47), p53 mutation (combined OR 1.35, 95% CI 1.06-1.72) or SMAD4 mutation (combined OR 2.04, 95% CI 1.41-2.95) were at a higher risk of distant metastasis. Subgroup analysis stratified by ethnic populations indicated that the BRAF mutation was related to CRC metastasis (combined OR 1.42, 95% CI 1.18-1.71) and distant metastasis (combined OR 1.51, 95% CI 1.20-1.91) in an Asian population. No significant association was found between mutations of APC or PIK3CA and CRC metastasis. In conclusion, mutations of KRAS, p53, SMAD4 and BRAF play significant roles in CRC metastasis and may be both potential biomarkers of CRC metastasis as well as therapeutic targets.
Collapse
Affiliation(s)
- Dongdong Huang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Wenjie Sun
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yuwei Zhou
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Peiwei Li
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, 310058, China
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Fang Chen
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hanwen Chen
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, 310058, China
| | - Dajing Xia
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, 310058, China
| | - Enping Xu
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Maode Lai
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yihua Wu
- Department of Toxicology, School of Public Health, Zhejiang University, Hangzhou, 310058, China.
| | - Honghe Zhang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
8
|
Khelwatty SA, Essapen S, Bagwan I, Green M, Seddon AM, Modjtahedi H. Co-expression and prognostic significance of putative CSC markers CD44, CD133, wild-type EGFR and EGFRvIII in metastatic colorectal cancer. Oncotarget 2019; 10:1704-1715. [PMID: 30899442 PMCID: PMC6422200 DOI: 10.18632/oncotarget.26722] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/15/2019] [Indexed: 12/24/2022] Open
Abstract
The presence of colorectal cancer stem cells (CSCs) have been associated with tumour initiation and resistance to therapy. This study investigated the co-expression and prognostic significance of the CSCs biomarkers CD44 and CD133 with wild-type EGFR (wtEGFR) and EGFRvIII in colorectal cancer (CRC). The expression of these biomarkers were determined in tumours from 70 patients with metastatic CRC by immunohistochemistry, and in a panel of human CRC cell lines, and their variants with acquired-resistance to EGFR inhibitors, by flow cytometry. The expression of CD44, CD133, wtEGFR and EGFRvIII were present in 17%, 23%, 26% and 13% of cases and the co-expression of CD44/CD133 with wtEGFR and EGFRvIII were present in 9% and 3% of the cases respectively. Only co-expression of CSCs/EGFRvIII (P = 0.037), and amphiregulin (P = 0.017) were associated with worse overall survival. Interestingly, disease-free survival was improved in BTC expressing patients (P = 0.025). In vitro CD133 expression and its co-expression with CD44 were associated with primary-resistance to irinotecan and acquired-resistance to anti-EGFR inhibitors respectively. Our results suggest co-expression of CSCs and EGFRvIII could be potential biomarkers of worse overall survival and resistance to therapy in patients with mCRC and warrants further validation in a larger cohort.
Collapse
Affiliation(s)
| | - Sharadah Essapen
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston, UK.,St. Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Izhar Bagwan
- Department of Histopathology, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Margaret Green
- Department of Histopathology, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Alan M Seddon
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston, UK
| | - Helmout Modjtahedi
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston, UK
| |
Collapse
|
9
|
Saint-Jean A, Reguart N, Eixarch A, Adán A, Castellà C, Sánchez-Dalmau B, Sainz-de-la-Maza M. Ocular surface adverse events of systemic epidermal growth factor receptor inhibitors (EGFRi): A prospective trial. J Fr Ophtalmol 2018; 41:955-962. [PMID: 30473235 DOI: 10.1016/j.jfo.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/20/2018] [Indexed: 11/26/2022]
Abstract
PURPOSE Controversy exists regarding the safety of agents that systemically inhibit epidermal growth factor receptor (EGFRi) in oncologic patients in terms of toxicity to the ocular surface. We performed a prospective clinical study comparing the ocular surface toxicity of systemic EGFRi between a case and a control group. METHODS Patients with lung or colon cancer were divided in two groups: 25 patients treated with systemic EGFRi and 25 control patients without EGFRi treatment. Patients in both groups were chemotherapy naive. Four visits were scheduled in a one year period comparing signs and symptoms in terms of symptom questionnaires (SIDEQ, OSDI and AVS), corneal fluorescein staining (Oxford test), tear production (Schirmer's test) and a quantitative evaluation of conjunctival chemosis and hyperemia. Basal epithelial cell density (CEBD) and corneal subepithelial nerve fiber density (CNFD) were measured and compared using confocal microscopy (Heidelberg Engineering, Germany). The differences in each variable were compared with the analysis of variance (ANOVA). A P value<0.05 was considered significant for all comparisons. RESULTS No statistically significant differences were found between patients under EGFRi treatment and the age-matched controls in the variables analyzed. When cases and controls were evaluated separately, the case group showed a significantly worse progression of signs (chemosis score, CFS, Schirmer's) as well as in terms of CEBD and CNFD (all P<0.05). CONCLUSION Systemic EGFRi may increase dry eye signs as well as decrease CEBD and CNFD. This study may help us to understand the true toxicity of EGFRi to the ocular surface.
Collapse
Affiliation(s)
- A Saint-Jean
- University of Barcelona, Clinica Baviera, Barcelona, Spain.
| | - N Reguart
- Thoracic Oncology, Medical Oncology Department, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | | | - A Adán
- Head Ophthalmology Department, Hospital Clinic, Barcelona, Spain
| | - C Castellà
- Head Glaucoma Department, Hospital Joan XXIII, Tarragona, Spain
| | | | | |
Collapse
|
10
|
Simple classifiers for molecular subtypes of colorectal cancer. Arab J Gastroenterol 2017; 18:191-200. [PMID: 29241727 DOI: 10.1016/j.ajg.2017.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 10/31/2017] [Accepted: 11/22/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND STUDY AIM Colorectal cancer (CRC) is a heterogeneous disease entity with a diverse biological pathogenesis. This study aims to validate the two studies published in 2013 which established a separate CRC molecular subtype classification by utilizing a rapidly accessible miniclassifier, and verify a simplified version thereof. PATIENTS AND METHODS Participants diagnosed with CRC (n = 568) were subtyped in three classifications for characteristic, and prognostic purposes. Colorectal cancer subtypes (CCS) were classified as: i) CCS1 (CDX2+, microsatellite stable (MSS)/microsatellite instability (MSI)-low), ii) CCS2 (MSI-high), and iii) CCS3 (FRMD6/ZEB1/HTR2B +, CDX2-, MSS/MSI-low]. Simplified CCS (SiCCS) subtypes were grouped as: i) CDX2 (CDX2+, MSS/MSI-low, ZEB1 ≤ 2), ii) MSI-H (MSI-high, CDX2/FRMD6/ZEB1/HTR2B +/-), and iii) ZEB1 (ZEB1 ≥ 2, CDX2-, MSS/MSI-low). New molecular classification (NMC) subtypes were defined as: i) enterocyte (E-C) (MUC2 +), ii) goblet-like (G-L) (MUC2 + and TFF3 +), iii) transit-amplifying (T-A) (CFTR +), and iv) stem-like (S-L) (ZEB1 +). RESULTS In total, 53.5% (n = 304) CCS, 58.3% (n = 331) SiCCS, and 37.7% (n = 214) NMC tumours could be evaluated. CCS2 and MSI-H CRCs had the most favourable survival outcome, whereas the CCS3, ZEB1 and S-L subtypes showed the poorest prognosis. A significant overlap between CCS3, ZEB1, and S-L tumours was demonstrated. CONCLUSION There is still a need for a consensus gene expression-based subtyping classification system for CRCs, thereby allowing the categorization of most CRC tumours. This study reveals that a simple and rapidly accessible process could replace the complicated, costly and mostly inapproachable methods clinical practices that have been introduced in the majority of previous studies.
Collapse
|