1
|
Pathak PS, Chan G, Deming DA, Chee CE. State-of-the-Art Management of Colorectal Cancer: Treatment Advances and Innovation. Am Soc Clin Oncol Educ Book 2024; 44:e438466. [PMID: 38768405 DOI: 10.1200/edbk_438466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Colorectal cancer (CRC) remains a significant global health challenge, ranking among the leading causes of cancer-related morbidity and mortality worldwide. Recent advancements in molecular characterization have revolutionized our understanding of the heterogeneity within colorectal tumors, particularly in the context of tumor sidedness. Tumor sidedness, referring to the location of the primary tumor in either the right or left colon, has emerged as a critical factor influencing prognosis and treatment responses in metastatic CRC. Molecular underpinnings of CRC, the impact of tumor sidedness, and how this knowledge guides therapeutic decisions in the era of precision medicine have led to improved outcomes and better quality of life in patients. The emergence of circulating tumor DNA as a prognostic and predictive tool in CRC heralds promising advancements in the diagnosis and monitoring of the disease. This innovation facilitates better patient selection for exploration of additional treatment options. As the field progresses, with investigational agents demonstrating potential as future treatments for refractory metastatic CRC, new avenues for enhancing outcomes in this challenging disease are emerging.
Collapse
Affiliation(s)
- Priyadarshini S Pathak
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gloria Chan
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore
| | - Dustin A Deming
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, Madison, WI
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Cheng Ean Chee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
2
|
Sichero L, Gonçalves MG, Bettoni F, Coser EM, Mota G, Nunes RAL, Mercante AMDC, Natalino R, Uno M, Ferreira Alves MJ, Matos LL, Kowalski LP, Kulcsar MAV, de Alvarenga GF, Höfler D, Schroeder L, Waterboer T, Tommasino M, Villa LL. Detection of serum biomarkers of HPV-16 driven oropharynx and oral cavity cancer in Brazil. Oral Oncol 2024; 149:106676. [PMID: 38150987 DOI: 10.1016/j.oraloncology.2023.106676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND HPV-16 driven oropharynx/oral cavity squamous cell carcinomas prevalence varies globally. We evaluated the presence of HPV-16 ctDNA and HPV-16 E6 antibodies in samples obtained from participants treated at the Instituto do Cancer do Estado de Sao Paulo, ICESP, and from whom tumoral HPV DNA, HPV-16 E6*I mRNA, and p16INK4a status was also accessed. METHODS HPV was genotyped by PCR-hybridization. All HPV DNA positive and ∼10 % HPV DNA negative cases underwent p16INK4a immunohistochemistry and E6*I RNA testing using a multiplex bead based protocol. HPV-16 ctDNA and anti-E6 antibodies were assessed by ddPCR (digital droplet PCR) and multiplex serology, respectively. RESULTS The prevalence of HPV-16 in oropharynx carcinoma (OPC) cases was low (8.7 %) when considering solely HPV-16 DNA detection, and even lower (5.2 %) when taken into consideration the concomitant detection of HPV-16 E6*I RNA and/or p16INK4 (HPV-16 attributable fraction - AF). None of the oral cavity cancer (OCC) cases were detected with HPV-16 DNA. HPV-16 ctDNA was more commonly detected than HPV-16 E6 antibodies (29.8 % versus 10.6 %). Both serum biomarkers attained 100 % sensitivity of detecting HPV-16 AF OPC, however the specificity of the HPV-16 anti-E6 biomarker was higher compared to ctDNA (93.2 % versus 75.0 %). Finally, when both HPV-16 ctDNA and anti-E6 biomarkers were considered together, the sensitivity and specificity for HPV-16 OPC detection was 100 % and about 70 %, respectively, independently of analyzing HPV-16 DNA positive or HPV-16 AF tumors. CONCLUSIONS Our findings corroborate that serum biomarkers are highly sensitive and specific biomarkers for detection of HPV-associated OPC.
Collapse
Affiliation(s)
- Laura Sichero
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil
| | - Milena Giulia Gonçalves
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil
| | - Fabiana Bettoni
- Molecular Oncology Center, Hospital Sírio-Libanês, São Paulo, SP, Brazil
| | | | - Giana Mota
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil
| | - Rafaella A L Nunes
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil
| | - Ana Maria da Cunha Mercante
- Department of Pathology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil
| | - Renato Natalino
- Department of Pathology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil
| | - Miyuki Uno
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil
| | - Maria José Ferreira Alves
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil
| | - Leandro Luongo Matos
- Department of Head and Neck Surgery, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | - Luiz Paulo Kowalski
- Department of Head and Neck Surgery, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, SP, Brazil
| | | | | | - Daniela Höfler
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lea Schroeder
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tim Waterboer
- Division of Infections and Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Massimo Tommasino
- Infections and Cancer Biology Group, International Agency for Research on Cancer (IARC), Lyon, France
| | - Luisa Lina Villa
- Center for Translational Research in Oncology, Instituto do Cancer do Estado de Sao Paulo ICESP, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo FMUSP HC, Sao Paulo, Brazil; Comprehensive Center for Precision Oncology, Universidade de Sao Paulo, São Paulo, Brazil; Department of Radiology and Oncology, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
3
|
Lin CY, Shen MY, Chen WTL, Yang CA. Evaluation of the Prognostic Value of Low-Frequency KRAS Mutation Detection in Circulating Tumor DNA of Patients with Metastatic Colorectal Cancer. J Pers Med 2023; 13:1051. [PMID: 37511664 PMCID: PMC10381461 DOI: 10.3390/jpm13071051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/24/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
KRAS mutation in tumor tissue is a well-known predictor of resistance to the treatment of anti-EGFR antibodies in metastatic colorectal cancers (mCRC). However, the prognostic value of low-frequency plasma circulating tumor DNA (ctDNA) KRAS mutation in predicting treatment resistance in pretreated mCRC patients remains controversial. This study retrospectively reviewed the clinical course, including response to anti-EGFR and anti-VEGF therapies, and changes in serum tumor marker levels along with image studies in mCRC patients with <1.5% KRAS mutations detected in plasma ctDNA by next-generation sequencing (NGS) at a single center in Taiwan. We identified six pretreated mCRC patients with low-frequency KRAS G12V/G12D/G12S/G13D mutations (variant allele frequency 0.26~1.23%) in plasma ctDNA. Co-occurring low-frequency ctDNA mutations in APC, TP53, MAP2K1, KEAP1, or CTNNB1 were also detected. Although all six patients had treatment adjustments within one month after the ctDNA genetic test, image-evident tumor progression was noted in all patients within a median of 4 months afterwards. Re-challenge therapy with a combination of anti-EGFR, anti-VEGF, and FOLFIRI chemotherapy was found to be ineffective in a patient with 0.38% KRAS G12D mutation in baseline ctDNA. Our study suggests that the detection of low-frequency KRAS mutations in ctDNA could be used as a predictor of treatment response in mCRC patients.
Collapse
Affiliation(s)
- Chien-Yu Lin
- Integrated Precision Health and Immunodiagnostic Center, Department of Laboratory Medicine, China Medical University Hsinchu Hospital, Zhubei City 302, Taiwan
| | - Ming-Yin Shen
- Department of Colorectal Surgery, China Medical University Hsinchu Hospital, Zhubei City 302, Taiwan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - William Tzu-Liang Chen
- Department of Colorectal Surgery, China Medical University Hsinchu Hospital, Zhubei City 302, Taiwan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| | - Chin-An Yang
- Integrated Precision Health and Immunodiagnostic Center, Department of Laboratory Medicine, China Medical University Hsinchu Hospital, Zhubei City 302, Taiwan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
- College of Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
4
|
de Abreu AR, Op de Beeck K, Laurent-Puig P, Taly V, Benhaim L. The Position of Circulating Tumor DNA in the Clinical Management of Colorectal Cancer. Cancers (Basel) 2023; 15:1284. [PMID: 36831626 PMCID: PMC9954551 DOI: 10.3390/cancers15041284] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer type worldwide, with over 1.9 million new cases and 935,000 related deaths in 2020. Within the next decade, the incidence of CRC is estimated to increase by 60% and the mortality by 80%. One of the underlying causes of poor prognosis is late detection, with 60 to 70% of the diagnoses occurring at advanced stages. Circulating cell-free DNA (ccfDNA) is probably the most promising tool for screening, diagnosis, prediction of therapeutic response, and prognosis. More specifically, the analysis of the tumor fraction within the ccfDNA (circulating tumor DNA, ctDNA) has great potential to improve the management of CRC. The present review provides an up-to-date and comprehensive overview of the various aspects related to ctDNA detection in CRC.
Collapse
Affiliation(s)
- Ana Regina de Abreu
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Ken Op de Beeck
- Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, 2650 Edegem, Belgium
- Center for Oncological Research, University of Antwerp and Antwerp University Hospital, Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Pierre Laurent-Puig
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
| | - Valerie Taly
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
| | - Leonor Benhaim
- UMR-S1138, CNRS SNC5096, Équipe labélisée Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Université de Paris, 75006 Paris, France
- Department of Visceral and Surgical Oncology, Gustave Roussy, Cancer Campus, 114 rue Edouard Vaillant, 94805 Villejuif, France
| |
Collapse
|
5
|
Gao Y, Qiao H, Pan V, Wang Z, Li J, Wei Y, Ke Y, Qi H. Accurate genotyping of fragmented DNA using a toehold assisted padlock probe. Biosens Bioelectron 2021; 179:113079. [PMID: 33636500 DOI: 10.1016/j.bios.2021.113079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 11/15/2022]
Abstract
Fragmented DNA from blood plasma, i.e., cell-free DNA, has received great interest as a noninvasive diagnostic biomarker for "point-of-care" testing or liquid biopsy. Here, we present a new approach for accurate genotyping of highly fragmented DNA. Based on toehold-mediated strand displacement, a toehold-assisted padlock probe and toehold blocker were designed and demonstrated with new controllability in significantly suppressing undesired cross-reaction, promoting target recycling and point mutation detection by tuning the thermodynamic properties. Furthermore, toehold-assisted padlock probe systems were elaborately designed for 14 different single-nucleotide variants (SNVs) and were demonstrated to be able to detect low concentration of variant alleles (0.1%). In addition, a target, spanning a narrow sequence window of 29 nucleotides on average is sufficient for the toehold-assisted padlock probe system, which is valuable for the analysis of highly fragmented DNA molecules from clinical samples. We further demonstrated that the toehold-assisted padlock probe, in combination with a unique asymmetric PCR technique, could detect more target SNVs at low allele fractions (1%) in highly fragmented cfDNA. This allows accurate genotyping and provides a new commercial approach for high-resolution analysis of genetic variation.
Collapse
Affiliation(s)
- Yanmin Gao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Hongyan Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Victor Pan
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, United States
| | - Zhaoguan Wang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Jiaojiao Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Yanan Wei
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300350, China
| | - Yonggang Ke
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, 30322, United States
| | - Hao Qi
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, 300350, China.
| |
Collapse
|
6
|
Fernandez-Rozadilla C, Simões AR, Lleonart ME, Carnero A, Carracedo Á. Tumor Profiling at the Service of Cancer Therapy. Front Oncol 2021; 10:595613. [PMID: 33505911 PMCID: PMC7832432 DOI: 10.3389/fonc.2020.595613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/27/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer treatment options have evolved significantly in the past few years. From the initial surgical procedures, to the latest next-generation technologies, we are now in the position to analyze and understand tumors in a one-by-one basis and use that to our advantage to provide with individualized treatment options that may increase patient survival. In this review, we will focus on how tumor profiling has evolved over the past decades to deliver more efficient and personalized treatment options, and how novel technologies can help us envisage the future of precision oncology toward a better management and, ultimately, increased survival.
Collapse
Affiliation(s)
- Ceres Fernandez-Rozadilla
- Grupo de Medicina Xenómica (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Ana Rita Simões
- Grupo de Medicina Xenómica (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain
| | - Matilde E Lleonart
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Madrid, Spain
| | - Amancio Carnero
- Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Madrid, Spain.,Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Ángel Carracedo
- Grupo de Medicina Xenómica (USC), Instituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, Spain.,Grupo de Medicina Xenómica (USC), Fundación Pública Galega de Medicina Xenómica, Santiago de Compostela, Spain
| |
Collapse
|
7
|
Nagy Á, Bátai B, Balogh A, Illés S, Mikala G, Nagy N, Kiss L, Kotmayer L, Matolcsy A, Alpár D, Masszi T, Masszi A, Bödör C. Quantitative Analysis and Monitoring of EZH2 Mutations Using Liquid Biopsy in Follicular Lymphoma. Genes (Basel) 2020; 11:genes11070785. [PMID: 32668764 PMCID: PMC7397208 DOI: 10.3390/genes11070785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/25/2020] [Accepted: 07/07/2020] [Indexed: 12/25/2022] Open
Abstract
Recent advances in molecular technologies enable sensitive and quantitative assessment of circulating tumor DNA, offering a noninvasive disease monitoring tool for patients with malignant disorders. Here, we demonstrated on four follicular lymphoma cases that circulating tumor DNA based EZH2 mutation analysis performed by a highly sensitive droplet digital PCR method may be a valuable treatment monitoring approach in EZH2 mutant follicular lymphoma. EZH2 variant allele frequencies changed in parallel with the volume of metabolically active tumor sites observed on 18F-fluorodeoxyglucose positron emission tomography combined with computer tomography (PET-CT) scans. Variant allele frequencies of EZH2 mutations decreased or were eliminated rapidly upon successful treatment, with treatment failure being associated with elevated EZH2 variant allele frequencies. We also demonstrated spatial heterogeneity in a patient with two different EZH2 mutations in distinct anatomical sites, with both mutations simultaneously detected in the liquid biopsy specimen. In summary, circulating tumor DNA based EZH2 mutation analysis offers a rapid, real-time, radiation-free monitoring tool for sensitive detection of EZH2 mutations deriving from different anatomical sites in follicular lymphoma patients receiving immunochemotherapy.
Collapse
Affiliation(s)
- Ákos Nagy
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
- Correspondence: (Á.N.); (C.B.)
| | - Bence Bátai
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
| | - Alexandra Balogh
- 3rd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary; (A.B.); (S.I.); (T.M.); (A.M.)
| | - Sarolta Illés
- 3rd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary; (A.B.); (S.I.); (T.M.); (A.M.)
| | - Gábor Mikala
- National Institute of Hematology and Infectious Diseases, Central Hospital of Southern Pest, 1097 Budapest, Hungary;
| | - Noémi Nagy
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
| | - Laura Kiss
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
| | - Lili Kotmayer
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
| | - András Matolcsy
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Karolinska University Hospital, 171 77 Stockholm, Sweden
| | - Donát Alpár
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
| | - Tamás Masszi
- 3rd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary; (A.B.); (S.I.); (T.M.); (A.M.)
| | - András Masszi
- 3rd Department of Internal Medicine, Semmelweis University, 1088 Budapest, Hungary; (A.B.); (S.I.); (T.M.); (A.M.)
| | - Csaba Bödör
- MTA-SE Lendület Molecular Oncohematology Research Group, 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, 1085 Budapest, Hungary; (B.B.); (N.N.); (L.K.); (L.K.); (A.M.); (D.A.)
- Correspondence: (Á.N.); (C.B.)
| |
Collapse
|
8
|
Cao H, Liu X, Chen Y, Yang P, Huang T, Song L, Xu R. Circulating Tumor DNA Is Capable of Monitoring the Therapeutic Response and Resistance in Advanced Colorectal Cancer Patients Undergoing Combined Target and Chemotherapy. Front Oncol 2020; 10:466. [PMID: 32318348 PMCID: PMC7154135 DOI: 10.3389/fonc.2020.00466] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/16/2020] [Indexed: 01/10/2023] Open
Abstract
Colorectal cancer (CRC) is a highly lethal disease worldwide. The majority of patients receiving targeted therapy or chemotherapy develop drug resistance, while its molecular mechanism remains to be elucidated. The plasma circulating tumor DNA (ctDNA) exhibited the potential in identifying gene variations and monitoring drug resistance in CRC treatment. In this study, we monitored the ctDNA mutational changes in advanced CRC patients underwent first-line therapy with bevacizumab and cetuximab combined with chemotherapy. The mutation spectrum of 43 patients was established by a 605-gene next-generation sequencing (NGS) panel. The baseline measurement shows that genes with the highest mutation frequency were TP53 (74%), APC (58%), KRAS (40%), SYNE1 (33%), LRP1B (23%), TOP1 (23%), and PIK3CA (21%). Mutations in TP53, APC, and KRAS were detected in 29 paired plasma and tissue samples with the consistency of 81, 67, and 42%, respectively. Clinically targetable gene mutations, such as APC, RNF43, SMAD4, BRAD1, KRAS, RAF1, and TP53, were also identified in ctDNA. The overall consistency between ctDNA and tissue samples was 54.6%. Alleviation of mutational burden in BRAF, KRAS, AMER1, and other major driving genes was observed following the first-line therapy. Patients with KRAS and TP53 mutations in tissues appeared to benefit more than the wild-type counterpart. The dynamic change of plasma mutation status was consistent with the tissue tumor burden and was closely correlated with disease progression. In conclusion, ctDNA monitoring is a useful method for molecular genotyping of colorectal cancer patients. Dynamic changes in resistance can be sensitively monitored by gene variation status, which potentially helps to develop treatment strategy.
Collapse
Affiliation(s)
- Hua Cao
- Department of Oncology, Shenzhen People's Hospital, The 2nd Clinical Medical School of Ji'nan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xinyi Liu
- HaploX Biotechnology, Co., Ltd., Shenzhen, China
| | - Yixin Chen
- Department of Oncology, Shenzhen People's Hospital, The 2nd Clinical Medical School of Ji'nan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Pan Yang
- HaploX Biotechnology, Co., Ltd., Shenzhen, China
| | | | - Lele Song
- HaploX Biotechnology, Co., Ltd., Shenzhen, China
| | - Ruilian Xu
- Department of Oncology, Shenzhen People's Hospital, The 2nd Clinical Medical School of Ji'nan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
9
|
Perspectives of the Application of Liquid Biopsy in Colorectal Cancer. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6843180. [PMID: 32258135 PMCID: PMC7085834 DOI: 10.1155/2020/6843180] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 02/25/2020] [Indexed: 02/06/2023]
Abstract
Colorectal cancer (CRC) is one of the most common gastrointestinal tumors and the second leading cause of cancer death worldwide. Since traditional biopsies are invasive and do not reflect tumor heterogeneity or monitor the dynamic progression of tumors, there is an urgent need for new noninvasive methods that can supplement and improve the current management strategies of CRC. Blood-based liquid biopsies are a promising noninvasive biomarker that can detect disease early, assist in staging, monitor treatment responses, and predict relapse and metastasis. Over time, an increasing number of experiments have indicated the clinical utility of liquid biopsies in CRC. In this review, we mainly focus on the development of circulating tumor cells and circulating tumor DNA as key components of liquid biopsies in CRC and introduce the potential of exosomal microRNAs as emerging liquid biopsy markers in clinical application for CRC.
Collapse
|
10
|
Oliveira KC, Ramos IB, Silva JM, Barra WF, Riggins GJ, Palande V, Pinho CT, Frenkel-Morgenstern M, Santos SE, Assumpcao PP, Burbano RR, Calcagno DQ. Current Perspectives on Circulating Tumor DNA, Precision Medicine, and Personalized Clinical Management of Cancer. Mol Cancer Res 2020; 18:517-528. [DOI: 10.1158/1541-7786.mcr-19-0768] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 11/13/2019] [Accepted: 01/23/2020] [Indexed: 11/16/2022]
|
11
|
Reece M, Saluja H, Hollington P, Karapetis CS, Vatandoust S, Young GP, Symonds EL. The Use of Circulating Tumor DNA to Monitor and Predict Response to Treatment in Colorectal Cancer. Front Genet 2019; 10:1118. [PMID: 31824558 PMCID: PMC6881479 DOI: 10.3389/fgene.2019.01118] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Colorectal cancer is one of the most common cancers worldwide and has a high mortality rate following disease recurrence. Treatment efficacy is maximized by providing tailored cancer treatment, ideally involving surgical resection and personalized neoadjuvant and adjuvant therapies, including chemotherapy, radiotherapy and increasingly, targeted therapy. Early detection of recurrence or disease progression results in more treatable disease and is essential to improving survival outcomes. Recent advances in the understanding of tumor genetics have resulted in the discovery of circulating tumor DNA (ctDNA). A growing body of evidence supports the use of these sensitive biomarkers in detecting residual disease and diagnosing recurrence as well as enabling targeted and tumor-specific adjuvant therapies. Methods: A literature search in Pubmed was performed to identify all original articles preceding April 2019 that utilize ctDNA for the purpose of monitoring response to colorectal cancer treatment. Results: Ninety-two clinical studies were included. These studies demonstrate that ctDNA is a reliable measure of tumor burden. Studies show the utility of ctDNA in assessing the adequacy of surgical tumor clearance and changes in ctDNA levels reflect response to systemic treatments. ctDNA can be used in the selection of targeted treatments. The reappearance or increase in ctDNA, as well as the emergence of new mutations, correlates with disease recurrence, progression, and resistance to therapy, with ctDNA measurement allowing more sensitive monitoring than currently used clinical tools. Conclusions: ctDNA shows enormous promise as a sensitive biomarker for monitoring response to many treatment modalities and for targeting therapy. Thus, it is emerging as a new way for guiding treatment decisions-initiating, altering, and ceasing treatments, or prompting investigation into the potential for residual disease. However, many potentially useful ctDNA markers are available and more work is needed to determine which are best suited for specific purposes and for improving specific outcomes.
Collapse
Affiliation(s)
- Mifanwy Reece
- Colorectal Surgery, Division of Surgery & Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Hariti Saluja
- Department of Medicine, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Paul Hollington
- Colorectal Surgery, Division of Surgery & Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Christos S Karapetis
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Department of Medical Oncology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Sina Vatandoust
- Department of Medical Oncology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Graeme P Young
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Erin L Symonds
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Bowel Health Service, Flinders Medical Centre, Bedford Park, SA, Australia
| |
Collapse
|
12
|
Nappi A, Nasti G, Romano C, Berretta M, Ottaiano A. Metastatic Colorectal Cancer: Prognostic and Predictive Factors. Curr Med Chem 2019; 27:2779-2791. [PMID: 31218949 DOI: 10.2174/0929867326666190620110732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 04/18/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
Abstract
Colorectal cancer represents the third most frequently occurring cancer worldwide. In the last decade, the survival of patients affected by metastatic colorectal cancer (mCRC) has improved through the introduction of biological drugs. However, in this new and dynamic therapeutic context, research about prognostic and predictive factors is important to guide the oncologists to effective therapies as well as to improve the understanding of colorectal cancer biology. Their identification is an intensive area of research and our future goal will be to depict tumour-specific "molecular signatures" in order to predict the clinical course of the disease and the best treatments. In this report, we describe clinical, pathological and molecular biomarkers that can play a role as prognostic or predictive factors in mCRC.
Collapse
Affiliation(s)
- Anna Nappi
- Abdominal medical oncology, National Cancer Institute, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Guglielmo Nasti
- SSD-Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Carmela Romano
- Abdominal medical oncology, National Cancer Institute, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | | | - Alessandro Ottaiano
- SSD-Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| |
Collapse
|
13
|
Circulating tumor DNA applications in monitoring the treatment of metastatic colorectal cancer patients. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2019. [PMCID: PMC7009314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal cancer is the third most common cancer worldwide. New cancer treatment strategies such as monoclonal antibodies against growth factor and angiogenesis receptors have improved the overall survival (OS) and progression-free survival (PFS) in metastatic colorectal cancer (mCRC) patients. However, acquired resistance could happen after these therapies. Circulating tumor DNA (ctDNA) is the DNA fraction derived from tumor cells which could be applied as a non-invasive method for detecting tumor mutations before, during, and after therapies. Here, we reviewed most of the studies examining ctDNA as treatment monitoring in mCRC patients who receive different target therapies. Also, we compared ctDNA with other existing cancer-treatment monitoring methods.
Collapse
|