1
|
Evrard C, Rochelle T, Martel M, Al Achkar A, Ferru A, Randrian V, Karayan-Tapon L, Tougeron D. Prognostic and predictive value of microsatellite instability analysis in ctDNA by digital droplet PCR for patients with MSI colorectal cancers. J Transl Med 2025:104176. [PMID: 40246285 DOI: 10.1016/j.labinv.2025.104176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Deficient MisMatch Repair (dMMR) and/or microsatellite instability (MSI) colorectal cancer (CRC) is highly sensitive to immune checkpoint inhibitors (ICI). It is thus becoming increasingly relevant to monitor circulating tumor DNA (ctDNA) and to determine MSI status (ctDNA-MSI) in CRC. So far, few studies have explored this, even though it could be particularly relevant in evaluating treatment efficacy in dMMR and/or MSI CRC patients. The ctDNA digestive cancers study (ADI-MSI) aims to assess the value of ctDNA-MSI as a predictor of ICI efficacy. Blood samples were collected prospectively in a single-center cohort to analyse circulating cell-free DNA (cfDNA) and ctDNA-MSI before the start and during treatment. ctDNA-MSI was measured using digital droplet PCR with the five microsatellite markers of the Pentaplex panel. The primary endpoint was to evaluate ctDNA-MSI levels as a predictor of progression-free survival (PFS). We included 54 dMMR and/or MSI CRC patients, most of whom had metastatic disease (77.8%) treated in the first (25.9%) or second-line (42.6%) with ICI. High baseline cfDNA and ctDNA-MSI were associated with worse PFS and worse overall survival (OS). ctDNA-MSI kinetics, but not cfDNA kinetics, was associated with treatment response (p=0.006), PFS (p=0.03) and OS (p=0.04). ctDNA-MSI kinetics divided into three groups (increase, decrease and negative) correlated strongly with PFS (PFS at 24 months was 0%, 53.0% and 77.0%, respectively; p<0.001) and remained significant in multivariate analysis (HR=7.93; 95% CI, 2.23-28.21; p=0.005). Since there is no strong predictor of ICI efficacy in dMMR and/or MSI CRC patients, these results suggest that ctDNA-MSI could help physicians in treatment decision-making in the future.
Collapse
Affiliation(s)
- Camille Evrard
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service d'Oncologie Médicale, Poitiers, France
| | - Tristan Rochelle
- CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - Marine Martel
- CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - Anis Al Achkar
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - Aurélie Ferru
- CHU de Poitiers, Service d'Oncologie Médicale, Poitiers, France
| | - Violaine Randrian
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service d'Hépato-Gastro-Entérologie, Poitiers, France
| | - Lucie Karayan-Tapon
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service de Cancérologie Biologique, Poitiers, France
| | - David Tougeron
- Université de Poitiers, PRoDiCeT, Poitiers, France; CHU de Poitiers, Service d'Hépato-Gastro-Entérologie, Poitiers, France.
| |
Collapse
|
2
|
Lucocq J, Trinder T, Homyer K, Baig H, Patil P, Muthukumarasamy G. Predicting disease-free survival following curative-intent resection of right-sided colon cancer using a pre- and post-operative nomogram: a prospective observational cohort study. Int J Surg 2025; 111:2886-2893. [PMID: 39909073 DOI: 10.1097/js9.0000000000002300] [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: 06/29/2024] [Accepted: 01/14/2025] [Indexed: 02/07/2025]
Abstract
INTRODUCTION Disease prognostication can be achieved through the derivation of biologically and clinically integrated prediction models. The present study reports 1-, 3-, and 5-year disease-free survival (DFS) in patients undergoing right hemicolectomy for curative intent and both derives and validates a pre- and post-operative prediction tool for DFS for prognostication and risk stratification purposes. METHOD Consecutive patients undergoing right-sided curative-intent resection for colorectal cancer (2010-2020) in a tertiary care unit were followed-up prospectively for recurrence and survival outcomes. Survival analyses were used to derive pre- and post-operative models predicting 1-, 3-, and 5-year DFS. Calibration was reported and internal validation was performed using bootstrapping. RESULTS A total of 822 patients underwent resection and 528 had ≥5-year follow-up. The 1-, 3-, and 5-year DFS rates were 85.6%, 72.5% and 57.6%, respectively. Variables associated with death/recurrence included: increasing age (HR > 1.95, P = 0.037), male gender (HR 1.62, P < 0.001), ASA ≥3 (HR 1.79, P < 0.001), low albumin (HR 1.54, P < 0.001), T4 stage (HR 2.35, P = 0.023), R1 status (HR 1.63, P = 0.024), ≥4 positive lymph nodes (HR > 1.74, P < 0.001) and Clavien-Dindo ≥3 (HR 2.83, P < 0.001). The pre- and post-operative models contained 9 and 13 demographic, clinical, biochemical, operative and pathological variables, respectively (C-index 0.75 and 0.79, respectively). Excluding demographic, clinical and operative variables significantly reduced the C-index of the pre- (0.62) and post-operative models (0.70). CONCLUSION The presented prediction tools for DFS will help clinicians stratify risk, offer appropriate adjuvant treatment and predict long-term DFS following curative-intent right-sided colon cancer resection.
Collapse
Affiliation(s)
- James Lucocq
- Department of Colorectal Surgery, Ninewells Hospital, Dundee, United Kingdom
- Department of General Surgery, Ayr Hospital, Ayr, United Kingdom
- Department of General Surgery, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Tom Trinder
- Department of General Surgery, Ayr Hospital, Ayr, United Kingdom
| | - Kate Homyer
- Department of General Surgery, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Hassan Baig
- Department of Colorectal Surgery, Ninewells Hospital, Dundee, United Kingdom
| | - Pradeep Patil
- Department of Colorectal Surgery, Ninewells Hospital, Dundee, United Kingdom
| | | |
Collapse
|
3
|
Shweikeh F, Zeng Y, Jabir AR, Whittenberger E, Kadatane SP, Huang Y, Mouchli M, Castillo DR. The emerging role of blood-based biomarkers in early detection of colorectal cancer: A systematic review. Cancer Treat Res Commun 2025; 42:100872. [PMID: 39892077 DOI: 10.1016/j.ctarc.2025.100872] [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: 03/10/2024] [Revised: 11/03/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Colorectal cancer (CRC), the third most commonly diagnosed and second most lethal cancer worldwide, necessitates efficient early detection strategies to improve patient outcomes. This review evaluates the promise of novel blood-based biomarkers for early detection of CRC. METHODS A systematic review, registered with PROSPERO (CRD42024513770) and adhering to PRISMA guidelines, was conducted across multiple databases from January 1st, 2020 to December 31st, 2022. The comprehensive search strategy centered on sensitivity, specificity, and AUC-ROC of multiple types of molecular blood biomarkers. RESULTS Of total of 142 included articles, 59 were on protein, 58 on RNA, and 21 on DNA. The investigation into DNA biomarkers revealed that cfDNA and ctDNA carry significant potential for early CRC diagnosis. For instance, methylation patterns in genes such as MYO1-G and NDRG4 exhibited high diagnostic accuracies with AUCs reaching up to 0.996. RNA biomarkers like miRNAs and circRNAs also showed promising results, with circ_0011536 achieving AUCs of 0.982. Protein biomarkers, contrasted with established cancer markers, unveiled notable candidates like Irisin and ANXA2, with AUCs surpassing 0.96. The review highlights several individual markers and panels with the potential to improve upon existing CRC screening tests. CONCLUSIONS Despite the promise shown by the novel biomarkers, challenges persist, including small sample sizes, potential selection biases, and a lack of comprehensive cost-effectiveness analysis. Future research should focus on large-scale, multicenter, prospective studies across diverse populations. The findings advocate for an integrated biomarker approach, potentially revolutionizing CRC screening and aligning it with clinical realities through rigorous validation.
Collapse
Affiliation(s)
- Faris Shweikeh
- Department of Internal Medicine, Cleveland Clinic Akron General, OH, USA
| | - Yuhao Zeng
- Department of Internal Medicine, Cleveland Clinic Akron General, OH, USA
| | - Abdur Rahman Jabir
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | | | - Saurav P Kadatane
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Yuting Huang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Mohamad Mouchli
- Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Dani Ran Castillo
- Department of Hematology and Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
4
|
Cardone C, Facchini S, de Stefano A, Rachiglio AM, Avallone A. Transient 'NeoBRAF wild type' state in a patient with BRAF V600E mutant metastatic colorectal cancer. Cytopathology 2024; 35:671-673. [PMID: 39086078 DOI: 10.1111/cyt.13429] [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: 04/05/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
We report the case of a patient with a BRAFV600E mutant mCRC, with evidence of acquired 'NeoBRAF wild-type' (wt) state. The patient, longitudinally assessed by liquid biopsy, obtained a remarkable clinical outcome with a multimodal approach including surgery, systemic treatment and targeted therapy. In patients with newly diagnosed RAS and BRAFV600E mutant mCRC, longitudinal assessment with liquid biopsy is not routinely used in clinical practice. We report the case of a patient with a BRAFV600E mutant mCRC, with evidence of acquired 'neoBRAF wild-type' (wt) state. The patient obtained a remarkable clinical outcome and has been longitudinally assessed by liquid biopsy.
Collapse
Affiliation(s)
- Claudia Cardone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Sergio Facchini
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Alfonso de Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| |
Collapse
|
5
|
Torresan S, de Scordilli M, Bortolot M, Di Nardo P, Foltran L, Fumagalli A, Guardascione M, Ongaro E, Puglisi F. Liquid biopsy in colorectal cancer: Onward and upward. Crit Rev Oncol Hematol 2024; 194:104242. [PMID: 38128627 DOI: 10.1016/j.critrevonc.2023.104242] [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: 10/05/2023] [Revised: 11/20/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer-related deaths worldwide. In recent years, liquid biopsy has emerged as one of the most interesting areas of research in oncology, leading to innovative trials and practical changes in all aspects of CRC management. RNAs and cell free DNA (cfDNA) methylation are emerging as promising biomarkers for early diagnosis. Post-surgical circulating tumour DNA (ctDNA) can aid in evaluating minimal residual disease and personalising adjuvant treatment. In rectal cancer, ctDNA could improve response assessment to neoadjuvant therapy and risk stratification, especially in the era of organ-preservation trials. In the advanced setting, ctDNA analysis offers the opportunity to monitor treatment response and identify driver and resistance mutations more comprehensively than traditional tissue analysis, providing prognostic and predictive information. The aim of this review is to provide a detailed overview of the clinical applications and future perspectives of liquid biopsy in CRC.
Collapse
Affiliation(s)
- Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Marco de Scordilli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy.
| | - Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Paola Di Nardo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Luisa Foltran
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Arianna Fumagalli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michela Guardascione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elena Ongaro
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Fabio Puglisi
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy; Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
6
|
Patelli G, Mauri G, Tosi F, Amatu A, Bencardino K, Bonazzina E, Pizzutilo EG, Villa F, Calvanese G, Agostara AG, Stabile S, Ghezzi S, Crisafulli G, Di Nicolantonio F, Marsoni S, Bardelli A, Siena S, Sartore-Bianchi A. Circulating Tumor DNA to Drive Treatment in Metastatic Colorectal Cancer. Clin Cancer Res 2023; 29:4530-4539. [PMID: 37436743 PMCID: PMC10643999 DOI: 10.1158/1078-0432.ccr-23-0079] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/28/2023] [Accepted: 06/23/2023] [Indexed: 07/13/2023]
Abstract
In the evolving molecular treatment landscape of metastatic colorectal cancer (mCRC), the identification of druggable alterations is pivotal to achieve the best therapeutic opportunity for each patient. Because the number of actionable targets is expanding, there is the need to timely detect their presence or emergence to guide the choice of different available treatment options. Liquid biopsy, through the analysis of circulating tumor DNA (ctDNA), has proven safe and effective as a complementary method to address cancer evolution while overcoming the limitations of tissue biopsy. Even though data are accumulating regarding the potential for ctDNA-guided treatments applied to targeted agents, still major gaps in knowledge exist as for their application to different areas of the continuum of care. In this review, we recapitulate how ctDNA information could be exploited to drive different targeted treatment strategies in mCRC patients, by refining molecular selection before treatment by addressing tumor heterogeneity beyond tumor tissue biopsy; longitudinally monitoring early-tumor response and resistance mechanisms to targeted agents, potentially leading to tailored, molecular-driven, therapeutic options; guiding the molecular triage towards rechallenge strategies with anti-EGFR agents, suggesting the best time for retreatment; and providing opportunities for an "enhanced rechallenge" through additional treatments or combos aimed at overcoming acquired resistance. Besides, we discuss future perspectives concerning the potential role of ctDNA to fine-tune investigational strategies such as immuno-oncology.
Collapse
Affiliation(s)
- Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- IFOM ETS – The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Gianluca Mauri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- IFOM ETS – The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elio Gregory Pizzutilo
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Villa
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Gabriele Calvanese
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alberto Giuseppe Agostara
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Stefano Stabile
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Silvia Ghezzi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Federica Di Nicolantonio
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino, Candiolo, Italy
| | - Silvia Marsoni
- IFOM ETS – The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia-IRCCS, Candiolo, Italy
- Department of Oncology, University of Torino, Candiolo, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
7
|
Bachet JB, Laurent-Puig P, Meurisse A, Bouché O, Mas L, Taly V, Cohen R, Gornet JM, Artru P, Louafi S, Thirot-Bidault A, Baumgaertner I, Coriat R, Tougeron D, Lecomte T, Mary F, Aparicio T, Marthey L, Blons H, Vernerey D, Taieb J. Circulating tumour DNA at baseline for individualised prognostication in patients with chemotherapy-naïve metastatic colorectal cancer. An AGEO prospective study. Eur J Cancer 2023; 189:112934. [PMID: 37390800 DOI: 10.1016/j.ejca.2023.05.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/19/2023] [Accepted: 05/27/2023] [Indexed: 07/02/2023]
Abstract
PURPOSE Baseline circulating tumour DNA (ctDNA) is a potential prognostic marker in metastatic colorectal cancer (mCRC) patients. However, few studies have compared ctDNA with the usual prognostic factors, and no ctDNA cut-off has been proposed for daily use in clinical practice. PATIENTS AND METHODS Chemotherapy-naive patients with mCRC were prospectively included. Plasma samples were collected at diagnosis and analysed centrally by both NGS and methylation digital PCR. Baseline patient and disease characteristics, treatment regimens, and secondary surgeries were collected. The restricted cubic spline method was used to define the optimal cut-off of ctDNA mutated allelic frequency (MAF). Prognostic values were assessed on overall survival (OS) using Cox models. RESULTS From July 2015 to December 2016, 412 patients were included. ctDNA was undetectable in 83 patients (20%). ctDNA was an independent prognostic marker for OS considering the whole study population. The optimal cut-off for ctDNA MAF was 20% with median OS of 16.0 and 35.8 months for patients with MAF ≥20% and<20%, respectively (hazard ratio = 0.40; 95% confidence intervals: 0.31-0.51; P < 0.0001). The independent prognostic value of ctDNA MAF at 20% was confirmed in subgroups defined by RAS/BRAF status or resectability of metastases. Combining ctDNA MAF and carcinoembryonic antigen levels allowed us to define three different prognostic groups with median OS of 14.2, 21.1, and 46.4 months (P < 0.0001). CONCLUSION ctDNA with a MAF cut-off of 20% improves prognostication of chemotherapy-naïve mCRC patients and may be useful in the future for individualised therapeutic decisions and as a stratification factor in clinical trials. TRIAL REGISTRATION Clinicaltrials.gov, NCT02502656.
Collapse
Affiliation(s)
- Jean-Baptiste Bachet
- Department of Hepato-gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris, France; AGEO (Association des Gastroentérologues Oncologues), Paris, France; Sorbonne Universités, UPMC Université, Paris 06, France; Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France; Institut du cancer Paris CARPEM, AP-HP, Hopital européen Georges Pompidou, Paris, France.
| | - Aurelia Meurisse
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France; INSERM, EFS BFC, UMR1098, RIGHT, University of Franche-Comté, Besançon, France
| | - Olivier Bouché
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Hepato-Gastroenterology, CHU Reims, Reims, France
| | - Léo Mas
- Department of Hepato-gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris, France; AGEO (Association des Gastroentérologues Oncologues), Paris, France
| | - Valérie Taly
- Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France
| | - Romain Cohen
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Sorbonne Universités, UPMC Université, Paris 06, France; Department of Oncology, Hôpital Saint-Antoine, Paris, France
| | - Jean-Marc Gornet
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Hôpital Saint-Louis, Paris, France
| | - Pascal Artru
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Ramsay Hôpital Privé Jean Mermoz, Lyon, France
| | - Samy Louafi
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Centre Hospitalier Sud Francilien, Corbeil-Essonnes, France; Department of Gastroenterology, Groupe Hospitalier Nord Essonne, Longjumeau, France
| | - Anne Thirot-Bidault
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Hôpital Kremlin Bicêtre, Le Kremlin-Bicêtre, France
| | - Isabelle Baumgaertner
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Oncology, Hôpital Henri Mondor, Créteil, France
| | - Romain Coriat
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Hôpital Cochin, Université Paris Cité, Paris, France
| | - David Tougeron
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology and Hepatology, Centre Hospitalo-universitaire de Poitiers, Poitiers, France
| | - Thierry Lecomte
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Hepatology and Digestive Oncology, Centre Hospitalo-universitaire de Tours, Tours, France
| | - Florence Mary
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Hôpital Avicenne, Bobigny, France
| | - Thomas Aparicio
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Gastroenterology, Hôpital Saint-Louis, Paris, France; Department of Gastroenterology, Hôpital Antoine Béclère, Clamart, France
| | - Lysiane Marthey
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Department of Biochemistry, Hôpital Européen Georges Pompidou, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France; Institut du cancer Paris CARPEM, AP-HP, Hopital européen Georges Pompidou, Paris, France
| | - Dewy Vernerey
- Methodology and Quality of Life Unit in Oncology, University Hospital of Besançon, Besançon, France; INSERM, EFS BFC, UMR1098, RIGHT, University of Franche-Comté, Besançon, France
| | - Julien Taieb
- AGEO (Association des Gastroentérologues Oncologues), Paris, France; Centre de Recherche des Cordeliers, INSERM, CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Paris, France; Institut du cancer Paris CARPEM, AP-HP, Hopital européen Georges Pompidou, Paris, France; Department of Gastroenterology and Digestive Oncology, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
8
|
Aoki Y, Nakamura Y, Denda T, Ohta T, Esaki T, Shiozawa M, Yamaguchi K, Yamazaki K, Sunakawa Y, Kato T, Okano N, Taniguchi H, Sato T, Oki E, Nishina T, Komatsu Y, Matsuhashi N, Goto M, Yasui H, Ohtsubo K, Moriwaki T, Takahashi N, Horita Y, Boku S, Wakabayashi M, Ikeno T, Mitani R, Yuasa M, Yoshino T. Clinical Validation of Plasma-Based Genotyping for RAS and BRAF V600E Mutation in Metastatic Colorectal Cancer: SCRUM-Japan GOZILA Substudy. JCO Precis Oncol 2023; 7:e2200688. [PMID: 37343204 PMCID: PMC10309506 DOI: 10.1200/po.22.00688] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/03/2023] [Accepted: 04/19/2023] [Indexed: 06/23/2023] Open
Abstract
PURPOSE Circulating tumor DNA (ctDNA) genotyping on the basis of next-generation sequencing (NGS) may guide targeted therapy for metastatic colorectal cancer (mCRC). However, the validity of NGS-based ctDNA genotyping for RAS/BRAF V600E mutation assessment and the efficacy of anti-EGFR and BRAF-targeted therapies on the basis of ctDNA results remains unclear. PATIENTS AND METHODS The performance of NGS-based ctDNA genotyping for RAS/BRAF V600E mutation assessment was compared with that of a validated polymerase chain reaction-based tissue testing in patients with mCRC enrolled in the GOZILA study, a nationwide plasma genotyping study. The primary end points were concordance rate, sensitivity, and specificity. The efficacy of anti-EGFR and BRAF-targeted therapies on the basis of ctDNA were also evaluated. RESULTS In 212 eligible patients, the concordance rate, sensitivity, and specificity were 92.9% (95% CI, 88.6 to 96.0), 88.7% (95% CI, 81.1 to 94.0), and 97.2% (95% CI, 92.0 to 99.4) for RAS and 96.2% (95% CI, 92.7 to 98.4), 88.0% (95% CI, 68.8 to 97.5), and 97.3% (95% CI, 93.9 to 99.1) for BRAF V600E, respectively. In patients with a ctDNA fraction of ≥1.0%, sensitivity rose to 97.5% (95% CI, 91.2 to 99.7) and 100% (95% CI, 80.5 to 100.0) for RAS and BRAF V600E mutations, respectively. In addition to a low ctDNA fraction, previous chemotherapy, lung and peritoneal metastases, and interval between dates of tissue and blood collection were associated with discordance. The progression-free survival of anti-EGFR therapy and BRAF-targeted treatment was 12.9 months (95% CI, 8.1 to 18.5) and 3.7 (95% CI, 1.3 to not evaluated) months, respectively, for matched patients with RAS/BRAF V600E results by ctDNA. CONCLUSION ctDNA genotyping effectively detected RAS/BRAF mutations, especially with sufficient ctDNA shedding. Clinical outcomes support ctDNA genotyping for determining the use of anti-EGFR and BRAF-targeted therapies in patients with mCRC.
Collapse
Affiliation(s)
- Yu Aoki
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Gastroenterology and Hepatology of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Tadamichi Denda
- Divisioin of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Manabu Shiozawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Kensei Yamaguchi
- Gastroenterological Chemotherapy Department, The Cancer Institute Hospital of JFCR, Tokyo, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizuma, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Taro Sato
- Palliative and Supportive Care Center, Osaka University Hospital, Suita, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomohiro Nishina
- Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Koushiro Ohtsubo
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Toshikazu Moriwaki
- Department of Gastroenterology, University of Tsukuba Hospital, Tsukuba, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Ina, Japan
| | - Yosuke Horita
- Department of Medical Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Shogen Boku
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan
| | - Masashi Wakabayashi
- Biostatistics Division, Center for Research Administration and Support, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takashi Ikeno
- Clinical Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Ryuta Mitani
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mihoko Yuasa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
9
|
Wang D, Zhao P, Lu T, Ren J, Zhu L, Han X, Zhang G, Dong X, Ma H, Yu M, Cai H. ctDNA as a prognostic biomarker in resectable CLM: Systematic review and meta-analysis. Open Life Sci 2023; 18:20220615. [PMID: 37250841 PMCID: PMC10224622 DOI: 10.1515/biol-2022-0615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/20/2023] [Accepted: 04/12/2023] [Indexed: 05/31/2023] Open
Abstract
Cell-free circulating tumor DNA (ctDNA) is synthesized by tumor cells, including metastatic tumors, and circulates in the bloodstream. Evidence suggests that ctDNA is a potential predictive and prognostic biomarker for colorectal cancer (CRC), but its predictive efficacy in detecting CRC liver metastasis (CLM) remains unclear. Additionally, its utility in the clinical setting needs further investigation. We conducted a meta-analysis to determine the utility of ctDNA as a biomarker for predicting the prognosis of CLM and investigate the relationship between CLM and ctDNA positivity. A literature search was performed in electronic databases to identify relevant studies published up to March 19, 2022. We retrieved data on overall survival (OS), disease-free survival (DFS), and recurrence-free survival (RFS) for both ctDNA-positive and ctDNA-negative colorectal liver metastasis (CLM) patients from the selected articles. Hazard ratios (HRs) were also calculated for these survival outcomes analysis was also performed. The stability of the combined meta-analysis was verified by sensitivity analysis and publication bias evaluation. Ten trials were included, and 615 patients were evaluated. In patients with CLM, pooled HRs revealed a substantial link between ctDNA positivity and RFS/DFS. Subgroup analysis revealed that ctDNA had a prospective detection value. Sensitivity analysis and publication bias evaluation indicated stable results. Although the results on pooled HR for OS suggested that ctDNA-positive patients had a shorter survival time, their pooled HRs had a relatively evident heterogeneity, and sensitivity analysis and publication bias evaluation indicated that pooled HRs were extremely unstable. In conclusion, our results demonstrate that ctDNA appears to be a prognostic biomarker for resectable CLM patients.
Collapse
Affiliation(s)
- Da Wang
- School of Medicine Jiangsu University, Zhenjiang, 212000, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Penglai Zhao
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Tingting Lu
- Institution of Clinical Research and Evidence Based Medicine, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Jingyao Ren
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
- School of Clinical Medicine Ning Xia Medical University, Yinchuan, Ning Xia, 750004, China
| | - Lihui Zhu
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
- School of Clinical Medicine Ning Xia Medical University, Yinchuan, Ning Xia, 750004, China
| | - Xiaoyong Han
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
- School of Clinical Medicine Ning Xia Medical University, Yinchuan, Ning Xia, 750004, China
| | - Guangming Zhang
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xiaohua Dong
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
- First Clinical College of Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Haizhong Ma
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Miao Yu
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hui Cai
- School of Medicine Jiangsu University, Zhenjiang, 212000, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, 730000, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, 730000, China
| |
Collapse
|
10
|
Liquid Biopsies in Colorectal Liver Metastases: Towards the Era of Precision Oncologic Surgery. Cancers (Basel) 2022; 14:cancers14174237. [PMID: 36077774 PMCID: PMC9455047 DOI: 10.3390/cancers14174237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/26/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Tumor mutational analysis has been incorporated into the management of patients with CRLM since it can provide valuable prognostic information as well as guide peri-operative systemic treatment. Unlike tumor biopsy, liquid biopsy has emerged as a promising, non-invasive alternative that can detect cell-derived markers from a variety of body fluids and might better characterize all subclones present at a specific time point and allow sequential monitoring of disease evolution. Although not currently considered standard of care, an increasing number of cancer centers are nowadays routinely using liquid biopsies in the treatment of CRLM patients with promising results. The current review provides an overview of liquid biopsies in cancer therapeutics and focuses on the application of this relatively new approach on patients with CRLM.
Collapse
|
11
|
Hao M, Wang K, Ding Y, Li H, Liu Y, Ding L. Which patients are prone to suffer liver metastasis? A review of risk factors of metachronous liver metastasis of colorectal cancer. Eur J Med Res 2022; 27:130. [PMID: 35879739 PMCID: PMC9310475 DOI: 10.1186/s40001-022-00759-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/09/2022] [Indexed: 12/07/2022] Open
Abstract
BACKGROUND In recent years, with the increasing incidence of colorectal cancer (CRC) and its high fatality rate, CRC has seized the attention of the world. And liver metastasis, as the main cause of death of CRC, has become the leading cause of treatment failure in CRC, especially metachronous liver metastasis, have caused patients who underwent bowel resection to experience multiple tortures. MAIN BODY Metachronous liver metastasis has severely affected the quality of life and prognosis of patients. Therefore, in this review, we discuss risk factors for metachronous liver metastasis of CRC, which is the premise for effective intervention for CRC patients who suffer metachronous liver metastasis after undergoing surgery, as well as the signaling pathways associated with CRC. CONCLUSION The occurrence of metachronous liver metastasis is closely related to histology-based prognostic biomarkers, serum-based biomarkers, tumor microenvironment, pre-metastatic niche, liquid biopsy and tissue-based biomarkers. Further research is required to explore the risk factors associated with liver metastasis of CRC.
Collapse
Affiliation(s)
- Mengdi Hao
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10 Yangfangdian, Haidian, Beijing, 100038, People's Republic of China
- Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Kun Wang
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10 Yangfangdian, Haidian, Beijing, 100038, People's Republic of China
- Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Yuhan Ding
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10 Yangfangdian, Haidian, Beijing, 100038, People's Republic of China
- Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Huimin Li
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10 Yangfangdian, Haidian, Beijing, 100038, People's Republic of China
- Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Yin Liu
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10 Yangfangdian, Haidian, Beijing, 100038, People's Republic of China
- Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China
| | - Lei Ding
- Department of Oncology Surgery, Beijing Shijitan Hospital, Capital Medical University, Tieyilu 10 Yangfangdian, Haidian, Beijing, 100038, People's Republic of China.
- Department of Oncology Surgery, Ninth School of Clinical Medicine, Peking University, Beijing, China.
| |
Collapse
|
12
|
Nazha B, Zhuang TZ, Dada HI, Drusbosky LM, Brown JT, Ravindranathan D, Carthon BC, Kucuk O, Goldman J, Master VA, Bilen MA. Blood-Based Next-Generation Sequencing in Adrenocortical Carcinoma. Oncologist 2022; 27:462-468. [PMID: 35462410 PMCID: PMC9177103 DOI: 10.1093/oncolo/oyac061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/16/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Adrenocortical carcinoma (ACC) is a rare and heterogeneous malignancy with poor prognosis. We aimed to evaluate the feasibility of next-generation sequencing (NGS) testing of circulating cell-free tumor DNA (ctDNA) in patients with ACC, to characterize the genomic landscape of alterations, and to identify potential clinically actionable mutations. METHODS Retrospective analysis of genomic data from 120 patients with ACC who had ctDNA testing between 12/2016 and 10/2021 using Guardant360 (Guardant Health, CA) was performed. ctDNA NGS analysis interrogated single nucleotide variants, fusions, indels, and copy number amplifications of up to 83 genes. The frequency of genomic alterations, landscape of co-occurring mutations, and pathogenic/likely pathogenic alterations with potential targeted therapies was identified. The prevalence of alterations identified in ctDNA was compared to those detected in tissue using a publicly available database (cBioPortal). RESULTS The median age of this cohort was 53 years (range 21-81), and 56% of patients were female. Ninety-six patients (80%) had ≥1 somatic alteration detected. TP53 (52%), EGFR (23%), CTNNB1 (18%), MET (18%), and ATM (14%) were found to be the most frequently altered genes in ACC samples. Pathogenic and/or likely pathogenic mutations in therapeutically relevant genes were observed in 56 patients (47%) and included EGFR, BRAF, MET, CDKN2A, CDK4/6, and ATM. The most frequent co-occurring mutations were EGFR + MET (9%), MET + CDK4 (7%), EGFR + CDK4 (7%), and BRAF + MET (7%). The frequencies of mutations detected in ctDNA were similar to those detected in tissue. CONCLUSIONS Utilizing blood-based NGS to characterize genomic alterations in advanced ACC is feasible in over 80% of patients. Almost half of the patients had actionable mutations with approved therapies in other cancers. This approach might inform the development of personalized treatment options or identify clinical trials available for this aggressive malignancy.
Collapse
Affiliation(s)
- Bassel Nazha
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Tony Z Zhuang
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Jacqueline T Brown
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Deepak Ravindranathan
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Bradley C Carthon
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Omer Kucuk
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Jamie Goldman
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Viraj A Master
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehmet Asim Bilen
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
13
|
Hamed RA, Marks S, Mcelligott H, Kalachand R, Ibrahim H, Atyani S, Korpanty G, Osman N. Inoperable de novo metastatic colorectal cancer with primary tumour in situ: Evaluating discordant responses to upfront systemic therapy of the primary tumours and metastatic sites and complications arising from primary tumours (experiences from an Irish Cancer Centre). Mol Clin Oncol 2022; 16:40. [PMID: 35003738 PMCID: PMC8739439 DOI: 10.3892/mco.2021.2472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
Systemic therapy is the mainstay of treatment for de novo metastatic colorectal cancer (mCRC). Heterogeneity between primary tumours and metastases may lead to discordant responses to systemic therapy at these sites. The aim of the present study was to examine these discrepancies and to evaluate the rates of complications arising from the primary tumour and the strategies employed to manage these complications. Electronic medical records were screened for patients eligible for data analysis between January 1st, 2014 and December 31st, 2019. All patients diagnosed with de novo mCRC with primary tumour in situ at the time of initial systemic therapy were included in data analysis. Responses in primary tumour and metastatic sites (according to the Response Evaluation Criteria In Solid Tumours v1.1), discrepancies in these responses and rates of complications arising from primary tumours were assessed along with patient, pathological or molecular factors that may be associated with these discrepant responses or primary tumour complications. A total of 50 patients were identified (median age, 62 years). Right-colon, left-colon and rectal primary tumours comprised 34, 44 and 22% of CRC cases, respectively. All patients received 5-fluorouracil-based chemotherapy (either alone or in combination with oxaliplatin or irinotecan). Disease response (DR), stable disease (SD) and progressive disease (PD) were observed as the first response to systemic therapy in 24, 62 and 12% of primary tumours and in 36, 18 and 44% of metastatic sites, respectively. Only 36% of patients demonstrated concordant responses between the primary tumours and metastases, while the remaining 62% demonstrated discordant responses between the primary tumour and distant metastases (22% had DR with SD; 36% had DR or SD with PD; and 4% had PD with SD in the primary tumour and metastases, respectively). Restaging images were not available for 2% of the patients. Approximately 30% of patients developed complications from primary tumours, including bowel obstruction (6.12%), perforation (6%), rectal pain (6%) and rectal bleeding (10%). Approximately 10% of patients underwent palliative stoma creation. Additionally, 12% required palliative radiotherapy to the primary tumour (due to localized complications arising from the tumour). Discordant responses to systemic therapy between primary tumours and metastases occurred in 60% of patients with de novo mCRC (with primary tumour in situ at the time of first systemic therapy). The observations of the present study have potential implications for molecular tissue analysis to help guide systemic therapy. Tissue from metastatic sites may be preferable to confirm biomarker status in mCRC based on this study.
Collapse
Affiliation(s)
- Ruba A Hamed
- Department of Oncology, Mid-Western Cancer Centre, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Sam Marks
- Department of Oncology, Mid-Western Cancer Centre, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Helen Mcelligott
- Department of Oncology, Mid-Western Cancer Centre, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Roshni Kalachand
- Department of Oncology, Mid-Western Cancer Centre, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Hawa Ibrahim
- Palliative Department, St. Francis Hospice, Dublin 5 D05 T9K8, Ireland
| | - Said Atyani
- Radiology Department, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Greg Korpanty
- Department of Oncology, Mid-Western Cancer Centre, University Hospital Limerick, Limerick V94 F858, Ireland
| | - Nemer Osman
- Department of Oncology, Mid-Western Cancer Centre, University Hospital Limerick, Limerick V94 F858, Ireland
| |
Collapse
|
14
|
Ye P, Cai P, Xie J, Zhang J. Reliability of BRAF mutation detection using plasma sample: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e28382. [PMID: 34941166 PMCID: PMC8701458 DOI: 10.1097/md.0000000000028382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/10/2021] [Accepted: 12/01/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Testing of B-Raf proto-oncogene (BRAF) mutation in tumor is necessary before targeted therapies are given. When tumor samples are not available, plasma samples are commonly used for the testing of BRAF mutation. The aim of this study was to investigate the diagnostic accuracy of BRAF mutation testing using plasma sample of cancer patients. METHODS Databases of Pubmed, Embase, and Cochrane Library were searched for eligible studies investigating BRAF mutation in paired tissue and plasma samples of cancer patients. A total of 798 publications were identified after database searching. After removing 229 duplicated publications, 569 studies were screened using the following exclusion criteria: (1) BRAF mutation not measured in plasma or in tumor sample; (2) lacking BRAF-wildtype or BRAF-mutated samples; (3) tissue and plasma samples not paired; (4) lacking tumor or plasma samples; (5) not plasma sample; (6) not cancer; (7) un-interpretable data. Accuracy data and relevant information were extracted from each eligible study by 2 independent researchers and analyzed using statistical software. RESULTS After pooling the accuracy data from 3943 patients of the 53 eligible studies, the pooled sensitivity, specificity, and diagnostic odds ratio of BRAF mutation testing using plasma sample were 69%, 98%, and 55.78, respectively. Area under curve of summary receiver operating characteristic curve was 0.9435. Subgroup analysis indicated that BRAF mutation testing using plasma had overall higher accuracy (diagnostic odds ratio of 89.17) in colorectal cancer, compared to melanoma and thyroid carcinoma. In addition, next-generation sequencing had an overall higher accuracy in detecting BRAF mutation using plasma sample (diagnostic odds ratio of 63.90), compared to digital polymerase chain reaction (PCR) and conventional PCR, while digital PCR showed the highest sensitivity (74%) among the 3 techniques. CONCLUSION BRAF testing using plasma sample showed an overall high accuracy compared to paired tumor tissue sample, which could be used for cancer genotyping when tissue sample is not available. Large prospective studies are needed to further investigate the accuracy of BRAF mutation testing in plasma sample.
Collapse
Affiliation(s)
- Peng Ye
- Department of Anatomy and Histology, School of Preclinical Medicine, Chengdu University, Chengdu, P.R. China
| | - Peiling Cai
- Department of Anatomy and Histology, School of Preclinical Medicine, Chengdu University, Chengdu, P.R. China
| | - Jing Xie
- Department of Pathology and Clinical Laboratory, Sichuan Provincial Fourth People's Hospital, Chengdu, P.R. China
| | - Jie Zhang
- Adverse Drug Reaction Monitoring Center, Chengdu, P.R. China
| |
Collapse
|
15
|
Ji J, Wang C, Fakih M. Rechallenge With BRAF and anti-EGFR Inhibitors in Patients With Metastatic Colorectal Cancer Harboring BRAF Mutation Who Progressed on Cetuximab and Encorafenib With or Without Binimetinib: A Case Series. Clin Colorectal Cancer 2021; 21:267-271. [DOI: 10.1016/j.clcc.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 12/07/2021] [Accepted: 12/11/2021] [Indexed: 11/27/2022]
|
16
|
Practices and expectations on the use of circulating tumor DNA in colorectal cancer patients: A bi-national AGEO/AIOM/GERCOR/FFCD/FRENCH survey. Clin Res Hepatol Gastroenterol 2021; 45:101681. [PMID: 33785445 DOI: 10.1016/j.clinre.2021.101681] [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] [Received: 10/25/2020] [Accepted: 03/22/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Increasing evidence shows that circulating tumor DNA (ctDNA) is a valuable tool in providing molecular, prognostic, predictive and dynamic information in colorectal cancer (CRC) patients. This study aimed to make a picture of knowledge, practice, attitudes and expectations about ctDNA in CRC patients. MATERIAL AND METHODS An online CRC-ctdna survey was distributed from November 2019 to January 2020 to French and Italian cooperative and scientific groups of Hepato-Gastroenterologists (HGE), Medical Oncologists (MO), Radiotherapists (RT) and Digestive Surgeons (DS). RESULTS Overall, 307 physicians completed the survey (57% Italian; 43% French). Most of them were MO (62%) and HGE (24%). Affiliations were University Hospital (48%), Cancer Center (21%), General Hospital (21%) and Private Hospital (10%). Notably, half of respondents declared to have access to ctDNA in their daily practice. Of them, 53% uses ctDNA to assess RAS/BRAF status only, 46% for RAS/BRAF with other mutations and 1% only for other mutations. MO and HGE identified quick RAS profiling (P = 0.031) as the main reason of interest in the use of ctDNA. Physicians from University Hospitals and Cancer Centers prescribed more ctDNA (P < 0.001) and more often in house (P < 0.001). The main future expectations concerning ctDNA were to guide therapeutic strategies in metastatic (78%) and adjuvant (73%) settings, and to better/quicker profile disease at baseline (56%). CONCLUSION Half of participants can order ctDNA in their daily practice. Molecular profiling of metastatic patients remains the main goal of ctDNA use and ctDNA-based therapeutic strategies are an expectation for the future in both adjuvant and metastatic settings.
Collapse
|
17
|
Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD, Oxnard GR. Circulating tumor DNA in advanced solid tumors: Clinical relevance and future directions. CA Cancer J Clin 2021; 71:176-190. [PMID: 33165928 DOI: 10.3322/caac.21650] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
The application of genomic profiling assays using plasma circulating tumor DNA (ctDNA) is rapidly evolving in the management of patients with advanced solid tumors. Diverse plasma ctDNA technologies in both commercial and academic laboratories are in routine or emerging use. The increasing integration of such testing to inform treatment decision making by oncology clinicians has complexities and challenges but holds significant potential to substantially improve patient outcomes. In this review, the authors discuss the current role of plasma ctDNA assays in oncology care and provide an overview of ongoing research that may inform real-world clinical applications in the near future.
Collapse
Affiliation(s)
- Michael L Cheng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Eirini Pectasides
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Heather A Parsons
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Atish D Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey R Oxnard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Schulte am Esch J, Windmöller BA, Hanewinkel J, Storm J, Förster C, Wilkens L, Krüger M, Kaltschmidt B, Kaltschmidt C. Isolation and Characterization of Two Novel Colorectal Cancer Cell Lines, Containing a Subpopulation with Potential Stem-Like Properties: Treatment Options by MYC/NMYC Inhibition. Cancers (Basel) 2020; 12:cancers12092582. [PMID: 32927768 PMCID: PMC7564713 DOI: 10.3390/cancers12092582] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The aim of this study was to gain a better understanding of cancer stem cells, which are a small subpopulation of tumor cells with high plasticity driving tumor growth and metastasis. Here we isolated two novel colorectal cancer cell lines originating from a rectal neuroendocrine carcinoma and a colorectal adenocarcinoma, depicting stem-like properties. These in vitro models offer the possibility to evaluate pathophysiological mechanisms in order to develop tailored therapeutic strategies for distinct colorectal malignancies. Investigations revealed gene copy number gain of the N-myc proto-oncogene for both. Accordingly, inhibition of the protein–protein interaction of myc and N-myc proto-oncogenes with the myc-associated factor X utilizing small molecule KJ-Pyr-9, exhibited a significant reduction in survival of both cell lines by the induction of apoptosis. Consequently, the blockage of these interactions may serve as a possible treatment strategy for colorectal cancer cell lines with gene copy number gain of the N-myc proto-oncogene. Abstract Cancer stem cells (CSC) are crucial mediators of cancer relapse. Here, we isolated two primary human colorectal cancer cell lines derived from a rectal neuroendocrine carcinoma (BKZ-2) and a colorectal adenocarcinoma (BKZ-3), both containing subpopulations with potential stem-like properties. Protein expression of CSC-markers prominin-1 and CD44 antigen was significantly higher for BKZ-2 and BKZ-3 in comparison to well-established colon carcinoma cell lines. High sphere-formation capacity further confirmed the existence of a subpopulation with potential stem-like phenotype. Epithelial–mesenchymal transition markers as well as immune checkpoint ligands were expressed more pronounced in BKZ-2. Both cell populations demonstrated N-myc proto-oncogene (NMYC) copy number gain. Myc proto-oncogene (MYC)/NMYC activity inhibitor all-trans retinoic acid (ATRA) significantly reduced the number of tumor spheres for both and the volume of BKZ-2 spheres. In contrast, the sphere volume of ATRA-treated BKZ-3 was increased, and only BKZ-2 cell proliferation was reduced in monolayer culture. Treatment with KJ-Pyr-9, a specific inhibitor of MYC/NMYC-myc-associated factor X interaction, decreased survival by the induction of apoptosis of both. In summary, here, we present the novel colorectal cancer cell lines BKZ-2 and BKZ-3 as promising cellular in vitro models for colorectal carcinomas and identify the MYC/NMYC molecular pathway involved in CSC-induced carcinogenesis with relevant therapeutic potential.
Collapse
Affiliation(s)
- Jan Schulte am Esch
- Department of General and Visceral Surgery, Protestant Hospital of Bethel Foundation, 33611 Bielefeld, Germany;
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
| | - Beatrice Ariane Windmöller
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
- Correspondence: ; Tel.: +49-0521-106-5629
| | - Johannes Hanewinkel
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
| | - Jonathan Storm
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
| | - Christine Förster
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Institute of Pathology, KRH Hospital Nordstadt, affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany
| | - Ludwig Wilkens
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Institute of Pathology, KRH Hospital Nordstadt, affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany
| | - Martin Krüger
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Internal Medicine and Gastroenterology, Protestant Hospital of Bethel Foundation, 33611 Bielefeld, Germany
| | - Barbara Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
- Molecular Neurobiology, University of Bielefeld, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Forschungsverbund BioMedizin Bielefeld (FBMB), 33611 Bielefeld, Germany; (J.S.); (C.F.); (L.W.); (M.K.); (B.K.); (C.K.)
- Department of Cell Biology, University of Bielefeld, 33611 Bielefeld, Germany;
| |
Collapse
|
19
|
Filip S, Vymetalkova V, Petera J, Vodickova L, Kubecek O, John S, Cecka F, Krupova M, Manethova M, Cervena K, Vodicka P. Distant Metastasis in Colorectal Cancer Patients-Do We Have New Predicting Clinicopathological and Molecular Biomarkers? A Comprehensive Review. Int J Mol Sci 2020; 21:E5255. [PMID: 32722130 PMCID: PMC7432613 DOI: 10.3390/ijms21155255] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) remains a serious health problem worldwide. Approximately half of patients will develop distant metastasis after CRC resection, usually with very poor prognosis afterwards. Because patient performance after distant metastasis surgery remains very heterogeneous, ranging from death within 2 years to a long-term cure, there is a clinical need for a precise risk stratification of patients to aid pre- and post-operative decisions. Furthermore, around 20% of identified CRC cases are at IV stage disease, known as a metastatic CRC (mCRC). In this review, we overview possible molecular and clinicopathological biomarkers that may provide prognostic and predictive information for patients with distant metastasis. These may comprise sidedness of the tumor, molecular profile and epigenetic characteristics of the primary tumor and arising metastatic CRC, and early markers reflecting cancer cell resistance in mCRC and biomarkers identified from transcriptome. This review discusses current stage in employment of these biomarkers in clinical practice as well as summarizes current experience in identifying predictive biomarkers in mCRC treatment.
Collapse
Affiliation(s)
- Stanislav Filip
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Veronika Vymetalkova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Jiri Petera
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Ludmila Vodickova
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Ondrej Kubecek
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Stanislav John
- Department of Oncology and Radiotherapy, Charles University, Faculty of Medicine in Hradec Kralove, Šimkova 870, 50001 Hradec Králové, Czech Republic; (J.P.); (O.K.); (S.J.)
| | - Filip Cecka
- Department of Surgery, University Hospital in Hradec Kralove, Sokolská 581, 50005 Hradec Králové, Czech Republic;
| | - Marketa Krupova
- The Fingerland Department of Pathology, University Hospital in Hradec Kralove, Sokolská 581, 50005 Hradec Králové, Czech Republic; (M.K.); (M.M.)
| | - Monika Manethova
- The Fingerland Department of Pathology, University Hospital in Hradec Kralove, Sokolská 581, 50005 Hradec Králové, Czech Republic; (M.K.); (M.M.)
| | - Klara Cervena
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| | - Pavel Vodicka
- Department of Molecular Biology of Cancer, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220 Prague, Czech Republic; (V.V.); (L.V.); (K.C.)
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655, 32300 Pilsen, Czech Republic
- Institute of Biology and Medical Genetics, First Faculty of Medicine, Charles University, Albertov 4, 12800 Prague, Czech Republic
| |
Collapse
|
20
|
Lièvre A, de la Fouchardière C, Samalin E, Benoist S, Phelip JM, André T, Lledo G. [BRAF V600E-mutant colorectal cancers: Where are we?]. Bull Cancer 2020; 107:881-895. [PMID: 32674932 DOI: 10.1016/j.bulcan.2020.04.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/14/2022]
Abstract
The BRAFV600E mutation, observed in 8 % of colorectal cancers (CRC), introduces a particular phenotype and a poor prognosis at the localized or metastatic stage. BRAF mutant CRCs are more often localized in the right colon, poorly differentiated and mucinous. They affect an older population (more often female) and are associated with a more frequent metastatic lymph node and peritoneal evolution. The BRAFV600E mutation is associated with a sporadic microsatellite instability (MSI) status in 20 to 40% of cases. In localized colon cancer, it does not imply any modification of the adjuvant treatment. In metastatic CRC, the first action must be the systematic search for an MSI phenotype, given its frequent association with the presence of a BRAF mutation, in order to propose immunotherapy that has been demonstrated to be very effective in MSI metastatic CRC. In non-MSI CRC, a first-line trichimiotherapy associated with bevacizumab is an option to be favored in patients in good general condition but the association with an anti-EGFR can be discussed, especially when the objective is tumor response. At the same time, surgical resection must be systematically discussed in the case of resectable hepatic metastases since the presence of a BRAFV600E mutation is not a risk factor for recurrence and that prolonged survival may be observed after surgery. In the second or third line, the triplet encorafenib, binimetinib and cetuximab, as well as the doublet encorafenib and cetuximab are superior to the association of irinotecan plus cetuximab in terms of response and survival (phase III study BEACON) and represent a new therapeutic standard. Their use on the front line is under study.
Collapse
Affiliation(s)
- Astrid Lièvre
- Service des maladies de l'appareil digestif, CHU Pontchaillou, université Rennes 1, Rennes, France.
| | | | - Emmanuelle Samalin
- Département d'oncologie, institut du cancer de Montpellier (ICM), University Montpellier, Montpellier et institut de génomique fonctionnelle, CNRS, inserm, university Montpellier, Montpellier, France
| | - Stéphane Benoist
- service de chirurgie digestive et oncologique, CHU Bicêtre, AP-HP, université Paris-Saclay, Le Kremlin Bicêtre, France
| | - Jean-Marc Phelip
- Service de gastroentérologie, CHU Saint-Étienne, Saint-Etienne, France
| | - Thierry André
- Département d'oncologie médicale, Sorbonne université, hôpital Saint-Antoine, AP-HP, 7512 Paris, France
| | | |
Collapse
|
21
|
Junca A, Tachon G, Evrard C, Villalva C, Frouin E, Karayan-Tapon L, Tougeron D. Detection of Colorectal Cancer and Advanced Adenoma by Liquid Biopsy (Decalib Study): The ddPCR Challenge. Cancers (Basel) 2020; 12:1482. [PMID: 32517177 PMCID: PMC7352444 DOI: 10.3390/cancers12061482] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND In most countries, participation in colorectal cancer (CRC) screening programs with the immunological fecal occult blood test (iFOBT) is low. Mutations of RAS and BRAF occur early in colorectal carcinogenesis and "liquid biopsy" allows detection of mutated circulating tumor DNA (ctDNA). This prospective study aims to evaluate the performance of RAS and BRAF-mutated ctDNA in detecting CRC and advanced adenomas (AA). METHODS One hundred and thirty patients who underwent colonoscopy for suspicion of colorectal lesion were included and divided into four groups: 20 CRC, 39 AA, 31 non-advanced adenoma and/or hyperplastic polyp(s) (NAA) and 40 with no lesion. Mutated ctDNA was analyzed by droplet digital PCR. RESULTS ctDNA was detected in 45.0% of CRC, in 2.6% of AA and none of the NAA and "no-lesion" groups. All patients with stage II to IV mutated CRC had detectable ctDNA (n = 8/8). Among the mutated AA, only one patient had detectable ctDNA (4.3%), maybe due to limited technical sensitivity or to a low rate of ctDNA or even the absence ctDNA in plasma. Specificity and sensitivity of KRAS- and BRAF-mutated ctDNA for the detection of all CRC and AA were 100% and 16.9%, respectively. CONCLUSIONS ctDNA had high sensitivity in detection of advanced mutated CRC but was unable to sensitively detect AA. ctDNA analysis was easy to perform and readily accepted by the population but requires combination with other circulating biomarkers before replacing iFOBT.
Collapse
Affiliation(s)
- Audelaure Junca
- University of Poitiers, 86000 Poitiers, France; (A.J.); (G.T.); (C.E.); (E.F.); (L.K.-T.)
- Department of Pathology, University Hospital of Poitiers, 86000 Poitiers, France
- Department of Cancer Biology, University Hospital of Poitiers, 86000 Poitiers, France;
| | - Gaëlle Tachon
- University of Poitiers, 86000 Poitiers, France; (A.J.); (G.T.); (C.E.); (E.F.); (L.K.-T.)
- Department of Cancer Biology, University Hospital of Poitiers, 86000 Poitiers, France;
- U1084, Institut national de la santé et de la recherche médicale (INSERM), 86000 Poitiers, France
| | - Camille Evrard
- University of Poitiers, 86000 Poitiers, France; (A.J.); (G.T.); (C.E.); (E.F.); (L.K.-T.)
- Department of Medical Oncology, University Hospital of Poitiers, 86000 Poitiers, France
| | - Claire Villalva
- Department of Cancer Biology, University Hospital of Poitiers, 86000 Poitiers, France;
| | - Eric Frouin
- University of Poitiers, 86000 Poitiers, France; (A.J.); (G.T.); (C.E.); (E.F.); (L.K.-T.)
- Department of Pathology, University Hospital of Poitiers, 86000 Poitiers, France
| | - Lucie Karayan-Tapon
- University of Poitiers, 86000 Poitiers, France; (A.J.); (G.T.); (C.E.); (E.F.); (L.K.-T.)
- Department of Cancer Biology, University Hospital of Poitiers, 86000 Poitiers, France;
- U1084, Institut national de la santé et de la recherche médicale (INSERM), 86000 Poitiers, France
| | - David Tougeron
- University of Poitiers, 86000 Poitiers, France; (A.J.); (G.T.); (C.E.); (E.F.); (L.K.-T.)
- Department of Medical Oncology, University Hospital of Poitiers, 86000 Poitiers, France
- Department of Gastroenterology, University Hospital of Poitiers, 86000 Poitiers, France
| |
Collapse
|
22
|
Allegretti M, Cottone G, Carboni F, Cotroneo E, Casini B, Giordani E, Amoreo CA, Buglioni S, Diodoro M, Pescarmona E, Zazza S, Federici O, Zeuli M, Conti L, Cigliana G, Fiorentino F, Valle M, Giacomini P, Spinella F. Cross-sectional analysis of circulating tumor DNA in primary colorectal cancer at surgery and during post-surgery follow-up by liquid biopsy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:69. [PMID: 32312295 PMCID: PMC7168847 DOI: 10.1186/s13046-020-01569-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Liquid biopsy (LB) in early-stage, non-metastatic colorectal cancer (CRC) must be sensitive enough to detect extremely low circulating tumor DNA (ctDNA) levels. This challenge has been seldom and non-systematically investigated. METHODS Next generation sequencing (NGS) and digital PCR (dPCR) were combined to test tumor DNAs (tDNAs) and paired ctDNAs collected at surgery from 39 patients, 12 of whom were also monitored during the immediate post-surgery follow up. Patients treated for metastatic disease (n = 14) were included as controls. RESULTS NGS and dPCR concordantly (100% agreement) called at least one single nucleotide variant (SNV) in 34 tDNAs, estimated differences in allelic frequencies being negligible (±1.4%). However, despite dPCR testing, SNVs were only detectable in 15/34 (44.1%) ctDNAs from patients at surgery, as opposed to 14/14 (100%) metastatic patients. This was likely due to striking differences (average 10 times, up to 500) in ctDNA levels between groups. NGS revealed blood-only SNVs, suggesting spatial heterogeneity since pre-surgery disease stages, and raising the combined NGS/dPCR sensitivity to 58.8%. ctDNA levels at surgery correlated with neither tumor size, stage, grade, or nodal status, nor with variant abundance in paired tDNA. LB sensitivity reached 63.6% when ctDNA was combined with CEA. Finally, persistence and absence of ctDNA on the first conventional (month 3) post-surgery follow-up were associated with fast relapse and a disease-free status in 3 and 7 patients, respectively. CONCLUSIONS A simple clinical NGS/dPCR/CEA combination effectively addresses the LB challenge in a fraction of non-metastatic CRC patients.
Collapse
Affiliation(s)
- Matteo Allegretti
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Giuliano Cottone
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy
| | - Fabio Carboni
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Ettore Cotroneo
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy
| | - Beatrice Casini
- Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Elena Giordani
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | | | | | - Maria Diodoro
- Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Settimio Zazza
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Orietta Federici
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Massimo Zeuli
- Medical Oncology 1, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Conti
- Clinical Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Cigliana
- Clinical Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Fiorentino
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy
| | - Mario Valle
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizio Giacomini
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy.
| | - Francesca Spinella
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy.
| |
Collapse
|
23
|
Prognostic and Predictive Molecular Biomarkers for Colorectal Cancer: Updates and Challenges. Cancers (Basel) 2020; 12:cancers12020319. [PMID: 32019056 PMCID: PMC7072488 DOI: 10.3390/cancers12020319] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death among cancer patients. This heterogeneous disease is characterized by alterations in multiple molecular pathways throughout its development. Mutations in RAS, along with the mismatch repair gene deficiency, are currently routinely tested in clinics. Such biomarkers provide information for patient risk stratification and for the choice of the best treatment options. Nevertheless, reliable and powerful prognostic markers that can identify “high-risk” CRC patients, who might benefit from adjuvant chemotherapy, in early stages, are currently missing. To bridge this gap, genomic information has increasingly gained interest as a potential method for determining the risk of recurrence. However, due to several limitations of gene-based signatures, these have not yet been clinically implemented. In this review, we describe the different molecular markers in clinical use for CRC, highlight new markers that might become indispensable over the next years, discuss recently developed gene expression-based tests and highlight the challenges in biomarker research.
Collapse
|