1
|
Zhou Q, Chen X, Zeng B, Zhang M, Guo N, Wu S, Zeng H, Sun F. Circulating tumor DNA as a biomarker of prognosis prediction in colorectal cancer: a systematic review and meta-analysis. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:167-178. [PMID: 40265088 PMCID: PMC12010414 DOI: 10.1016/j.jncc.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 04/18/2024] [Accepted: 05/14/2024] [Indexed: 02/03/2025] Open
Abstract
Objective Circulating tumor DNA (ctDNA) is increasingly being used as a potential biomarker in colorectal cancer (CRC) patients. However, the role of ctDNA in CRC prognosis prediction remains unclear. The objective is to systematically assess the clinical value of ctDNA in colorectal cancer prognosis prediction throughout the treatment cycle. Methods PubMed, Web of Science, Embase, Cochrane Library, Scopus, and clinical trials.gov database was searched from January 2016 to April 2023. Observational studies and randomized clinical trials reporting on ctDNA and prognostic outcomes in CRC patients were included. Pooled hazard risk ratios (HRs) were calculated for the primary outcomes, relapse-free survival (RFS), and overall survival (OS). Random-effects models were preferred considering the potential heterogeneity. Results Sixty-five cohort studies were included. Association between ctDNA and shorter RFS or OS was significant, especially after the full-course treatment recommended by the guidelines (HR = 8.92 [ 95 % CI: 6.02-13.22], P < 0.001, I2 = 73 %; HR = 3.05 [ 95 % CI: 1.72-5.41], P < 0.001, I2 = 48 %) for all types of CRC patients. Despite the presence of heterogeneity, subgroup analyses showed that the cancer type and ctDNA detection assays may be the underlying cause. Besides, ctDNA may detect recurrence earlier than radiographic progression, but no uniform sampling time point between studies might bring bias. However, ctDNA detection did not appear to correlate with pathological complete response achievement in patients with locally advanced rectal cancer. Conclusion ctDNA detection was significantly associated with poorer prognosis. The potential applications in prognostic prediction are promising and remain to be evaluated in other fields.
Collapse
Affiliation(s)
- Qingxin Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
- Tianjin Centers for Disease Control and Prevention, Tianjin, China
| | - Xiaowei Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Baoqi Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China
| | - Meng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
| | - Nana Guo
- Hebei Centers for Disease Control and Prevention, Hebei, China
| | - Shanshan Wu
- Clinical Epidemiology and EBM Unit, National Clinical Research Center for Digestive Diseases, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hongmei Zeng
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Centre, Beijing, China
- Key Laboratory of Major Disease Epidemiology, Ministry of Education (Peking University), Beijing, China
- Xinjiang Medical University, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
2
|
Ghidini M, Hahne JC, Senti C, Heide T, Proszek PZ, Shaikh R, Carter P, Hubank M, Trevisani F, Garrone O, Cappelletti MR, Generali D, Cattaneo M, Gnocchi N, Donati G, Gobbi A, Grizzi G, Lampis A, Elghadi R, Tanzi G, Tomasello G, Ratti M, Pinato DJ, Fassan M, Vlachogiannis G, Sottoriva A, Cortellini A, Passalacqua R, Valeri N. Circulating Tumor DNA Dynamics and Clinical Outcome in Metastatic Colorectal Cancer Patients Undergoing Front-Line Chemotherapy. Clin Cancer Res 2025; 31:707-718. [PMID: 39688961 DOI: 10.1158/1078-0432.ccr-24-0924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 08/01/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
PURPOSE We tested whether circulating tumor DNA (ctDNA) changes may be used to assess early response and clinical outcomes in patients with metastatic colorectal cancer (mCRC) undergoing first-line systemic anticancer therapy (SACT). EXPERIMENTAL DESIGN Eight hundred sixty-two plasma samples were collected 4-weekly from baseline (BL) until disease progression in patients with mCRC receiving first-line SACT. ctDNA was tested using tissue-agnostic next-generation sequencing panels. ctDNA normalization was defined as ≥99% clearance after 1 month of therapy (Mo1) in the three variants with the highest allele frequency in BL ctDNA. RESULTS Eighty-three paired samples from 75 patients were available for analysis. Twelve pairs (14.4%) showed no variants in either BL or Mo1. In the remaining 71 comparisons (65 patients), 37 (52.1%) showed ctDNA normalization at Mo1. Patients who cleared ctDNA had significantly longer overall (45.6 months) and progression-free survival (13.9 months) compared with nonnormalized patients [overall survival = 22.6 months (log-rank P = 0.01) and progression-free survival = 10.7 months (log-rank P = 0.036), respectively]. In addition, a higher response rate was observed in patients with ctDNA clearance (72.9%) compared with nonnormalized cases (38.2%). Longitudinal sequencing of at least four time points in patients with a progression-free survival of >10 months showed emerging variants in 47.8% of cases; in all these patients, the trajectory of these new "outlier" variants seemed in stark contrast with the clinical-radiological course of disease and the trend in other mutations. CONCLUSIONS ctDNA clearance represents an early indicator of benefit from SACT in patients with mCRC; serial tracking of multiple variants is warranted to improve specificity and avoid misleading information due to the emergence of mutations of unknown clinical significance.
Collapse
Affiliation(s)
- Michele Ghidini
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jens Claus Hahne
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- CRUK Experimental Cancer Centre and NIHR Biomedical Research Centre, University of Leicester, Leicester, United Kingdom
| | - Chiara Senti
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | | | - Paula Z Proszek
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Ridwan Shaikh
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Paul Carter
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Mike Hubank
- Clinical Genomics Translational Research Laboratory, Royal Marsden Hospital, London, United Kingdom
| | - Francesco Trevisani
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Ornella Garrone
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Rosa Cappelletti
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Daniele Generali
- Multidisciplinary Unit of Breast Pathology and Translational Research, Cremona Hospital, Cremona, Italy
| | - Monica Cattaneo
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Nicoletta Gnocchi
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Gianvito Donati
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Angela Gobbi
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Giulia Grizzi
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Andrea Lampis
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Raghad Elghadi
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Giulia Tanzi
- Division of Pathology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Gianluca Tomasello
- Division of Medical Oncology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Margherita Ratti
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - David J Pinato
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Matteo Fassan
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Georgios Vlachogiannis
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Human Technopole, Milan, Italy
| | - Alessio Cortellini
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Rodolfo Passalacqua
- Division of Medical Oncology, ASST of Cremona, Hospital of Cremona, Cremona, Italy
| | - Nicola Valeri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Mirandola A, Kudriavtsev A, Cofre Muñoz CI, Navarro RC, Macagno M, Daoud S, Sanchez C, Pastor B, Pisareva E, Marin MS, Ruiz JG, Piris A, Rodriguez AG, Gonzalez NS, Vivancos A, Quarà V, Mellano A, Borghi F, Corti G, Marchiò C, Sapino A, Bartolini A, Crisafulli G, Bardelli A, Di Maio M, Lossaint G, Frayssinoux F, Crapez E, Ychou M, Soler RS, Fenocchio E, Fernandez Calotti PX, Mazard T, Vivas CS, Elez E, Di Nicolantonio F, Thierry AR. Post-surgery sequelae unrelated to disease progression and chemotherapy revealed in follow-up of patients with stage III colon cancer. EBioMedicine 2024; 108:105352. [PMID: 39303668 PMCID: PMC11437914 DOI: 10.1016/j.ebiom.2024.105352] [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: 12/07/2023] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND We studied the poorly-known dynamics of circulating DNA (cir-nDNA), as monitored prospectively over an extended post-surgery period, in patients with cancer. METHODS On patients with stage III colon cancer (N = 120), using personalised molecular tags we carried out the prospective, multicenter, blinded cohort study of the post-surgery serial analysis of cir-nDNA concentration. 74 patients were included and 357 plasma samples tested. FINDINGS During post-operative follow-up, the patients' median cir-nDNA concentration was greater (P < 0.0001 in the [43-364 days range]) than both the median value in healthy individuals and the pre-surgery value. These cir-nDNA levels were highly associated with NETs markers (P-value associating MPO and cir-nDNA, and NE and cir-nDNA are 6.6 x 10-17, and 1.9 x 10-7), in accordance with previous reports which indicate that cir-nDNA are NETs by-products. Unexpectedly, in 34 out of 50 patients we found that NETs continued to be formed for an extended duration post-surgery, even in patients without disease progression. Given that this phenomenon was observed in patients without adjuvant CT, and in patients >18 months post-surgery, the data suggest that the persistence of NETs formation is not due to the adjuvant CT. INTERPRETATION (1), Given the inter-patient heterogeneity, the post-surgery cir-nDNA level cannot be considered a reliable value, and caution must be exercised when determining mutation allele frequency or the mutation status; and (2), specific studies must be undertaken to investigate the possible clinical impact of the persistent, low-grade inflammation resulting from elevated NETs levels, such as observed in these post-surgery patients, given that such levels are known to potentially induce adverse cardiovascular or thrombotic events. FUNDING This work was supported by the H2020 European ERA-NET grant on Translational Cancer Research (TRANSCAN-2).
Collapse
Affiliation(s)
- Alexia Mirandola
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Andrei Kudriavtsev
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | | | - Raquel Comas Navarro
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Marco Macagno
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Saidi Daoud
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Cynthia Sanchez
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Brice Pastor
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Ekaterina Pisareva
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Mireia Sanchis Marin
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Javier Gonzalo Ruiz
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Alejandro Piris
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | | | - Nadia Saoudi Gonzalez
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Ana Vivancos
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Virginia Quarà
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Alfredo Mellano
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Felice Borghi
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Giorgio Corti
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Caterina Marchiò
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy; Department of Medical Sciences, University of Torino, Turin, Italy
| | - Anna Sapino
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy; Department of Medical Sciences, University of Torino, Turin, Italy
| | - Alice Bartolini
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | - Giovanni Crisafulli
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology, University of Torino, Turin, Italy
| | - Alberto Bardelli
- IFOM, The AIRC Institute of Molecular Oncology, Milan, Italy; Department of Oncology, University of Torino, Turin, Italy
| | | | - Gerald Lossaint
- ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Florence Frayssinoux
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France
| | - Evelyne Crapez
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Marc Ychou
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Ramon Salazar Soler
- Medical Oncology Department, Institut Català d'Oncologia (ICO) - IDIBELL, Barcelona, Spain
| | - Elisabetta Fenocchio
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy
| | | | - Thibault Mazard
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France
| | - Cristina Santos Vivas
- Medical Oncology Department, Institut Català d'Oncologia (ICO) - IDIBELL, Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain
| | - Elena Elez
- VHIO Vall d'Hebron Institute of Oncology, Medical Oncology Department, Barcelona, Spain
| | - Federica Di Nicolantonio
- Istituto di Candiolo - Fondazione del Piemonte per l'Oncologia - IRCCS, Candiolo, Torino, Italy; Department of Oncology, University of Torino, Turin, Italy
| | - Alain R Thierry
- IRCM, Montpellier Cancer Research Institute, INSERM U1194, Montpellier University, Montpellier, F-34298, France; ICM, Institut Régional du Cancer de Montpellier, Montpellier, F-34298, France.
| |
Collapse
|
4
|
Virga A, Gianni C, Palleschi M, Angeli D, Merloni F, Maltoni R, Ulivi P, Martinelli G, De Giorgi U, Bravaccini S. A Novel AKT1, ERBB2, ESR1, KRAS, PIK3CA, and TP53 NGS Assay: A Non-Invasive Tool to Monitor Resistance Mechanisms to Hormonal Therapy and CDK4/6 Inhibitors. Biomedicines 2024; 12:2183. [PMID: 39457497 PMCID: PMC11505462 DOI: 10.3390/biomedicines12102183] [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: 08/22/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Patients with hormone receptor-positive (HR+)/HER2- metastatic breast cancer (mBC) generally receive hormonal therapy (HT) combined with CDK4/6 inhibitors (CDK4/6i). Despite this treatment, resistance mechanisms to CDK4/6i emerge and the majority of these patients experience disease progression (PD). This highlight the necessity to uncover the resistance mechanism to CDK4/6i through the identification of specific biomarkers. The primary objective is to assess the accuracy and feasibility of a novel multi-gene target panel NGS assay on circulating tumor DNA (ctDNA) to detect molecular alterations of AKT1, ERBB2, ESR1, KRAS, PIK3CA, and TP53 genes in women with BC undergoing HT plus CDK4/6i treatment. Secondarily, the study aims to explore the relationship between genomic profiling and clinical outcomes. Materials and Methods: Plasma samples were collected from 16 patients diagnosed with advanced/locally advanced HR+/HER2- BC at 2 time points: T0 (baseline) and at T1 (3 months after CDK4/6i treatment). Starting from 2 mL of plasma, ctDNA was isolated and libraries were set up using the Plasma-SeqSensei (PQS)® Breast Cancer IVD Kit, sequenced on Nextseq 550 and analyzed using the Plasma-SeqSensei™ IVD Software®. Results: Among the five patients who presented PD, three had PIK3CA mutations and, of these, two showed a higher mutant allele frequency (MAF) at T1. In three patients with stable disease and in eight patients with partial response, the MAF of the detected alterations decreased dramatically or disappeared during CDK4/6i treatment. Conclusions: Based on our findings, the liquid biopsy analysis using the PQS panel seems to be both feasible and accurate, demonstrating a strong sensitivity in detecting mutations. This exploratory analysis of the clinical outcome associated to the mutational status of patients highlights the potential of molecular analysis on liquid biopsy for disease monitoring, although further validation with a larger patient cohort is necessary to confirm these preliminary observations.
Collapse
Affiliation(s)
- Alessandra Virga
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (P.U.); (S.B.)
| | - Caterina Gianni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.G.); (M.P.); (F.M.); (R.M.); (U.D.G.)
| | - Michela Palleschi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.G.); (M.P.); (F.M.); (R.M.); (U.D.G.)
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Filippo Merloni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.G.); (M.P.); (F.M.); (R.M.); (U.D.G.)
| | - Roberta Maltoni
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.G.); (M.P.); (F.M.); (R.M.); (U.D.G.)
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (P.U.); (S.B.)
| | - Giovanni Martinelli
- Department of Hematology and Sciences Oncology, Institute of Haematology “L. and A. Seràgnoli”, S. Orsola University Hospital, 40138 Bologna, Italy;
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.G.); (M.P.); (F.M.); (R.M.); (U.D.G.)
| | - Sara Bravaccini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (P.U.); (S.B.)
| |
Collapse
|
5
|
Devalle S, Aran V, Bastos Júnior CDS, Pannain VL, Brackmann P, Gregório ML, Ferreira Manso JE, Moura Neto V. A panorama of colon cancer in the era of liquid biopsy. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100148. [PMID: 40027146 PMCID: PMC11863817 DOI: 10.1016/j.jlb.2024.100148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/05/2025]
Abstract
Colon cancer (CC) is one of the most frequent cancers worldwide being responsible for over 500 thousand deaths in 2022. Its financial and human burden is expected to increase in the next decades accompanying the growing and aging of the global population. Much of this burden could be alleviated considering that the lethality of CC is mostly due to its late diagnosis and failure in the individualized management of patients. Coordinated government actions and implementation of better diagnostic tools capable of detecting CC earlier and of tracking tumoral evolution are mandatory to achieve a reduction in CC's social impact. CtDNA-based liquid biopsy (LB) has great potential to contribute to patients' screening adhesion, CC earlier detection, and to longitudinal tumor follow-up. In this review, we will discuss the latest epidemiological data on CC disease, diagnostic, subtypes, genetics, and treatment management focusing on the advantages and limitations of ctDNA-based LB, including important bottlenecks and solutions necessary for its clinical translation. The latest ctDNA-directed CC clinical trials will also be examined.
Collapse
Affiliation(s)
- Sylvie Devalle
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | - Veronica Aran
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| | | | - Vera Lucia Pannain
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paulo Brackmann
- Clínica de Coloproctologia do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - Marcelo Leal Gregório
- Instituto de Pesquisas Biomédicas do Hospital Naval Marcílio Dias - IPB/HNMD, Rio de Janeiro, Brazil
| | - José Eduardo Ferreira Manso
- Departamento de Cirurgia, Faculdade de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vivaldo Moura Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria de Estado de Saúde, Rio de Janeiro, Brazil
| |
Collapse
|
6
|
Anayyat U, Ahad F, Muluh TA, Zaidi SAA, Usmani F, Yang H, Li M, Hassan HA, Wang X. Immunotherapy: Constructive Approach for Breast Cancer Treatment. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:925-951. [PMID: 38116189 PMCID: PMC10729681 DOI: 10.2147/bctt.s424624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
A novel and rapid therapeutic approach is the treatment of human breast cancer by enhancing the host's immune system. In initial findings, program death one (PD-1) and program cell death ligand one (PD-L1) showed positive results towards solid tumors, but tumor relapse and drug resistance are the major concerns. Breast cancer therapy has been transformed by the advent of immune checkpoint blockades (ICBs). Triple-negative breast cancers (TNBCs) have exhibited enduring responses to clinical usage of immune checkpoint inhibitors (ICBs) like atezolizumab and pembrolizumab. Nonetheless, a notable proportion of individuals with TNBC do not experience advantages from these treatments, and there is limited comprehension of the resistance mechanisms. Another approach to overcome resistance is cancer stem cells (CSCs), as these cells are crucial for the initiation and growth of tumors in the body. Various cancer vaccines are created using stem cells (dendritic, whole cell, bacterial) and focus primarily on targeting tumor-related antigens. The ultimate objective of cancer vaccines is to immunize the patients by active artificial immunity against cancer, though. In this review, we primarily focused on existing immunotherapeutic options, immune checkpoint blockers, the latest progress in understanding the molecular mechanisms underlying resistance to immune checkpoint inhibitors (ICBs), advanced strategies to overcome resistance to ICBs, cancer stem cell antigens and molecular markers, ongoing clinical trials for BCs and cancer vaccines for breast cancer.
Collapse
Affiliation(s)
- Umer Anayyat
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Faiza Ahad
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Tobias Achu Muluh
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Syed Aqib Ali Zaidi
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Faiza Usmani
- Department of Biotechnology, University of Karachi, Karachi, Pakistan
| | - Hua Yang
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Mengqing Li
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Hammad Ali Hassan
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Xiaomei Wang
- Department of Physiology, School of Basic Medical Sciences, Health Sciences Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518055, People’s Republic of China
| |
Collapse
|
7
|
Lavacchi D, Gelmini S, Calabri A, Rossi G, Simi L, Caliman E, Mancini I, Salvianti F, Petroni G, Guidolin A, Scolari F, Messerini L, Pillozzi S, Pinzani P, Antonuzzo L. Early changes in circulating tumor DNA (ctDNA) predict treatment response in metastatic KRAS-mutated colorectal cancer (mCRC) patients. Heliyon 2023; 9:e21853. [PMID: 38027900 PMCID: PMC10663919 DOI: 10.1016/j.heliyon.2023.e21853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
The detection of RAS mutations and co-mutations in liquid biopsy offers a novel paradigm for the dynamic management of metastatic colorectal cancer (mCRC) patients. Expanding the results of the prospective OMITERC (OMIcs application from solid to liquid biopsy for a personalized ThERapy of Cancer) project, we collected blood samples at specific time points from patients who received a first-line chemotherapy (CT) for KRAS-mutated mCRC. CTC quantification was performed by CellSearch® system. Libraries from cfDNA were prepared using the Oncomine™ Colon cfDNA Assay to detect tumour-derived DNA in cfDNA. The analysis involved >240 hotspots in 14 genes. Twenty patients with KRAS-mutated mCRC treated at the Medical Oncology Unit of Careggi University Hospital were prospectively enrolled. Nine patients had available data for longitudinal monitoring of cfDNA. After 6 weeks of first-line CT an increase of KRAS-mutated clone was reported in the only patient who did not obtain disease control, while all patients with decrease of KRAS clones obtained disease control. Overall, in patients with a short (<9 months) progression-free survival (PFS) we registered, at 6 weeks, an increase in cfDNA levels and in KRAS mutations or other co-mutations, i.e. PIK3CA, FBXW7, GNAS, and TP53. In selected cases, co-mutations were able to better anticipate radiological progressive disease (PD) than the increase of KRAS-mutated clones. In conclusion, our study confirms plasma ctDNA as a crucial tool for anticipating PD at an early time point and highlights the value of a comprehensive assessment of clonal dynamics to improve the management of patients with mCRC.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Stefania Gelmini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Adele Calabri
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Gemma Rossi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Lisa Simi
- Clinical and Molecular Biochemistry Careggi University Hospital, Florence, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Irene Mancini
- Clinical and Molecular Biochemistry Careggi University Hospital, Florence, Italy
| | - Francesca Salvianti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Giulia Petroni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessia Guidolin
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Federico Scolari
- Department of Health Science, University of Florence, Florence, Italy
| | - Luca Messerini
- Pathology Unit, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Pamela Pinzani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
- Clinical and Molecular Biochemistry Careggi University Hospital, Florence, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
8
|
Verlicchi A, Canale M, Chiadini E, Cravero P, Urbini M, Andrikou K, Pasini L, Flospergher M, Burgio MA, Crinò L, Ulivi P, Delmonte A. The Clinical Significance of Circulating Tumor DNA for Minimal Residual Disease Identification in Early-Stage Non-Small Cell Lung Cancer. Life (Basel) 2023; 13:1915. [PMID: 37763318 PMCID: PMC10532754 DOI: 10.3390/life13091915] [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: 07/07/2023] [Revised: 08/18/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Lung cancer (LC) is the deadliest malignancy worldwide. In an operable stage I-III patient setting, the detection of minimal residual disease (MRD) after curative treatment could identify patients at higher risk of relapse. In this context, the study of circulating tumor DNA (ctDNA) is emerging as a useful tool to identify patients who could benefit from an adjuvant treatment, and patients who could avoid adverse events related to a more aggressive clinical management. On the other hand, ctDNA profiling presents technical, biological and standardization challenges before entering clinical practice as a decisional tool. In this paper, we review the latest advances regarding the role of ctDNA in identifying MRD and in predicting patients' prognosis, with a particular focus on clinical trials investigating the potential of ctDNA, the technical challenges to address and the biological parameters that influence the MRD detection.
Collapse
Affiliation(s)
- Alberto Verlicchi
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| | - Matteo Canale
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.C.); (M.U.); (L.P.); (P.U.)
| | - Elisa Chiadini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.C.); (M.U.); (L.P.); (P.U.)
| | - Paola Cravero
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.C.); (M.U.); (L.P.); (P.U.)
| | - Kalliopi Andrikou
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| | - Luigi Pasini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.C.); (M.U.); (L.P.); (P.U.)
| | - Michele Flospergher
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| | - Marco Angelo Burgio
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.C.); (M.U.); (L.P.); (P.U.)
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.V.); (P.C.); (K.A.); (M.F.); (M.A.B.); (L.C.); (A.D.)
| |
Collapse
|
9
|
Saoudi González N, Salvà F, Ros J, Baraibar I, Rodríguez-Castells M, García A, Alcaráz A, Vega S, Bueno S, Tabernero J, Elez E. Unravelling the Complexity of Colorectal Cancer: Heterogeneity, Clonal Evolution, and Clinical Implications. Cancers (Basel) 2023; 15:4020. [PMID: 37627048 PMCID: PMC10452468 DOI: 10.3390/cancers15164020] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Colorectal cancer (CRC) is a global health concern and a leading cause of death worldwide. The disease's course and response to treatment are significantly influenced by its heterogeneity, both within a single lesion and between primary and metastatic sites. Biomarkers, such as mutations in KRAS, NRAS, and BRAF, provide valuable guidance for treatment decisions in patients with metastatic CRC. While high concordance exists between mutational status in primary and metastatic lesions, some heterogeneity may be present. Circulating tumor DNA (ctDNA) analysis has proven invaluable in identifying genetic heterogeneity and predicting prognosis in RAS-mutated metastatic CRC patients. Tumor heterogeneity can arise from genetic and non-genetic factors, affecting tumor development and response to therapy. To comprehend and address clonal evolution and intratumoral heterogeneity, comprehensive genomic studies employing techniques such as next-generation sequencing and computational analysis are essential. Liquid biopsy, notably through analysis of ctDNA, enables real-time clonal evolution and treatment response monitoring. However, challenges remain in standardizing procedures and accurately characterizing tumor subpopulations. Various models elucidate the origin of CRC heterogeneity, highlighting the intricate molecular pathways involved. This review focuses on intrapatient cancer heterogeneity and genetic clonal evolution in metastatic CRC, with an emphasis on clinical applications.
Collapse
Affiliation(s)
- Nadia Saoudi González
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Francesc Salvà
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Javier Ros
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Iosune Baraibar
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Marta Rodríguez-Castells
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Ariadna García
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
| | - Adriana Alcaráz
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Sharela Vega
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Sergio Bueno
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Josep Tabernero
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| | - Elena Elez
- Vall d’Hebron Institute of Oncology, 08035 Barcelona, Spain; (N.S.G.)
- Oncology Department, Vall d’Hebron Hospital, 08035 Barcelona, Spain
| |
Collapse
|
10
|
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
|
11
|
Rao W, Liu Y, Li Y, Guo L, Qiu T, Dong L, Ying J, Li W. Potential unreliability of ALK variant allele frequency in the efficacy prediction of targeted therapy in NSCLC. Front Med 2023; 17:493-502. [PMID: 37010729 DOI: 10.1007/s11684-022-0946-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/28/2022] [Indexed: 04/04/2023]
Abstract
Anaplastic lymphoma kinase (ALK) is the most common fusion gene involved in non-small cell lung cancer (NSCLC), and remarkable response has been achieved with the use of ALK tyrosine kinase inhibitors (ALK-TKIs). However, the clinical efficacy is highly variable. Pre-existing intratumoral heterogeneity (ITH) has been proven to contribute to the poor treatment response and the resistance to targeted therapies. In this work, we investigated whether the variant allele frequencies (VAFs) of ALK fusions can help assess ITH and predict targeted therapy efficacy. Through the application of next-generation sequencing (NGS), 7.2% (326/4548) of patients were detected to be ALK positive. On the basis of the adjusted VAF (adjVAF, VAF normalization for tumor purity) of four different threshold values (adjVAF < 50%, 40%, 30%, or 20%), the association of ALK subclonality with crizotinib efficacy was assessed. Nonetheless, no statistical association was observed between median progression-free survival (PFS) and ALK subclonality assessed by adjVAF, and a poor correlation of adjVAF with PFS was found among the 85 patients who received first-line crizotinib. Results suggest that the ALK VAF determined by hybrid capture-based NGS is probably unreliable for ITH assessment and targeted therapy efficacy prediction in NSCLC.
Collapse
Affiliation(s)
- Wei Rao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
12
|
Ros J, Salva F, Dopazo C, López D, Saoudi N, Baraibar I, Charco R, Tabernero J, Elez E. Liver transplantation in metastatic colorectal cancer: are we ready for it? Br J Cancer 2023; 128:1797-1806. [PMID: 36879000 PMCID: PMC10147684 DOI: 10.1038/s41416-023-02213-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/10/2023] [Accepted: 02/17/2023] [Indexed: 03/08/2023] Open
Abstract
Colorectal cancer (CRC) is a prevalent disease worldwide, with more than 50% of patients developing metastases to the liver. Five-year overall survival remains modest among patients with metastatic CRC (mCRC) treated with conventional therapies however, liver transplantation in a highly selected population can improve clinical outcomes with an impressive 5-year overall survival of 83%. Despite liver transplantation appearing to be a promising therapeutical option for well-selected patients with mCRC with the liver-limited disease, these data come from small monocentric trials which included a heterogeneous population. Currently, several clinical trials are evaluating liver transplantation in this scenario, aiming for a more accurate patient selection by integrating liquid biopsy, tissue profiling, and nuclear medicine to the already known clinical biomarkers that eventually may lead to a survival improvement. In this paper, the clinical outcomes and inclusion criteria from the most relevant clinical trials and clinical series involving liver transplantation in patients with liver-limited disease colorectal cancer are reviewed as well as the trials currently recruiting.
Collapse
Affiliation(s)
- Javier Ros
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain.
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131, Naples, Italy.
| | - Francesc Salva
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Cristina Dopazo
- Department of HPB Surgery and Transplants, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Daniel López
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Nadia Saoudi
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Iosune Baraibar
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Ramon Charco
- Department of HPB Surgery and Transplants, Vall d'Hebron Hospital Universitari, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Elena Elez
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| |
Collapse
|
13
|
Economopoulou P, Spathis A, Kotsantis I, Maratou E, Anastasiou M, Moutafi MK, Kirkasiadou M, Pantazopoulos A, Giannakakou M, Edelstein DL, Sloane H, Fredebohm J, Jones FS, Kyriazoglou A, Gavrielatou N, Foukas P, Panayiotides I, Psyrri A. Next-generation sequencing (NGS) profiling of matched tumor and circulating tumor DNA (ctDNA) in head and neck squamous cell carcinoma (HNSCC). Oral Oncol 2023; 139:106358. [PMID: 36871349 DOI: 10.1016/j.oraloncology.2023.106358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023]
Abstract
OBJECTIVES The aim of this pilot study was to evaluate the presence of somatic mutations in matched tumor and circulating DNA (ctDNA) samples from patients with primary head and neck squamous cell carcinoma (HNSCC) and assess the association of changes in ctDNA levels with survival. MATERIALS AND METHODS Our study included 62 patients with stage I-IVB HNSCC treated with surgery or radical chemoradiotherapy with curative intent. Plasma samples were obtained at baseline, at the end of treatment (EOT), and at disease progression. Tumor DNA was extracted from plasma (ctDNA) and tumor tissue (tDNA). The Safe Sequencing System was used assess the presence of pathogenic variants in four genes (TP53, CDKN2A, HRAS and PI3KCA) in both ctDNA and tDNA. RESULTS Forty-five patients had available tissue and plasma samples. Concordance of genotyping results between tDNA and ctDNA at baseline was 53.3%. TP53 mutations were most commonly identified at baseline in both ctDNA (32.6%) and tDNA (40%). The presence of mutations in this restricted set of 4 genes in tissue samples at baseline was associated with decreased overall survival (OS) [median 58.3 months for patients with mutations vs. 89 months for patients without mutations, p < 0.013]. Similarly, patients presenting with mutations in ctDNA had shorter OS [median 53.8 vs. 78.6 months, p < 0.037]. CtDNA clearance at EOT did not show any association with PFS or OS. CONCLUSIONS Liquid biopsy enables real-time molecular characterization of HNSCC and might predict survival. Larger studies are needed to validate the utility of ctDNA as a biomarker in HNSCC.
Collapse
Affiliation(s)
- Panagiota Economopoulou
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Aris Spathis
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Ioannis Kotsantis
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Eirini Maratou
- Second Department of Internal Medicine and Research Institute, School of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Maria Anastasiou
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Myrto K Moutafi
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Maria Kirkasiadou
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Anastasios Pantazopoulos
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Maria Giannakakou
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Daniel L Edelstein
- Medical Affairs, Sysmex Inostics Inc., 1812 Ashland Ave #500, Baltimore, MD 21205, USA
| | - Hillary Sloane
- Medical Affairs, Sysmex Inostics Inc., 1812 Ashland Ave #500, Baltimore, MD 21205, USA
| | - Johannes Fredebohm
- Research and Innovation, Sysmex Inostics GmbH, Alkenried 88, 20251 Hamburg, Germany.
| | - Frederick S Jones
- Research and Innovation, Sysmex Inostics GmbH, Alkenried 88, 20251 Hamburg, Germany.
| | - Anastasios Kyriazoglou
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Niki Gavrielatou
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece.
| | - Periklis Foukas
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Ioannis Panayiotides
- Second Department of Pathology, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece
| | - Amanda Psyrri
- Section of Medical Oncology, Second Department of Internal Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, 1(st) Rimini St, 12462 Haidari, Athens, Greece.
| |
Collapse
|
14
|
Vidal J, Fernández-Rodríguez MC, Casadevall D, García-Alfonso P, Páez D, Guix M, Alonso V, Cano MT, Santos C, Durán G, Elez E, Manzano JL, Garcia-Carbonero R, Ferreiro R, Losa F, Pineda E, Sastre J, Rivera F, Bellosillo B, Tabernero J, Aranda E, Salazar R, Montagut C. Liquid Biopsy Detects Early Molecular Response and Predicts Benefit to First-Line Chemotherapy plus Cetuximab in Metastatic Colorectal Cancer: PLATFORM-B Study. Clin Cancer Res 2023; 29:379-388. [PMID: 36074154 DOI: 10.1158/1078-0432.ccr-22-1696] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/22/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
PURPOSE Chemotherapy plus anti-EGFR is standard first-line therapy in RAS wild-type (wt) metastatic colorectal cancer (mCRC), but biomarkers of early response are clinically needed. We aimed to define the utility of ctDNA to assess early response in patients with mCRC receiving first-line anti-EGFR therapy. EXPERIMENTAL DESIGN Prospective multicentric study of tissue patients with RAS wt mCRC treated with first-line chemotherapy plus cetuximab undergoing sequential liquid biopsies. Baseline and early (C3) ctDNA were analyzed by NGS. Trunk mutations were assessed as surrogate marker of total tumor burden. RAS/BRAF/MEK/EGFR-ECD were considered mutations of resistance. ctDNA results were correlated with clinical outcome. RESULTS One hundred patients were included. ctDNA was detected in 72% of patients at baseline and 34% at C3. Decrease in ctDNA trunk mutations correlated with progression-free survival (PFS; HR, 0.23; P = 0.001). RAS/BRAF were the only resistant mutations detected at C3. An increase in the relative fraction of RAS/BRAF at C3 was followed by an expansion of the RAS clone until PD, and was associated with shorter PFS (HR, 10.5; P < 0.001). The best predictor of response was the combined analysis of trunk and resistant mutations at C3. Accordingly, patients with "early molecular response" (decrease in trunk and decrease in resistant mutations) had better response (77.5% vs. 25%, P = 0.008) and longer PFS (HR, 0.18; P < 0.001) compared with patients with "early molecular progression" (increase in trunk and/or increase in resistant mutations). CONCLUSIONS ctDNA detects early molecular response and predicts benefit to chemotherapy plus cetuximab. A comprehensive NGS-based approach is recommended to integrate information on total disease burden and resistant mutations. See related commentary by Eluri et al., p. 302.
Collapse
Affiliation(s)
- Joana Vidal
- Medical Oncology Department, Hospital del Mar, Institut Mar Investigacions Mèdiques (IMIM), Universitat Pompeu Fabra, CIBERONC, Barcelona, Spain
| | | | - David Casadevall
- Medical Oncology Department, Hospital del Mar, Institut Mar Investigacions Mèdiques (IMIM), Universitat Pompeu Fabra, CIBERONC, Barcelona, Spain
| | | | - David Páez
- Medical Oncology Department, H. Santa Creu i Sant Pau, Barcelona, Spain
| | - Marta Guix
- Medical Oncology Department, Hospital del Mar, Institut Mar Investigacions Mèdiques (IMIM), Universitat Pompeu Fabra, CIBERONC, Barcelona, Spain
| | - Vicente Alonso
- Medical Oncology Department, H. Miguel Servet, Zaragoza, Spain
| | - Maria Teresa Cano
- Medical Oncology Department, IMIBIC, Reina Sofía Hospital, University of Cordoba, CIBERONC, Cordoba, Spain
| | - Cristina Santos
- Medical Oncology Department, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL)-CIBERONC, Barcelona, Spain
| | - Gema Durán
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Elena Elez
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - José Luís Manzano
- Medical Oncology Department, ICO, H. Germans Trias i Pujol, Barcelona, Spain
| | - Rocío Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (Imas12), UCM, Madrid, Spain
| | - Reyes Ferreiro
- Medical Oncology Department, Hospital Ramón y Cajal, Madrid, Spain
| | - Ferran Losa
- Medical Oncology Department, Hospital Sant Joan Despí - Moisès Broggi, ICO-Hospitalet, Barcelona. Spain
| | - Estela Pineda
- Medical Oncology Department Hospital Clínic, Barcelona, Spain
| | - Javier Sastre
- Medical Oncology Department, Hospital Universitario Clínico San Carlos, CIBERONC, Madrid, Spain
| | - Fernando Rivera
- Medical Oncology Department Hospital Marqués de Valdecilla, IDIVAL, Santander, Spain
| | | | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Enrique Aranda
- Medical Oncology Department, IMIBIC, Reina Sofía Hospital, University of Cordoba, CIBERONC, Cordoba, Spain
| | - Ramon Salazar
- Medical Oncology Department, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL)-CIBERONC, Barcelona, Spain
| | - Clara Montagut
- Medical Oncology Department, Hospital del Mar, Institut Mar Investigacions Mèdiques (IMIM), Universitat Pompeu Fabra, CIBERONC, Barcelona, Spain
| |
Collapse
|
15
|
Spiliopoulou P, Vornicova O, Genta S, Spreafico A. Shaping the Future of Immunotherapy Targets and Biomarkers in Melanoma and Non-Melanoma Cutaneous Cancers. Int J Mol Sci 2023; 24:1294. [PMID: 36674809 PMCID: PMC9862040 DOI: 10.3390/ijms24021294] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Recent advances in treating cutaneous melanoma have resulted in impressive patient survival gains. Refinement of disease staging and accurate patient risk classification have significantly improved our prognostic knowledge and ability to accurately stratify treatment. Undoubtedly, the most important step towards optimizing patient outcomes has been the advent of cancer immunotherapy, in the form of immune checkpoint inhibition (ICI). Immunotherapy has established its cardinal role in the management of both early and late-stage melanoma. Through leveraging outcomes in melanoma, immunotherapy has also extended its benefit to other types of skin cancers. In this review, we endeavor to summarize the current role of immunotherapy in melanoma and non-melanoma skin cancers, highlight the most pertinent immunotherapy-related molecular biomarkers, and lastly, shed light on future research directions.
Collapse
Affiliation(s)
- Pavlina Spiliopoulou
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Olga Vornicova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
- Mount Sinai Hospital, University Health Network, Toronto, ON M5G 1X5, Canada
| | - Sofia Genta
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada
| |
Collapse
|
16
|
A phase II study to explore biomarkers for the use of mFOLFOX6/XELOX plus bevacizumab as a first-line chemotherapy in patients with metastatic colorectal cancer (WJOG7612GTR). ESMO Open 2022; 7:100592. [PMID: 36502778 PMCID: PMC9808456 DOI: 10.1016/j.esmoop.2022.100592] [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: 08/03/2021] [Revised: 08/07/2022] [Accepted: 08/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The purpose of this prospective study was to assess the ability of plasma vascular endothelial growth factor-A short isoforms (pVEGF-Asi) to predict bevacizumab (BV) efficacy and to explore other circulating biomarkers in metastatic colorectal cancer (mCRC) patients treated with modified FOLFOX6/XELOX plus BV (mFOLFOX6/XELOX + BV). PATIENTS AND METHODS Pre-treatment plasma samples were collected from 100 mCRC patients receiving first-line chemotherapy with mFOLFOX6/XELOX + BV. The plasma levels of 11 angiogenesis-associated molecules, including pVEGF-Asi and 22 cancer-associated gene mutations in circulating tumor DNA, were analyzed. For the primary endpoint, we assumed that the hazard ratio (HR) for progression-free survival (PFS) calculated using a Cox proportional hazards model was <1.15, comparing patients with a high versus those with a low pVEGF-Asi level divided according to the median pVEGF-Asi value. RESULTS The median value of pVEGF-Asi was 37 (range 6.5-262) pg/ml. The HR for PFS between the high and low pVEGF-Asi patient groups was 1.3 [95% confidence interval (CI) 0.8-2.1; log rank, P = 0.25], which was larger than the predefined threshold of 1.15. The multivariate analysis demonstrated that PFS was significantly associated with plasma intercellular adhesion molecule-1 (pICAM-1) (≥190.0 versus <190.0 ng/ml; HR 2.1; 95% CI 1.3-3.5), RAS (mutant versus wild; HR 2.5; 95% CI 1.5-4.3), and FBXW7 (mutant versus wild; HR 2.8; 95% CI 1.2-6.8), whereas overall survival was significantly associated with pICAM-1 (HR 2.0; 95% CI 1.1-3.7) and RAS (HR 2.6; 95% CI 1.5-4.6). CONCLUSIONS The addition of BV was unable to compensate for the poor PFS associated with a high pVEGF-Asi level, suggesting that pVEGF-Asi is unlikely to be a good predictive biomarker of the efficacy of mFOLFOX6/XELOX + BV therapy. The clinical significance of circulating ICAM-1, mutant RAS, and mutant FBXW7 levels should be studied further.
Collapse
|
17
|
Callesen LB, Hamfjord J, Boysen AK, Pallisgaard N, Guren TK, Kure EH, Spindler KLG. Circulating tumour DNA and its clinical utility in predicting treatment response or survival in patients with metastatic colorectal cancer: a systematic review and meta-analysis. Br J Cancer 2022; 127:500-513. [PMID: 35440666 PMCID: PMC9345951 DOI: 10.1038/s41416-022-01816-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We investigate the current knowledge on circulating tumour DNA (ctDNA) and its clinical utility in predicting outcomes in patients with metastatic colorectal cancer (mCRC). METHODS PubMed, Embase, Cochrane Database of Systematic Reviews and Cochrane Central Register of Controlled Trials were searched. Last search 16/12/2020. We included studies on patients with mCRC reporting the predictive or prognostic value of ctDNA. We performed separate random-effects meta-analyses to investigate if baseline ctDNA and early changes in ctDNA levels during treatment were associated with survival. The risk of bias was assessed according to the Quality in Prognosis Studies tool. RESULTS Seventy-one studies were included with 6930 patients. Twenty-four studies were included in meta-analyses. High baseline ctDNA level was associated with short progression-free survival (PFS) (HR = 2.2; 95% CI 1.8-2.8; n = 509) and overall survival (OS) (HR = 2.4; 95% CI 1.9-3.1; n = 1336). A small or no early decrease in ctDNA levels during treatment was associated with short PFS (HR = 3.0; 95% CI 2.2-4.2; n = 479) and OS (HR = 2.8; 95% CI 2.1-3.9; n = 583). Results on clonal evolution and lead-time were inconsistent. A majority of included studies (n = 50/71) had high risk of bias in at least one domain. CONCLUSIONS Plasma ctDNA is a strong prognostic biomarker in mCRC. However, true clinical utility is lacking.
Collapse
Affiliation(s)
- Louise B Callesen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Julian Hamfjord
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anders K Boysen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Pallisgaard
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Tormod K Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Elin H Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Natural Sciences and Environmental Health, Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Campus Bø, Bø, Norway
| | - Karen-Lise G Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
18
|
Boccaccino A, Borelli B, Intini R, Antista M, Bensi M, Rossini D, Passardi A, Tamberi S, Giampieri R, Antonuzzo L, Noto L, Roviello G, Zichi C, Salati M, Puccini A, Noto C, Parisi A, Rihawi K, Persano M, Crespi V, Libertini M, Giordano M, Moretto R, Lonardi S, Cremolini C. Encorafenib plus cetuximab with or without binimetinib in patients with BRAF V600E-mutated metastatic colorectal cancer: real-life data from an Italian multicenter experience. ESMO Open 2022; 7:100506. [PMID: 35696748 PMCID: PMC9271503 DOI: 10.1016/j.esmoop.2022.100506] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/03/2022] [Accepted: 04/06/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Encorafenib plus cetuximab with or without binimetinib showed increased objective response rate (ORR), progression-free survival (PFS), and overall survival (OS) compared with chemotherapy plus anti-EGFR in previously treated patients with BRAF V600E-mutated (mut) metastatic colorectal cancer (mCRC). Although no formal comparison was planned, addition of binimetinib to encorafenib plus cetuximab did not provide significant efficacy advantage. PATIENTS AND METHODS This real-life study was aimed at evaluating safety, activity, and efficacy of encorafenib plus cetuximab with or without binimetinib in patients with BRAF V600E-mut mCRC treated at 21 Italian centers within a nominal use program launched in May 2019. RESULTS Out of 133 patients included, 97 (73%) received encorafenib plus cetuximab (targeted doublet) and 36 (27%) the same therapy plus binimetinib (targeted triplet). Most patients had Eastern Cooperative Group Performance Status (ECOG-PS) of 0 or 1 (86%), right-sided primary tumor (69%), and synchronous disease (66%). Twenty (15%) tumors were DNA mismatch repair deficiency (dMMR)/microsatellite instability (MSI)-high. As many as 44 (34%) patients had received two or more prior lines of therapy, 122 (92%) were previously exposed to oxaliplatin, and 109 (82%) to anti-vascular endothelial growth factor (anti-VEGF). Most frequent adverse events were asthenia (62%) and anti-EGFR-related skin rash (52%). Any grade nausea (P = 0.03), vomiting (P = 0.04), and diarrhea (P = 0.07) were more frequent with the triplet therapy, while melanocytic nevi were less common (P = 0.06). Overall, ORR and disease control rate (DCR) were 23% and 69%, respectively, with numerically higher rates in the triplet group (ORR 31% versus 17%, P = 0.12; DCR 78% versus 65%, P = 0.23). Median PFS and OS were 4.5 and 7.2 months, respectively. Worse ECOG-PS, peritoneal metastases, and more than one prior treatment were independent poor prognostic factors for PFS and OS. Clonality of BRAF mutation measured as adjusted mutant allele fraction in tumor tissue was not associated with clinical outcome. CONCLUSIONS Our real-life data are consistent with those from the BEACON trial in terms of safety, activity, and efficacy. Patients in good general condition and not heavily pretreated are those more likely to derive benefit from the targeted treatment.
Collapse
Affiliation(s)
- A Boccaccino
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - B Borelli
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - R Intini
- Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology IOV-IRCCSP, Padova, Italy
| | - M Antista
- Medical Oncology Department, ENETS Center of Excellence, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - M Bensi
- Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Università Cattolica del Sacro Cuore, Roma, Italy
| | - D Rossini
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - A Passardi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - S Tamberi
- UOC Oncologia Ravenna, AUSL Romagna, Ravenna, Italy
| | - R Giampieri
- Clinica Oncologica, Dipartimento Scienze Cliniche e Molecolari, Università Politecnica delle Marche, Ancona, Italy
| | - L Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - L Noto
- UOC Oncologia Medica, Policlinico "G.Rodolico" AOU Policlinico - San Marco, Catania, Italy
| | - G Roviello
- Department of Health Sciences, University of Florence, Florence, Italy
| | - C Zichi
- Oncologia Medica, A.O. Ordine Mauriziano - Umberto I, Torino, Italy
| | - M Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy; PhD Program Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - A Puccini
- Università degli Studi di Genova, Ospedale Policlinico San Martino IRCCS, Genova, Italy
| | - C Noto
- Università degli Studi di Udine, Dipartimento di Area Medica, Udine, Italy; Azienda Sanitaria Universitaria Friuli Centrale, Dipartimento di Oncologia medica, Udine, Italy
| | - A Parisi
- Medical Oncology, St. Salvatore Hospital, L'Aquila, Italy; Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - K Rihawi
- IRCSS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - M Persano
- Medical Oncology, University of Cagliari, Cagliari, Italy
| | - V Crespi
- Department of Oncology, University of Turin, Torino, Italy
| | - M Libertini
- Oncology Unit, Poliambulanza Foundation, Brescia, Italy
| | - M Giordano
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - R Moretto
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - S Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy.
| | - C Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology 2, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| |
Collapse
|
19
|
Li M, Yang L, Hughes J, van den Hout A, Burns C, Woodhouse R, Dennis L, Hegde P, Oxnard GR, Vietz C. Driver Mutation Variant Allele Frequency in Circulating Tumor DNA and Association with Clinical Outcome in Patients with Non-Small Cell Lung Cancer and EGFR- and KRAS-Mutated Tumors. J Mol Diagn 2022; 24:543-553. [PMID: 35301118 DOI: 10.1016/j.jmoldx.2022.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/21/2021] [Accepted: 02/04/2022] [Indexed: 10/18/2022] Open
Abstract
Clinical implementation of mutational analysis for next-generation sequencing-based assays has largely been of a binary nature, with pathogenic mutations being reported as either positive or negative. Actionability on the continuous output of variant allele frequency (VAF) has not been well characterized in the clinical setting, thus limiting its use. In this study, analytical validity and performance of the short variant VAF based on a next-generation sequencing-based liquid biopsy assay, FoundationOne Liquid CDx, were evaluated through assessment of precision, analytical accuracy, limit of blank, and limit of detection. Analytical validation of VAF values measured by using FoundationOne Liquid CDx supports that these values are accurate, precise, robust, and linear with the true VAF value. The association of allele frequency of clinically relevant short variants in circulating-free DNA and the association with overall survival for patients using real-world data were also assessed. The results of the association analysis indicate that VAF of the predictive biomarker mutation negatively correlates with overall survival of patients with non-small cell lung cancer.
Collapse
Affiliation(s)
- Meijuan Li
- Foundation Medicine Inc., Cambridge, Massachusetts.
| | - Lei Yang
- Foundation Medicine Inc., Cambridge, Massachusetts
| | - Jason Hughes
- Foundation Medicine Inc., Cambridge, Massachusetts
| | | | | | | | - Lucas Dennis
- Foundation Medicine Inc., Cambridge, Massachusetts
| | - Priti Hegde
- Foundation Medicine Inc., Cambridge, Massachusetts
| | | | | |
Collapse
|
20
|
Jiang M, Zhou H, Jiang S, Yu H. A Review of Circulating Tumor DNA in the Diagnosis and Monitoring of Esophageal Cancer. Med Sci Monit 2022; 28:e934106. [PMID: 35210388 PMCID: PMC8886734 DOI: 10.12659/msm.934106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Circulating tumor DNA (ctDNA) is a type of cell-free DNA released by tumor cells after necrosis and apoptosis, and it can be actively secreted by tumor cells. Since ctDNA is derived from various tumor sites, it can provide far more comprehensive genomic and epigenomic information than a single-site biopsy. Therefore, ctDNA can overcome tumor heterogeneity, which is the major limitation of a traditional tissue biopsy approach. Noninvasive ctDNA assays allow continuous real-time monitoring of the molecular status of cancers. Recently, ctDNA assays have been widely used in clinical practice, including cancer diagnosis, evaluation of therapeutic efficacy and prognosis, and monitoring of relapse and metastasis. Although ctDNA shows a high diagnostic performance in advanced esophageal cancer, it is far from satisfactory for early diagnosis of esophageal cancer. Monitoring the dynamic changes of ctDNA is beneficial for the evaluation of therapeutic efficacy and prediction of early recurrence in esophageal cancer. It is necessary to establish standards for individualized ctDNA detection in the evaluation of treatment response and surveillance of esophageal cancer and to develop clinical practice guideline for the systemic treatment of patients with "ctDNA recurrence." This review aims to provide an update on the role of ctDNA in the diagnosis and monitoring of esophageal cancer.
Collapse
Affiliation(s)
- Min Jiang
- Department of Pathology, Taizhou People’s Hospital, Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, Jiangsu, PR China
| | - Huilin Zhou
- Department of Pathology, Taizhou People’s Hospital, Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, Jiangsu, PR China
| | - Su Jiang
- Department of Rehabilitation, Taizhou People’s Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, Jiangsu, PR China
| | - Hong Yu
- Department of Pathology, Taizhou People’s Hospital, Affiliated to Nanjing University of Traditional Chinese Medicine, Taizhou, Jiangsu, PR China
| |
Collapse
|
21
|
Yang W, Zou J, Li Y, Liu R, Yan Z, Chen S, Zhao X, Guo W, Huang M, Li W, Zhu X, Chen Z. Longitudinal Circulating Tumor DNA Profiling in Metastatic Colorectal Cancer During Anti-EGFR Therapy. Front Oncol 2022; 12:830816. [PMID: 35280779 PMCID: PMC8908369 DOI: 10.3389/fonc.2022.830816] [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: 12/07/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundMetastatic colorectal cancer (mCRC) is a heterogenous disease with limited precision medicine and targeted therapy options. Monoclonal antibodies against epidermal growth factor receptor (EGFR) have been a crucial treatment option for mCRC. However, proper biomarkers for predicting therapeutic response remain unknown. As a non-invasive test, circulating tumor DNA (ctDNA) is appropriately positioned to reveal tumor heterogeneity and evolution, as it can be used in real-time genomic profiling. To evaluate the significance of ctDNA in monitoring the dynamic therapeutic response and prognosis of mCRC, we detected the baseline and dynamic changes of ctDNA in mCRC patients receiving anti-EGFR therapies.MethodsA single-center study was conducted retrospectively. Plasma samples from mCRC patients who received anti-EGFR therapies were collected at baseline and continuous treatment points. The ctDNA was extracted and sequenced with a target panel of tumor-related genes via next-generation sequencing (NGS). Clinical information was also collected and analyzed.ResultsWe conducted dynamic sampling of 22 mCRC patients, analyzed 130 plasma samples, obtained a baseline genomic mutation profile of the patients. In total, 54 variations were detected in 22 plasma samples, with a positive rate of 77.3% (17/22). TP53 was the most mutated gene (59.1%, 13/22), followed by APC (18.2%, 4/22). There was a high concordance rate of genomic characteristics between the tumor tissue test by polymerase chain reaction and ctDNA test by NGS. The mutation discrepancy increased with an extended course of treatment. During remission TP53 and APC were the most frequently decreased clonal mutations and KRAS, NRAS, ERBB2 and PIK3CA were the most decreased subclonal mutations. Both mutation types were increased during progression. The ctDNA decreased earlier than did the responses of computed tomography and traditional tumor markers (carbohydrate antigen 19-9 and carcinoembryonic antigen [CEA]). Lactate dehydrogenase level (P = 0.041), CEA level (P = 0.038), and primary lesion site (P = 0.038) were independent risk factors that influenced overall survival. Moreover, patients with RAS mutations tended to have a worse prognosis (P = 0.072).ConclusionsThis study demonstrates that ctDNA is a promising biomarker for monitoring the dynamic response to treatment and determining the prognosis of mCRC.
Collapse
Affiliation(s)
- Wentao Yang
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianling Zou
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ye Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Rujiao Liu
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengqing Yan
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Shiqing Chen
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Xiaoying Zhao
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weijian Guo
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mingzhu Huang
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenhua Li
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaodong Zhu
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyu Chen
- Department of Gastrointestinal Medical Oncology Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Zhiyu Chen,
| |
Collapse
|
22
|
Manca P, Corallo S, Lonardi S, Fucà G, Busico A, Leone AG, Corti F, Antoniotti C, Procaccio L, Smiroldo V, Ratti M, Murialdo R, Racca P, Pagani F, Randon G, Martinetti A, Sottotetti E, Prisciandaro M, Ambrosini M, Raimondi A, Morano F, Pietrantonio F. Variant allele frequency in baseline circulating tumour DNA to measure tumour burden and to stratify outcomes in patients with RAS wild-type metastatic colorectal cancer: a translational objective of the Valentino study. Br J Cancer 2022; 126:449-455. [PMID: 34811502 PMCID: PMC8810873 DOI: 10.1038/s41416-021-01591-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/21/2021] [Accepted: 10/06/2021] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION In patients with metastatic colorectal cancer (mCRC), baseline circulating tumour DNA (ctDNA) variant allele fraction (VAF) might serve as a surrogate of disease burden and should be evaluated in comparison with CEA and RECIST-defined sum of target lesions. METHODS In this pre-planned analysis of the VALENTINO trial, we included patients with RAS wild-type mCRC receiving upfront FOLFOX/panitumumab with available baseline liquid biopsy. CtDNA was analysed by means of a 14-gene NGS panel. For each patient, the gene with the highest VAF in ctDNA was selected. RESULTS The final cohort included 135 patients. The median VAF was 12.6% (IQR: 2.0-45.2%). Higher VAF was observed in patients with liver metastases and with synchronous metastases presentation. Patients with high VAF had poorer median OS compared to those with low VAF (21.8 vs 36.5 months; HR: 1.82, 95%CI: 1.20-2.76; p = 0.005). VAF outperformed baseline CEA and target lesion diameter in the prognostic stratification and remained significantly correlated with OS (p = 0.003) in a multivariate model. VAF was not significantly correlated with dimensional response and PFS. CONCLUSION CtDNA measured by VAF is prognostic in patients with RAS wild-type mCRC. Response and PFS after an anti-EGFR-based first-line strategy are independent from initial tumour burden.
Collapse
Affiliation(s)
- Paolo Manca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Corallo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology - IRCCS, Padova, Italy
- Oncology Unit 3, Department of Oncology, Veneto Institute of Oncology - IRCCS, Padova, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Adele Busico
- Pathology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alberto Giovanni Leone
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesca Corti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Carlotta Antoniotti
- Unit of Medical Oncology, Azienda Ospedaliero-Universitaria (AOU) Pisana, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Letizia Procaccio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology - IRCCS, Padova, Italy
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, Padova, Italy
| | - Valeria Smiroldo
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center, Rozzano, Italy
| | - Margherita Ratti
- Medical Oncology Unit, Azienda Socio-Sanitaria Territoriale (ASST) Ospedale di Cremona, Cremona, Italy
| | - Roberto Murialdo
- Department of Internal Medicine, University of Genoa and IRCCS AOU San Martino-IST, Genoa, Italy
| | - Patrizia Racca
- ColoRectal Cancer Unit - Department of oncology, AOU Città della Salute e della Scienza, Torino, Italy
| | - Filippo Pagani
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Randon
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Prisciandaro
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Margherita Ambrosini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandra Raimondi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Morano
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
23
|
Sanz-Garcia E, Zhao E, Bratman SV, Siu LL. Monitoring and adapting cancer treatment using circulating tumor DNA kinetics: Current research, opportunities, and challenges. SCIENCE ADVANCES 2022; 8:eabi8618. [PMID: 35080978 PMCID: PMC8791609 DOI: 10.1126/sciadv.abi8618] [Citation(s) in RCA: 88] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Circulating tumor DNA (ctDNA) has emerged as a biomarker with wide-ranging applications in cancer management. While its role in guiding precision medicine in certain tumors via noninvasive detection of susceptibility and resistance alterations is now well established, recent evidence has pointed to more generalizable use in treatment monitoring. Quantitative changes in ctDNA levels over time (i.e., ctDNA kinetics) have shown potential as an early indicator of therapeutic efficacy and could enable treatment adaptation. However, ctDNA kinetics are complex and heterogeneous, affected by tumor biology, host physiology, and treatment factors. This review outlines the current preclinical and clinical knowledge of ctDNA kinetics in cancer and how early on-treatment changes in ctDNA levels could be applied in clinical research to collect evidence to support implementation in daily practice.
Collapse
Affiliation(s)
- Enrique Sanz-Garcia
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Eric Zhao
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Scott V. Bratman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Lillian L. Siu
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Corresponding author.
| |
Collapse
|
24
|
Kastrisiou M, Zarkavelis G, Kougioumtzi A, Sakaloglou P, Kostoulas C, Georgiou I, Batistatou A, Pentheroudakis G, Magklara A. Development and Validation of a Targeted ‘Liquid’ NGS Panel for Treatment Customization in Patients with Metastatic Colorectal Cancer. Diagnostics (Basel) 2021; 11:diagnostics11122375. [PMID: 34943612 PMCID: PMC8700616 DOI: 10.3390/diagnostics11122375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/12/2021] [Accepted: 12/13/2021] [Indexed: 01/22/2023] Open
Abstract
The detection of actionable mutations in tumor tissue is a prerequisite for treatment customization in patients with metastatic colorectal cancer (mCRC). Analysis of circulating tumor DNA (ctDNA) for the identification of such mutations in patients’ plasma is an attractive alternative to invasive tissue biopsies. Despite having the high analytical sensitivity required for ctDNA analysis, digital polymerase chain reaction (dPCR) technologies can only detect a very limited number of hotspot mutations, whilst a broader mutation panel is currently needed for clinical decision making. Recent advances in next-generation sequencing (NGS) have led to high-sensitivity platforms that allow screening of multiple genes at a single assay. Our goal was to develop a small, cost- and time-effective NGS gene panel that could be easily integrated in the day-to-day clinical routine in the management of patients with mCRC. We designed a targeted panel comprising hotspots in six clinically relevant genes (KRAS, NRAS, MET, BRAF, ERBB2 and EGFR) and validated it in a total of 68 samples from 30 patients at diagnosis, first and second disease progression. Results from our NGS panel were compared against plasma testing with BEAMing dPCR regarding the RAS gene status. The overall percent of agreement was 83.6%, with a positive and negative percent agreement of 74.3% and 96.2%, respectively. Further comparison of plasma NGS with standard tissue testing used in the clinic showed an overall percent agreement of 86.7% for RAS status, with a positive and negative percent agreement of 81.2% and 92.8%, respectively. Thus, our study strongly supports the validity and efficiency of an affordable targeted NGS panel for the detection of clinically relevant mutations in patients with mCRC.
Collapse
Affiliation(s)
- Myrto Kastrisiou
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (M.K.); (A.K.)
- Department of Medical Oncology, University General Hospital of Ioannina, 45500 Ioannina, Greece;
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45444 Ioannina, Greece
| | - George Zarkavelis
- Department of Medical Oncology, University General Hospital of Ioannina, 45500 Ioannina, Greece;
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45444 Ioannina, Greece
| | - Anastasia Kougioumtzi
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (M.K.); (A.K.)
| | - Prodromos Sakaloglou
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (P.S.); (C.K.); (I.G.)
| | - Charilaos Kostoulas
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (P.S.); (C.K.); (I.G.)
| | - Ioannis Georgiou
- Laboratory of Medical Genetics in Clinical Practice, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (P.S.); (C.K.); (I.G.)
| | - Anna Batistatou
- Department of Pathology, Faculty of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - George Pentheroudakis
- Department of Medical Oncology, University General Hospital of Ioannina, 45500 Ioannina, Greece;
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45444 Ioannina, Greece
- Correspondence: (G.P.); (A.M.)
| | - Angeliki Magklara
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, 45110 Ioannina, Greece; (M.K.); (A.K.)
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, 45110 Ioannina, Greece
- Institute of Biosciences, University Research Center of Ioannina (URCI), 45110 Ioannina, Greece
- Correspondence: (G.P.); (A.M.)
| |
Collapse
|
25
|
Benavides M, Gómez-España A, García-Alfonso P, González CG, Viéitez JM, Rivera F, Safont MJ, Abad A, Sastre J, Valladares-Ayerbes M, Carrato A, González-Flores E, Robles L, Salud A, Alonso-Orduña V, Montagut C, Asensio E, Díaz-Rubio E, Aranda E. Upfront primary tumour resection and survival in synchronous metastatic colorectal cancer according to primary tumour location and RAS status: Pooled analysis of the Spanish Cooperative Group for the Treatment of Digestive Tumours (TTD). Eur J Surg Oncol 2021; 48:1123-1132. [PMID: 34872775 DOI: 10.1016/j.ejso.2021.11.122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 09/14/2021] [Accepted: 11/17/2021] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Retrospective studies and meta-analyses suggest that upfront primary tumour resection (UPTR) confers a survival benefit in patients with asymptomatic unresectable metastatic colorectal cancer (mCRC) undergoing chemotherapy, however a consensus of its role in routine clinical practice in the current era of targeted therapies is lacking. This retrospective study aimed to analyse the survival benefit of UPTR in terms of tumour location and mutational status, in patients with synchronous mCRC receiving chemotherapy and targeted therapy. PATIENTS AND METHODS Survival was analysed in a pooled cohort of synchronous mCRC patients treated with a first-line anti-VEGF or anti-EGFR inhibitor in seven trials of the Spanish TTD group, according to UPTR, tumour-sidedness and mutational profiling. RESULTS Of 1334 eligible patients, 642 (48%) had undergone UPTR. UPTR was associated with significantly longer overall survival (OS; 25.0 vs 20.3 months; HR 1.30, 95%CI 1.15-1.48; p < 0.0001). UPTR was associated with significant OS benefit in both left-sided (HR 1.38, 95%CI 1.13-1.69; p = 0.002) and right-sided (HR 1.39, 95%CI 1.00-1.94; p = 0.049) tumours, RASwt (HR 1.29, 95%CI 1.05-1.60; p = 0.016) and BRAFwt (HR 1.49, 95%CI 1.21-1.84; p = 0.0002) tumours, and treatment with anti-EGFRs (HR 1.47, 95%CI 1.13-1.92; p = 0.004) and anti-VEGFs (HR 1.25, 95%CI 1.08-1.44; p = 0.003). Multivariate analysis identified number of metastatic sites, RAS status, primary tumour location and UPTR as independent prognostic factors for OS. CONCLUSION Considering the selection bias inherent to this study, our results support UPTR before first-line anti-EGFR or anti-VEGF targeted therapy in right and left-sided asymptomatic unresectable synchronous mCRC patients. RAS/BRAF mutational status may also influence UPTR function.
Collapse
Affiliation(s)
- Manuel Benavides
- UGC Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain.
| | - Auxiliadora Gómez-España
- Department of Medical Oncology, IMIBIC, Universidad de Córdoba, CIBERONC, Instituto de Salud Carlos III, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Pilar García-Alfonso
- Department of Medical Oncology, Hospital Universitario Gregorio Marañón, Madrid, Spain
| | - Cristina García González
- UGC Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, IBIMA, Málaga, Spain
| | - Jose María Viéitez
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Fernando Rivera
- Department of Medical Oncology, University Hospital Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - María José Safont
- Department of Medical Oncology, Hospital General Universitario Valencia, Universidad de Valencia, CIBERONC, Valencia, Spain
| | - Albert Abad
- Department of Medical Oncology, Instituto Oncológico Dr. Rosell, Barcelona, Spain
| | - Javier Sastre
- Department of Medical Oncology, Hospital Clínico San Carlos, Instituto de Investigación Hospital Clínico San Carlos (IdISSC), University Complutense, CIBERONC, Madrid, Spain
| | | | - Alfredo Carrato
- Department of Medical Oncology, IRYCIS, CIBERONC, Alcalá University, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | | | - Luis Robles
- Department of Medical Oncology, Hospital 12 de Octubre, Madrid, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Hospital de Lleida Arnau de Vilanova, Lérida, Spain
| | - Vicente Alonso-Orduña
- Department of Medical Oncology, Hospital Universitario Miguel Servet, Instituto de Investigación Sanitaria de Aragón (IISA), Zaragoza, Spain
| | - Clara Montagut
- Department of Medical Oncology, Hospital del Mar Medical Research Institute, CIBERONC, Barcelona, Spain
| | - Elena Asensio
- Department of Medical Oncology, Hospital General Universitario de Elche, Alicante, Spain
| | - Eduardo Díaz-Rubio
- Department of Medical Oncology, Hospital Clínico San Carlos, Instituto de Investigación Hospital Clínico San Carlos (IdISSC), University Complutense, CIBERONC, Madrid, Spain
| | - Enrique Aranda
- Department of Medical Oncology, IMIBIC, Universidad de Córdoba, CIBERONC, Instituto de Salud Carlos III, Hospital Universitario Reina Sofía, Córdoba, Spain
| | | |
Collapse
|
26
|
Procaccio L, Bergamo F, Daniel F, Rasola C, Munari G, Biason P, Crucitta S, Barsotti G, Zanella G, Angerilli V, Magro C, Paccagnella S, Di Antonio V, Loupakis F, Danesi R, Zagonel V, Del Re M, Lonardi S, Fassan M. A Real-World Application of Liquid Biopsy in Metastatic Colorectal Cancer: The Poseidon Study. Cancers (Basel) 2021; 13:cancers13205128. [PMID: 34680277 PMCID: PMC8533756 DOI: 10.3390/cancers13205128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND First-line decision making is the key to the successful care of mCRC patients and RAS/BRAF status is crucial to select the best targeted agent. In hub centers, a relevant proportion of patients referred from small volume centers may not have standard tissue-based (STB) molecular results available at the time of the first visit (T0). Liquid biopsy (LB) may help circumvent these hurdles. METHODS A monoinstitutional prospective head-to-head comparison of LB versus (vs.) STB testing was performed in a real-world setting. Selection criteria included: mCRC diagnosis with unknown RAS/BRAF status at T0, tumoral tissue archived in external centers, no previous treatment with anti-EGFR. At T0, patients underwent plasma sampling for LB testing and procedure for tissue recovery. RAS/BRAF genotyping was carried out by droplet digital PCR on circulating-tumoral (ct) DNA. The primary endpoint was the comparison of time to LB (T1) vs. STB (T2) results using the Mann-Whitney U test. Secondary endpoints were the concordance between LB and STB defined as overall percent agreement and the accuracy of LB in terms of specificity, sensitivity, positive and negative predictive value. We also performed an exploratory analysis on urinary (u) ctDNA. RESULTS A total of 33 mCRC patients were included. Mean T1 and T2 was 7 and 22 days (d), respectively (p < 0.00001). T2 included a mean time for archival tissue recovery of 17 d. The overall percent agreement between LB and STB analysis was 83%. Compared to STB testing, LB specificity and sensitivity were 90% and 80%, respectively, with a positive predictive value of 94% and negative one of 69%. In detail, at STB and LB testing, RAS mutation was found in 45% and 42% of patients, respectively; BRAF mutation in 15%. LB results included one false positive and four false negative. False negative cases showed a significantly lower tumor burden at basal CT scan. Concordance between STB and uctDNA testing was 89%. CONCLUSIONS Faster turnaround time, high concordance and accuracy are three key points supporting the adoption of LB in routinary mCRC care, in particular when decision on first-line therapy is urgent and tissue recovery from external centers may require a long time. Results should be interpreted with caution in LB wild-type cases with low tumor burden.
Collapse
Affiliation(s)
- Letizia Procaccio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Francesca Bergamo
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Francesca Daniel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Cosimo Rasola
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Giada Munari
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padova, Italy
| | - Paola Biason
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Stefania Crucitta
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Giulia Barsotti
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Giulia Zanella
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
| | - Cristina Magro
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Silvia Paccagnella
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
| | - Veronica Di Antonio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Fotios Loupakis
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Vittorina Zagonel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Sara Lonardi
- Oncology Unit 3, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-0498215953
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padova, Italy
| |
Collapse
|
27
|
Clinical Applications of Minimal Residual Disease Assessments by Tumor-Informed and Tumor-Uninformed Circulating Tumor DNA in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184547. [PMID: 34572774 PMCID: PMC8471730 DOI: 10.3390/cancers13184547] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Circulating tumor DNA, or ctDNA, are fragments of tumor DNA that can be detected in the blood of patients with colorectal cancer. Measuring ctDNA levels in the blood has shown the potential to provide important information that can be helpful in the clinical care of patients with colorectal cancer. For example, in patients with colon cancer that has been removed by surgery, measuring ctDNA in the blood can predict the likelihood of cancer recurrence, while in those with metastatic colorectal cancer, measuring ctDNA can inform the clinician whether chemotherapy is effective at earlier timepoints than currently available tests. In this review, we discuss the results from ongoing studies describing the utility of ctDNA measurements across all stages of colorectal cancer. We also discuss the various clinical scenarios that ctDNA may have the most immediate impact in colorectal cancer management. Abstract Emerging data suggest that circulating tumor DNA (ctDNA) can detect colorectal cancer (CRC)-specific signals across both non-metastatic and metastatic settings. With the development of multiple platforms, including tumor-informed and tumor-agnostic ctDNA assays and demonstration of their provocative analytic performance to detect minimal residual disease, there are now ongoing, phase III randomized clinical trials to evaluate their role in the management paradigm of CRC. In this review, we highlight landmark studies that have formed the basis for ongoing studies on the clinically applicability of plasma ctDNA assays in resected, stage I–III CRC and metastatic CRC. We discuss clinical settings by which ctDNA may have the most immediate impact in routine clinical practice. These include the potential for ctDNA to (1) guide surveillance and intensification or de-intensification strategies of adjuvant therapy in resected, stage I–III CRC, (2) predict treatment response to neoadjuvant therapy in locally advanced rectal cancer inclusive of total neoadjuvant therapy (TNT), and (3) predict response to systemic and surgical therapies in metastatic disease. We end by considering clinical variables that can influence our ability to reliably interpret ctDNA dynamics in the clinic.
Collapse
|
28
|
Garrido P, Paz-Ares L, Majem M, Morán T, Trigo JM, Bosch-Barrera J, Garcίa-Campelo R, González-Larriba JL, Sánchez-Torres JM, Isla D, Viñolas N, Camps C, Insa A, Juan Ó, Massuti B, Paredes A, Artal Á, López-Brea M, Palacios J, Felip E. LungBEAM: A prospective multicenter study to monitor stage IV NSCLC patients with EGFR mutations using BEAMing technology. Cancer Med 2021; 10:5878-5888. [PMID: 34296539 PMCID: PMC8419773 DOI: 10.1002/cam4.4135] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVES The aim of LungBEAM was to determine the value of a novel epidermal growth factor receptor (EGFR) mutation test in blood based on BEAMing technology to predict disease progression in advanced non-small cell lung cancer (NSCLC) patients treated with first- or second-generation EGFR-tyrosine kinase inhibitors (EGFR-TKIs). Another goal was to monitor the dynamics of EGFR mutations, as well as to track EGFR exon 20 p.T790M (p.T790M) resistance during treatment, as critical indicators of therapeutic efficacy and patient survival. METHODS Stage IV NSCLC patients with locally confirmed EGFR-TKI sensitizing mutations (ex19del and/or L858R) in biopsy tissue who were candidates to receive first- or second-generation EGFR-TKI as first-line therapy were included. Plasma samples were obtained at baseline and every 4 weeks during treatment until a progression-free survival (PFS) event or until study completion (72-week follow-up). The mutant allele fraction (MAF) was determined for each identified mutation using BEAMing. RESULTS A total of 68 of the 110 (61.8%) patients experienced a PFS event. Twenty-six patients (23.6%) presented with an emergent p.T790M mutation in plasma at some point during follow-up, preceding radiologic progression with a median of 76 (interquartile ratio: 54-111) days. Disease progression correlated with the appearance of p.T790M in plasma with a hazard ratio (HR) of 1.94 (95% confidence interval [CI], 1.48-2.54; p < 0.001). The HR for progression in patients showing increasing plasma sensitizing mutation levels (positive MAF slope) versus patients showing either decreasing or unchanged plasma mutation levels (negative or null MAF slopes) was 3.85 (95% CI, 2.01-7.36; p < 0.001). CONCLUSION Detection and quantification of EGFR mutations in circulating tumor DNA using the highly sensitive BEAMing method should greatly assist in optimizing treatment decisions for advanced NSCLC patients.
Collapse
Affiliation(s)
- Pilar Garrido
- Medical Oncology Department, IRYCIS Hospital Universitario Ramón y Cajal, Universidad Alcalá, Madrid, Spain.,CIBERONC, Madrid, Spain
| | - Luis Paz-Ares
- CIBERONC, Madrid, Spain.,Medical Oncology Department, Hospital Universitario 12 de Octubre and i+12 Research Institute, Madrid, Spain.,Lung Cancer Group, Clinical Research Program, Spanish National Cancer Research Center (CNIO), Madrid, Spain.,Complutense University, Madrid, Spain
| | - Margarita Majem
- Medical Oncology Department, Hospital De La Santa Creu I Sant Pau, Barcelona, Spain.,Spanish Lung Cancer Group (GECP), Barcelona, Spain
| | - Teresa Morán
- Spanish Lung Cancer Group (GECP), Barcelona, Spain.,ICO Badalona, Hospital Germans Trias i Pujol, Barcelona, Spain
| | - José Manuel Trigo
- Medical Oncology Department, Hospital Universitario Virgen de la Victoria, Málaga, Spain
| | - Joaquim Bosch-Barrera
- Medical Oncology, Catalan Institute of Oncology (ICO), Dr. Josep Trueta Hospital of Girona, Girona, Spain
| | | | | | | | - Dolores Isla
- Medical Oncology Department, Hospital Universitario Lozano Blesa, Zaragoza, Spain
| | - Núria Viñolas
- Medical Oncology Department, Hospital Clinic i Provincial, Barcelona, Spain
| | - Carlos Camps
- CIBERONC, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario de Valencia, Universidad de Valencia, Valencia, Spain
| | - Amelia Insa
- Medical Oncology Department, Hospital Clínico Universitario, Valencia, Spain
| | - Óscar Juan
- Medical Oncology Department, Hospital Universitario La Fe, Valencia, Spain
| | - Bartomeu Massuti
- Medical Oncology Department, Hospital General Universitario de Alicante, Alicante, Spain
| | - Alfredo Paredes
- Medical Oncology Department, Hospital Universitario Donostia, Donostia-San Sebastián, Spain
| | - Ángel Artal
- Medical Oncology, Hospital Universitario Miguel Servet, Zaragoza, Spain
| | - Marta López-Brea
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - José Palacios
- CIBERONC, Madrid, Spain.,Pathology Department, IRYCIS Hospital Universitario Ramón y Cajal, Universidad Alcalá, Madrid, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Hospital Universitario Vall d'Hebron, Barcelona, Spain
| |
Collapse
|
29
|
Zhang Y, Zhao Y, Li Q, Wang Y. Macrophages, as a Promising Strategy to Targeted Treatment for Colorectal Cancer Metastasis in Tumor Immune Microenvironment. Front Immunol 2021; 12:685978. [PMID: 34326840 PMCID: PMC8313969 DOI: 10.3389/fimmu.2021.685978] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/24/2021] [Indexed: 12/16/2022] Open
Abstract
The tumor immune microenvironment plays a vital role in the metastasis of colorectal cancer. As one of the most important immune cells, macrophages act as phagocytes, patrol the surroundings of tissues, and remove invading pathogens and cell debris to maintain tissue homeostasis. Significantly, macrophages have a characteristic of high plasticity and can be classified into different subtypes according to the different functions, which can undergo reciprocal phenotypic switching induced by different types of molecules and signaling pathways. Macrophages regulate the development and metastatic potential of colorectal cancer by changing the tumor immune microenvironment. In tumor tissues, the tumor-associated macrophages usually play a tumor-promoting role in the tumor immune microenvironment, and they are also associated with poor prognosis. This paper reviews the mechanisms and stimulating factors of macrophages in the process of colorectal cancer metastasis and intends to indicate that targeting macrophages may be a promising strategy in colorectal cancer treatment.
Collapse
Affiliation(s)
- Yingru Zhang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiyang Zhao
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
30
|
Personalized Therapy and Liquid Biopsy-A Focus on Colorectal Cancer. J Pers Med 2021; 11:jpm11070630. [PMID: 34357097 PMCID: PMC8305103 DOI: 10.3390/jpm11070630] [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: 06/03/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
(1) Background: Resistance mechanisms represent a barrier to anti-cancer therapies. Liquid biopsies would allow obtaining additional information in order to develop targeted therapies to thwart the resistance phenomena but also to follow in time real response to treatment and be able to adapt it the most quickly possible way in case of resistance. (2) Methods: herein we summarize the different liquid biopsies which are currently under research; we then review the literature and focalize on one of their potential roles: the theranostic one and especially in the cases of colorectal cancers. (3) Results: few studies targeting liquid biopsy as a potential tool to adapt cancer treatments are present in the literature and encompass few patients. (4) Conclusions: further research is needed to prove the efficiency of LB. Indeed, it seems a promising tool to guide treatment by targeting actionable mutations with detection of resistant mutations.
Collapse
|
31
|
Petrillo A, Salati M, Trapani D, Ghidini M. Circulating Tumor DNA as a Biomarker for Outcomes Prediction in Colorectal Cancer Patients. Curr Drug Targets 2021; 22:1010-1020. [PMID: 33155906 DOI: 10.2174/1389450121999201103194248] [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: 05/23/2020] [Revised: 08/07/2020] [Accepted: 08/23/2020] [Indexed: 12/24/2022]
Abstract
Circulating tumour DNA (ctDNA) is a novel tool that has been investigated in several types of tumours, including colorectal cancer (CRC). In fact, the techniques based on liquid biopsies are proposed as appealing non-invasive alternatives to tissue biopsy, adding more insights into tumour molecular profile, heterogeneity and for cancer detection and monitoring. Additionally, some analysis showed that in CRC patients, ctDNA seems to act as a biomarker able to predict the outcome (prognostic role) and the response to treatments (predictive role). In particular, in the early stage CRC (stage I-III), it could represent a time marker of adjuvant therapy as well as a marker of minimal residual disease and recurrence risk in addition to the already recognized risk factors. In metastatic CRC, the analysis of molecular tumour profile by ctDNA has shown to have high concordance with the tissue biopsy at diagnosis. Additionally, some studies demonstrated that ctDNA level during the treatment was linked with the early response to treatment and prognosis. Finally, the quantitative analysis of ctDNA and copy number alterations may be useful in order to detect resistance to therapy at the time of progression of disease and to help in finding new therapeutic targets.
Collapse
Affiliation(s)
| | - Massimiliano Salati
- Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Dario Trapani
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
32
|
Hamfjord J, Guren TK, Glimelius B, Sorbye H, Pfeiffer P, Dajani O, Lingjaerde OC, Tveit KM, Pallisgaard N, Spindler KLG, Kure EH. Clinicopathological factors associated with tumour-specific mutation detection in plasma of patients with RAS-mutated or BRAF-mutated metastatic colorectal cancer. Int J Cancer 2021; 149:1385-1397. [PMID: 33961700 DOI: 10.1002/ijc.33672] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/09/2021] [Accepted: 04/20/2021] [Indexed: 12/09/2022]
Abstract
Detection of tumour-specific circulating cell-free DNA in plasma (ctDNA) fails in a significant number of cases depending on the clinical context. The primary aim was to investigate clinicopathological factors associated with detection of ctDNA in patients with RAS-/BRAF-mutated metastatic colorectal cancer (mCRC) prior to first-line therapy. A secondary aim was to evaluate the prognostic impact of ctDNA compared to other biomarkers. Patients were included from the NORDIC-VII study (N = 253). ctDNA was sampled prior to treatment and analysed for hotspot tissue mutations (KRAS, NRAS, and BRAF) using droplet digital PCR. Multivariable regression models were constructed to predict the probability of mutation detection and survival. Increasing radiological size of target lesions by increments of 1 cm (odds ratio [OR] = 1.18; 95% confidence interval [CI] 1.09-1.27; P < .001), intact primary tumour (OR = 3.17; 95% CI 1.22-8.22; P = .018) and more than one metastatic site (OR = 3.08; 95% CI 1.32-7.19; P = .009) were associated with mutation detection in plasma. Metastatic involvement of the lung was associated with non-detection (OR = 0.26; 95% CI 0.12-0.58; P = .001). Preanalytical and analytical factors modulated detection. High allele frequencies of ctDNA indicated poor prognosis independently of CEA and CA19-9 (hazard ratio [HR] = 2.38; 95% CI 1.74-3.26; P < .001; N = 206). Clinicopathological characteristics should be carefully considered when evaluating ctDNA results from mCRC patients, especially when confronted with a plasma negative result. ctDNA may prove to be a clinically useful marker in the evaluation of mCRC treatment.
Collapse
Affiliation(s)
- Julian Hamfjord
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Tormod Kyrre Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Bengt Glimelius
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark.,Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Olav Dajani
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Ole Christian Lingjaerde
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Department of Computer Science, University of Oslo, Oslo, Norway
| | - Kjell Magne Tveit
- Department of Oncology, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,K.G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Oslo, Norway
| | - Niels Pallisgaard
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Karen-Lise Garm Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Elin H Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.,Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Campus Bø, Norway
| |
Collapse
|
33
|
Provencio M, Serna-Blasco R, Franco F, Calvo V, Royuela A, Auglytė M, Sánchez-Hernández A, de Julián Campayo M, García-Girón C, Dómine M, Blasco A, Sánchez JM, Oramas J, Bosch-Barrera J, Sala MÁ, Sereno M, Ortega AL, Chara L, Hernández B, Padilla A, Coves J, Blanco R, Balsalobre J, Mielgo X, Bueno C, Jantus-Lewintre E, Molina-Vila MÁ, Romero A. Analysis of circulating tumour DNA to identify patients with epidermal growth factor receptor-positive non-small cell lung cancer who might benefit from sequential tyrosine kinase inhibitor treatment. Eur J Cancer 2021; 149:61-72. [PMID: 33831609 DOI: 10.1016/j.ejca.2021.02.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/05/2021] [Accepted: 02/24/2021] [Indexed: 01/09/2023]
Abstract
BACKGROUND Survival data support the use of first-line osimertinib as the standard of care for epidermal growth factor receptor (EGFR)-positive non-small cell lung cancer (NSCLC). However, it remains unclear whether upfront osimertinib is superior to sequential first- or second-generation tyrosine kinase inhibitors (TKIs) followed by osimertinib for all patients. It is impossible to predict which patients are at high risk of progression, and this constitutes a major limitation of the sequential TKI approach. PATIENTS AND METHODS A total of 830 plasma samples from 228 patients with stage IV, EGFR-positive NSCLC who were treated with first-line TKIs were analysed by digital polymerase chain reaction (dPCR). RESULTS The circulating tumour DNA (ctDNA) levels helped to identify patients with significantly improved survival rate, regardless of the treatment. Patients treated with first- or second-generation TKIs (N = 189) with EGFR mutations in plasma at a mutant allele frequency (MAF) <7% before treatment initiation (low-risk patients) or who were ctDNA negative after 3 or 6 months of treatment and with an MAF <7% at diagnosis (high responders) had two-thirds lower risk of death than patients in the opposite situation (adjusted hazard ratio [HR] = 0.38; 95% confidence interval [CI]: 0.23-0.64 and HR = 0.22; 95% CI: 0.12-0.42, respectively). The median overall survival (OS) for low-risk patients and high responders treated with first- or second-generation TKIs was 34.2 months and not reached, respectively, regardless of second-line treatment. There were no significant difference in OS between low-risk or high-responder patients treated upfront with osimertinib (N = 39) and those treated under a sequential approach with osimertinib (N = 60). Median OS was not reached in both cases. CONCLUSIONS Pre-treatment ctDNA levels identify low-risk patients, who may benefit from sequential TKI treatment. Information regarding EGFR mutation clearance can help to improve patient selection.
Collapse
Affiliation(s)
- Mariano Provencio
- Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain.
| | - Roberto Serna-Blasco
- Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Fabio Franco
- Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Virgina Calvo
- Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Ana Royuela
- Biostatistics Unit, Hospital Universitario Puerta de Hierro- Majadahonda, CIBERESP, Majadahonda, Madrid, Spain
| | - Milda Auglytė
- Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | | | - María de Julián Campayo
- Medical Oncology, Hospital Provincial Centre de Castelló, Castellón de La Plana, Castellón, Spain
| | | | - Manuel Dómine
- Medical Oncology, Fundación Jiménez Díaz, Madrid, Madrid, Spain
| | - Ana Blasco
- Medical Oncology, Hospital General Universitario Valencia, Valencia, Valencia, Spain
| | - José M Sánchez
- Medical Oncology, Hospital de La Princesa, Madrid, Madrid, Spain
| | - Juana Oramas
- Medical Oncology, Hospital Universitario de Canarias, La Laguna, Santa Cruz de Tenerife, Spain
| | | | - María Á Sala
- Medical Oncology, Hospital Basurto, Bilbao, Vizcaya, Spain
| | - María Sereno
- Medical Oncology, Hospital Universitario Infanta Sofía, San Sebastián de Los Reyes, Madrid, Spain
| | - Ana L Ortega
- Medical Oncology, Consorcio Hospitalario de Jaén, Jaén, Jaén, Spain
| | - Luis Chara
- Medical Oncology, Hospital Universitario de Guadalajara, Guadalajara, Guadalajara, Spain
| | - Berta Hernández
- Medical Oncology, Complejo Hospitalario de Navarra, Pamplona, Navarra, Spain
| | - Airam Padilla
- Medical Oncology, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Santa Cruz de Tenerife, Spain
| | - Juan Coves
- Medical Oncology, Hospital Son Llàtzer, Palma, Islas Baleares, Spain
| | - Remedios Blanco
- Medical Oncology, Consorci Sanitari Terrassa, Terrassa, Barcelona, Spain
| | - José Balsalobre
- Medical Oncology, Hospital General Universitario Santa Lucia, Cartagena, Murcia, Spain
| | - Xabier Mielgo
- Medical Oncology, Hospital Universitario Fundación Alcorcón, Alcorcón, Madrid, Spain
| | - Coralia Bueno
- Medical Oncology, Hospital Universitario Infanta Cristina, Parla, Madrid, Spain
| | - Eloisa Jantus-Lewintre
- Mixed Unit TRIAL, Príncipe Felipe Research Center & General University Hospital of Valencia Research Foundation, Valencia, Spain
| | - Miguel Á Molina-Vila
- Laboratory of Oncology/Pangaea Oncology, Quirón-Dexeus University Hospital, Barcelona, Spain
| | - Atocha Romero
- Medical Oncology, Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain.
| |
Collapse
|
34
|
Kagawa Y, Elez E, García-Foncillas J, Bando H, Taniguchi H, Vivancos A, Akagi K, García A, Denda T, Ros J, Nishina T, Baraibar I, Komatsu Y, Ciardiello D, Oki E, Kudo T, Kato T, Yamanaka T, Tabernero J, Yoshino T. Combined Analysis of Concordance between Liquid and Tumor Tissue Biopsies for RAS Mutations in Colorectal Cancer with a Single Metastasis Site: The METABEAM Study. Clin Cancer Res 2021; 27:2515-2522. [PMID: 33602686 DOI: 10.1158/1078-0432.ccr-20-3677] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 12/04/2020] [Accepted: 02/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE OncoBEAM™ is a circulating tumor DNA (ctDNA) test that uses the BEAMing digital PCR technology. We clarified the association between the baseline tumor burden and discordance in the RAS status by metastatic sites in patients with a single metastatic site. EXPERIMENTAL DESIGN Data from previous Spanish and Japanese studies investigating the concordance of the RAS status between OncoBEAM™ and tissue biopsy in 221 patients with metastatic colorectal cancer (mCRC) were used. We collected data from patients with liver, peritoneal, or lung metastases and evaluated the concordance rates according to the metastatic site and the association between the concordance rate and tumor burden. RESULTS Patients had metastases in the liver (n = 151), peritoneum (n = 25), or lung (n = 45) with concordance rates of 91% (95% confidence interval, 85%-95%), 88% (68%-97%), and 64% (49%-78%), respectively. Factors associated with concordance included the baseline longest diameter and lesion number (P = 0.004), and sample collection interval (P = 0.036). Concordance rates ≥90% were observed in the following groups: liver metastases alone, regardless of the baseline longest diameter and lesion number; peritoneal metastases alone in patients with a baseline longest diameter ≥20 mm; and lung metastases alone in patients with a baseline longest diameter ≥20 mm and/or number of lesions ≥10. CONCLUSIONS Plasma ctDNA-based liquid biopsy in patients with mCRC may be useful depending on the metastatic site. The maximum diameter and lesion number should be carefully considered when determining patients' RAS status with only peritoneal or lung metastases.
Collapse
Affiliation(s)
- Yoshinori Kagawa
- Department of Surgery, Kansai Rosai Hospital, Amagasaki, Hyogo, Japan
| | - Elena Elez
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Jesús García-Foncillas
- Department of Oncology, Fundacion Jimenez Diaz University Hospital, Autonomous University, Madrid, Spain
| | - Hideaki Bando
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Hiroya Taniguchi
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Kiwamu Akagi
- Division of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Ina, Saitama, Japan
| | - Ariadna García
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chuo, Chiba, Japan
| | - Javier Ros
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Iosune Baraibar
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo, Hokkaido, Japan
| | - Davide Ciardiello
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain.,Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Toshihiro Kudo
- Department of Frontier Science for Cancer and Chemotherapy, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Takeshi Kato
- Department of Surgery, National Hospital Organization Osaka National Hospital, Chuo, Osaka, Japan
| | - Takeharu Yamanaka
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Kanagawa, Japan
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron University Hospital (HUVH), Vall d'Hebron Institute of Oncology (VHIO), IOB-Quiron, UVic-UCC, Barcelona, Spain
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan.
| |
Collapse
|
35
|
Jones RP, Pugh SA, Graham J, Primrose JN, Barriuso J. Circulating tumour DNA as a biomarker in resectable and irresectable stage IV colorectal cancer; a systematic review and meta-analysis. Eur J Cancer 2021; 144:368-381. [PMID: 33422803 DOI: 10.1016/j.ejca.2020.11.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/01/2020] [Accepted: 11/15/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND For patients with metastatic colorectal cancer, stratification for treatment (surgery or chemotherapy) is often based on crude clinicopathological characteristics like tumour size and number of lesions. Circulating tumour DNA (ctDNA) acts as a potential biomarker of disease trajectory and biology, allowing better stratification. This study aims to systematically review ctDNA in stage IV colorectal cancer to assess its potential role as a prospective biomarker to guide management decisions. METHODS A literature search was performed to identify studies where the measurement of ctDNA in stage IV colorectal cancer was correlated with a clinical outcome (radiological response, secondary resection rate, PFS, DFS or OS). RESULTS Twenty-eight studies were included, reporting on 2823 patients. Circulating tumour DNA was detectable in between 80% and 90% of patients prior to treatment. Meta-analysis identified a strong correlation between detectable ctDNA after treatment (surgery or chemotherapy) and overall survival (HR 2.2, 95% CI 1.79-2.69, p < 0.00001), as well as progression-free survival (HR 3.15, 95% CI 2.10-4.73, p < 0.00001). ctDNA consistently offered an early marker of long-term prognosis in irresectable disease, with changes after one cycle of systemic therapy demonstrating prognostic value. In resectable disease treated with curative intent, detection of ctDNA offered a lead time over radiological recurrence of 10 months. CONCLUSION Circulating tumour DNA is detectable in the majority of resectable and irresectable patients. The presence of ctDNA is clearly associated with shorter overall survival, with changes in ctDNA an early biomarker of adverse disease behaviour. Prospective trials are essential to test its clinical efficacy.
Collapse
Affiliation(s)
- Robert P Jones
- School of Cancer Studies, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Hepatobiliary Surgery, Liverpool University Teaching Hospitals NHS Foundation Trust, Liverpool, UK.
| | | | - Janet Graham
- Beatson West of Scotland Cancer Centre, Glasgow, UK; University of Glasgow, UK
| | | | - Jorge Barriuso
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
36
|
Clinical Utility of Plasma KRAS, NRAS and BRAF Mutational Analysis with Real Time PCR in Metastatic Colorectal Cancer Patients-The Importance of Tissue/Plasma Discordant Cases. J Clin Med 2020; 10:jcm10010087. [PMID: 33383664 PMCID: PMC7794782 DOI: 10.3390/jcm10010087] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/22/2020] [Accepted: 12/25/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Tumor tissue (T) mutational analysis represents the standard for metastatic colorectal cancer (mCRC); however, circulating tumor DNA (ctDNA) detected by liquid biopsy in plasma (PL) can better represent tumor heterogeneity. METHODS mCRC patients undergoing standard first-line chemotherapy with known T-KRAS/NRAS/BRAF status were enrolled in the present prospective study. PL mutations were assessed within 2 weeks before chemotherapy start with real time PCR and correlated with T status and Progression free survival (PFS). Clinical and biochemical variables including also total number of tumor lesions (TNL) and the sum of maximum diameter (SMD) of all lesions were assessed as potential predictors of T/PL discordance. RESULTS Among 45 enrolled patients, all BRAF mutations were concordant between T and PL and there were 20% of patients RAS discordant: 9% wild type in T and mutated in PL and 11% mutated in T and wild type in PL. T mutations were significantly associated to median PFS (mPFS of 4.5, 8.3 and 22.9 months for T-BRAF mutated, T-RAS mutated, and T-wild type patients, respectively, p for trend 0.00014). PL mutations further refined prognosis: RAS wild type in T and mutated in PL had significantly shorter PFS than concordant RAS wild type in T and PL: mPFS 9.6 vs. 23.3 months, respectively, p = 0.02. Patients RAS mutated in T and wild type in PL had longer PFS than concordant RAS mutated in T and PL: 24.4 vs. 7.8 months, respectively, p = 0.008. At a multivariate cox regression analysis for PFS, PL mutations were independent prognostic factor superior to T analysis (HR 0.13, p = 0.0008). At multivariate logistic regression analysis TNL and SMD were significant predictors of discordant cases. CONCLUSIONS PL mutational analysis allows a better prognostication than T analysis alone and could help in mCRC treatment management.
Collapse
|
37
|
Ottaiano A, Caraglia M, Di Mauro A, Botti G, Lombardi A, Galon J, Luce A, D’Amore L, Perri F, Santorsola M, Hermitte F, Savarese G, Tatangelo F, Granata V, Izzo F, Belli A, Scala S, Delrio P, Circelli L, Nasti G. Evolution of Mutational Landscape and Tumor Immune-Microenvironment in Liver Oligo-Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:cancers12103073. [PMID: 33096795 PMCID: PMC7589866 DOI: 10.3390/cancers12103073] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/14/2020] [Accepted: 10/18/2020] [Indexed: 12/17/2022] Open
Abstract
Simple Summary About 10% of colorectal cancer patients presents with oligo-metastatic disease. The aim of our study was to assess genetic and immunologic dynamics underlying the oligo-metastatic status, evaluating genotype-phenotype correlations in a clean and homogeneous clinical model of liver-limited metastatic colorectal cancer. We show that loss of KRAS and SMAD4 mutations characterizes the oligo-metastatic disease while a progressive mutational evolution (gain in KRAS, PI3KCA, BRAF and SMAD4) is observed in poly-metastatic evolving disease. Furthermore, high granzyme-B+ T-cells infiltration is found in oligo-metastatic lesions. This study can support innovative strategies to monitor clinical evolution and to induce regressive genetic trajectories in cancer. Abstract Genetic dynamics underlying cancer progression are largely unknown and several genes involved in highly prevalent illnesses (e.g., hypertension, obesity, and diabetes) strongly concur to cancer phenotype heterogeneity. To study genotype-phenotype relationships contributing to the mutational evolution of colorectal cancer (CRC) with a focus on liver metastases, we performed genome profiling on tumor tissues of CRC patients with liver metastatic disease and no co-morbidities. We studied 523 cancer-related genes and tumor-immune microenvironment characteristics in primary and matched metastatic tissues. We observed a loss of KRAS and SMAD4 alterations and a high granzyme-B+ T-cell infiltration when the disease did not progress. Conversely, gain in KRAS, PIK3CA and SMAD4 alterations and scarce granzyme-B+ T-cells infiltration were observed when the tumor evolved towards a poly-metastatic spread. These findings provide novel insights into the identification of tumor oligo-metastatic status, indicating that some genes are on a boundary line between these two clinical settings (oligo- vs. poly-metastatic CRC). We speculate that the identification of these genes and modification of their evolution could be a new approach for anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Department of Abdominal Oncology, SSD-Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.)
- Correspondence: ; Tel.: +39-081-590-3510; Fax: +39-081-771-4224
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (M.C.); (A.L.); (A.L.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031 Ariano Irpino, Italy
| | - Annabella Di Mauro
- Department of Pathology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (A.D.M.); (G.B.); (F.T.)
| | - Gerardo Botti
- Department of Pathology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (A.D.M.); (G.B.); (F.T.)
| | - Angela Lombardi
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (M.C.); (A.L.); (A.L.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031 Ariano Irpino, Italy
| | - Jerome Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Université Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Centre de Recherche des Cordeliers, F-75006 Paris, France;
| | - Amalia Luce
- Department of Precision Medicine, University of Campania “L. Vanvitelli”, Via L. De Crecchio 7, 80138 Naples, Italy; (M.C.); (A.L.); (A.L.)
- Biogem Scarl, Institute of Genetic Research, Laboratory of Precision and Molecular Oncology, 83031 Ariano Irpino, Italy
| | - Luigi D’Amore
- AMES-Centro Polidiagnostico Strumentale, Srl, 80013 Naples, Italy; (L.D.); (G.S.); (L.C.)
| | - Francesco Perri
- Head and Neck Cancer Medical Oncology Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Mariachiara Santorsola
- Department of Abdominal Oncology, SSD-Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.)
| | | | - Giovanni Savarese
- AMES-Centro Polidiagnostico Strumentale, Srl, 80013 Naples, Italy; (L.D.); (G.S.); (L.C.)
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (A.D.M.); (G.B.); (F.T.)
| | - Vincenza Granata
- Department of Radiology, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Francesco Izzo
- Hepatic Surgery Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.I.); (A.B.)
| | - Andrea Belli
- Hepatic Surgery Unit, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (F.I.); (A.B.)
| | - Stefania Scala
- Functional Genomics, Istituto Nazionale Tumori, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Paolo Delrio
- Colorectal Abdominal Surgery Division, Istituto Nazionale Tumori, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy;
| | - Luisa Circelli
- AMES-Centro Polidiagnostico Strumentale, Srl, 80013 Naples, Italy; (L.D.); (G.S.); (L.C.)
| | - Guglielmo Nasti
- Department of Abdominal Oncology, SSD-Innovative Therapies for Abdominal Cancers, Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, Via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.)
| |
Collapse
|
38
|
Bouchahda M, Saffroy R, Karaboué A, Hamelin J, Innominato P, Saliba F, Lévi F, Bosselut N, Lemoine A. Undetectable RAS-Mutant Clones in Plasma: Possible Implication for Anti-EGFR Therapy and Prognosis in Patients With RAS-Mutant Metastatic Colorectal Cancer. JCO Precis Oncol 2020; 4:1900400. [PMID: 33015528 PMCID: PMC7529530 DOI: 10.1200/po.19.00400] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Combining cetuximab with chemotherapy provides clinical benefit to 60% of the patients with RAS wild-type (RAS-wt) metastatic colorectal cancer (mCRC). This pilot study investigated the efficacy of cetuximab-based chemotherapy in a sample of patients (40%) with RAS mutation (RAS-mt) in their primary tumor whose circulating tumor DNA (ctDNA) was RAS-wt. MATERIALS AND METHODS The occurrence of Kirsten rat sarcoma viral oncogene homolog (KRAS), neuroblastoma rat sarcoma viral oncogene homolog (NRAS), V-raf murine sarcoma viral oncogene homolog B1 (BRAF), and PI3KCA mutations was determined in ctDNA by using a new ultrasensitive analysis based on mass spectrometry detection. All consenting patients with confirmed RAS-mt mCRC had disease progression on previous chemotherapy that contained no anti–epidermal growth factor receptor (EGFR). The patients with RAS-wt ctDNA received cetuximab + fluorouracil, leucovorin, and irinotecan (FOLFIRI), whereas those with RAS-mt ctDNA were treated with the oncologist’s choice of therapy. RESULTS Of 16 registered patients, 11 were male and five female. They were age 48 to 81 years, and they had unresectable metastatic adenocarcinoma from the colon (n = 11) or rectum (n = 5), with a median of two metastatic sites. They had received a median number of three previous chemotherapy protocols. Plasma genotyping identified RAS-mt in seven patients (44%) and RAS-wt in nine patients (56%). In the patients with wt ctDNA, objective tumor response rate was 50.0%, including one complete response and four partial responses after a median number of 6 courses of cetuximab + FOLFIRI (range, 1 to 16 courses). Two of the nine patients had stable disease, and two had progressive disease. No grade 3 to 4 toxicities were encountered. One-year survival rates were 60.0% for the patients with RAS-wt ctDNA and 17.9% for those with RAS-mt ctDNA. Median overall survival times were not reached and 4.7 months, respectively. CONCLUSION Patients with RAS-mt mCRC whose plasma biopsies contained RAS-wt could benefit from cetuximab-based therapy, a hypothesis to be tested in a prospective randomized trial.
Collapse
Affiliation(s)
- Mohamed Bouchahda
- Medical Oncology Department, Paul Brousse Hospital, Villejuif, France.,Medical Oncology Unit, Clinique du Mousseau, Evry, France
| | - Raphael Saffroy
- Oncogenetics Department, Assistance Publique-Hôpitaux de Paris, Paul Brousse Hospital, Université Paris-Saclay, Villejuif, France
| | - Abdoulaye Karaboué
- French National Institute for Health and Medical Research (INSERM), Unit 935, Villejuif, France.,Medical Oncology Unit, Groupe Hospitalier Intercommunal Le Raincy-Montfermeil, Montfermeil, France
| | - Jocelyne Hamelin
- Oncogenetics Department, Assistance Publique-Hôpitaux de Paris, Paul Brousse Hospital, Université Paris-Saclay, Villejuif, France
| | - Pasquale Innominato
- French National Institute for Health and Medical Research (INSERM), Unit 935, Villejuif, France.,North Wales Cancer Centre, Ysbyty Gwynedd, Betsi Cadwaladr University Health Board, Bangor, United Kingdom.,Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical Sciences, Warwick Medical School, Coventry, United Kingdom
| | - Faouzi Saliba
- Centre Hépato Biliaire, Assistance Publique-Hôpitaux de Paris, Hôpital Paul Brousse, Villejuif, France
| | - Francis Lévi
- Medical Oncology Department, Paul Brousse Hospital, Villejuif, France.,French National Institute for Health and Medical Research (INSERM), Unit 935, Villejuif, France.,Cancer Chronotherapy Team, Cancer Research Centre, Division of Biomedical Sciences, Warwick Medical School, Coventry, United Kingdom
| | - Nelly Bosselut
- Oncogenetics Department, Assistance Publique-Hôpitaux de Paris, Paul Brousse Hospital, Université Paris-Saclay, Villejuif, France
| | - Antoinette Lemoine
- Oncogenetics Department, Assistance Publique-Hôpitaux de Paris, Paul Brousse Hospital, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
39
|
Circulating Tumor DNA in KRAS positive colorectal cancer patients as a prognostic factor - a systematic review and meta-analysis. Crit Rev Oncol Hematol 2020; 154:103065. [PMID: 32763752 DOI: 10.1016/j.critrevonc.2020.103065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/13/2020] [Accepted: 07/20/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Liquid biopsy is a novel tool in oncology. It provides minimally invasive detection of tumor specific DNA. This review summarizes data on presence of circulating tumor DNA in serum or plasma of CRC patients as a potential negative prognostic factor. MATERIALS AND METHODS The systematic review was performed according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). The search was performed using PubMed, Web of Science and Scopus. RESULTS In total 18 articles with a total of 1779 patients met the inclusion criteria. Six out of 8 studies found that presence of ctDNA in plasma/serum was associated with inferior overall survival. All 6 studies found that high concentrations of ctDNA in plasma/serum was associated with inferior overall survival. CONCLUSIONS Presence or high concentrations of KRAS mutation in plasma or serum were associated with inferior prognosis. Establishing cut-off concentrations is warranted for further clinical implementation of liquid biopsy.
Collapse
|
40
|
You W, Yan L, Cai Z, Xie L, Sheng N, Wang G, Wu X, Wang Z. Clinical Significances of Positive Postoperative Serum CEA and Post-preoperative CEA Increment in Stage II and III Colorectal Cancer: A Multicenter Retrospective Study. Front Oncol 2020; 10:671. [PMID: 32509572 PMCID: PMC7251078 DOI: 10.3389/fonc.2020.00671] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/09/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Carcinoembryonic antigen (CEA) is the most common serum tumor marker in colorectal cancer (CRC). Nevertheless, few previous studies demonstrated the impacts of postoperative CEA and post-preoperative CEA increment on prognosis of CRC. Methods: Patients with stage II and III CRC were included from January 2009 to December 2015. All clinical and follow-up data were collected. Patients were divided into four different groups according to the levels of postoperative serum CEA and post-preoperative CEA trends. Chi-square test was used to analyze the relationship between clinical variables and categorized postoperative CEA and CEA increment. Cox proportional hazard regression was used for univariate and multivariable analyses. The log-rank test was performed to compare PFS and OS among groups. Results: Patients, 1,008, who underwent radical surgery, were enrolled. Our results showed that positive postoperative CEA and CEA increment were related to clinical stage, T stage, N stage, tumor differentiation, and lymphatic invasion (p < 0.05). Univariate and multivariable analysis results suggested that positive postoperative CEA and CEA increment were independent prognostic factors for PFS (HR = 3.149, 95% CI, 2.426–4.088, p = 0.000 for postoperative CEA; HR = 2.708, 95% CI, 2.106–3.482, p = 0.000 for CEA increment) and OS (HR = 3.414, 95% CI, 2.549–4.574, p = 0.000 for postoperative CEA; HR = 2.373, 95% CI, 1.783–3.157, p = 0.000 for CEA increment). The survival analyses revealed positive postoperative CEA, and CEA increment predicted worse prognosis. Furthermore, our results indicated that the 3- and 5-year PFS rates were 86.6 and 78.4% in group A, but decreased to 25.3 and 7.2% in group D (p < 0.001). Similarly, the 3- and 5-year OS rates for group A were 92.5 and 83.9%, much higher than group D (p < 0.001). In other words, patients with both postoperative CEA elevation and CEA increment had the worst prognosis. Conclusions: Positive postoperative CEA and CEA increment were independent prognostic factors for stage II and III CRC. Additionally, postoperative CEA and CEA increment had significant impacts on PFS and OS of CRC.
Collapse
Affiliation(s)
- Weiqiang You
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Li Yan
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zerong Cai
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lei Xie
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Nengquan Sheng
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guiyu Wang
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaojian Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Colorectal Surgery, Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhigang Wang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
41
|
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: 11] [Impact Index Per Article: 2.2] [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
|
42
|
Martini G, Ciardiello D, Vitiello PP, Napolitano S, Cardone C, Cuomo A, Troiani T, Ciardiello F, Martinelli E. Resistance to anti-epidermal growth factor receptor in metastatic colorectal cancer: What does still need to be addressed? Cancer Treat Rev 2020; 86:102023. [PMID: 32474402 DOI: 10.1016/j.ctrv.2020.102023] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022]
Abstract
Colorectal cancer (CRC) represents a global health problem, being one of the most diagnosed and aggressive tumors. Cetuximab and panitumumab monoclonal antibodies (mAbs) in combination with chemotherapy are an effective strategy for patients with RAS Wild Type (WT) metastatic colorectal cancer (mCRC). However, tumors are often unresponsive or develop resistance. In the last years, molecular alterations in principal oncogenes (RAS, BRAF, PI3KCA, PTEN) in the downstream pathway of the epidermal growth factor receptor (EGFR) and in other receptors (HER2, MET) that converge on MAPK-ERK signalling have been identified as novel mechanisms of resistance to anti-EGFR strategies. However, further efforts are needed to better stratify CRCs and ensure more individualized treatments. Herein, we describe the consolidated molecular drivers of resistance and the therapeutic strategies available so far, with an overview on potential biomarkers of response that could be integrated in clinical practice.
Collapse
Affiliation(s)
- Giulia Martini
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Davide Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Pietro Paolo Vitiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Claudia Cardone
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Antonio Cuomo
- Gastroenterology Unit, Ospedale Umberto I, Nocera Inferiore, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
43
|
Vitiello PP, De Falco V, Giunta EF, Ciardiello D, Cardone C, Vitale P, Zanaletti N, Borrelli C, Poliero L, Terminiello M, Arrichiello G, Caputo V, Famiglietti V, Mattera Iacono V, Marrone F, Di Liello A, Martini G, Napolitano S, Caraglia M, Lombardi A, Franco R, De Vita F, Morgillo F, Troiani T, Ciardiello F, Martinelli E. Clinical Practice Use of Liquid Biopsy to Identify RAS/BRAF Mutations in Patients with Metastatic Colorectal Cancer (mCRC): A Single Institution Experience. Cancers (Basel) 2019; 11:1504. [PMID: 31597339 PMCID: PMC6827157 DOI: 10.3390/cancers11101504] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/21/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022] Open
Abstract
Tumor heterogeneity represents a possible cause of error in detecting predictive genetic alterations on tumor tissue and can be overcome by testing alterations in circulating tumor DNA (ctDNA) using liquid biopsy. We assessed 72 consecutive patients with a diagnosis of metastatic colorectal cancer (mCRC) using Idylla™ Biocartis, a fully automated platform that evaluates the most frequent mutations of KRAS, NRAS and BRAF genes. We correlated the results of liquid biopsy and standard tissue-based next generation sequencing (NGS) analyses to patient clinical features. The overall agreement was 81.94%. Concordance was 85.71% and 96.15% in treatment-naïve patients and in the patient subgroup with liver metastases, respectively. In liver metastases positive, treatment-naïve patients, sensitivity, specificity and positive predictive value (PPV) were 92.31%, 100% and 100%, respectively. Circulating mutational fraction (CMF) was significantly higher in patients with liver metastases and high carcinoembryonic antigen (CEA) levels. In a subgroup of patients pre-treated with anti-Epidermal Growth Factor Receptor (EGFR) agents, emerging KRAS mutations were evidenced in 33% of cases. Testing RAS/BRAF mutations on plasma using the Idylla™ Biocartis platform is feasible and reliable in mCRC patients in clinical practice.
Collapse
Affiliation(s)
- Pietro Paolo Vitiello
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Vincenzo De Falco
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Emilio Francesco Giunta
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Davide Ciardiello
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Claudia Cardone
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Pasquale Vitale
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Nicoletta Zanaletti
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Carola Borrelli
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Luca Poliero
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Marinella Terminiello
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Gianluca Arrichiello
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Vincenza Caputo
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Vincenzo Famiglietti
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Valentina Mattera Iacono
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Francesca Marrone
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Alessandra Di Liello
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Giulia Martini
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
- Centro Cellex, Vall D'Hebron Institute of Oncology (VHIO), 08035 Barcelona, Spain.
| | - Stefania Napolitano
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
- Department of Gastrointestinal Medical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Michele Caraglia
- Department of Experimental Medicine, Università della Campania "Luigi Vanvitelli", 80138 Napoli, Italy.
| | - Angela Lombardi
- Department of Experimental Medicine, Università della Campania "Luigi Vanvitelli", 80138 Napoli, Italy.
| | - Renato Franco
- Department of Mental and Physical Health and Preventive Medicine, Pathology Unit, Università della Campania "Luigi Vanvitelli", 80138 Napoli, Italy.
| | - Ferdinando De Vita
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Floriana Morgillo
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Teresa Troiani
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| | - Fortunato Ciardiello
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy
| | - Erika Martinelli
- Department of Precision Medicine, Università della Campania "Luigi Vanvitelli", 80131 Napoli, Italy.
| |
Collapse
|