1
|
Ros J, Matito J, Villacampa G, Comas R, Garcia A, Martini G, Baraibar I, Saoudi N, Salvà F, Martin Á, Antista M, Toledo R, Martinelli E, Pietrantonio F, Boccaccino A, Cremolini C, Dientsmann R, Tabernero J, Vivancos A, Elez E. Plasmatic BRAF-V600E allele fraction as a prognostic factor in metastatic colorectal cancer treated with BRAF combinatorial treatments. Ann Oncol 2023; 34:543-552. [PMID: 36921693 DOI: 10.1016/j.annonc.2023.02.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/24/2023] [Accepted: 02/24/2023] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND Combination of a BRAF inhibitor (BRAFi) and an anti-epidermal growth factor receptor (EGFR), with or without a MEK inhibitor (MEKi), improves survival in BRAF-V600E-mutant metastatic colorectal cancer (mCRC) over standard chemotherapy. However, responses are heterogeneous and there are no available biomarkers to assess patient prognosis or guide doublet- or triplet-based regimens. In order to better characterize the clinical heterogeneity observed, we assessed the prognostic and predictive role of the plasmatic BRAF allele fraction (AF) for these combinations. PATIENTS AND METHODS A prospective discovery cohort including 47 BRAF-V600E-mutant patients treated with BRAFi-anti-EGFR ± MEKi in clinical trials and real-world practice was evaluated. Results were validated in an independent multicenter cohort (n= 29). Plasmatic BRAF-V600E AF cut-off at baseline was defined in the discovery cohort with droplet digital PCR (ddPCR). All patients had tissue-confirmed BRAF-V600E mutations. RESULTS Patients with high AF have major frequency of liver metastases and more metastatic sites. In the discovery cohort, median progression-free survival (PFS) and overall survival (OS) were 4.4 and 10.1 months, respectively. Patients with high BRAF AF (≥2%, n = 23) showed worse PFS [hazard ratio (HR) 2.97, 95% confidence interval (CI) 1.55-5.69; P = 0.001] and worse OS (HR 3.28, 95% CI 1.58-6.81; P = 0.001) than low-BRAF AF patients (<2%, n = 24). In the multivariable analysis, BRAF AF levels maintained independent significance. In the validation cohort, high BRAF AF was associated with worse PFS (HR 3.83, 95% CI 1.60-9.17; P = 0.002) and a trend toward worse OS was observed (HR 1.86, 95% CI 0.80-4.34; P = 0.15). An exploratory analysis of predictive value showed that high-BRAF AF patients (n = 35) benefited more from triplet therapy than low-BRAF AF patients (n = 41; PFS and OS interaction tests, P < 0.01). CONCLUSIONS Plasmatic BRAF AF determined by ddPCR is a reliable surrogate of tumor burden and aggressiveness in BRAF-V600E-mutant mCRC treated with a BRAFi plus an anti-EGFR with or without a MEKi and identifies patients who may benefit from treatment intensification. Our results warrant further validation of plasmatic BRAF AF to refine clinical stratification and guide treatment strategies.
Collapse
Affiliation(s)
- J Ros
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Matito
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - G Villacampa
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; The Institute of Cancer Research, London, UK
| | - R Comas
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Garcia
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - G Martini
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - I Baraibar
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - N Saoudi
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - F Salvà
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Á Martin
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - M Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - R Toledo
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - E Martinelli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
| | - F Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - A Boccaccino
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - C Cremolini
- Medical Oncology Unit, Azienda Ospedaliero-Universitaria Pisana, Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - R Dientsmann
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - J Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - A Vivancos
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - E Elez
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
2
|
Liang L, Li X, Nong L, Cai W, Zhang J, Liu P, Li T. An innovative single-base extension method for synchronous detection of point mutations and MSI status in colorectal cancer. Cancer Med 2022; 12:8367-8377. [PMID: 36583506 PMCID: PMC10134345 DOI: 10.1002/cam4.5557] [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: 03/31/2022] [Revised: 07/14/2022] [Accepted: 12/09/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND An accurate genotyping analysis is one of the critical prerequisites for patients with colorectal cancer receiving matched therapies. Conventional genotyping analysis is currently used to detect either gene mutations or MSI status, delaying the detection of critical tumor biomarkers and thus the optimal time for treatment. An assay that analyzes both biomarkers in a streamlined process is eagerly needed. METHODS We developed an assay combining Multiplex PCR Amplification, Single-base Extension and capillary electrophoresis (CE) analysis (MASE-CE) for synchronous detection of KRAS/NRAS/BRAF mutations and MSI status. In a 190 colorectal cancer cohort, we identified seven somatic mutations in KRAS, NRAS and BRAF as well as five MSI loci (D2S123/D5S346/D17S250/BAT-25/BAT-26) simultaneously. KRAS/NRAS/BRAF mutations were detected by NGS and MASE-CE, and MSI status were detected by PCR-CE and MASE-CE methods. RESULTS The MASE-CE method showed high consistency with NGS for mutation detection (Kappa value ≥0.8) and PCR-CE (Kappa value = 0.79). In addition, the limits of detection (LOD) of MASE-CE assay for MSI and somatic mutation were 5% and 2%, respectively. CONCLUSIONS In somatic mutation detection and MSI detection, the LOD of MASE-CE assay was superior to that of qPCR and NGS. MASE-CE assay is a highly sensitive, time-saving and specimen-saving method, which can greatly avoid the cumbersome testing process and provide clinical decision for doctors in time.
Collapse
Affiliation(s)
- Li Liang
- Peking University First Hospital, Beijing, China
| | - Xin Li
- Peking University First Hospital, Beijing, China
| | - Lin Nong
- Peking University First Hospital, Beijing, China
| | - Weijing Cai
- Shanghai Tongshu Biotechnology Co., Ltd, Shanghai, China
| | - Jixin Zhang
- Peking University First Hospital, Beijing, China
| | - Ping Liu
- Peking University First Hospital, Beijing, China
| | - Ting Li
- Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Bidshahri R, Fakhfakh K, McNeil K, Won JR, Wolber R, Hughesman C, Haynes C. Analysis of
KRAS
G12
/
G13
in colorectal cancer using an economical digital
PCR
assay that unequivocally differentiates missense and synonymous alleles. CAN J CHEM ENG 2021. [DOI: 10.1002/cjce.24243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Roza Bidshahri
- Michael Smith Laboratories University of British Columbia Vancouver British Columbia Canada
- Biomedical Engineering Program University of British Columbia Vancouver British Columbia Canada
| | - Kareem Fakhfakh
- Michael Smith Laboratories University of British Columbia Vancouver British Columbia Canada
- Department of Chemical and Biological Engineering University of British Columbia Vancouver British Columbia Canada
| | - Kelly McNeil
- Department of Genetics and Molecular Diagnostics British Columbia Cancer Agency Vancouver British Columbia Canada
| | - Jennifer R. Won
- Canadian Immunohistochemistry Quality Control, Department of Pathology and Laboratory Medicine University of British Columbia Vancouver British Columbia Canada
| | - Robert Wolber
- Canadian Immunohistochemistry Quality Control, Department of Pathology and Laboratory Medicine University of British Columbia Vancouver British Columbia Canada
- Department of Pathology Lion's Gate Hospital North Vancouver British Columbia Canada
| | - Curtis Hughesman
- Cancer Genetics and Genomics Lab British Columbia Cancer Agency Vancouver British Columbia Canada
| | - Charles Haynes
- Michael Smith Laboratories University of British Columbia Vancouver British Columbia Canada
- Biomedical Engineering Program University of British Columbia Vancouver British Columbia Canada
- Department of Chemical and Biological Engineering University of British Columbia Vancouver British Columbia Canada
- Genome Sciences and Technology Program Vancouver British Columbia Canada
| |
Collapse
|
4
|
Brunsell TH, Sveen A, Bjørnbeth BA, Røsok BI, Danielsen SA, Brudvik KW, Berg KCG, Johannessen B, Cengija V, Abildgaard A, Guren MG, Nesbakken A, Lothe RA. High Concordance and Negative Prognostic Impact of RAS/BRAF/PIK3CA Mutations in Multiple Resected Colorectal Liver Metastases. Clin Colorectal Cancer 2019; 19:e26-e47. [PMID: 31982351 DOI: 10.1016/j.clcc.2019.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 07/11/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence and clinical implications of genetic heterogeneity in patients with multiple colorectal liver metastases remain largely unknown. In a prospective series of patients undergoing resection of colorectal liver metastases, the aim was to investigate the inter-metastatic and primary-to-metastatic heterogeneity of mutations in KRAS, NRAS, BRAF, and PIK3CA and their prognostic impact. PATIENTS AND METHODS We analyzed the mutation status among 372 liver metastases and 78 primary tumors from 106 patients by methods used in clinical routine testing, by Sanger sequencing, by next-generation sequencing (NGS), and/or by droplet digital polymerase chain reaction. The 3-year cancer-specific survival (CSS) was analyzed using the Kaplan-Meier method. RESULTS Although Sanger sequencing indicated inter-metastatic mutation heterogeneity in 14 of 97 patients (14%), almost all cases were refuted by high-sensitive NGS. Also, heterogeneity among metastatic deposits was concluded only for PIK3CA in 2 patients. Similarly, primary-to-metastatic heterogeneity was indicated in 8 of 78 patients (10%) using Sanger sequencing but for only 2 patients after NGS, showing the emergence of 1 KRAS and 1 PIK3CA mutation in the metastatic lesions. KRAS mutations were present in 53 of 106 patients (50%) and were associated with poorer 3-year CSS after liver resection (37% vs. 61% for KRAS wild-type; P = .004). Poor prognostic associations were found also for the combination of KRAS/NRAS/BRAF mutations compared with triple wild-type (P = .002). CONCLUSION Intra-patient mutation heterogeneity was virtually undetected, both between the primary tumor and the liver metastases and among the metastatic deposits. KRAS mutations separately, and KRAS/NRAS/BRAF mutations combined, were associated with poor patient survival after partial liver resection.
Collapse
Affiliation(s)
- Tuva Høst Brunsell
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjørn Atle Bjørnbeth
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Bård I Røsok
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Stine Aske Danielsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | - Kristoffer Watten Brudvik
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Kaja C G Berg
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bjarne Johannessen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vanja Cengija
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Radiology and Nuclear Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andreas Abildgaard
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Radiology and Nuclear Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marianne Grønlie Guren
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Department of Oncology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Arild Nesbakken
- K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Department of Gastrointestinal Surgery, Oslo University Hospital, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; K. G. Jebsen Colorectal Cancer Research Centre, Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
5
|
Ylli D, Patel A, Jensen K, Li ZZ, Mendonca-Torres MC, Costello J, Gomes-Lima CJ, Wartofsky L, Burman KD, Vasko VV. Microfluidic Droplet Digital PCR Is a Powerful Tool for Detection of BRAF and TERT Mutations in Papillary Thyroid Carcinomas. Cancers (Basel) 2019; 11:cancers11121916. [PMID: 31810221 PMCID: PMC6966523 DOI: 10.3390/cancers11121916] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/15/2022] Open
Abstract
We examined the utility of microfluidic digital PCR (dPCR) for detection of BRAF and TERT mutations in thyroid tumors. DNA extracted from 100 thyroid tumors (10 follicular adenomas, 10 follicular cancers, 5 medullary cancers, and 75 papillary thyroid cancer (PTC) were used for detection of BRAF and TERT mutations. Digital PCRs were performed using rare mutation SNP genotyping assays on QuantStudio 3D platform. In PTCs, BRAFV600E was detected by dPCR and Sanger sequencing in 42/75 (56%) and in 37/75 (49%), respectively. BRAFV600E was not detected in other tumors. The ratio of mutant/total BRAF alleles varied from 4.7% to 47.5%. These ratios were higher in classical PTCs (27.1%) as compared to follicular variant PTCs (9.4%) p = 0.001. In PTCs with and without metastases, the ratios of mutant/total BRAF alleles were 27.6% and 18.4%, respectively, (p = 0.03). In metastatic lesions percentages of mutant/total BRAF alleles were similar to those detected in primary tumors. TERTC228T and TERTC250T were found in two and one cases, respectively, and these tumors concomitantly harbored BRAFV600E. These tumors exhibited gross extra-thyroidal extension, metastases to lymph nodes, and pulmonary metastases (one case). Our results showed that dPCR allows quantitative assessment of druggable targets in PTCs and could be helpful in a molecular-based stratification of prognosis in patients with thyroid cancer.
Collapse
Affiliation(s)
- Dorina Ylli
- Thyroid Cancer Research Center, MedStar Health Research Institute, 100 Irving St NW, Washington, DC 2010, USA; (D.Y.); (C.J.G.-L.); (L.W.); (K.D.B.)
- Division of Endocrinology, Department of Internal Medicine, MedStar Washington Hospital Center, 110 Irving St NW, Washington, DC 2010, USA
- Department of Imaging and Clinical Semeiotic, Faculty of Medicine, University of Medicine Tirana, 371 Dibra St, 1005 Tirana, Albania
| | - Aneeta Patel
- Department of Pediatrics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge, Bethesda, MD 20814, USA; (A.P.); (K.J.); (M.C.M.-T.); (J.C.)
| | - Kirk Jensen
- Department of Pediatrics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge, Bethesda, MD 20814, USA; (A.P.); (K.J.); (M.C.M.-T.); (J.C.)
| | - Zhao-Zhang Li
- Biomedical instrumentation center, Uniformed Services University of the Health Sciences, 4301 Jones Bridge, Bethesda, MD 20814, USA;
| | - Maria Cecilia Mendonca-Torres
- Department of Pediatrics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge, Bethesda, MD 20814, USA; (A.P.); (K.J.); (M.C.M.-T.); (J.C.)
| | - John Costello
- Department of Pediatrics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge, Bethesda, MD 20814, USA; (A.P.); (K.J.); (M.C.M.-T.); (J.C.)
| | - Cristiane Jeyce Gomes-Lima
- Thyroid Cancer Research Center, MedStar Health Research Institute, 100 Irving St NW, Washington, DC 2010, USA; (D.Y.); (C.J.G.-L.); (L.W.); (K.D.B.)
- Division of Endocrinology, Department of Internal Medicine, MedStar Washington Hospital Center, 110 Irving St NW, Washington, DC 2010, USA
| | - Leonard Wartofsky
- Thyroid Cancer Research Center, MedStar Health Research Institute, 100 Irving St NW, Washington, DC 2010, USA; (D.Y.); (C.J.G.-L.); (L.W.); (K.D.B.)
- Division of Endocrinology, Department of Internal Medicine, MedStar Washington Hospital Center, 110 Irving St NW, Washington, DC 2010, USA
| | - Kenneth Dale Burman
- Thyroid Cancer Research Center, MedStar Health Research Institute, 100 Irving St NW, Washington, DC 2010, USA; (D.Y.); (C.J.G.-L.); (L.W.); (K.D.B.)
- Division of Endocrinology, Department of Internal Medicine, MedStar Washington Hospital Center, 110 Irving St NW, Washington, DC 2010, USA
| | - Vasyl V. Vasko
- Department of Pediatrics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge, Bethesda, MD 20814, USA; (A.P.); (K.J.); (M.C.M.-T.); (J.C.)
- Correspondence: ; Tel.: +1-(240)-423-1051
| |
Collapse
|
6
|
Elez E, Chianese C, Sanz-García E, Martinelli E, Noguerido A, Mancuso FM, Caratù G, Matito J, Grasselli J, Cardone C, Esposito Abate R, Martini G, Santos C, Macarulla T, Argilés G, Capdevila J, Garcia A, Mulet N, Maiello E, Normanno N, Jones F, Tabernero J, Ciardello F, Salazar R, Vivancos A. Impact of circulating tumor DNA mutant allele fraction on prognosis in RAS-mutant metastatic colorectal cancer. Mol Oncol 2019; 13:1827-1835. [PMID: 31322322 PMCID: PMC6717744 DOI: 10.1002/1878-0261.12547] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 05/11/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022] Open
Abstract
Despite major advances in the treatment of metastatic colorectal cancer (mCRC), the survival rate remains very poor. This study aims at exploring the prognostic value of RAS-mutant allele fraction (MAF) in plasma in mCRC. Forty-seven plasma samples from 37 RAS-mutated patients with nonresectable metastases were tested for RAS in circulating tumor DNA using BEAMing before first- and/or second-line treatment. RAS MAF was correlated with several clinical parameters (number of metastatic sites, hepatic volume, carcinoembryonic antigen, CA19-9 levels, primary site location, and treatment line) and clinical outcome [progression-free survival (PFS) and overall survival (OS)]. An independent cohort of 32 patients from the CAPRI-GOIM trial was assessed for clinical outcome based on plasma baseline MAF. RAS MAF analysis at baseline revealed a significant correlation with longer OS [Hazard ratios (HR) = 3.514; P = 0.00066]. Patients with lower MAF also showed a tendency to longer PFS, although not statistically significant. Multivariate analysis showed RAS MAFs as an independent prognostic factor in both OS (HR = 2.73; P = 0.006) and first-line PFS (HR = 3.74; P = 0.049). Tumor response to treatment in patients with higher MAF was progression disease (P = 0.007). Patients with low MAFs at baseline in the CAPRI-GOIM group also showed better OS [HR = 3.84; 95% confidence intervals (CI) 1.5-9.6; P = 0.004] and better PFS (HR = 2.5; 95% CI: 1.07-5.62; P = 0.033). This minimally invasive test may help in adding an independent factor to better estimate outcomes before initiating treatment. Further prospective studies using MAF as a stratification factor could further validate its utility in clinical practice.
Collapse
Affiliation(s)
- Elena Elez
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Chiara Chianese
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Enrique Sanz-García
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Erica Martinelli
- Medical Oncology, Department of Clinical and Experimental Medicine 'F. Magrassi', Università della Campania 'L. Vanvitelli', Napoli, Italy
| | - Alba Noguerido
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | | | - Ginevra Caratù
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Judit Matito
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Julieta Grasselli
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Spain
| | - Claudia Cardone
- Medical Oncology, Department of Clinical and Experimental Medicine 'F. Magrassi', Università della Campania 'L. Vanvitelli', Napoli, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori 'Fondazione Giovanni Pascale' IRCCS, Napoli, Italy
| | - Giulia Martini
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Cristina Santos
- Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Spain
| | - Teresa Macarulla
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Guillem Argilés
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Jaume Capdevila
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Ariadna Garcia
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Nuria Mulet
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain.,Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Spain
| | - Evaristo Maiello
- Medical Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (Foggia), Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori 'Fondazione Giovanni Pascale' IRCCS, Napoli, Italy
| | | | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona, Spain.,Department of Medical Oncology, Vall d' Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Fortunato Ciardello
- Medical Oncology, Department of Clinical and Experimental Medicine 'F. Magrassi', Università della Campania 'L. Vanvitelli', Napoli, Italy
| | - Ramon Salazar
- Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
7
|
Santos C, Azuara D, Viéitez JM, Páez D, Falcó E, Élez E, López-López C, Valladares M, Robles-Díaz L, García-Alfonso P, Bugés C, Durán G, Salud A, Navarro V, Capellá G, Aranda E, Salazar R. Phase II study of high-sensitivity genotyping of KRAS, NRAS, BRAF and PIK3CA to ultra-select metastatic colorectal cancer patients for panitumumab plus FOLFIRI: the ULTRA trial. Ann Oncol 2019; 30:796-803. [PMID: 30840064 DOI: 10.1093/annonc/mdz082] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Several studies show the importance of accurately quantifying not only KRAS and other low-abundant mutations because benefits of anti-EGFR therapies may depend on certain sensitivity thresholds. We assessed whether ultra-selection of patients using a high-sensitive digital PCR (dPCR) to determine KRAS, NRAS, BRAF and PIK3CA status can improve clinical outcomes of panitumumab plus FOLFIRI. PATIENTS AND METHODS This was a single-arm phase II trial that analysed 38 KRAS, NRAS, BRAF and PIK3CA hotspots in tumour tissues of irinotecan-resistant metastatic colorectal cancer patients who received panitumumab plus FOLFIRI until disease progression or early withdrawal. Mutation profiles were identified by nanofluidic dPCR and correlated with clinical outcomes (ORR, overall response rate; PFS, progression-free survival; OS, overall survival) using cut-offs from 0% to 5%. A quantitative PCR (qPCR) analysis was also performed. RESULTS Seventy-two evaluable patients were enrolled. RAS (KRAS/NRAS) mutations were detected in 23 (32%) patients and RAS/BRAF mutations in 25 (35%) by dPCR, while they were detected in 7 (10%) and 11 (15%) patients, respectively, by qPCR. PIK3CA mutations were not considered in the analyses as they were only detected in 2 (3%) patients by dPCR and in 1 (1%) patient by qPCR. The use of different dPCR cut-offs for RAS (KRAS/NRAS) and RAS/BRAF analyses translated into differential clinical outcomes. The highest ORR, PFS and OS in wild-type patients with their lowest values in patients with mutations were achieved with a 5% cut-off. We observed similar outcomes in RAS/BRAF wild-type and mutant patients defined by qPCR. CONCLUSIONS High-sensitive dPCR accurately identified patients with KRAS, NRAS, BRAF and PIK3CA mutations. The optimal RAS/BRAF mutational cut-off for outcome prediction is 5%, which explains that the predictive performance of qPCR was not improved by dPCR. The biological and clinical implications of low-frequent mutated alleles warrant further investigations. CLINICALTRIALS.GOV NUMBER NCT01704703. EUDRACT NUMBER 2012-001955-38.
Collapse
Affiliation(s)
- C Santos
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat; Department of Medical Oncology, Institut Català d'Oncologia Oncobell Program-IDIBELL, CIBERONC, L'Hospitalet de Llobregat
| | - D Azuara
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat
| | - J M Viéitez
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo
| | - D Páez
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - E Falcó
- Department of Medical Oncology, Hospital Son Llàtzer, Palma de Mallorca
| | - E Élez
- Department of Medical Oncology, Hospital Vall d'Hebrón, Barcelona
| | - C López-López
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander
| | - M Valladares
- Department of Medical Oncology, Hospital Universitario de A Coruña, A Coruña
| | - L Robles-Díaz
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid
| | - P García-Alfonso
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid
| | - C Bugés
- Department of Medical Oncology, Institut Català d'Oncologia-Hospital Germans Trias i Pujol, Badalona, Institut Català d'Oncologia-Hospital Germans Trias i Pujol
| | - G Durán
- Department of Medical Oncology, Hospital Universitario Virgen de la Victoria, Málaga
| | - A Salud
- Department of Medical Oncology, Hospital Universitari Arnau de Vilanova, Lleida
| | - V Navarro
- Clinical Research Unit, Institut Català d'Oncologia, L'Hospitalet de Llobregat
| | - G Capellá
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat
| | - E Aranda
- Department of Medical Oncology, IMIBIC, Hospital Universitario Reina Sofía, Universidad de Córdoba, CIBERONC, Córdoba, Spain
| | - R Salazar
- Translational Research Laboratory, Institut Català d'Oncologia Oncobell Program-IDIBELL, L'Hospitalet de Llobregat; Department of Medical Oncology, Institut Català d'Oncologia Oncobell Program-IDIBELL, CIBERONC, L'Hospitalet de Llobregat.
| |
Collapse
|
8
|
Rodriguez-Freixinos V, Ruiz-Pace F, Fariñas-Madrid L, Garrido-Castro AC, Villacampa G, Nuciforo P, Vivancos A, Dienstmann R, Oaknin A. Genomic heterogeneity and efficacy of PI3K pathway inhibitors in patients with gynaecological cancer. ESMO Open 2019. [PMID: 30962959 DOI: 10.1136/esmoopen-2018-000444] [] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objectives Aberrant PI3K/AKT/mTOR activation is common in gynaecological malignancies. However, predictive biomarkers of response to PI3K pathway inhibitors (PAMi) have yet to be identified. Methods We analysed the outcomes of patients with advanced gynaecological cancer with available genomic data, treated with PAMi as single agents or in combination in phase I clinical trials. Clinical relevance of the PIK3CA mutant allele fraction (MAF) was investigated. MAF of each variant was normalised for tumour purity in the sample (adjMAFs) to infer clonality of PIK3CA mutations, defined as clonal (≥0.4) or subclonal (<0.4). Results A total of 50 patients with gynaecological cancer (24 ovarian; 15 endometrial; 11 cervical) with available targeted mutation profiling were selected. PAMi therapy was matched to PIK3CA/PTEN mutation in 30 patients (60%). The overall response rate, median time to progression (mTTP) and clinical benefit rate (CBR) of the entire population were 10% (N=5), 3.57 months (2.57-4.4) and 40% (N=18), respectively. Genotype-matched therapy did not lead to a favourable CBR (OR 0.91, p=1 (0.2-3.7)) or mTTP (3.57 months (2.6-4.4) vs 3.73 months (1.9-13.2); HR 1.41; p=0.29). We did not detect differences in mTTP according to therapy or PIK3CA codon mutation (HR 1.71, p=0.24). Overall, 41% of patients had a TTP ratio (TTP PAMi/TTP on immediately prior or subsequent palliative chemotherapy) ≥1.3, without statistically significant differences according to tumour type (p=0.39), molecular alteration status (p=0.13) or therapy (p=0.54). In univariate analysis, genotype-matched therapy in patients with PIK3CA clonal events was associated with improved mTTP (HR 3.6; p=0.03). Conclusions Our study demonstrates that patients with advanced gynaecological cancer, refractory to standard therapies, achieved meaningful clinical benefit from PAMi. The impact of PI3KCA clonality on response to selected PAMi in patients with gynaecological cancer deserves further investigation.
Collapse
Affiliation(s)
- Victor Rodriguez-Freixinos
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Fiorella Ruiz-Pace
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lorena Fariñas-Madrid
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Christina Garrido-Castro
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Guillermo Villacampa
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Oaknin
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
9
|
Rodriguez-Freixinos V, Ruiz-Pace F, Fariñas-Madrid L, Garrido-Castro AC, Villacampa G, Nuciforo P, Vivancos A, Dienstmann R, Oaknin A. Genomic heterogeneity and efficacy of PI3K pathway inhibitors in patients with gynaecological cancer. ESMO Open 2019. [PMID: 30962959 DOI: 10.1136/esmoopen-2018-000444]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
OBJECTIVES Aberrant PI3K/AKT/mTOR activation is common in gynaecological malignancies. However, predictive biomarkers of response to PI3K pathway inhibitors (PAMi) have yet to be identified. METHODS We analysed the outcomes of patients with advanced gynaecological cancer with available genomic data, treated with PAMi as single agents or in combination in phase I clinical trials. Clinical relevance of the PIK3CA mutant allele fraction (MAF) was investigated. MAF of each variant was normalised for tumour purity in the sample (adjMAFs) to infer clonality of PIK3CA mutations, defined as clonal (≥0.4) or subclonal (<0.4). RESULTS A total of 50 patients with gynaecological cancer (24 ovarian; 15 endometrial; 11 cervical) with available targeted mutation profiling were selected. PAMi therapy was matched to PIK3CA/PTEN mutation in 30 patients (60%). The overall response rate, median time to progression (mTTP) and clinical benefit rate (CBR) of the entire population were 10% (N=5), 3.57 months (2.57-4.4) and 40% (N=18), respectively. Genotype-matched therapy did not lead to a favourable CBR (OR 0.91, p=1 (0.2-3.7)) or mTTP (3.57 months (2.6-4.4) vs 3.73 months (1.9-13.2); HR 1.41; p=0.29). We did not detect differences in mTTP according to therapy or PIK3CA codon mutation (HR 1.71, p=0.24). Overall, 41% of patients had a TTP ratio (TTP PAMi/TTP on immediately prior or subsequent palliative chemotherapy) ≥1.3, without statistically significant differences according to tumour type (p=0.39), molecular alteration status (p=0.13) or therapy (p=0.54). In univariate analysis, genotype-matched therapy in patients with PIK3CA clonal events was associated with improved mTTP (HR 3.6; p=0.03). CONCLUSIONS Our study demonstrates that patients with advanced gynaecological cancer, refractory to standard therapies, achieved meaningful clinical benefit from PAMi. The impact of PI3KCA clonality on response to selected PAMi in patients with gynaecological cancer deserves further investigation.
Collapse
Affiliation(s)
- Victor Rodriguez-Freixinos
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Fiorella Ruiz-Pace
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lorena Fariñas-Madrid
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Christina Garrido-Castro
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Guillermo Villacampa
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.,Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Oaknin
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
10
|
Rodriguez-Freixinos V, Ruiz-Pace F, Fariñas-Madrid L, Garrido-Castro AC, Villacampa G, Nuciforo P, Vivancos A, Dienstmann R, Oaknin A. Genomic heterogeneity and efficacy of PI3K pathway inhibitors in patients with gynaecological cancer. ESMO Open 2019; 4:e000444. [PMID: 30962959 PMCID: PMC6435251 DOI: 10.1136/esmoopen-2018-000444] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Aberrant PI3K/AKT/mTOR activation is common in gynaecological malignancies. However, predictive biomarkers of response to PI3K pathway inhibitors (PAMi) have yet to be identified. METHODS We analysed the outcomes of patients with advanced gynaecological cancer with available genomic data, treated with PAMi as single agents or in combination in phase I clinical trials. Clinical relevance of the PIK3CA mutant allele fraction (MAF) was investigated. MAF of each variant was normalised for tumour purity in the sample (adjMAFs) to infer clonality of PIK3CA mutations, defined as clonal (≥0.4) or subclonal (<0.4). RESULTS A total of 50 patients with gynaecological cancer (24 ovarian; 15 endometrial; 11 cervical) with available targeted mutation profiling were selected. PAMi therapy was matched to PIK3CA/PTEN mutation in 30 patients (60%). The overall response rate, median time to progression (mTTP) and clinical benefit rate (CBR) of the entire population were 10% (N=5), 3.57 months (2.57-4.4) and 40% (N=18), respectively. Genotype-matched therapy did not lead to a favourable CBR (OR 0.91, p=1 (0.2-3.7)) or mTTP (3.57 months (2.6-4.4) vs 3.73 months (1.9-13.2); HR 1.41; p=0.29). We did not detect differences in mTTP according to therapy or PIK3CA codon mutation (HR 1.71, p=0.24). Overall, 41% of patients had a TTP ratio (TTP PAMi/TTP on immediately prior or subsequent palliative chemotherapy) ≥1.3, without statistically significant differences according to tumour type (p=0.39), molecular alteration status (p=0.13) or therapy (p=0.54). In univariate analysis, genotype-matched therapy in patients with PIK3CA clonal events was associated with improved mTTP (HR 3.6; p=0.03). CONCLUSIONS Our study demonstrates that patients with advanced gynaecological cancer, refractory to standard therapies, achieved meaningful clinical benefit from PAMi. The impact of PI3KCA clonality on response to selected PAMi in patients with gynaecological cancer deserves further investigation.
Collapse
Affiliation(s)
- Victor Rodriguez-Freixinos
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Fiorella Ruiz-Pace
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Lorena Fariñas-Madrid
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Christina Garrido-Castro
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Guillermo Villacampa
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Oaknin
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
11
|
Prospective multicenter real-world RAS mutation comparison between OncoBEAM-based liquid biopsy and tissue analysis in metastatic colorectal cancer. Br J Cancer 2018; 119:1464-1470. [PMID: 30467411 PMCID: PMC6288144 DOI: 10.1038/s41416-018-0293-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 02/06/2023] Open
Abstract
Background Liquid biopsy offers a minimally invasive alternative to tissue-based evaluation of mutational status in cancer. The goal of the present study was to evaluate the aggregate performance of OncoBEAM RAS mutation analysis in plasma of colorectal cancer (CRC) patients at 10 hospital laboratories in Spain where this technology is routinely implemented. Methods Circulating cell-free DNA from plasma was examined for RAS mutations using the OncoBEAM platform at each hospital laboratory. Results were then compared to those obtained from DNA extracted from tumour tissue from the same patient. Results The overall percentage agreement between plasma-based and tissue-based RAS mutation testing of the 236 participants was 89% (210/236; kappa, 0.770 (95% CI: 0.689–0.852)). Re-analysis of tissue from all discordant cases by BEAMing revealed two false negative and five false positive tumour tissue RAS results, with a final concordance of 92%. Plasma false negative results were found more frequently in patients with exclusive lung metastatic disease. Conclusions In this first prospective real-world RAS mutation performance comparison study, a high overall agreement was observed between results obtained from plasma and tissue samples. Overall, these findings indicate that the plasma-based BEAMing assay is a viable solution for rapid delivery of RAS mutation status to determine mCRC patient eligibility for anti-EGFR therapy.
Collapse
|
12
|
Grasselli J, Elez E, Caratù G, Matito J, Santos C, Macarulla T, Vidal J, Garcia M, Viéitez JM, Paéz D, Falcó E, Lopez Lopez C, Aranda E, Jones F, Sikri V, Nuciforo P, Fasani R, Tabernero J, Montagut C, Azuara D, Dienstmann R, Salazar R, Vivancos A. Concordance of blood- and tumor-based detection of RAS mutations to guide anti-EGFR therapy in metastatic colorectal cancer. Ann Oncol 2018; 28:1294-1301. [PMID: 28368441 PMCID: PMC5834108 DOI: 10.1093/annonc/mdx112] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Circulating tumor DNA (ctDNA) is a potential source for tumor genome analysis. We explored the concordance between the mutational status of RAS in tumor tissue and ctDNA in metastatic colorectal cancer (mCRC) patients to establish eligibility for anti-epidermal growth factor receptor (EGFR) therapy. Patients and methods A prospective-retrospective cohort study was carried out. Tumor tissue from 146 mCRC patients was tested for RAS status with standard of care (SoC) PCR techniques, and Digital PCR (BEAMing) was used both in plasma and tumor tissue. Results ctDNA BEAMing RAS testing showed 89.7% agreement with SoC (Kappa index 0.80; 95% CI 0.71 − 0.90) and BEAMing in tissue showed 90.9% agreement with SoC (Kappa index 0.83; 95% CI 0.74 − 0.92). Fifteen cases (10.3%) showed discordant tissue-plasma results. ctDNA analysis identified nine cases of low frequency RAS mutations that were not detected in tissue, possibly due to technical sensitivity or heterogeneity. In six cases, RAS mutations were not detected in plasma, potentially explained by low tumor burden or ctDNA shedding. Prediction of treatment benefit in patients receiving anti-EGFR plus irinotecan in second- or third-line was equivalent if tested with SoC PCR and ctDNA. Forty-eight percent of the patients showed mutant allele fractions in plasma below 1%. Conclusions Plasma RAS determination showed high overall agreement and captured a mCRC population responsive to anti-EGFR therapy with the same predictive level as SoC tissue testing. The feasibility and practicality of ctDNA analysis may translate into an alternative tool for anti-EGFR treatment selection.
Collapse
Affiliation(s)
- J Grasselli
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona.,Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Barcelona
| | - E Elez
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona.,Department of Medical Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona
| | - G Caratù
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - J Matito
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - C Santos
- Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Barcelona
| | - T Macarulla
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona.,Department of Medical Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona
| | - J Vidal
- Department of Medical Oncology, Del Mar University Hospital, Barcelona
| | - M Garcia
- Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Barcelona
| | - J M Viéitez
- Department of Medical Oncology, Asturias University Hospital, Oviedo
| | - D Paéz
- Department of Medical Oncology, Santa Creu i Sant Pau University Hospital, Barcelona
| | - E Falcó
- Department of Medical Oncology, Son Llatzer University Hospital, Palma de Mallorca
| | - C Lopez Lopez
- Department of Medical Oncology, Marques de Valdecilla University Hospital, Santander
| | - E Aranda
- Department of Medical Oncology, Reina Sofía University Hospital, Córdoba, Spain
| | - F Jones
- Sysmex Inostics, Mundelein, USA
| | - V Sikri
- Sysmex Inostics, Mundelein, USA
| | - P Nuciforo
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - R Fasani
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology, Barcelona
| | - J Tabernero
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona.,Department of Medical Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona
| | - C Montagut
- Department of Medical Oncology, Del Mar University Hospital, Barcelona
| | - D Azuara
- Traslational Research Laboratory, Catalan Institute of Oncology, L'Hospitalet, Barcelona
| | - R Dienstmann
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Barcelona.,Oncology Data Science Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - R Salazar
- Department of Medical Oncology, Catalan Institute of Oncology, Universitat de Barcelona, L'Hospitalet, Barcelona
| | - A Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology, Barcelona
| |
Collapse
|
13
|
Vidal J, Muinelo L, Dalmases A, Jones F, Edelstein D, Iglesias M, Orrillo M, Abalo A, Rodríguez C, Brozos E, Vidal Y, Candamio S, Vázquez F, Ruiz J, Guix M, Visa L, Sikri V, Albanell J, Bellosillo B, López R, Montagut C. Plasma ctDNA RAS mutation analysis for the diagnosis and treatment monitoring of metastatic colorectal cancer patients. Ann Oncol 2018; 28:1325-1332. [PMID: 28419195 PMCID: PMC5834035 DOI: 10.1093/annonc/mdx125] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background RAS assessment is mandatory for therapy decision in metastatic colorectal cancer (mCRC) patients. This determination is based on tumor tissue, however, genotyping of circulating tumor (ct)DNA offers clear advantages as a minimally invasive method that represents tumor heterogeneity. Our study aims to evaluate the use of ctDNA as an alternative for determining baseline RAS status and subsequent monitoring of RAS mutations during therapy as a component of routine clinical practice. Patients and methods RAS mutational status in plasma was evaluated in mCRC patients by OncoBEAM™ RAS CRC assay. Concordance of results in plasma and tissue was retrospectively evaluated. RAS mutations were also prospectively monitored in longitudinal plasma samples from selected patients. Results Analysis of RAS in tissue and plasma samples from 115 mCRC patients showed a 93% overall agreement. Plasma/tissue RAS discrepancies were mainly explained by spatial and temporal tumor heterogeneity. Analysis of clinico-pathological features showed that the site of metastasis (i.e. peritoneal, lung), the histology of the tumor (i.e. mucinous) and administration of treatment previous to blood collection negatively impacted the detection of RAS in ctDNA. In patients with baseline mutant RAS tumors treated with chemotherapy/antiangiogenic, longitudinal analysis of RAS ctDNA mirrored response to treatment, being an early predictor of response. In patients RAS wt, longitudinal monitoring of RAS ctDNA revealed that OncoBEAM was useful to detect emergence of RAS mutations during anti-EGFR treatment. Conclusion The high overall agreement in RAS mutational assessment between plasma and tissue supports blood-based testing with OncoBEAM™ as a viable alternative for genotyping RAS of mCRC patients in routine clinical practice. Our study describes practical clinico-pathological specifications to optimize RAS ctDNA determination. Moreover, OncoBEAM™ is useful to monitor RAS in patients undergoing systemic therapy to detect resistance and evaluate the efficacy of particular treatments.
Collapse
Affiliation(s)
- J Vidal
- Cancer Research Program, FIMIM Hospital del Mar, Barcelona, Spain.,Medical Oncology Department, Hospital del Mar, Barcelona
| | - L Muinelo
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - A Dalmases
- Pathology Department, Hospital del Mar, Barcelona
| | - F Jones
- Sysmex Inostics Inc., Mundelein, USA
| | | | - M Iglesias
- Cancer Research Program, FIMIM Hospital del Mar, Barcelona, Spain.,Pathology Department, Hospital del Mar, Barcelona
| | - M Orrillo
- Medical Oncology Department, Hospital del Mar, Barcelona
| | - A Abalo
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - C Rodríguez
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - E Brozos
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - Y Vidal
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - S Candamio
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - F Vázquez
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - J Ruiz
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - M Guix
- Medical Oncology Department, Hospital del Mar, Barcelona
| | - L Visa
- Medical Oncology Department, Hospital del Mar, Barcelona
| | - V Sikri
- Sysmex Inostics Inc., Mundelein, USA
| | - J Albanell
- Cancer Research Program, FIMIM Hospital del Mar, Barcelona, Spain.,Medical Oncology Department, Hospital del Mar, Barcelona.,Universitat Pompeu Fabra, Barcelona, Spain
| | - B Bellosillo
- Medical Oncology Department, Hospital del Mar, Barcelona.,Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - R López
- Traslational Medical Oncology Group (Oncomet)/Liquid Biopsy Analysis Unit, Health Research Institute of Santiago (IDIS), Complexo Hospitalario Universitario de Santiago de Compostela (SERGAS) CIBERONC, Santiago de Compostela
| | - C Montagut
- Cancer Research Program, FIMIM Hospital del Mar, Barcelona, Spain.,Medical Oncology Department, Hospital del Mar, Barcelona
| |
Collapse
|
14
|
Dienstmann R, Elez E, Argiles G, Matos I, Sanz-Garcia E, Ortiz C, Macarulla T, Capdevila J, Alsina M, Sauri T, Verdaguer H, Vilaro M, Ruiz-Pace F, Viaplana C, Garcia A, Landolfi S, Palmer HG, Nuciforo P, Rodon J, Vivancos A, Tabernero J. Analysis of mutant allele fractions in driver genes in colorectal cancer - biological and clinical insights. Mol Oncol 2017; 11:1263-1272. [PMID: 28618197 PMCID: PMC5579330 DOI: 10.1002/1878-0261.12099] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/29/2017] [Accepted: 04/17/2017] [Indexed: 12/21/2022] Open
Abstract
Sequencing of tumors is now routine and guides personalized cancer therapy. Mutant allele fractions (MAFs, or the ‘mutation dose’) of a driver gene may reveal the genomic structure of tumors and influence response to targeted therapies. We performed a comprehensive analysis of MAFs of driver alterations in unpaired primary and metastatic colorectal cancer (CRC) at our institution from 2010 to 2015 and studied their potential clinical relevance. Of 763 CRC samples, 622 had detailed annotation on overall survival in the metastatic setting (OSmet) and 89 received targeted agents matched to KRAS (MEK inhibitors), BRAF (BRAF inhibitors), or PIK3CA mutations (PI3K pathway inhibitors). MAFs of each variant were normalized for tumor purity in the sample (adjMAFs). We found lower adjMAFs for BRAFV600E and PIK3CA than for KRAS,NRAS, and BRAF non‐V600 variants. TP53 and BRAFV600E adjMAFs were higher in metastases as compared to primary tumors, and high KRAS adjMAFs were found in CRC metastases of patients with KRAS wild‐type primary tumors previously exposed to EGFR antibodies. Patients with RAS‐ or BRAFV600E‐mutated tumors, irrespective of adjMAFs, had worse OSmet. There was no significant association between adjMAFs and time to progression on targeted therapies matched to KRAS,BRAF, or PIK3CA mutations, potentially related to the limited antitumor activity of the employed drugs (overall response rate of 4.5%). In conclusion, the lower BRAFV600E and PIK3CA adjMAFs in subsets of primary CRC tumors indicate subclonality of these driver genes. Differences in adjMAFs between metastases and primary tumors suggest that approved therapies may result in selection of BRAFV600E‐ and KRAS‐resistant clones and an increase in genomic heterogeneity with acquired TP53 alterations. Despite significant differences in prognosis according to mutations in driver oncogenes, adjMAFs levels did not impact on survival and did not help predict benefit with matched targeted agents in the metastatic setting.
Collapse
Affiliation(s)
- Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Elena Elez
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Guillem Argiles
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ignacio Matos
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Enrique Sanz-Garcia
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Carolina Ortiz
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Teresa Macarulla
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Alsina
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Tamara Sauri
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Helena Verdaguer
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Marta Vilaro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Fiorella Ruiz-Pace
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Cristina Viaplana
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ariadna Garcia
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Stefania Landolfi
- Pathology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | - Hector G Palmer
- Stem Cells and Cancer Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jordi Rodon
- Molecular Therapeutics Research Unit, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
15
|
Wong YK, Tsang HF, Xue VW, Chan CM, Au TC, Cho WC, Chan LW, Wong SC. Applications of digital PCR in precision medicine. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1347482] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Y. K. Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - H. F. Tsang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - V. W. Xue
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - C. M. Chan
- Department of Clinical Oncology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - T. C. Au
- Department of Clinical Oncology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - W. C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region, China
| | - L. W. Chan
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - S. C. Wong
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
16
|
Santos C, Azuara D, Garcia-Carbonero R, Alfonso PG, Carrato A, Elez ME, Gomez A, Losa F, Montagut C, Massuti B, Navarro V, Varela M, Lopez-Doriga A, Moreno V, Valladares M, Manzano JL, Vieitez JM, Aranda E, Sanjuan X, Tabernero J, Capella G, Salazar R. Optimization of RAS/BRAF Mutational Analysis Confirms Improvement in Patient Selection for Clinical Benefit to Anti-EGFR Treatment in Metastatic Colorectal Cancer. Mol Cancer Ther 2017. [DOI: 10.1158/1535-7163.mct-17-0153] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|