1
|
Patrignani P, Tacconelli S, Contursi A, Piazuelo E, Bruno A, Nobili S, Mazzei M, Milillo C, Hofling U, Hijos-Mallada G, Sostres C, Lanas A. Optimizing aspirin dose for colorectal cancer patients through deep phenotyping using novel biomarkers of drug action. Front Pharmacol 2024; 15:1362217. [PMID: 38495101 PMCID: PMC10941341 DOI: 10.3389/fphar.2024.1362217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
Background: Low-dose aspirin's mechanism of action for preventing colorectal cancer (CRC) is still debated, and the optimal dose remains uncertain. We aimed to optimize the aspirin dose for cancer prevention in CRC patients through deep phenotyping using innovative biomarkers for aspirin's action. Methods: We conducted a Phase II, open-label clinical trial in 34 CRC patients of both sexes randomized to receive enteric-coated aspirin 100 mg/d, 100 mg/BID, or 300 mg/d for 3 ± 1 weeks. Biomarkers were evaluated in blood, urine, and colorectal biopsies at baseline and after dosing with aspirin. Novel biomarkers of aspirin action were assessed in platelets and colorectal tissues using liquid chromatography-mass spectrometry to quantify the extent of cyclooxygenase (COX)-1 and COX-2 acetylation at Serine 529 and Serine 516, respectively. Results: All aspirin doses caused comparable % acetylation of platelet COX-1 at Serine 529 associated with similar profound inhibition of platelet-dependent thromboxane (TX)A2 generation ex vivo (serum TXB2) and in vivo (urinary TXM). TXB2 was significantly reduced in CRC tissue by aspirin 300 mg/d and 100 mg/BID, associated with comparable % acetylation of COX-1. Differently, 100 mg/day showed a lower % acetylation of COX-1 in CRC tissue and no significant reduction of TXB2. Prostaglandin (PG)E2 biosynthesis in colorectal tumors and in vivo (urinary PGEM) remained unaffected by any dose of aspirin associated with the variable and low extent of COX-2 acetylation at Serine 516 in tumor tissue. Increased expression of tumor-promoting genes like VIM (vimentin) and TWIST1 (Twist Family BHLH Transcription Factor 1) vs. baseline was detected with 100 mg/d of aspirin but not with the other two higher doses. Conclusion: In CRC patients, aspirin 300 mg/d or 100 mg/BID had comparable antiplatelet effects to aspirin 100 mg/d, indicating similar inhibition of the platelet's contribution to cancer. However, aspirin 300 mg/d and 100 mg/BID can have additional anticancer effects by inhibiting cancerous tissue's TXA2 biosynthesis associated with a restraining impact on tumor-promoting gene expression. EUDRACT number: 2018-002101-65. Clinical Trial Registration: ClinicalTrials.gov, identifier NCT03957902.
Collapse
Affiliation(s)
- Paola Patrignani
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Stefania Tacconelli
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Annalisa Contursi
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Elena Piazuelo
- Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Annalisa Bruno
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Stefania Nobili
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Matteo Mazzei
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Cristina Milillo
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Psychological Sciences, Health, and Territory, “G. d’Annunzio” University, Chieti, Italy
| | - Ulrika Hofling
- Systems Pharmacology and Translational Therapeutics Laboratory, at the Center for Advanced Studies and Technology (CAST), “G. d’Annunzio” University, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Science, “G. d’Annunzio” University Medical School, Chieti, Italy
| | - Gonzalo Hijos-Mallada
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| | - Carlos Sostres
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| | - Angel Lanas
- University Hospital LB, Aragon Health Research Institute (IISAragon), CIBERehd, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
2
|
Maes-Carballo M, Gómez-Fandiño Y, García-García M, Martín-Díaz M, De-Dios-de-Santiago D, Khan KS, Bueno-Cavanillas A. Colorectal cancer treatment guidelines and shared decision making quality and reporting assessment: Systematic review. PATIENT EDUCATION AND COUNSELING 2023; 115:107856. [PMID: 37451055 DOI: 10.1016/j.pec.2023.107856] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 06/07/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Physicians must share decisions and choose personalised treatments regarding patients´ beliefs and values. OBJECTIVE To analyse the quality of the recommendations about shared decision making (SDM) in colorectal (CRC) and anal cancer treatment clinical practice guidelines (CPGs) and consensus statements (CSs). METHODS Guidelines were systematically reviewed following prospective registration (Prospero: CRD42021286146) without language restrictions searching 15 databases and 59 professional society websites from January 2010 to November 2021. A validated 31-item SDM quality assessment tool was employed to extract data in duplicate. RESULTS We identified 134 guidelines. Only 46/134 (34.3 %) mentioned SDM. Fifteen (11.1 %) made clear, precise and actionable recommendations, while 9/134 (6.7 %) indicated the strength of the SDM-related recommendations. CPGs underpinned by systematic reviews reported SDM more often than those based on consensus or reviews (35.9 % vs 32.0 %; p = 0.031). Guidelines that reported following quality tools (i.e., AGREE II) more commonly commented about SDM than when it was not reported (75.0 % vs 32.0 %; p = 0.003). CONCLUSION AND PRACTICE IMPLICATIONS Most of the CRC and anal treatment guidelines did not mention SDM and it was superficial. Guidelines based on systematic reviews and those using quality tools demonstrated better reporting of SDM. Recommendations about SDM in these guidelines merit urgent improvement.
Collapse
Affiliation(s)
- Marta Maes-Carballo
- Department of General Surgery, Breast Cancer Unit, Complexo Hospitalario de Ourense, Ourense, Spain; Hospital Público de Verín, Ourense, Spain; Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain; University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Yolanda Gómez-Fandiño
- Department of General Surgery, Breast Cancer Unit, Complexo Hospitalario de Ourense, Ourense, Spain
| | - Manuel García-García
- Department of General Surgery, Breast Cancer Unit, Complexo Hospitalario de Ourense, Ourense, Spain; University of Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | - Khalid Saeed Khan
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria IBS, Granada, Spain
| | - Aurora Bueno-Cavanillas
- Department of Preventive Medicine and Public Health, University of Granada, Granada, Spain; Instituto de Investigación Biosanitaria IBS, Granada, Spain; CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
3
|
Podolsky E, Hudek N, McCudden C, Presseau J, Yanikomeroglu S, Brouwers M, Brehaut JC. Choosing which in-hospital laboratory tests to target for intervention: a scoping review. Clin Chem Lab Med 2023; 61:388-401. [PMID: 36410390 PMCID: PMC9876731 DOI: 10.1515/cclm-2022-0910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Some laboratory testing practices may be of low value, leading to wasted resources and potential patient harm. Our scoping review investigated factors and processes that developers report using to inform decisions about what tests to target for practice improvement. METHODS We searched Medline on May 30th, 2019 and June 28th, 2021 and included guidelines, recommendation statements, or empirical studies related to test ordering practices. Studies were included if they were conducted in a tertiary care setting, reported making a choice about a specific test requiring intervention, and reported at least one factor informing that choice. We extracted descriptive details, tests chosen, processes used to make the choice, and factors guiding test choice. RESULTS From 114 eligible studies, we identified 30 factors related to test choice including clinical value, cost, prevalence of test, quality of test, and actionability of test results. We identified nine different processes used to inform decisions regarding where to spend intervention resources. CONCLUSIONS Intervention developers face difficult choices when deciding where to put scarce resources intended to improve test utilization. Factors and processes identified here can be used to inform a framework to help intervention developers make choices relevant to improving testing practices.
Collapse
Affiliation(s)
- Eyal Podolsky
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Centre for Practice Changing Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Natasha Hudek
- Clinical Epidemiology Program, Centre for Practice Changing Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Christopher McCudden
- Clinical Epidemiology Program, Centre for Practice Changing Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
- Division of Biochemistry, Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, ON, Canada
- Eastern Ontario Regional Laboratory Association, Ottawa, ON, Canada
| | - Justin Presseau
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Centre for Practice Changing Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Sezgi Yanikomeroglu
- Clinical Epidemiology Program, Centre for Practice Changing Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| | - Melissa Brouwers
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Jamie C. Brehaut
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Clinical Epidemiology Program, Centre for Practice Changing Research, Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
4
|
Luchini C, Bibeau F, Ligtenberg MJL, Singh N, Nottegar A, Bosse T, Miller R, Riaz N, Douillard JY, Andre F, Scarpa A. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: a systematic review-based approach. Ann Oncol 2020; 30:1232-1243. [PMID: 31056702 DOI: 10.1093/annonc/mdz116] [Citation(s) in RCA: 582] [Impact Index Per Article: 145.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cancers with a defective DNA mismatch repair (dMMR) system contain thousands of mutations most frequently located in monomorphic microsatellites and are thereby defined as having microsatellite instability (MSI). Therefore, MSI is a marker of dMMR. MSI/dMMR can be identified using immunohistochemistry to detect loss of MMR proteins and/or molecular tests to show microsatellite alterations. Together with tumour mutational burden (TMB) and PD-1/PD-L1 expression, it plays a role as a predictive biomarker for immunotherapy. METHODS To define best practices to implement the detection of dMMR tumours in clinical practice, the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project, based on a systematic review-approach, to generate consensus recommendations on the: (i) definitions related to the concept of MSI/dMMR; (ii) methods of MSI/dMMR testing and (iii) relationships between MSI, TMB and PD-1/PD-L1 expression. RESULTS The MSI-related definitions, for which a consensus frame-work was used to establish definitions, included: 'microsatellites', 'MSI', 'DNA mismatch repair' and 'features of MSI tumour'. This consensus also provides recommendations on MSI testing; immunohistochemistry for the mismatch repair proteins MLH1, MSH2, MSH6 and PMS2 represents the first action to assess MSI/dMMR (consensus with strong agreement); the second method of MSI/dMMR testing is represented by polymerase chain reaction (PCR)-based assessment of microsatellite alterations using five microsatellite markers including at least BAT-25 and BAT-26 (strong agreement). Next-generation sequencing, coupling MSI and TMB analysis, may represent a decisive tool for selecting patients for immunotherapy, for common or rare cancers not belonging to the spectrum of Lynch syndrome (very strong agreement). The relationships between MSI, TMB and PD-1/PD-L1 expression are complex, and differ according to tumour types. CONCLUSIONS This ESMO initiative is a response to the urgent questions raised by the growing success of immunotherapy and provides also important insights on the relationships between MSI, TMB and PD-1/PD-L1.
Collapse
Affiliation(s)
- C Luchini
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - F Bibeau
- Department of Pathology, Caen University Hospital, Caen, France
| | - M J L Ligtenberg
- Departments of Human Genetics Radboud university medical center, Nijmegen, The Netherlands; Departments of Pathology, Radboud university medical center, Nijmegen, The Netherlands
| | - N Singh
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - A Nottegar
- Department of Surgery, San Bortolo Hospital, Vicenza, Italy
| | - T Bosse
- Department of Pathology, Leiden University Medical Centre, Leiden, The Netherlands
| | - R Miller
- Department of Oncology, University College London, London, UK
| | - N Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J-Y Douillard
- European Society for Medical Oncology, Lugano, Switzerland
| | - F Andre
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France.
| | - A Scarpa
- ARC-Net Research Centre, University of Verona, Verona, Italy
| |
Collapse
|
5
|
Unim B, Pitini E, De Vito C, D'Andrea E, Marzuillo C, Villari P. Cost-Effectiveness of RAS Genetic Testing Strategies in Patients With Metastatic Colorectal Cancer: A Systematic Review. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2020; 23:114-126. [PMID: 31952666 DOI: 10.1016/j.jval.2019.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/13/2019] [Accepted: 07/22/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Monoclonal antibodies against epidermal growth factor receptor (EGFR) have proved beneficial for the treatment of metastatic colorectal cancer (mCRC), particularly when combined with predictive biomarkers of response. International guidelines recommend anti-EGFR therapy only for RAS (NRAS,KRAS) wild-type tumors because tumors with RAS mutations are unlikely to benefit. OBJECTIVES We aimed to review the cost-effectiveness of RAS testing in mCRC patients before anti-EGFR therapy and to assess how well economic evaluations adhere to guidelines. METHODS A systematic review of full economic evaluations comparing RAS testing with no testing was performed for articles published in English between 2000 and 2018. Study quality was assessed using the Quality of Health Economic Studies scale, and the British Medical Journal and the Philips checklists. RESULTS Six economic evaluations (2 cost-effectiveness analyses, 2 cost-utility analyses, and 2 combined cost-effectiveness and cost-utility analyses) were included. All studies were of good quality and adopted the perspective of the healthcare system/payer; accordingly, only direct medical costs were considered. Four studies presented testing strategies with a favorable incremental cost-effectiveness ratio under the National Institute for Clinical Excellence (£20 000-£30 000/QALY) and the US ($50 000-$100 000/QALY) thresholds. CONCLUSIONS Testing mCRC patients for RAS status and administering EGFR inhibitors only to patients with RAS wild-type tumors is a more cost-effective strategy than treating all patients without testing. The treatment of mCRC is becoming more personalized, which is essential to avoid inappropriate therapy and unnecessarily high healthcare costs. Future economic assessments should take into account other parameters that reflect the real world (eg, NRAS mutation analysis, toxicity of biological agents, genetic test sensitivity and specificity).
Collapse
Affiliation(s)
- Brigid Unim
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
| | - Erica Pitini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Corrado De Vito
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Elvira D'Andrea
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy; Brigham & Women's Hospital, Department of Medicine, Division of Pharmacoepidemiology & Pharmacoeconomics, Boston, MA, USA
| | - Carolina Marzuillo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Paolo Villari
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Kim S, Yun YM, Chae HJ, Cho HJ, Ji M, Kim IS, Wee KA, Lee W, Song SH, Woo HI, Lee SY, Chun S. Clinical Pharmacogenetic Testing and Application: Laboratory Medicine Clinical Practice Guidelines. Ann Lab Med 2017; 37:180-193. [PMID: 28029011 PMCID: PMC5204002 DOI: 10.3343/alm.2017.37.2.180] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/07/2016] [Accepted: 12/11/2016] [Indexed: 12/15/2022] Open
Abstract
Pharmacogenetic testing for clinical applications is steadily increasing. Correct and adequate use of pharmacogenetic tests is important to reduce unnecessary medical costs and adverse patient outcomes. This document contains recommended pharmacogenetic testing guidelines for clinical application, interpretation, and result reporting through a literature review and evidence-based expert opinions for the clinical pharmacogenetic testing covered by public medical insurance in Korea. This document aims to improve the utility of pharmacogenetic testing in routine clinical settings.
Collapse
Affiliation(s)
- Sollip Kim
- Department of Laboratory Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Yeo Min Yun
- Department of Laboratory Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Korea
| | - Hyo Jin Chae
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun Jung Cho
- Department of Laboratory Medicine, Konyang University Hospital, College of Medicine, Konyang University, Daejeon, Korea
| | - Misuk Ji
- Department of Laboratory Medicine, Veterans Health Service Medical Center, Seoul, Korea
| | - In Suk Kim
- Department of Laboratory Medicine, School of Medicine, Pusan National University, Busan, Korea
| | - Kyung A Wee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Woochang Lee
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea
| | - Sang Hoon Song
- Department of Laboratory Medicine, Seoul National University Hospital and College of Medicine, Seoul, Korea
| | - Hye In Woo
- Department of Laboratory Medicine, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, Korea
| | - Soo Youn Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Sail Chun
- Department of Laboratory Medicine, University of Ulsan College of Medicine and Asan Medical Center, Seoul, Korea.
| |
Collapse
|
7
|
Estevez-Garcia P, Rivera F, Molina-Pinelo S, Benavent M, Gómez J, Limón ML, Pastor MD, Martinez-Perez J, Paz-Ares L, Carnero A, Garcia-Carbonero R. Gene expression profile predictive of response to chemotherapy in metastatic colorectal cancer. Oncotarget 2016; 6:6151-9. [PMID: 25730906 PMCID: PMC4467428 DOI: 10.18632/oncotarget.3152] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 01/15/2015] [Indexed: 01/24/2023] Open
Abstract
Fluoropyrimidine-based chemotherapy (CT) has been the mainstay of care of metastatic colorectal cancer (mCRC) for years. Response rates are only observed, however, in about half of treated patients, and there are no reliable tools to prospectively identify patients more likely to benefit from therapy. The purpose of our study was to identify a gene expression profile predictive of CT response in mCRC. Whole genome expression analyses (Affymetrix GeneChip® HG-U133 Plus 2.0) were performed in fresh frozen tumor samples of 37 mCRC patients (training cohort). Differential gene expression profiles among the two study conditions (responders versus non-responders) were assessed using supervised class prediction algorithms. A set of 161 differentially expressed genes in responders (23 patients; 62%) versus non-responders (14 patients; 38%) was selected for further assessment and validation by RT-qPCR (TaqMan®Low Density Arrays (TLDA) 7900 HT Micro Fluidic Cards) in an independent multi-institutional cohort (53 mCRC patients). Seven of these genes were confirmed as significant predictors of response. Patients with a favorable predictive signature had significantly greater response rate (58% vs 13%, p = 0.024), progression-free survival (61% vs 13% at 1 year, HR = 0.32, p = 0.009) and overall survival (32 vs 16 months, HR = 0.21, p = 0.003) than patients with an unfavorable gene signature. This is the first study to validate a gene-expression profile predictive of response to CT in mCRC patients. Larger and prospective confirmatory studies are required, however, in order to successfully provide oncologists with adequate tools to optimize treatment selection in routine clinical practice.
Collapse
Affiliation(s)
- Purificacion Estevez-Garcia
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain.,Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Fernando Rivera
- Medical Oncology Department, Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Sonia Molina-Pinelo
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Marta Benavent
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain.,Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Javier Gómez
- Pathology Department, Hospital Universitario Marques de Valdecilla, Santander, Spain
| | - Maria Luisa Limón
- Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Maria Dolores Pastor
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Julia Martinez-Perez
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain.,Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Luis Paz-Ares
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain.,Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Amancio Carnero
- Laboratorio de Biología Molecular del Cáncer, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Rocio Garcia-Carbonero
- Laboratorio de Oncología Molecular y Nuevas Terapias, Instituto de Biomedicina de Sevilla (IBIS) (HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain.,Medical Oncology Department, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| |
Collapse
|
8
|
Bolton L, Reiman A, Lucas K, Timms J, Cree IA. KRAS mutation analysis by PCR: a comparison of two methods. PLoS One 2015; 10:e0115672. [PMID: 25568935 PMCID: PMC4287618 DOI: 10.1371/journal.pone.0115672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 11/26/2014] [Indexed: 01/04/2023] Open
Abstract
Background KRAS mutation assays are important companion diagnostic tests to guide anti-EGFR antibody treatment of metastatic colorectal cancer. Direct comparison of newer diagnostic methods with existing methods is an important part of validation of any new technique. In this this study, we have compared the Therascreen (Qiagen) ARMS assay with Competitive Allele-Specific TaqMan PCR (castPCR, Life Technologies) to determine equivalence for KRAS mutation analysis. Methods DNA was extracted by Maxwell (Promega) from 99 colorectal cancers. The ARMS-based Therascreen and a customized castPCR assay were performed according to the manufacturer’s instructions. All assays were performed on either an Applied Biosystems 7500 Fast Dx or a ViiA7 real-time PCR machine (both from Life Technologies). The data were collected and discrepant results re-tested with newly extracted DNA from the same blocks in both assay types. Results Of the 99 tumors included, Therascreen showed 62 tumors to be wild-type (WT) for KRAS, while 37 had KRAS mutations on initial testing. CastPCR showed 61 tumors to be wild-type (WT) for KRAS, while 38 had KRAS mutations. Thirteen tumors showed BRAF mutation in castPCR and in one of these there was also a KRAS mutation. The custom castPCR plate included several other KRAS mutations and BRAF V600E, not included in Therascreen, explaining the higher number of mutations detected by castPCR. Re-testing of discrepant results was required in three tumors, all of which then achieved concordance for KRAS. CastPCR assay Ct values were on average 2 cycles lower than Therascreen. Conclusion There was excellent correlation between the two methods. Although castPCR assay shows lower Ct values than Therascreen, this is unlikely to be clinically significant.
Collapse
Affiliation(s)
- Louise Bolton
- Department of Pathology, Queen Alexandra Hospital, Portsmouth, United Kingdom
| | - Anne Reiman
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Katie Lucas
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Judith Timms
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
| | - Ian A. Cree
- Department of Pathology and Warwick Medical School, University Hospitals Coventry and Warwickshire, Coventry, United Kingdom
- * E-mail:
| |
Collapse
|
9
|
Updated guidelines for biomarker testing in colorectal carcinoma: a national consensus of the Spanish Society of Pathology and the Spanish Society of Medical Oncology. Clin Transl Oncol 2014; 17:264-73. [DOI: 10.1007/s12094-014-1252-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/17/2014] [Indexed: 12/12/2022]
|
10
|
Wong NACS, Gonzalez D, Salto-Tellez M, Butler R, Diaz-Cano SJ, Ilyas M, Newman W, Shaw E, Taniere P, Walsh SV. RAS testing of colorectal carcinoma—a guidance document from the Association of Clinical Pathologists Molecular Pathology and Diagnostics Group. J Clin Pathol 2014; 67:751-7. [PMID: 24996433 DOI: 10.1136/jclinpath-2014-202467] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Analysis of colorectal carcinoma (CRC) tissue for KRAS codon 12 or 13 mutations to guide use of anti-epidermal growth factor receptor (EGFR) therapy is now considered mandatory in the UK. The scope of this practice has been recently extended because of data indicating that NRAS mutations and additional KRAS mutations also predict for poor response to anti-EGFR therapy. The following document provides guidance on RAS (i.e., KRAS and NRAS) testing of CRC tissue in the setting of personalised medicine within the UK and particularly within the NHS. This guidance covers issues related to case selection, preanalytical aspects, analysis and interpretation of such RAS testing.
Collapse
|
11
|
Tembuyser L, Ligtenberg MJL, Normanno N, Delen S, van Krieken JH, Dequeker EMC. Higher quality of molecular testing, an unfulfilled priority: Results from external quality assessment for KRAS mutation testing in colorectal cancer. J Mol Diagn 2014; 16:371-7. [PMID: 24631467 DOI: 10.1016/j.jmoldx.2014.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 01/20/2014] [Accepted: 01/24/2014] [Indexed: 01/09/2023] Open
Abstract
Precision medicine is now a key element in clinical oncology. RAS mutational status is a crucial predictor of responsiveness to anti-epidermal growth factor receptor agents in metastatic colorectal cancer. In an effort to guarantee high-quality testing services in molecular pathology, the European Society of Pathology has been organizing an annual KRAS external quality assessment program since 2009. In 2012, 10 formalin-fixed, paraffin-embedded samples, of which 8 from invasive metastatic colorectal cancer tissue and 2 artificial samples of cell line material, were sent to more than 100 laboratories from 26 countries with a request for routine KRAS testing. Both genotyping and clinical reports were assessed independently. Twenty-seven percent of the participants genotyped at least 1 of 10 samples incorrectly. In total, less than 5% of the distributed specimens were genotyped incorrectly. Genotyping errors consisted of false negatives, false positives, and incorrectly genotyped mutations. Twenty percent of the laboratories reported a technical error for one or more samples. A review of the written reports showed that several essential elements were missing, most notably a clinical interpretation of the test result, the method sensitivity, and the use of a reference sequence. External quality assessment serves as a valuable educational tool in assessing and improving molecular testing quality and is an important asset for monitoring quality assurance upon incorporation of new biomarkers in diagnostic services.
Collapse
Affiliation(s)
- Lien Tembuyser
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven-University of Leuven, Leuven, Belgium
| | | | - Nicola Normanno
- Cell Biology and Biotherapy Unit, National Institute for the Study and Treatment of Cancer "Fondazione Giovanni Pascale" - IRCCS, Naples, Italy
| | - Sofie Delen
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven-University of Leuven, Leuven, Belgium
| | - J Han van Krieken
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elisabeth M C Dequeker
- Biomedical Quality Assurance Research Unit, Department of Public Health and Primary Care, KU Leuven-University of Leuven, Leuven, Belgium.
| |
Collapse
|
12
|
Thiel A, Ristimäki A. Toward a Molecular Classification of Colorectal Cancer: The Role of BRAF. Front Oncol 2013; 3:281. [PMID: 24298448 PMCID: PMC3828559 DOI: 10.3389/fonc.2013.00281] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/31/2013] [Indexed: 12/24/2022] Open
Abstract
Different genetic aberrations of BRAF have been reported in various malignancies. BRAF is member of the RAS/RAF/MEK/ERK pathway and constitutive activity of this pathway can lead to increased cellular growth, invasion, and metastasis. The most common activating BRAF mutation in colorectal cancer is the V600E mutation, which is present in 5–15% of all tumors, and up to 80% of tumors with high microsatellite instability (MSI) harbor this mutation. BRAF mutation is associated with proximal location, higher age, female gender, MSI-H, high grade, and mucinous histology, and is a marker of poor prognosis in colorectal cancer. The role of BRAF mutation as a predictive marker in respect of EGFR targeted treatments is controversial. BRAF V600 selective inhibitors have been approved for the treatment of V600 mutation positive metastatic melanoma, but the response rates in colorectal cancer are poor. This might be due to innate resistance mechanisms of colorectal cancers against the treatment solely targeting BRAF. To overcome resistance the combination of treatments, simultaneous inhibition of BRAF and MEK or PI3K/mTOR, might emerge as a successful therapeutic concept.
Collapse
Affiliation(s)
- Alexandra Thiel
- Division of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital, University of Helsinki , Helsinki , Finland ; Genome-Scale Biology, Research Programs Unit, University of Helsinki , Helsinki , Finland
| | | |
Collapse
|
13
|
Malapelle U, Carlomagno C, de Luca C, Bellevicine C, Troncone G. KRAS testing in metastatic colorectal carcinoma: challenges, controversies, breakthroughs and beyond. J Clin Pathol 2013; 67:1-9. [DOI: 10.1136/jclinpath-2013-201835] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
14
|
Hottenrott C. From single protein to colorectal cancer genome landscape and network biology-based biomarkers. Surg Endosc 2013; 27:3047-8. [DOI: 10.1007/s00464-013-2852-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 01/31/2013] [Indexed: 10/27/2022]
|
15
|
Katsios CS, Papaloukas C, Roukos DH, Baltogiannis G. Gene expression 'signature' limitations and genome architecture-based perspectives for robust cancer biomarkers. Biomark Med 2013; 7:79-82. [PMID: 23387487 DOI: 10.2217/bmm.12.102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|