Implementing Pharmacogenetic Testing in Gastrointestinal Cancers (IMPACT-GI): Study Protocol for a Pragmatic Implementation Trial for Establishing DPYD and UGT1A1 Screening to Guide Chemotherapy Dosing

Lisa A Varughese, Madhuri Bhupathiraju, Glenda Hoffecker, Shannon Terek, Margaret Harr, Hakon Hakonarson, Christine Cambareri, Jessica Marini, Jeffrey Landgraf, Jinbo Chen, Genevieve Kanter, Kelsey S Lau-Min, Ryan C Massa, Nevena Damjanov, Nandi J Reddy, Randall A Oyer, Ursina R Teitelbaum, Sony Tuteja, Lisa A Varughese, Madhuri Bhupathiraju, Glenda Hoffecker, Shannon Terek, Margaret Harr, Hakon Hakonarson, Christine Cambareri, Jessica Marini, Jeffrey Landgraf, Jinbo Chen, Genevieve Kanter, Kelsey S Lau-Min, Ryan C Massa, Nevena Damjanov, Nandi J Reddy, Randall A Oyer, Ursina R Teitelbaum, Sony Tuteja

Abstract

Background: Fluoropyrimidines (fluorouracil [5-FU], capecitabine) and irinotecan are commonly prescribed chemotherapy agents for gastrointestinal (GI) malignancies. Pharmacogenetic (PGx) testing for germline DPYD and UGT1A1 variants associated with reduced enzyme activity holds the potential to identify patients at high risk for severe chemotherapy-induced toxicity. Slow adoption of PGx testing in routine clinical care is due to implementation barriers, including long test turnaround times, lack of integration in the electronic health record (EHR), and ambiguity in test cost coverage. We sought to establish PGx testing in our health system following the Exploration, Preparation, Implementation, Sustainment (EPIS) framework as a guide. Our implementation study aims to address barriers to PGx testing.

Methods: The Implementing Pharmacogenetic Testing in Gastrointestinal Cancers (IMPACT-GI) study is a non-randomized, pragmatic, open-label implementation study at three sites within a major academic health system. Eligible patients with a GI malignancy indicated for treatment with 5-FU, capecitabine, or irinotecan will undergo PGx testing prior to chemotherapy initiation. Specimens will be sent to an academic clinical laboratory followed by return of results in the EHR with appropriate clinical decision support for the care team. We hypothesize that the availability of a rapid turnaround PGx test with specific dosing recommendations will increase PGx test utilization to guide pharmacotherapy decisions and improve patient safety outcomes. Primary implementation endpoints are feasibility, fidelity, and penetrance. Exploratory analyses for clinical effectiveness of genotyping will include assessing grade ≥3 treatment-related toxicity using available clinical data, patient-reported outcomes, and quality of life measures.

Conclusion: We describe the formative work conducted to prepare our health system for DPYD and UGT1A1 testing. Our prospective implementation study will evaluate the clinical implementation of this testing program and create the infrastructure necessary to ensure sustainability of PGx testing in our health system. The results of this study may help other institutions interested in implementing PGx testing in oncology care.

Clinical trial registration: https://ichgcp.net/clinical-trials-registry/NCT04736472, identifier [NCT04736472].

Keywords: DPYD; UGT1A1; cancer; chemotherapy; implementation science; pharmacogenetics; pragmatic trial; toxicity.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Varughese, Bhupathiraju, Hoffecker, Terek, Harr, Hakonarson, Cambareri, Marini, Landgraf, Chen, Kanter, Lau-Min, Massa, Damjanov, Reddy, Oyer, Teitelbaum and Tuteja.

Figures

Figure 1
Figure 1
Exploration, Preparation, Implementation, Sustainment (EPIS) framework used as a guide for implementing DPYD/UGT1A1 pharmacogenetic testing. EHR, electronic health record; PGx, pharmacogenomics; CDS, clinical decision support; QC/QI, quality control/quality improvement.
Figure 2
Figure 2
Discrete DPYD/UGT1A1 genotype results in a patient chart.
Figure 3
Figure 3
In-line warning at fluorouracil order entry informing provider of actionable DPYD results, clinical implication, and dose recommendation. © 2021 Epic Systems Corporation
Figure 4
Figure 4
Study schema for the IMPACT-GI study. (1) Patient provides informed consent at initial evaluation (baseline) visit in the gastrointestinal oncology clinic. (2) A laboratory order for pharmacogenetic testing is placed in the EHR. (3) A specimen is collected alongside routine laboratory collections by phlebotomy. (4) The specimen is sent to an external CLIA laboratory for genotyping and report generation. (5) Pharmacogenetic results are entered by the institutional lab into the precision medicine section of the EHR as discrete result components. (6a) When the care team signs and verifies chemotherapy orders, (6b) an alert indicating increased toxicity risk appears in the patient’s chart for individuals with actionable results. Clinical decision support provides recommendations for dose adjustments. (7) Following order verification, chemotherapy is prepared and dispensed to the patient in the oncology infusion suite. Fidelity is demonstrated when the prescriber adjusts dosing according to the patient’s genotype. Adverse event data is collected for the first six cycles of treatment. Patients complete a symptom questionnaire at each of these cycles and a one-time survey assessing attitudes towards pharmacogenetic testing after receiving at least two cycles. Tumor outcomes (progression-free survival, overall survival) are assessed at approximately six months following treatment initiation.

References

    1. Volpi S, Bult CJ, Chisholm RL, Deverka PA, Ginsburg GS, Jacob HJ, et al. . Research Directions in the Clinical Implementation of Pharmacogenomics: An Overview of US Programs and Projects. Clin Pharmacol Ther (2018) 103:778–86. doi: 10.1002/cpt.1048
    1. Dunnenberger HM, Crews KR, Hoffman JM, Caudle KE, Broeckel U, Howard SC, et al. . Preemptive Clinical Pharmacogenetics Implementation: Current Programs in Five US Medical Centers. Annu Rev Pharmacol Toxicol (2015) 55:89–106. doi: 10.1146/annurev-pharmtox-010814-124835
    1. Rasmussen-Torvik LJ, Stallings SC, Gordon AS, Almoguera B, Basford MA, Bielinski SJ, et al. . Design and Anticipated Outcomes of the eMERGE-PGx Project: A Multicenter Pilot for Preemptive Pharmacogenomics in Electronic Health Record Systems. Clin Pharmacol Ther (2014) 96:482–9. doi: 10.1038/clpt.2014.137
    1. Shuldiner AR, Palmer K, Pakyz RE, Alestock TD, Maloney KA, O’Neill C, et al. . Implementation of Pharmacogenetics: The University of Maryland Personalized Anti-Platelet Pharmacogenetics Program. Am J Med Genet C Semin Med Genet (2014) 166C:76–84. doi: 10.1002/ajmg.c.31396
    1. Deenen MJ, Meulendijks D, Cats A, Sechterberger MK, Severens JL, Boot H, et al. . Upfront Genotyping of DPYD*2A to Individualize Fluoropyrimidine Therapy: A Safety and Cost Analysis. J Clin Oncol (2016) 34:227–34. doi: 10.1200/JCO.2015.63.1325
    1. Rivers Z, Stenehjem DD, Jacobson P, Lou E, Nelson A, Kuntz KM. A Cost-Effectiveness Analysis of Pretreatment DPYD and UGT1A1 Screening in Patients With Metastatic Colorectal Cancer (mCRC) Treated With FOLFIRI+bevacizumab (FOLFIRI+Bev). JCO (2020) 38:168–8. doi: 10.1200/JCO.2020.38.4_suppl.168
    1. Murphy C, Byrne S, Ahmed G, Kenny A, Gallagher J, Harvey H, et al. . Cost Implications of Reactive Versus Prospective Testing for Dihydropyrimidine Dehydrogenase Deficiency in Patients With Colorectal Cancer: A Single-Institution Experience. Dose Response (2018) 16:1559325818803042. doi: 10.1177/1559325818803042
    1. Cremolini C, Del Re M, Antoniotti C, Lonardi S, Bergamo F, Loupakis F, et al. . DPYD and UGT1A1 Genotyping to Predict Adverse Events During First-Line FOLFIRI or FOLFOXIRI Plus Bevacizumab in Metastatic Colorectal Cancer. Oncotarget (2018) 9:7859–66. doi: 10.18632/oncotarget.23559
    1. Van Kuilenburg AB, Vreken P, Abeling NG, Bakker HD, Meinsma R, Van Lenthe H, et al. . Genotype and Phenotype in Patients With Dihydropyrimidine Dehydrogenase Deficiency. Hum Genet (1999) 104:1–9. doi: 10.1007/pl00008711
    1. Sharma BB, Rai K, Blunt H, Zhao W, Tosteson TD, Brooks GA. Pathogenic DPYD Variants and Treatment-Related Mortality in Patients Receiving Fluoropyrimidine Chemotherapy: A Systematic Review and Meta-Analysis. Oncologist (2021) 26:1008–16. doi: 10.1002/onco.13967
    1. Steiner M, Seule M, Steiner B, Bauer I, Freund M, Köhne CH, et al. . 5-Fluorouracil/Irinotecan Induced Lethal Toxicity as a Result of a Combined Pharmacogenetic Syndrome: Report of a Case. J Clin Pathol (2005) 58:553–5. doi: 10.1136/jcp.2004.022319
    1. Henricks LM, Lunenburg CATC, de Man FM, Meulendijks D, Frederix GWJ, Kienhuis E, et al. . DPYD Genotype-Guided Dose Individualisation of Fluoropyrimidine Therapy in Patients With Cancer: A Prospective Safety Analysis. Lancet Oncol (2018) 19:1459–67. doi: 10.1016/S1470-2045(18)30686-7
    1. Offer SM, Lee AM, Mattison LK, Fossum C, Wegner NJ, Diasio RB. A DPYD Variant (Y186C) in Individuals of African Ancestry Is Associated With Reduced DPD Enzyme Activity. Clin Pharmacol Ther (2013) 94:158–66. doi: 10.1038/clpt.2013.69
    1. Saif MW, Lee AM, Offer SM, McConnell K, Relias V, Diasio RB. A DPYD Variant (Y186C) Specific to Individuals of African Descent in a Patient With Life-Threatening 5-FU Toxic Effects: Potential for an Individualized Medicine Approach. Mayo Clin Proc (2014) 89:131–6. doi: 10.1016/j.mayocp.2013.09.008
    1. Amstutz U, Henricks LM, Offer SM, Barbarino J, Schellens JHM, Swen JJ, et al. . Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Dihydropyrimidine Dehydrogenase Genotype and Fluoropyrimidine Dosing: 2017 Update. Clin Pharmacol Ther (2018) 103:210–6. doi: 10.1002/cpt.911
    1. Nelson RS, Seligson ND, Bottiglieri S, Carballido E, Cueto AD, Imanirad I, et al. . UGT1A1 Guided Cancer Therapy: Review of the Evidence and Considerations for Clinical Implementation. Cancers (Basel) (2021) 13:1566. doi: 10.3390/cancers13071566
    1. CAMPTOSAR (Irinotecan) Injection, Intravenous Infusion . Available at: (Accessed November 4, 2021).
    1. Schmoll HJ, Van Cutsem E, Stein A, Valentini V, Glimelius B, Haustermans K, et al. . ESMO Consensus Guidelines for Management of Patients With Colon and Rectal Cancer. A Personalized Approach to Clinical Decision Making. Ann Oncol (2012) 23:2479–516. doi: 10.1093/annonc/mds236
    1. Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, et al. . ESMO Consensus Guidelines for the Management of Patients With Metastatic Colorectal Cancer. Ann Oncol (2016) 27:1386–422. doi: 10.1093/annonc/mdw235
    1. PharmGKB . Annotation of PMDA Label for Irinotecan and UGT1A1 . PharmGKB. Available at: (Accessed November 4, 2021).
    1. PharmGKB . Annotation of DPWG Guideline for Irinotecan and UGT1A1 . PharmGKB. Available at: (Accessed November 5, 2021).
    1. Luczak TS, Schillo PJ, Renier CM, Waring SC, Friday BB. Feasibility of Preemptive Pharmacogenetic Testing in Colorectal Cancer Patients Within a Community Oncology Setting. J Oncol Pharm Pract (2021), 28:10781552211005528. doi: 10.1177/10781552211005529
    1. Kasi PM, Koep T, Schnettler E, Shahjehan F, Kamatham V, Baldeo C, et al. . Feasibility of Integrating Panel-Based Pharmacogenomics Testing for Chemotherapy and Supportive Care in Patients With Colorectal Cancer. Technol Cancer Res Treat (2019) 18:1533033819873924. doi: 10.1177/1533033819873924
    1. Catenacci DVT, Chase L, Lomnicki S, Karrison T, de Wilton Marsh R, Rampurwala MM, et al. . Evaluation of the Association of Perioperative UGT1A1 Genotype-Dosed gFOLFIRINOX With Margin-Negative Resection Rates and Pathologic Response Grades Among Patients With Locally Advanced Gastroesophageal Adenocarcinoma: A Phase 2 Clinical Trial. JAMA Netw Open (2020) 3:e1921290. doi: 10.1001/jamanetworkopen.2019.21290
    1. Brooks GA, Waleed M, McGrath EB, Beloin K, Walsh SK, Benoit PR, et al. . Sustainability and Clinical Outcomes of Routine Screening for Pathogenic DPYD Gene Variants Prior to Fluoropyrimidine (FP) Chemotherapy for Gastrointestinal (GI) Cancer. JCO (2021) 39:216–6. doi: 10.1200/JCO.2020.39.28_suppl.216
    1. Bauer MS, Damschroder L, Hagedorn H, Smith J, Kilbourne AM. An Introduction to Implementation Science for the Non-Specialist. BMC Psychol (2015) 3:32. doi: 10.1186/s40359-015-0089-9
    1. Damschroder LJ, Aron DC, Keith RE, Kirsh SR, Alexander JA, Lowery JC. Fostering Implementation of Health Services Research Findings Into Practice: A Consolidated Framework for Advancing Implementation Science. Implement Sci (2009) 4:50. doi: 10.1186/1748-5908-4-50
    1. Moullin JC, Dickson KS, Stadnick NA, Rabin B, Aarons GA. Systematic Review of the Exploration, Preparation, Implementation, Sustainment (EPIS) Framework. Implement Sci (2019) 14:1. doi: 10.1186/s13012-018-0842-6
    1. Aarons GA, Hurlburt M, Horwitz SM. Advancing a Conceptual Model of Evidence-Based Practice Implementation in Public Service Sectors. Adm Policy Ment Health (2011) 38:4–23. doi: 10.1007/s10488-010-0327-7
    1. Lau-Min KS, Varughese LA, Nelson MN, Cambareri C, Reddy NJ, Oyer RA, et al. . Preemptive Pharmacogenetic Testing to Guide Chemotherapy Dosing in Patients With Gastrointestinal Malignancies: A Qualitative Study of Barriers to Implementation. BMC Cancer (2022) 22:47. doi: 10.1186/s12885-022-09171-6
    1. Gammal RS, Court MH, Haidar CE, Iwuchukwu OF, Gaur AH, Alvarellos M, et al. . Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for UGT1A1 and Atazanavir Prescribing. Clin Pharmacol Ther (2016) 99:363–9. doi: 10.1002/cpt.269
    1. Lau-Min KS, Asher SB, Chen J, Domchek SM, Feldman M, Joffe S, et al. . Real-World Integration of Genomic Data Into the Electronic Health Record: The PennChart Genomics Initiative. Genet Med (2021) 23:603–5. doi: 10.1038/s41436-020-01056-y
    1. National Cancer Institute . Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0 (2017). Available at: (Accessed December 6, 2021).
    1. Kluetz PG, Chingos DT, Basch EM, Mitchell SA. Patient-Reported Outcomes in Cancer Clinical Trials: Measuring Symptomatic Adverse Events With the National Cancer Institute’s Patient-Reported Outcomes Version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). Am Soc Clin Oncol Educ Book (2016) 35:67–73. doi: 10.1200/EDBK_159514
    1. Yanez B, Pearman T, Lis CG, Beaumont JL, Cella D. The FACT-G7: A Rapid Version of the Functional Assessment of Cancer Therapy-General (FACT-G) for Monitoring Symptoms and Concerns in Oncology Practice and Research. Ann Oncol (2013) 24:1073–8. doi: 10.1093/annonc/mds539
    1. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research Electronic Data Capture (REDCap)–A Metadata-Driven Methodology and Workflow Process for Providing Translational Research Informatics Support. J BioMed Inform (2009) 42:377–81. doi: 10.1016/j.jbi.2008.08.010
    1. Patel HN, Ursan ID, Zueger PM, Cavallari LH, Pickard AS. Stakeholder Views on Pharmacogenomic Testing. Pharmacotherapy (2014) 34:151–65. doi: 10.1002/phar.1364
    1. Empey PE, Pratt VM, Hoffman JM, Caudle KE, Klein TE. Expanding Evidence Leads to New Pharmacogenomics Payer Coverage. Genet Med (2021) 23:830–2. doi: 10.1038/s41436-021-01117-w
    1. LCD - MolDX: Pharmacogenomics Testing (L38294). Available at: (Accessed November 4, 2021).
    1. Huddart R, Sangkuhl K, Whirl-Carrillo M, Klein TE. Are Randomized Controlled Trials Necessary to Establish the Value of Implementing Pharmacogenomics in the Clinic? Clin Pharmacol Ther (2019) 106:284–6. doi: 10.1002/cpt.1420
    1. Moaddeb J, Haga SB. Pharmacogenetic Testing: Current Evidence of Clinical Utility. Ther Adv Drug Saf (2013) 4:155–69. doi: 10.1177/2042098613485595
    1. Frueh FW. Back to the Future: Why Randomized Controlled Trials Cannot be the Answer to Pharmacogenomics and Personalized Medicine. Pharmacogenomics (2009) 10:1077–81. doi: 10.2217/pgs.09.62
    1. Hernández-Guío A, Bernabéu-Martínez Ma Á, Corno-Caparrós A, Aznar-Saliente Ma T, Bonete-Sánchez M, Calleja-Hernández MÁ. DPYD Variant Testing in Candidates for Fluoropyrimidine Treatment: A Study Protocol. Farm Hosp (2021) 45:155–9. doi: 10.7399/fh.11607
    1. Roberto M, Rossi A, Panebianco M, Pomes LM, Arrivi G, Ierinò D, et al. . Drug–Drug Interactions and Pharmacogenomic Evaluation in Colorectal Cancer Patients: The New Drug-PIN® System Comprehensive Approach. Pharmaceuticals (2021) 14:67. doi: 10.3390/ph14010067
    1. Maggiore RJ, Gross CP, Hurria A. Polypharmacy in Older Adults With Cancer. Oncologist (2010) 15:507–22. doi: 10.1634/theoncologist.2009-0290

Source: PubMed

3
Se inscrever