Individualized Dosing of Fluoropyrimidine-Based Chemotherapy to Prevent Severe Fluoropyrimidine-Related Toxicity: What Are the Options?

Jonathan E Knikman, Hans Gelderblom, Jos H Beijnen, Annemieke Cats, Henk-Jan Guchelaar, Linda M Henricks, Jonathan E Knikman, Hans Gelderblom, Jos H Beijnen, Annemieke Cats, Henk-Jan Guchelaar, Linda M Henricks

Abstract

Fluoropyrimidines are widely used in the treatment of several types of solid tumors. Although most often well tolerated, severe toxicity is encountered in ~ 20-30% of the patients. Individualized dosing for these patients can reduce the incidence of severe fluoropyrimidine-related toxicity. However, no consensus has been achieved on which dosing strategy is preferred. The most established strategy for individualized dosing of fluoropyrimidines is upfront genotyping of the DPYD gene. Prospective research has shown that DPYD-guided dose-individualization significantly reduces the incidence of severe toxicity and can be easily applied in routine daily practice. Furthermore, the measurement of the dihydropyrimidine dehydrogenase (DPD) enzyme activity has shown to accurately detect patients with a DPD deficiency. Yet, because this assay is time-consuming and expensive, it is not widely implemented in routine clinical care. Other methods include the measurement of pretreatment endogenous serum uracil concentrations, the uracil/dihydrouracil-ratio, and the 5-fluorouracil (5-FU) degradation rate. These methods have shown mixed results. Next to these methods to detect DPD deficiency, pharmacokinetically guided follow-up of 5-FU could potentially be used as an addition to dosing strategies to further improve the safety of fluoropyrimidines. Furthermore, baseline characteristics, such as sex, age, body composition, and renal function have shown to have a relationship with the development of severe toxicity. Therefore, these baseline characteristics should be considered as a dose-individualization strategy. We present an overview of the current dose-individualization strategies and provide perspectives for a future multiparametric approach.

Trial registration: ClinicalTrials.gov NCT04194957.

Conflict of interest statement

The authors declared no competing interests for this work.

© 2020 The Authors. Clinical Pharmacology & Therapeutics published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Metabolism of fluoropyrimidines. 5′‐dFCR, 5′‐deoxy‐5‐fluorocytidine; 5′‐dFUR, 5′‐deoxy‐5‐fluorouridine; 5‐FU, 5‐fluorouracil; 5‐FUH2, 5,6‐dihydro‐5‐fluorouracil; B‐AL, β‐ alanine; B‐UP, β‐ureidopropionate; DHU, Dihydrouracil; FBAL, α‐fluoro‐β‐alanine; FdUDP, 5‐fluoro‐2′‐deoxyuridine 5′‐diphosphate; FdUMP, 5‐fluoro‐2′‐deoxyuridine 5′‐monophosphate; FdUrd, 5‐fluoro‐2'‐deoxyuridine; FdUTP, 5‐fluoro‐2′‐deoxyuridine 5‐’triphosphate; FUDP, 5‐fluorouridine 5′‐diphosphate; FUMP, 5‐fluorouridine 5′‐monophosphate; FUPA, α‐fluoro‐β‐ureidopropionic acid; FUrd, 5‐fluorouridine; FUTP, 5‐fluorouridine 5′‐triphosphate. [Colour figure can be viewed at wileyonlinelibrary.com]
Figure 2
Figure 2
Overview of the current dosing strategy and a suggestion for a potential future dosing strategy. (a) Current dosing strategy. (b) Potential future dosing strategy in which upfront screening is performed which includesDPYD‐genotyping, DPD‐phenotyping and screening of baseline characteristics and PK‐guided follow‐up. 5‐FU, 5‐fluorouracil; DPD, dihydropyrimidine dehydrogenase; PK, pharmacokinetic. [Colour figure can be viewed at wileyonlinelibrary.com]

References

    1. Longley, D.B. , Harkin, D.P. & Johnston, P.G. 5‐Fluorouracil: mechanisms of action and clinical strategies. Nat. Rev. Cancer 3, 330–338 (2003).
    1. Rosmarin, D. et al. Genetic markers of toxicity from capecitabine and other fluorouracil‐based regimens: Investigation in the QUASAR2 study, systematic review, and meta‐analysis. J. Clin. Oncol. 32, 1031–1039 (2014).
    1. Twelves, C. et al. Capecitabine as adjuvant treatment for stage III colon cancer. N. Engl. J. Med. 352, 2696–2704 (2005).
    1. Hoff, P.M. et al. Comparison of oral capecitabine versus intravenous fluorouracil plus leucovorin as first‐line treatment in 605 patients with metastatic colorectal cancer: results of a randomized phase III study. J. Clin. Oncol. 19, 2282–2292 (2017).
    1. Van Kuilenburg, A.B.P. Dihydropyrimidine dehydrogenase and the efficacy and toxicity of 5‐fluorouracil. Eur. J. Cancer 40, 939–950 (2004).
    1. European Medicines Agency (EMA) . Capecitabine Scientific Discussion <> 2005.
    1. Diasio, R.B. & Harris, B.E. Clinical pharmacology of 5‐fluorouracil. Clin. Pharmacokinet. 16, 215–237 (1989).
    1. Jacobs, B.A.W. et al. Pronounced between‐subject and circadian variability in thymidylate synthase and dihydropyrimidine dehydrogenase enzyme activity in human volunteers. Br. J. Clin. Pharmacol. 82, 706–716 (2016).
    1. Reilly, J.J. & Workman, P. Normalisation of anti‐cancer drug dosage using body weight and surface area: is it worthwhile? Cancer Chemother. Pharmacol. 32, 411–418 (1993).
    1. Gamelin, B.E. et al. Correlation between uracil and dihydrouracil plasma ratio, fluorouracil (5‐FU) pharmacokinetic parameters, and tolerance in patients with advanced colorectal cancer: a potential interest for predicting 5‐FU toxicity and determining optimal 5‐FU dosage. J. Clin. Oncol. 17, 1105–1110 (1999).
    1. Ratain, M.J. Dear doctor: we really are not sure what dose of capecitabine you should prescribe for your patient. J. Clin. Oncol. 20, 1434–1435 (2002).
    1. Reigner, B. , Blesch, K. & Weidekamm, E. Clinical pharmacokinetics of capecitabine. Clin. Pharmacokinet. 40, 85–104 (2001).
    1. Xeloda [SmPC] . Hoffmann‐La Roche Inc [Internet] <> (2017).
    1. de Man, F.M. et al. Comparison of toxicity and effectiveness between fixed‐dose and body surface area‐based dose capecitabine. Ther. Adv. Vaccines 11, 1–14 (2019).
    1. Henricks, L.M. et al. Translating DPYD genotype into DPD phenotype: using the DPYD gene activity score. Pharmacogenomics 16, 1277–1286 (2015).
    1. Henricks, L.M. et al. DPYD genotype‐guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol. 19, 1459–1467 (2018).
    1. Mattison, L.K. , Fourie, J. , Desmond, R.A. , Modak, A. , Saif, M.W. & Diasio, R.B. Increased prevalence of dihydropyrimidine dehydrogenase deficiency in African‐Americans compared with Caucasians. Clin. Cancer Res. 12, 5491–5495 (2006).
    1. Wei, X. , McLeod, H.L. , McMurrough, J. , Gonzalez, F.J. & Fernandez‐Salguero, P. Molecular basis of the human dihydropyrimidine dehydrogenase deficiency and 5‐fluorouracil toxicity. J. Clin. Invest. 98, 610–615 (1996).
    1. Meulendijks, D. et al. Clinical relevance of DPYD variants c.1679T>G, c.1236G>A/HapB3, and c.1601G>A as predictors of severe fluoropyrimidine‐associated toxicity: a systematic review and meta‐analysis of individual patient data. Lancet Oncol. 16, 1639–1650 (2015).
    1. Lunenburg, C.A.T.C. et al. Diagnostic and therapeutic strategies for fluoropyrimidine treatment of patients carrying multiple DPYD variants. Genes (Basel) 9, 1–13 (2018).
    1. Meulendijks, D. , Cats, A. , Beijnen, J.H. & Schellens, J.H.M. Improving safety of fluoropyrimidine chemotherapy by individualizing treatment based on dihydropyrimidine dehydrogenase activity – ready for clinical practice? Cancer Treat. Rev. 50, 23–34 (2016).
    1. Terrazino, S. , Cargnin, S. , Del Re, M. , Danesi, R. , Canonico, P.L. & Genazzani, A.A. DPYD IVS14 + 1G > A and 2846A > T genotyping for the prediction of severe fluoropyrimidine‐related toxicity: a meta‐analysis. Pharmacogenomics 14, 1255–1272 (2013).
    1. Deenen, M.J. et al. Upfront genotyping of DPYD∗2A to individualize fluoropyrimidine therapy: a safety and cost analysis. J. Clin. Oncol. 34, 227–234 (2016).
    1. Henricks, L.M. et al. A cost analysis of upfront DPYD genotype–guided dose individualisation in fluoropyrimidine‐based anticancer therapy. Eur. J. Cancer 107, 60–67 (2019).
    1. Henricks, L.M. et al. Effectiveness and safety of reduced‐dose fluoropyrimidine therapy in patients carrying the DPYD*2A variant: a matched pair analysis. Int. J. Cancer 144, 2347–2354 (2019).
    1. Amstutz, U. et al. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing: 2017 update. Clin. Pharmacol. Ther. 103, 210–216 (2018).
    1. Lunenburg, C.A.T.C. et al. Dutch Pharmacogenetics Working Group (DPWG) guideline for the gene–drug interaction of DPYD and fluoropyrimidines. Eur. J. Hum. Genet. 28, 508–517 (2020).
    1. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for Fluoropyrimidines and DPYD [Internet] <> (2018).
    1. European Medicines Agency . EMA recommendations on DPD testing prior to treatment with fluorouracil, capecitabine, tegafur and flucytosine <> (2020).
    1. US Food and Drug Administration (FDA) . Fluorouracil Highlights of Prescribing Information [Internet] <> (2016).
    1. Lunenburg, C.A.T.C. , Van Staveren, M.C. , Gelderblom, H. & Swen, J.J. Pharmacogenomics of prospective DPYD genotyping in. Pharmacogenomics 17, 721–729 (2016).
    1. Falvella, F.S. et al. DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Br. J. Clin. Pharmacol. 80, 581–588 (2015).
    1. Elraiyah, T. et al. Novel deleterious dihydropyrimidine dehydrogenase variants may contribute to 5‐fluorouracil sensitivity in an East African population. Clin. Pharmacol. Ther. 101, 382–390 (2017).
    1. Offer, S.M. , Lee, A.M. , Mattison, L.K. , Fossum, C. , Wegner, N.J. & Diasio, R.B. A DPYD variant (Y186C) in individuals of African ancestry associated with reduced DPD enzyme activity. Clin. Pharmacol. Ther. 94, 158–166 (2013).
    1. Hariprakash, J.M. et al. Pharmacogenetic landscape of DPYD variants in south Asian populations by integration of genome‐scale data. Pharmacogenomics 19, 227–241 (2018).
    1. Meulendijks, D. et al. Pretreatment serum uracil concentration as a predictor of severe and fatal fluoropyrimidine‐associated toxicity. Br. J. Cancer 116, 1415–1424 (2017).
    1. Etienne‐Grimaldi, M.C. et al. New advances in DPYD genotype and risk of severe toxicity under capecitabine. PLoS One 12, 1–19 (2017).
    1. Boisdron‐Celle, M. et al. 5‐Fluorouracil‐related severe toxicity: a comparison of different methods for the pretherapeutic detection of dihydropyrimidine dehydrogenase deficiency. Cancer Lett. 249, 271–282 (2007).
    1. Etienne‐Grimaldi, M.C. et al. A French prospective pilot study for identifying dihydropyrimidine dehydrogenase (DPD) deficiency in breast cancer patients receiving capecitabine. J. Clin. Oncol. 31, e13519 (2013).
    1. Kristensen, M.H. , Pedersen, P. & Mejer, J. The value of dihydrouracil/uracil plasma ratios in predicting 5‐fluorouracilrelated toxicity in colorectal cancer patients. J. Int. Med. Res. 38, 1313–1323 (2010).
    1. Jiang, H. , Lu, J. , Jiang, J. & Hu, P. Important role of the dihydrouracil/uracil ratio in marked interpatient variations of fluoropyrimidine pharmacokinetics and pharmacodynamics. J. Clin. Pharmacol. 44, 1260–1272 (2004).
    1. Zhou, Z.W. et al. The dihydrouracil/uracil ratios in plasma and toxicities of 5‐fluorouracil‐based adjuvant chemotherapy in colorectal cancer patients. Chemotherapy 53, 127–131 (2007).
    1. Ben Fredj, R. , Gross, E. , Ben Ahmed, S. , Hassine, H. & Saguem, S. The dihydrouracil/uracil ratio in plasma, clinical and genetic analysis for screening of dihydropyrimidine dehydrogenase deficiency in colorectal cancer patients treated with 5‐fluorouracil. Pathol. Biol. 57, 470–476 (2009).
    1. French National Authority for Health . Screening for dihydropyrimidine dehydrogenase deficiency to decrease the risk of severe toxicities related to fluoropyrimidines (5‐fluorouracil or capecitabine. Ina Br <https://www.has‐> (2018).
    1. Capitain, O. et al. Screening patients for fluoropyrimidine‐related toxicity risk: the most effective method to save lives <> (2019).
    1. Coudoré, F. et al. Validation of an ultra‐high performance liquid chromatography tandem mass spectrometric method for quantifying uracil and 5,6‐dihydrouracil in human plasma. J. Chromatogr. Sci. 50, 877–884 (2012).
    1. Jacobs, B.A.W. et al. Journal of pharmaceutical and biomedical analysis development and validation of a rapid and sensitive UPLC – MS/MS method for determination of uracil and dihydrouracil in human plasma. J. Pharm. Biomed. Anal. 126, 75–82 (2016).
    1. Henricks, L.M. et al. Food‐effect study on uracil and dihydrouracil plasma levels as marker for dihydropyrimidine dehydrogenase activity in human volunteers. Br. J. Clin. Pharmacol. 84, 2761–2769 (2018).
    1. Van Staveren, M.C. , Theeuwes‐Oonk, B. , Guchelaar, H.J. , Van Kuilenburg, A.B.P. & Maring, J.G. Pharmacokinetics of orally administered uracil in healthy volunteers and in DPD‐deficient patients, a possible tool for screening of DPD deficiency. Cancer Chemother. Pharmacol. 68, 1611–1617 (2011).
    1. Van Staveren, M.C. et al. Evaluation of an oral uracil loading test to identify DPD‐deficient patients using a limited sampling strategy. Br. J. Clin. Pharmacol. 81, 553–561 (2016).
    1. van Kuilenburg, A.B.P. et al. Phenotypic and clinical implications of variants in the dihydropyrimidine dehydrogenase gene. Biochim. Biophys. Acta 1862, 754–762 (2016).
    1. Mattison, L.K. , Ezzeldin, H. , Carpenter, M. , Modak, A. , Johnson, M.R. & Diasio, R.B. Rapid identification of dihydropyrimidine dehydrogenase deficiency by using a novel 2–13C‐uracil breath test. Clin. Cancer Res. 10, 2652–2658 (2004).
    1. Opdam, F.L. , Modak, A.S. , Gelderblom, H. & Guchelaar, H.J. Breath tests to phenotype drug disposition in oncology. Clin. Pharmacokinet. 52, 919–926 (2013).
    1. Mattison, L.K. et al. The uracil breath test in the assessment of dihydropyrimidine dehydrogenase activity: Pharmacokinetic relationship between expired13CO2and plasma [2‐13C] dihydrouracil. Clin. Cancer Res. 12, 549–555 (2006).
    1. Cunha‐Junior, G.F. et al. 13C‐uracil breath test to predict 5‐fluorouracil toxicity in gastrointestinal cancer patients. Cancer Chemother. Pharmacol. 72, 1273–1282 (2013).
    1. Van Kuilenburg, A.B.P. , Van Lenthe, H. , Blom, M.J. , Mul, E.P.J. & Van Gennip, A.H. The activity of dihydropyrimidine dehydrogenase in human blood cells. Adv. Exp. Med. Biol. 431, 823–826 (1998).
    1. Chazal, M. , Etienne, M.C. , Renée, N. , Bourgeon, A. , Richelme, H. & Milano, G. Link between dihydropyrimidine dehydrogenase activity in peripheral blood mononuclear cells and liver. Clin. Cancer Res. 2, 507–510 (1996).
    1. Van Kuilenburg, A.B.P. , De Abreu, R.A. & Van Gennip, A.H. Pharmacogenetic and clinical aspects of dihydropyrimidine dehydrogenase deficiency. Ann. Clin. Biochem. 40, 41–45 (2003).
    1. Milano, G. , Etienne, M.C. , Pierrefite, V. , Barberi‐Heyob, M. , Deporte‐Fety, R. & Renée, N. Dihydropyrimidine dehydrogenase deficiency and fluorouracil‐related toxicity. Br. J. Cancer 79, 627–630 (1999).
    1. Henricks, L.M. et al. Treatment algorithm for homozygous or compound heterozygous DPYD variant allele carriers with low‐dose capecitabine. JCO Precis. Oncol. 1, 1–10 (2017).
    1. Lostia, A.M. et al. A liquid chromatography‐tandem mass spectrometry method for the determination of 5‐fluorouracil degradation rate by intact peripheral blood mononuclear cells. Ther. Drug Monit. 31, 482–488 (2009).
    1. Mazzuca, F. et al. Pre‐treatment evaluation of 5‐fluorouracil degradation rate: association of poor and ultra‐rapid metabolism with severe toxicity in a colorectal cancer patients cohort. Oncotarget 7, 20612–20620 (2016).
    1. Onesti, C.E. et al. 5‐Fluorouracil degradation rate could predict toxicity in stages II‐III colorectal cancer patients undergoing adjuvant FOLFOX. Anticancer Drugs 28, 322–326 (2016).
    1. Roberto, M. et al. Evaluation of 5‐fluorouracil degradation rate and pharmacogenetic profiling to predict toxicity following adjuvant capecitabine. Eur. J. Clin. Pharmacol. 73, 157–164 (2017).
    1. Borro, M. et al. Pre‐treatment assay of 5‐fluorouracil degradation rate (5‐ FUDR) to improve prediction of 5‐fluorouracil toxicity in gastroesophageal cancer. Oncotarget 8, 14050–14057 (2017).
    1. Gamelin, E.C. et al. Intensity and therapeutic response in patients with advanced colorectal cancer receiving infusional therapy containing 5‐FU. Cancer 77, 441–451 (1996).
    1. Gamelin, E. et al. Individual fluorouracil dose adjustment based on pharmacokinetic follow‐up compared with conventional dosage: results of a multicenter randomized trial of patients with metastatic colorectal cancer. J. Clin. Oncol. 26, 2099–2105 (2008).
    1. Milano, G. et al. Relationship between fluorouracil systemic exposure and tumor response and patient survival. J. Clin. Oncol. 12, 1291–1295 (1994).
    1. Patel, J.N. et al. A community‐based multicenter trial of pharmacokinetically guided 5‐fluorouracil dosing for personalized colorectal cancer therapy. Oncologist 19, 959–965 (2014).
    1. Kaldate, R.R. , Haregewoin, A. , Grier, C.E. , Hamilton, S.A. & McLeod, H.L. Modeling the 5‐fluorouracil area under the curve versus dose relationship to develop a pharmacokinetic dosing algorithm for colorectal cancer patients receiving FOLFOX6. Oncologist 17, 296–302 (2012).
    1. Wilhelm, M. et al. Prospective, multicenter study of 5‐fluorouracil therapeutic drug monitoring in metastatic colorectal cancer treated in routine clinical practice. Clin. Colorectal Cancer 15, 381–388 (2016).
    1. Fety, R. et al. Clinical patients impact with of pharmacokinetically‐guided results locally from advanced a multicentric head dose randomized neck trial in of carcinomas. Clin. Cancer Res. 4, 2039–2045 (1998).
    1. Gamelin, E.C. et al. Relationship between 5‐fluorouracil (5‐FU) dose intensity and therapeutic response in patients with advanced colorectal cancer receiving infusional therapy containing 5‐FU. Cancer 77, 441–451 (1996).
    1. Gamelin, E. et al. Long‐term weekly treatment of colorectal metastatic cancer with fluorouracil and leucovorin: Results of a multicentric prospective trial of fluorouracil dosage optimization by pharmacokinetic monitoring in 152 patients. J. Clin. Oncol. 16, 1470–1478 (1998).
    1. Goldstein, D.A. et al. Cost effectiveness analysis of pharmacokinetically‐guided 5‐fluorouracil in FOLFOX chemotherapy for metastatic colorectal cancer. Clin. Colorectal Cancer 13, 219–225 (2014).
    1. Bocci, G. et al. Comparative pharmacokinetic analysis of 5‐fluorouracil and its major metabolite 5‐fluoro‐5,6‐dihydrouracil after conventional and reduced test dose in cancer patients. Clin. Cancer Res. 6, 3032–3037 (2000).
    1. Bocci, G. et al. A pharmacokinetic‐based test to prevent severe 5‐fluorouracil toxicity. Clin. Pharmacol. Ther. 80, 384–395 (2006).
    1. Arnold, A.P. , Chen, X. & Itoh, Y. Sex differences in drug therapy for oncologic diseases [Internet]. Sex Gender Diff. Pharmacol 214, 67–88 (2012).
    1. Milano, G. et al. Influence of sex and age on fluorouracil clearance. J. Clin. Oncol. 10, 1171–1175 (1992).
    1. Mueller, F. et al. Gender‐specific elimination of continuous‐infusional 5‐fluorouracil in patients with gastrointestinal malignancies: results from a prospective population pharmacokinetic study. Cancer Chemother. Pharmacol. 71, 361–370 (2013).
    1. Stein, B.N. , Petrelli, N.J. , Douglass, H.O. , Driscoll, D.L. , Arcangeli, G. & Meropol, N.J. Age and sex are independent predictors of 5‐fluorouracil toxicity. Analysis of a large scale phase III trial. Cancer 75, 11–17 (1995).
    1. Sloan, B.J.A. , Loprinzi, C.L. , Novotny, P.J. , Okuno, S. , Nair, S. & Barton, D.L. Sex differences in fluorouracil‐induced stomatitis. J. Clin. Oncol. 18, 412–420 (2000).
    1. Sloan, J.A. et al. Women experience greater toxicity with fluorouracil‐based chemotherapy for colorectal cancer. J. Clin. Oncol. 20, 1491–1498 (2002).
    1. Wettergren, Y. , Carlsson, G. , Odin, E. & Gustavsson, B. Pretherapeutic uracil and dihydrouracil levels of colorectal cancer patients are associated with sex and toxic side effects during adjuvant 5‐fluorouracil–based chemotherapy. Cancer 118, 2953–3043 (2012).
    1. Abdel‐Rahman, O. Impact of sex on chemotherapy toxicity and efficacy among patients with metastatic colorectal cancer: pooled analysis of 5 randomized trials. Clin. Colorectal Cancer 18, 110–115.e2 (2019).
    1. Lim, H. et al. Sex‐dependent adverse drug reactions to 5‐fluorouracil in colorectal cancer. Biol. Pharm. Bull. 42, 594–600 (2019).
    1. Meulendijks, D. et al. Renal function, body surface area, and age are associated with risk of early‐onset fluoropyrimidine‐associated toxicity in patients treated with capecitabine‐based anticancer regimens in daily clinical care. Eur. J. Cancer 54, 120–130 (2016).
    1. Abdel‐Rahman, O. & Karachiwala, H. Impact of age on toxicity and efficacy of 5‐FU‐based combination chemotherapy among patients with metastatic colorectal cancer; a pooled analysis of five randomized trials. Int. J. Colorectal Dis. 34, 1741–1747 (2019).
    1. Gusella, M. , Toso, S. , Ferrazzi, E. , Ferrari, M. & Padrini, R. Relationships between body composition parameters and fluorouracil pharmacokinetics. Br. J. Clin. Pharmacol. 54, 131–139 (2002).
    1. Prado, C.M.M. et al. Body composition as an independent determinant of 5‐fluorouracil‐based chemotherapy toxicity. Clin. Cancer Res. 13, 3264–3268 (2007).
    1. Williams, G.R. et al. The impact of skeletal muscle on the pharmacokinetics and toxicity of 5‐fluorouracil in colorectal cancer. Cancer Chemother. Pharmacol. 81, 413–417 (2018).
    1. Kurk, S. et al. Skeletal muscle mass loss and dose‐limiting toxicities in metastatic colorectal cancer patients. J. Cachexia Sarcopenia Muscle 10, 803–813 (2019).
    1. Jung, H.W. et al. Effect of muscle mass on toxicity and survival in patients with colon cancer undergoing adjuvant chemotherapy. Support Care Cancer 23, 687–694 (2014).
    1. Cassidy, J. et al. First‐line oral capecitabine therapy in metastatic colorectal cancer: a favorable safety profile compared with intravenous 5‐fluorouracil/leucovorin. Ann. Oncol. 13, 566–575 (2002).
    1. Boisdron‐Celle, M. et al. Prevention of 5‐fluorouracil‐induced early severe toxicity by pre‐therapeutic dihydropyrimidine dehydrogenase deficiency screening: assessment of a multiparametric approach. Semin. Oncol. 44, 13–23 (2017).
    1. Etienne‐Grimaldi, M.C. et al. Prevention of 5‐fluorouracil‐induced early severe toxicity by pre‐therapeutic dihydropyrimidine dehydrogenase (DPD) deficiency screening: The multiparametric approach is not convincing. Semin. Oncol. 44, 13–23 (2017).
    1. Botticelli, A. et al. A nomogram to predict 5‐fluorouracil toxicity: when pharmacogenomics meets the patient. Anticancer Drugs 28, 551–556 (2017).
    1. Etienne‐Grimaldi, M.C. et al. FUSAFE individual patient data meta‐analysis (MA) to assess the performance of dihydropyrimidine dehydrogenase (DPD) gene polymorphisms for predicting grade 4–5 fluoropyrimidine (FP) toxicity (Abstract). Ann. Oncol. 30, 4–5 (2019).

Source: PubMed

3
订阅