Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Chem Lab Med ; 61(8): 1490-1496, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-36856054

RESUMEN

OBJECTIVES: Plasma uracil is a new biomarker to assess the activity of dihydropyrimidine dehydrogenase before cancer treatment with fluoropyrimidine drugs. Knowledge on the biological variation of plasma uracil is important to assess the applicability of plasma uracil as a biomarker of drug tolerance and efficacy. METHODS: A total of 33 apparently healthy individuals were submitted to sequential blood draws for three days. On the second day, blood draws were performed every third hour for 12 h. Plasma uracil was quantified by LC-MS/MS. The within-subject (CVI) and between-subject (CVG) biological variation estimates were calculated using linear mixed-effects models. RESULTS: The overall median value of plasma uracil was 10.6 ng/mL (range 5.6-23.1 ng/mL). The CVI and CVG were 13.5 and 22.1%, respectively. Plasma uracil remained stable during the day, and there was no day-to-day variation observed. No differences in biological variation components were found between sex and no correlation to age was found. Four samples were calculated to be required to estimate the homeostatic set-point ±15% with 95% confidence. CONCLUSIONS: Plasma uracil is subject to tight homeostatic regulation without semidiurnal and day-to-day variation, however between-subject variation exists. This emphasizes plasma uracil as a well-suited biomarker for evaluation of dihydropyrimidine dehydrogenase activity, but four samples are required to establish the homeostatic set-point in a patient.


Asunto(s)
Fluorouracilo , Uracilo , Humanos , Dihidrouracilo Deshidrogenasa (NADP) , Cromatografía Liquida , Espectrometría de Masas en Tándem , Biomarcadores
2.
J Oncol Pharm Pract ; 29(1): 5-13, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34797200

RESUMEN

OBJECTIVE: To investigate if dihydropyrimidine dehydrogenase phenotyping has added value when combined with DPYD genotyping in predicting fluoropyrimidine-related toxicity. METHODS: Retrospective cohort study in which treatment and toxicity data were collected of 228 patients genotyped for four DPYD variants and phenotyped using an ex vivo peripheral blood mononuclear cell assay. RESULTS: Severe toxicity occurred in 25% of patients with a variant and normal dihydropyrimidine dehydrogenase activity, in 21% of patients without a variant and with decreased dihydropyrimidine dehydrogenase activity, and in 29% of patients without a variant and with normal dihydropyrimidine dehydrogenase activity (controls). The majority of patients with a variant or a decreased dihydropyrimidine dehydrogenase activity received an initial dose reduction (68% and 53% vs 19% in controls) and had a lower mean dose intensity (75% and 81% vs 91% in controls). Fifty percent of patients with a variant and decreased enzyme activity experienced severe toxicity, despite the lowest initial dose and whole treatment dose intensity. They also experienced more grade 4/5 toxicities. CONCLUSIONS: Our results indicate that a combined genotype-phenotype approach could be useful to identify patients at increased risk for fluoropyrimidine-associated toxicity (e.g. patients with a variant and decreased dihydropyrimidine dehydrogenase activity). Because the group sizes are too small to demonstrate statistically significant differences, this warrants further research in a prospective study in a larger cohort.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP) , Leucocitos Mononucleares , Dihidrouracilo Deshidrogenasa (NADP)/genética , Capecitabina/efectos adversos , Genotipo , Estudios Prospectivos , Estudios Retrospectivos , Fluorouracilo/efectos adversos , Antimetabolitos Antineoplásicos/efectos adversos
3.
J Oncol Pharm Pract ; 28(4): 850-869, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33832365

RESUMEN

Although therapeutically actionable molecular alterations are widely distributed across many cancer types, only a handful of them show evidence of clinical utility and are recommended for routine clinical practice in the management of cancers of colon and rectum (CRC). This 2021 update aims to provide a succinct summary on the use of prognostic and/or predictive biomarkers (expanded RAS, BRAF, microsatellite-high [MSI-H] or deficient mismatch repair [dMMR], neurotrophic tyrosine receptor kinase [NTRK] fusion genes, and human epidermal growth factor receptor type II [HER2] gene amplification) associated with CRC. Therapeutic implications of each relevant predictive or prognostic biomarker for patients with CRC are described, along with discussion on new developments on (1) biomarker-driven therapies such as testing of BRAF, MLH1 promoter methylation and MMR germline genes in differentiating sporadic CRC or hereditary conditions such as Lynch syndrome; (2) first-line use of immune checkpoint inhibitors in metastatic CRC; (3) risk stratification and therapy selection based on primary tumor location (left-sided vs. right-sided colon cancer); (3) atypical BRAF mutations; (4) use of EGFR directed therapy in the perioperative oligometastatic disease setting; (5) re-challenge of EGFR directed therapy and (6) personalizing therapy of fluoropyrimidine and irinotecan based on new evidence in pharmacogenomic testing. Data are collected and analyzed from available systematic reviews and meta-analyses of treatments with known therapeutic targets in CRC, which may be associated with predictive and/or prognostic biomarkers. Discussions are presented in an application-based format, with goal to empower pharmacists or other clinicians to gain awareness and understanding in biomarker-driven cancer therapy issues.


Asunto(s)
Neoplasias Colorrectales , Inestabilidad de Microsatélites , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptores ErbB/genética , Humanos , Pronóstico , Proteínas Proto-Oncogénicas B-raf/genética
5.
Tumour Biol ; 39(6): 1010428317701629, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28618970

RESUMEN

Zhao et al. investigated the association between germline genetic polymorphisms in DPYD, the gene encoding dihydropyrimidine dehydrogenase, and (1) the risk of developing pediatric acute lymphoblastic leukemia and (2) outcome of acute lymphoblastic leukemia following the treatment with 5-fluorouracil plus oxaliplatin (FOLFOX). The authors found that the common DPYD variant c.85T>C (rs1801265, DPYD*9A) was significantly associated with (1) risk of developing pediatric acute lymphoblastic leukemia, (2) complete response rate, (3) event-free survival, and (4) treatment-related toxicity. The authors conclude that patients carrying the c.85T>C C allele have an increased risk of developing acute lymphoblastic leukemia and have inferior outcome, and that DPYD c.85T>C can be used as a guide for individualized treatment and the decision to utilize 5-fluorouracil in acute lymphoblastic leukemia patients. In our view, the published article gives rise to multiple critical issues regarding the study's rationale and the methodology used, which strongly question the validity of the authors' conclusions.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP)/genética , Fluorouracilo , Antimetabolitos Antineoplásicos , Niño , Humanos , Polimorfismo Genético , Leucemia-Linfoma Linfoblástico de Células Precursoras , Pronóstico
7.
Urologie ; 63(4): 367-372, 2024 Apr.
Artículo en Alemán | MEDLINE | ID: mdl-38270604

RESUMEN

Penile squamous cell carcinoma is a rare, highly aggressive cancer of older men. The metastatic stage has significant therapeutic and prognostic features. Treatment of penile cancer is significantly influenced by the operation, in which an R0 situation must be achieved to ensure a realistic chance of cure. Other local therapeutic procedures such as radiotherapy are often of secondary importance. Neoadjuvant and adjuvant chemotherapy are relevant components of multimodal therapy. Post-therapeutically, patients require lifelong, risk-adapted follow-up care.


Asunto(s)
Neoplasias del Pene , Masculino , Humanos , Anciano , Neoplasias del Pene/diagnóstico , Escisión del Ganglio Linfático/métodos , Estadificación de Neoplasias , Metástasis Linfática/patología , Ganglios Linfáticos/patología
8.
ESMO Open ; 8(1): 100782, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36791638

RESUMEN

BACKGROUND: In 2020, the European Medicines Agency recommended testing patients for dihydropyrimidine dehydrogenase (DPD) deficiency before systemic treatment with fluoropyrimidines (FP). DPD activity testing identifies patients at elevated risk of severe FP-related toxicity (FP-TOX). The two most used methods for DPD testing are DPYD genotyping and DPD phenotyping (plasma uracil concentration). The primary objective of this study was to compare the overall frequency of overall grade ≥3 FP-TOX before and after the implementation of DPYD genotyping. PATIENTS AND METHODS: Two hundred thirty Danish, primarily gastrointestinal cancer patients, were DPYD-genotyped before their first dose of FP, and blood was sampled for post hoc assessment of P-uracil. The initial dose was reduced for variant carriers. Grade ≥3 FP-TOX was registered after the first three treatment cycles of FP. The frequency of toxicity was compared to a historical cohort of 492 patients with post hoc determined DPYD genotype from a biobank. RESULTS: The frequency of overall grade ≥3 FP-TOX was 27% in the DPYD genotype-guided group compared to 24% in the historical cohort. In DPYD variant carriers, DPYD genotyping reduced the frequency of FP-related hospitalization from 19% to 0%. In the control group, 4.8% of DPYD variant carriers died due to FP-TOX compared to 0% in the group receiving DPYD genotype-guided dosing of FP. In the intervention group, wild-type patients with uracil ≥16 ng/ml had a higher frequency of FP-TOX than wild-type patients with uracil <16 ng/ml (55% versus 28%). CONCLUSIONS: We found no population-level benefit of DPYD genotyping when comparing the risk of grade ≥3 FP-TOX before and after clinical implementation. We observed no deaths or FP-related hospitalizations in patients whose FP treatment was guided by a variant DPYD genotype. The use of DPD phenotyping may add valuable information in DPYD wild-type patients.


Asunto(s)
Deficiencia de Dihidropirimidina Deshidrogenasa , Neoplasias Gastrointestinales , Humanos , Antimetabolitos Antineoplásicos/efectos adversos , Capecitabina/efectos adversos , Dinamarca , Deficiencia de Dihidropirimidina Deshidrogenasa/inducido químicamente , Deficiencia de Dihidropirimidina Deshidrogenasa/tratamiento farmacológico , Deficiencia de Dihidropirimidina Deshidrogenasa/genética , Dihidrouracilo Deshidrogenasa (NADP)/genética , Neoplasias Gastrointestinales/tratamiento farmacológico , Genotipo , Uracilo/uso terapéutico
9.
Clin Chim Acta ; 538: 87-90, 2023 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-36347332

RESUMEN

BACKGROUND AND AIMS: Measurement of plasma uracil is used before cancer treatment with fluoropyrimidines to determine if patients tolerate a full dose. Incorrect preanalytical handling may cause falsely elevated concentration and result in suboptimal cancer treatment. We aimed to examine the stability of uracil in whole blood stored at room temperature (RT) and the effect of centrifugation temperature. MATERIALS AND METHODS: EDTA tubes (6x4 mL) were collected from 25 healthy volunteers. Five samples were stored 0, 1.5, 2, 3, and 4 h at RT and centrifuged at 4 °C. The sixth sample was centrifuged at RT after 1.5 h. Uracil was measured using an in-house LC-MS/MS method. RESULTS: Storage of whole blood at RT followed by centrifugation at 4 °C caused a rapid increase in uracil concentration. Already after 1.5 h, the mean change (20.5 % (95 % CI: 11.9-29.2 %)) exceeded the maximum permissible difference. Centrifugation at RT instead of 4 °C after 1.5 h resulted in a smaller increase (7.0 % (95 % CI: 0.7-13.4 %)), although not statistically significant (p = 0.0527). CONCLUSION: Uracil was unstable in samples processed according to current recommendations. Our data indicates better stability when centrifugation is performed at RT compared with 4 °C but further research into this is necessary.


Asunto(s)
Neoplasias , Uracilo , Humanos , Recolección de Muestras de Sangre/métodos , Cromatografía Liquida , Espectrometría de Masas en Tándem/métodos , Neoplasias/tratamiento farmacológico , Temperatura , Factores Inmunológicos
10.
Front Oncol ; 13: 1187052, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37409256

RESUMEN

Esophageal cancer is a cancer with poor prognosis and the standard 1st line treatment for metastatic or recurrent EC is systemic chemotherapy with doublet chemotherapy based on platinum and 5-fluorouracil (5-FU). However, 5-FU could be a source of severe treatment-related toxicities due to deficiency of dihydropyrimidine dehydrogenase (DPD). In this case report, a 74-year-old man with metastatic esophageal cancer was found to have partial DPD deficiency based on uracilemia measurements (about 90 ng/mL). Despite this, 5-FU was safely administered thanks to therapeutic drug monitoring (TDM). The case report highlights the importance of TDM in administering 5-FU to patients with partial DPD deficiency, as it allows individualized dosing and prevents severe toxicity.

11.
ESMO Open ; 8(2): 101197, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36989883

RESUMEN

BACKGROUND: The main cause for fluoropyrimidine-related toxicity is deficiency of the metabolizing enzyme dihydropyrimidine dehydrogenase (DPD). In 2020, the European Medicines Agency (EMA) recommended two methods for pre-treatment DPD deficiency testing in clinical practice: phenotyping using endogenous uracil concentration or genotyping for DPYD risk variant alleles. This study assessed the DPD testing implementation status in Europe before (2019) and after (2021) the release of the EMA recommendations. METHODS: The survey was conducted from 16 March 2022 to 31 July 2022. An electronic form with seven closed and three open questions was e-mailed to 251 professionals with DPD testing expertise of 34 European countries. A descriptive analysis was conducted. RESULTS: We received 79 responses (31%) from 23 countries. Following publication of the EMA recommendations, 87% and 75% of the countries reported an increase in the amount of genotype and phenotype testing, respectively. Implementation of novel local guidelines was reported by 21 responders (27%). Countries reporting reimbursement of both tests increased in 2021, and only four (18%) countries reported no coverage for any testing type. In 2019, major implementation drivers were 'retrospective assessment of fluoropyrimidine-related toxicity' (39%), and in 2021, testing was driven by 'publication of guidelines' (40%). Although the major hurdles remained the same after EMA recommendations-'lack of reimbursement' (26%; 2019 versus 15%; 2021) and 'lack of recognizing the clinical relevance by medical oncologists' (25%; 2019 versus 8%; 2021)-the percentage of specialists citing these decreased. Following EMA recommendations, 25% of responders reported no hurdles at all in the adoption of the new testing practice in the clinics. CONCLUSIONS: The EMA recommendations have supported the implementation of DPD deficiency testing in Europe. Key factors for successful implementation were test reimbursement and clear clinical guidelines. Further efforts to improve the oncologists' awareness of the clinical relevance of DPD testing in clinical practice are needed.


Asunto(s)
Deficiencia de Dihidropirimidina Deshidrogenasa , Humanos , Deficiencia de Dihidropirimidina Deshidrogenasa/diagnóstico , Deficiencia de Dihidropirimidina Deshidrogenasa/genética , Deficiencia de Dihidropirimidina Deshidrogenasa/tratamiento farmacológico , Fluorouracilo/uso terapéutico , Antimetabolitos Antineoplásicos/uso terapéutico , Estudios Retrospectivos , Dihidrouracilo Deshidrogenasa (NADP)/genética , Europa (Continente)
12.
Cancer Chemother Pharmacol ; 91(5): 435-439, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36890284

RESUMEN

BACKGROUND: Fluoropyrimidines (FPs) form still nowadays the backbone of chemotherapic schemes in colorectal cancer (CRC). Inter-patient variability of the toxicity profile of FPs may be partially accounted for by variable expression of dihydropyrimidine dehydrogenase (DPD). DPD rate activity is genetically determined by its extremely polymorphic coding gene DPYD. In spite of pharmacogenetic guideline-directed-dosing of FPs based regimens treating carrier of multiple variants of DPYD gene remains still challenging. CASE PRESENTATION: We present a case of a 48-year-old Caucasian man, compound heterozygous variant carrier of the DPYD gene (HapB3 and c.2194G>A) who had a diagnosis of adenocarcinoma of the left colon and was safely treated with a pharmacogenetic-guided 25% dose reduction of the standard CAP adjuvant treatment. Compound heterozygosis may have been responsible for an earlier over exposure to CAP resulting into low-grade toxicity with an anticipated median time to toxicity of the c.2194G>A variant to the 4th vs. 6th cycles. Some haplotypes of DPYD variants may have an advantage in terms of survival compared to wild-type patients. Our patient may also have benefitted from compound heterozygosis, as shown by no evidence of disease (NED) at 6-month follow-up. CONCLUSION: Pharmacogenetic-guided dosing of DPYD intermediate metabolizer compound heterozygous HapB3 and c.2194G>A variant carries should be managed by a multidisciplinary team with a dose reduction ranging from 25 to 50% to maintain effectiveness and close clinical monitoring for early detection of ADRs.


Asunto(s)
Antimetabolitos Antineoplásicos , Fluorouracilo , Masculino , Humanos , Persona de Mediana Edad , Capecitabina , Antimetabolitos Antineoplásicos/uso terapéutico , Dihidrouracilo Deshidrogenasa (NADP)/genética , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Antimetabolitos
13.
Clin Colorectal Cancer ; 21(3): e189-e195, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35668003

RESUMEN

BACKGROUND: Adjuvant fluoropyrimidine-based chemotherapy substantially reduces recurrence and mortality after resection of stage 3 colon cancer. While standard doses of 5-fluorouracil and capecitabine are safe for most patients, the risk of severe toxicity is increased for the approximately 6% of patients with dihydropyimidine dehydrogenase (DPD) deficiency caused by pathogenic DPYD gene variants. Pre-treatment screening for pathogenic DPYD gene variants reduces severe toxicity but has not been widely adopted in the United States. METHODS: We conducted a cost-effectiveness analysis of DPYD genotyping prior to fluoropyrimidine-based adjuvant chemotherapy for stage 3 colon cancer, covering the c.1129-5923C>G (HapB3), c.1679T>G (*13), c.1905+1G>A (*2A), and c.2846A>T gene variants. We used a Markov model with a 5-year horizon, taking a United States healthcare perspective. Simulated patients with pathogenic DPYD gene variants received reduced-dose fluoropyrimidine chemotherapy. The primary outcome was the incremental cost-effectiveness ratio (ICER) for DPYD genotyping. RESULTS: Compared with no screening for DPD deficiency, DPYD genotyping increased per-patient costs by $78 and improved survival by 0.0038 quality-adjusted life years (QALYs), leading to an ICER of $20,506/QALY. In 1-way sensitivity analyses, The ICER exceeded $50,000 per QALY when the cost of the DPYD genotyping assay was greater than $286. In probabilistic sensitivity analysis using a willingness-to-pay threshold of $50,000/QALY DPYD genotyping was preferred to no screening in 96.2% of iterations. CONCLUSION: Among patients receiving adjuvant chemotherapy for stage 3 colon cancer, screening for DPD deficiency with DPYD genotyping is a cost-effective strategy for preventing infrequent but severe and sometimes fatal toxicities of fluoropyrimidine chemotherapy.


Asunto(s)
Neoplasias del Colon , Deficiencia de Dihidropirimidina Deshidrogenasa , Capecitabina/efectos adversos , Quimioterapia Adyuvante , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , Análisis Costo-Beneficio , Deficiencia de Dihidropirimidina Deshidrogenasa/tratamiento farmacológico , Deficiencia de Dihidropirimidina Deshidrogenasa/genética , Dihidrouracilo Deshidrogenasa (NADP)/genética , Fluorouracilo/efectos adversos , Genotipo , Humanos
14.
Am J Med Sci ; 363(4): 364-370, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35114182

RESUMEN

Capecitabine is an anticancer drug related to 5-fluorouracil (5-FU) that is used to treat multiple cancers. Little is known about the central nervous system toxicity of capecitabine owing to the low frequency of occurrence. In this report we describe a rare case of capecitabine-related toxic encephalopathy involving the cerebellum and subcortical white matter. A review of the literature showed that most reported cases have shown excellent recovery within a few days of capecitabine termination. Whether uridine triacetate is a reasonable treatment choice for patients with life-threatening toxic encephalopathy depends on the availability of reliable clinical data. Prescreening for dihydropyrimidine dehydrogenase genotype variants and detection of 5-FU degradation rate prior to capecitabine treatment may become an effective way to avoid toxic reactions by regulating the therapeutic dose for each patient, which remains to be investigated and needs more clinical data to support.


Asunto(s)
Síndromes de Neurotoxicidad , Sustancia Blanca , Antimetabolitos Antineoplásicos/efectos adversos , Capecitabina/efectos adversos , Cerebelo , Fluorouracilo/efectos adversos , Humanos , Síndromes de Neurotoxicidad/diagnóstico por imagen , Síndromes de Neurotoxicidad/tratamiento farmacológico , Síndromes de Neurotoxicidad/etiología , Sustancia Blanca/diagnóstico por imagen
15.
Intern Med ; 61(16): 2449-2455, 2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-35110482

RESUMEN

Dihydropyrimidine dehydrogenase (DPD) deficiency induces severe adverse events in patients receiving fluoropyrimidines. We encountered a 64-year-old DPD-deficient man with a severe capecitabine-related gastrointestinal disorder. He received capecitabine-containing chemotherapy after rectal cancer resection. During the first course of chemotherapy, he developed severe diarrhea, a fever, and hematochezia. Endoscopy revealed mucosal shedding with bleeding throughout the gastrointestinal tract. DPD deficiency was suspected because he developed many severe adverse events of capecitabine early and was finally confirmed based on the finding of a low DPD activity level in peripheral blood mononuclear cells. After one month of intensive care, hemostasis and mucosal healing were noted, although his gastrointestinal function did not improve, and he had persistent nutritional management issues.


Asunto(s)
Deficiencia de Dihidropirimidina Deshidrogenasa , Neoplasias del Recto , Antimetabolitos Antineoplásicos/efectos adversos , Capecitabina/efectos adversos , Deficiencia de Dihidropirimidina Deshidrogenasa/inducido químicamente , Deficiencia de Dihidropirimidina Deshidrogenasa/complicaciones , Deficiencia de Dihidropirimidina Deshidrogenasa/tratamiento farmacológico , Fluorouracilo/efectos adversos , Humanos , Leucocitos Mononucleares , Masculino , Persona de Mediana Edad , Neoplasias del Recto/complicaciones , Neoplasias del Recto/tratamiento farmacológico
16.
Cancer Chemother Pharmacol ; 87(5): 657-663, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33544210

RESUMEN

Capecitabine is a fluoropyrimidine that is widely used as a cancer drug for the treatment of patients with a variety of cancers. Unfortunately, early onset, severe or life-threatening toxicity is observed in 19-32% of patients treated with capecitabine and 5FU. Dihydropyrimidine dehydrogenase (DPD) is the rate-limiting enzyme in the degradation of 5FU and a DPD deficiency has been shown to be a major determinant of severe fluoropyrimidine-associated toxicity. DPD is encoded by the DPYD gene and some of the identified variants have been described to cause DPD deficiency. Preemptive screening for DPYD gene alterations enables the identification of DPD-deficient patients before administering fluoropyrimidines. In this article, we describe the application of upfront DPD screening in Finnish patients, as a part of daily clinical practice, which was based on a comprehensive DPYD gene analysis, measurements of enzyme activity and plasma uracil concentrations. Almost 8% of the patients (13 of 167 patients) presented with pathogenic DPYD variants causing DPD deficiency. The DPD deficiency in these patients was further confirmed via analysis of the DPD activity and plasma uracil levels. Interestingly, we identified a novel intragenic deletion in DPYD which includes exon 4 in four patients (31% of patients carrying a pathogenic variant). The high prevalence of the exon 4 deletion among Finnish patients highlights the importance of full-scale DPYD gene analysis. Based on the literature and our own experience, genotype preemptive screening should always be used to detect DPD-deficient patients before fluoropyrimidine therapy.


Asunto(s)
Dihidrouracilo Deshidrogenasa (NADP)/genética , Exones , Adulto , Anciano , Anciano de 80 o más Años , Deficiencia de Dihidropirimidina Deshidrogenasa/diagnóstico , Femenino , Finlandia , Eliminación de Gen , Genotipo , Humanos , Masculino , Persona de Mediana Edad
17.
Cancer Chemother Pharmacol ; 87(4): 579-583, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33587160

RESUMEN

We report the case of a 44-year-old patient who experienced severe toxicity while being treated with capecitabine at standard dose for metastatic breast cancer. As the patient had already received 5-FU within the FEC protocol (5-FU 500 mg/m2, epirubicin 100 mg/m2, and cyclophosphamide 500 mg/m2) 10 years ago without experiencing any severe adverse event, no DPD deficiency testing was performed before capecitabine treatment. Nevertheless, she experienced severe diarrhea and grade 2 hand-foot syndrome from the first cycle, forcing her to stop the treatment. Phenotypic and genotypic investigation of DPD activity revealed that the patient had a partial deficiency and had therefore been exposed to a higher risk of developing severe toxicities on fluoropyrimidines. This case proves that tolerance to low-dose fluoropyrimidines does not preclude DPD deficiency and the occurrence of severe toxicities if higher doses of fluoropyrimidines are used as a second-line treatment. It emphasizes the role of DPD phenotyping testing based on uracilemia in patients scheduled for fluoropyrimidine drugs, even if previous courses with low-dose 5-FU were safely administered.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Capecitabina/efectos adversos , Deficiencia de Dihidropirimidina Deshidrogenasa/complicaciones , Fluorouracilo/efectos adversos , Adulto , Femenino , Humanos
18.
Dose Response ; 18(3): 1559325820951367, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973417

RESUMEN

BACKGROUND: Fluoropyrimidines (FPs) carry around 20% risk of G3-5 toxicity and 0.2-1% risk of death, due to dihydropyrimidine dehydrogenase (DPD) deficiency. Several screening approaches exist for predicting toxicity, however there is ongoing debate over which method is best. This study compares 4 screening approaches. METHOD: 472 patients treated for colorectal, head-and-neck, breast, or pancreatic cancers, who had not been tested pre-treatment for FP toxicity risk, were screened using: DPYD genotyping (G); phenotyping via plasma Uracil (U); phenotyping via plasma-dihydrouracil/uracil ratio (UH2/U); and a Multi-Parametric Method (MPM) using genotype, phenotype, and epigenetic data. Performance was compared, particularly the inability to detect at-risk patients (false negatives). RESULTS: False negative rates for detecting G5 toxicity risk were 51.2%, 19.5%, 9.8% and 2.4%, for G, U, UH2/U and MPM, respectively. False negative rates for detecting G4-5 toxicity risk were 59.8%, 36.1%, 21.3% and 4.7%, respectively. MPM demonstrated significantly (p < 0.001) better prediction performance. CONCLUSION: MPM is the most effective method for limiting G4-5 toxicity. Its systematic implementation is cost-effective and significantly improves the risk-benefit ratio of FP-treatment. The use of MPM, rather than G or U testing, would avoid nearly 8,000 FP-related deaths per year globally (500 in France), and spare hundreds of thousands from G4 toxicity.

19.
Artículo en Inglés | MEDLINE | ID: mdl-32169798

RESUMEN

Fluoropyrimidines-based chemotherapies are the backbone in the treatment of many cancers. However, the use of 5-fluorouracil and its oral pre-prodrug, capecitabine, is associated with an important risk of toxicity. This toxicity is mainly due to a deficiency of dihydropyrimidine dehydrogenase (DPD). This deficiency may be detected by using a phenotypic approach that consists in the measurement of uracilemia or the calculation of dihydrouracil (UH2)/uracil (U) ratio. For uracilemia, a threshold value of 16 ng/ml has been proposed for partial deficiency, while a value of 150 ng/ml has been proposed for complete deficiency. We have developed a rapid, accurate and fully-automated procedure for the quantification of U and UH2 in plasma. Sample extraction was carried out by a programmable liquid handler directly coupled to a liquid chromatography - tandem mass spectrometry (LC-MS/MS) system. The method was validated according to the EMA guidelines and ISO 15189 requirements and was applied to real patient samples (n = 64). The limit of quantification was 5 and 10 ng/ml for U and UH2 respectively. Imprecision and inaccuracy were less than 15% for inter and intra-assay tests. Comparison with dedicated routine method showed excellent correlation. An automated procedure perfectly fulfills the need of low inaccuracy and CVs at the threshold values (less than 5% at 16 ng/ml) and is highly suitable for the characterization of DPD deficiency. Automatization should guaranty reliable and robust performances by minimizing the sources of variation such as volume inaccuracies, filtration or manual extraction related errors.


Asunto(s)
Deficiencia de Dihidropirimidina Deshidrogenasa/diagnóstico , Uracilo/sangre , Automatización , Capecitabina/metabolismo , Cromatografía Líquida de Alta Presión , Fluorouracilo/metabolismo , Humanos , Límite de Detección , Reproducción , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem , Uracilo/análogos & derivados , Uracilo/metabolismo
20.
Int J Surg Case Rep ; 56: 55-58, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30831507

RESUMEN

INTRODUCTION: 5-Fluorouracil (5-FU) is widely used for cancer treatment. The reduced activity of dihydropyrimidine dehydrogenase (DPD), the key enzyme in 5-FU inactivation, increases a patient's risk of developing severe 5-FU related toxicity. However, screening for DPD deficiency is rarely performed before 5-FU administration. PRESENTATION OF CASE: Our patient was a 69-year-old man with rectal cancer (T2N1bM0 stage IIIA) who underwent laparoscopic low anterior resection. He developed severe neutropenia and diarrhea 15 days after the administration of capecitabine for adjuvant chemotherapy, and was admitted to our hospital. Four days after admission, he was transferred to the intensive care unit for sepsis. DPD protein screening revealed DPD deficiency. On day 27, massive melena suddenly appeared. He died of continual bleeding 41 days after admission. Pathological autopsy revealed cytomegalovirus enterocolitis. DISCUSSION: The administration of 5-FU to patients with DPD deficiency is lethal. Genotypic and phenotypic assessments are reliable tests for DPD deficiency. A genetic study can effectively screen for DPD deficiency; however, its use has not been established in the national insurance system. Patients with DPD deficiency tend to develop severe neutropenia, so clinicians should pay attention to opportunistic infections such as cytomegalovirus enterocolitis. CONCLUSION: Screening for DPD deficiency is necessary prior to 5-FU administration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA