Next Article in Journal
Personalized Dosimetry in Targeted Radiation Therapy: A Look to Methods, Tools and Critical Aspects
Previous Article in Journal
Applicability of Oculomotor Tests for Predicting Central Vestibular Disorder Using Principal Component Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Implementing Pre-Therapeutic UGT1A1 Genotyping in Clinical Practice: A Real-Life Study

by
Nicola Personeni
1,2,
Laura Giordano
3,
Angelica Michelini
1,2,
Antonio D’Alessio
1,4,
Antonella Cammarota
1,2,
Silvia Bozzarelli
2,
Tiziana Pressiani
2,
Maria Giuseppina Prete
2,
Maria Teresa Sandri
5,6,
Sabine Stioui
5,7,
Luca Germagnoli
5,
Armando Santoro
1,2,
Lorenza Rimassa
1,2,* and
Rossana Mineri
5,*
1
Department of Biomedical Sciences, Humanitas University, Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
2
Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
3
Biostatistic Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
4
Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London W12 0HS, UK
5
Medical Genetics Section, Laboratory Medicine, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
6
Bianalisi Laboratory, Via Mattavelli 3, 20841 Carate Brianza, Italy
7
CDI—Genetic and Cytogenetic Laboratory, Via Saint Bon 20, 20147 Milan, Italy
*
Authors to whom correspondence should be addressed.
J. Pers. Med. 2022, 12(2), 204; https://doi.org/10.3390/jpm12020204
Submission received: 28 December 2021 / Revised: 25 January 2022 / Accepted: 27 January 2022 / Published: 2 February 2022
(This article belongs to the Section Pharmacogenetics)

Abstract

:
Current guidelines recommend pre-therapeutic UGT1A1 genotyping to guide irinotecan dosing, but the usefulness of this approach remains to be clarified. In 247 patients with advanced gastrointestinal cancers undergoing irinotecan-based chemotherapy, we prospectively performed UGT1A1*28 genotyping and we analyzed the incidence of severe neutropenia according to genotype-guided dose reductions. Overall, 28 (11.3%) and 92 (37.2%) patients were homozygous or heterozygous UGT1A1*28 carriers, respectively. Grade ≥ 3 neutropenia was reported in 39% of homozygous patients receiving an upfront dose reduction of irinotecan (median 40%, range 22–58%), in 20% of heterozygous or wild-type patients receiving full dose (ORvs*28/*28 genotype = 0.38; 95% CI: 0.14–1.03; p = 0.058), and in 15.3% of those receiving a reduced dose for clinical reasons (OR vs*28/*28 genotype = 0.28, 95% IC: 0.12–0.67; p = 0.004). Occurrence of severe neutropenia was inversely associated with dose reduction in UGT1A1*28 homozygous carriers (ORx10 unit = 0.62, 95% CI: 0.27–1.40, p = 0.249) and UGT1A1 heterozygous or wild-type patients (ORx10 unit = 0.87, 95% CI: 0.59–1.28, p = 0.478). Incidence of severe neutropenia was related to irinotecan doses and UGT1A1 polymorphisms. Upfront irinotecan dose reductions do not reduce the burden of grade ≥ 3 neutropenia in UGT1A1*28 homozygous carriers.

1. Introduction

Fluoropyrimidines (FP), irinotecan, and oxaliplatin are a mainstay in the systemic treatment of metastatic colorectal cancer [1] and other gastrointestinal malignancies including gastric, pancreatic, and biliary cancers. From 35 to 50% of patients receiving combination regimens with FP and irinotecan experience unpredictable and sometimes clinically relevant treatment-related toxicities [2]. In past years, interindividual differences in the occurrence and/or seriousness of FP- and irinotecan-related toxicities were ascribed to clinical factors, including age, sex, and performance status [3]. Irinotecan is a mildly active prodrug converted into the topoisomerase I inhibitory metabolite SN-38, eventually inactivated by the uridine diphosphate-glycosyltransferase 1 (UGT1A1) enzyme. Polymorphisms of the UGT1A1 gene occur within its promoter sequence (TATA box), and may lead to a deficient irinotecan metabolism, resulting in drug accumulation and increased risks of toxicities. In particular, the UGT1A1*28 allele is associated with a decreased gene expression and lower glucuronidation efficacy of the irinotecan metabolites. Associations between irinotecan toxicity and the UGT1A1*28/*28 genotype have been reported in cancer patients [4,5,6], suggesting a significantly higher risk of severe neutropenia [7]. Given these premises, a pre-therapeutic screening for the homozygous carriers of the UGT1A1*28 allele before the administration of irinotecan has been recommended to reduce the risk of disproportionate and sometimes fatal toxicity [8]. In addition, retrospective studies investigating the possibility to predict irinotecan toxicity with UGT1A1 genotyping have suggested that alternative UGT1A1 alleles/haplotypes could be more predictive of hematologic toxicity than UGT1A1*28 [9,10]. Additionally, more recently in patients undergoing UGT1A1 genotyping, sex, age, irinotecan dose, and treatment schedule were identified as relevant predictors of toxicity, thereby limiting the scope of UGT1A1 polymorphisms screening [11,12,13,14]. Guidelines published by the Dutch Pharmacogenomics Working Group recommend dose reductions by 30% in patients with a known UGT1A1*28 homozygous genotype, who are candidates to irinotecan therapy at doses of >150 mg/m2 [15]. Similar recommendations are found in the Italian [16] and French guidelines [17]. While most of the evidence gathered was generated on the basis of retrospective genotyping, it is still unclear whether upfront irinotecan dose reduction may mitigate irinotecan-related toxicities in individuals carrying the *28/*28 genotype. As a result, UGT1A1 genotyping has not been routinely integrated into clinical practice yet. To this end, we undertook an analysis on the safety of irinotecan dose reductions following UGT1A1 genotyping. In particular, here we explored the incidence of severe neutropenia in patients with gastrointestinal malignancies being treated in a palliative setting with irinotecan-containing chemotherapy regimens.

2. Materials and Methods

2.1. Patient Population

In this prospective study, we included consecutive patients who were treated at IRCCS Humanitas Research Hospital, between 2012 and 2019. The list of patients was extracted from an in-house web-based application for the management of the hospital medication life cycle, from its prescription, preparation, and its administration. Eligibility criteria included a histologically confirmed diagnosis of advanced or metastatic gastrointestinal malignancy; administration of at least one dose of irinotecan either as a single agent or in combination with other cytotoxic and/or biologic agents; and availability of clinical information and laboratory data. As of March 2016, UGT1A1 genotyping became a standard practice in our Institution, following the publication of national AIOM-SIF recommendations [16], which recommend the pharmacogenetic analysis of UGT1A1 before starting irinotecan therapy. This indication principally concerns patients whose clinical characteristics may lead to an increased risk–benefit ratio from irinotecan-based chemotherapy regimens. All patients carrying the homozygous UGT1A1*28 allele polymorphism underwent irinotecan dose reduction in accordance with the guidelines [16]. On the other hand, patients with UGT1A1*1/*1 (wild-type) and *1/*28 genotypes might receive full irinotecan doses, or reduced doses based on clinical grounds. Besides UGT1A1, concomitant dihydropyrimidine dehydrogenase (DPYD) genotyping was performed in all FP-naïve patients who were candidates to irinotecan in combination with FP. Specifically, the following DPYD polymorphisms were investigated: c.1905+1G>A, c.2846A>T, and c.1679T>G. Neutropenia was graded according to the National Cancer Institute Common Toxicity Criteria for Adverse Events, Version 4.3 (NCI-CTC 4.3 criteria, http://ctep.cancer.gov/reporting/ctc.html (accessed on 29 November 2021)). This study received Institutional Review Board approval (IRB No ONC-OSS-03-2020), and all patients provided written informed consent for genotyping.

2.2. DNA Extraction and Genotyping of UGT1A1 and DPYD

Three mL of blood were taken from all patients, stored in EDTA, and DNA was extracted from 200 μL using the Genomic DNA isolation kit (Roche Life Science, Penzberg, Germany) on a MagNA Pure LC 2.0 Instrument (Roche Life Science, Penzberg, Germany). The DPYD and UGT1A1 variants were analyzed through pyrosequencing. Assays of samples for candidate gene polymorphisms were performed using the CE-IVD “Irinotecan response®” and “Fluoropyrimidines response®” kit (Diatech Pharmacogenetics®, Jesi, Italy), according to the manufacturer’s instructions. DNA was first amplified through RT-PCR on a RotorGene Q (Qiagen®, Valencia, CA, USA), and reaction products were then run on a PyroMark Q96 ID (Qiagen®, Valencia, CA, USA).

2.3. Statistical Analysis

The objective of this study was to describe the association between UGT1A1 genotyping and grade ≥ 3 neutropenia or febrile neutropenia. Specifically, we compared the incidence of severe neutropenia in patients receiving irinotecan with and without dose reduction. Data were summarized as frequencies and proportions or as medians and ranges, and differences were estimated by the chi-square test (or the Fisher exact test when appropriate) and the t-test (the Wilcoxon–Mann–Whitney U test when appropriate). A logistic model was used to estimate the odds ratio (OR) and their corresponding 95% confidence intervals (CI) in univariable and multivariable analyses. The p value was reported considering all evaluations as explorative in nature. All analyses were performed using SAS version 9.4 (SAS Institute Inc., Cary, NC, USA).

3. Results

3.1. Baseline Patient Characteristics

Overall, 247 patients underwent UGT1A1 genotyping, with allele frequencies summarized in Table 1.
Additionally, 179 underwent concomitant DPYD genotyping (c.1905+1G>A, c.2846A>T and c.1679T>G) when a FP-based treatment was planned in FP-naive patients (Table 2).
This latter assessment allowed us to safely treat two patients with reduced FP doses in accordance to national guidelines [16]. All patients who were homozygous carriers of the UGT1A1*28 allele (n = 28) received an upfront median irinotecan dose reduction equal to 40% (range 22–58%). On the other hand, among 219 patients who were heterozygous carriers of either the UGT1A1*28 allele or the wild-type allele, only 56 were deemed clinically fit to receive full irinotecan doses. Their clinical characteristics are summarized in Table 3. Significant differences between the two subgroups of patients (*28/*28 vs. *1/*1 and *1/*28) were observed in terms of primary tumor location (p = 0.016) and baseline bilirubin levels (p < 0.001).
The remaining 163 heterozygous or wild-type patients who were not deemed clinically fit for full irinotecan doses received a median dose reduction equal to 29% (range 12–55%), which was justified on the basis of a poor performance status.

3.2. Factors Associated with Grade ≥ 3 Neutropenia in the Whole Cohort

Grade ≥ 3 neutropenia was detected in 47/247 patients (19.0%), including five patients developing febrile neutropenia. In univariate analyses the risk of grade ≥ 3 neutropenia was higher in female patients as compared to male patients (OR = 2.94, 95% CI: 1.96–3.85; p = 0.030). Other clinical parameters, including age, body surface area, performance status, and UGT1A1 genotyping were not significantly associated with the development of severe neutropenia.

3.3. Grade ≥ 3 Neutropenia Associated with *28/*28, *1/*1, and *1/*28 Genotypes

Eleven patients (39.2%) carrying the homozygous UGT1A1*28 allele developed grade ≥ 3 neutropenia despite upfront reduced irinotecan doses, as compared to 19.6% of heterozygous or wild-type patients treated with full doses (ORvs*28/*28 genotype = 0.38; 95% CI: 0.14–1.03; p = 0.058). Of note, in *28*28 carriers, the risk of developing grade ≥ 3 neutropenia was inversely associated on a continuous scale to the irinotecan dose reduction that had been administered (ORx10 unit = 0.62, 95% CI: 0.27–1.40, p = 0.249).

3.4. Grade ≥ 3 Neutropenia Associated with *1/*28 or *1/*1 Genotypes in Patients Receiving Reduced Doses of Irinotecan

Among 163 patients carrying the UGT1A1*1*28 or UGT1A1*1*1 genotypes treated with upfront reduced doses of irinotecan following clinical judgment, 25 (15.3%) developed grade ≥ 3 neutropenia. Compared with UGT1A1*28 homozygous patients, the rate of grade ≥ 3 neutropenia was significantly lower (OR vs*28/*28 genotype = 0.28, 95% CI: 0.12–0.67; p = 0.004). Again, the risk of developing grade ≥ 3 neutropenia was inversely associated on a continuous scale to the upfront irinotecan dose reduction (ORx10 unit = 0.87, 95% CI: 0.59–1.28, p = 0.478).

4. Discussion

The UGT1A1 enzyme activity is reduced in patients carrying the homozygous mutation in the UGT1A1 gene (*28/*28 alleles); dose reductions have therefore been recommended to prevent severe adverse events associated with irinotecan [14]. Given these premises, this study evaluated the incidence of neutropenia following pre-therapeutic irinotecan dose modifications driven by the UGT1A1 genotype. The most relevant finding is that the UGT1A1*28*28 genotype is associated with a substantial risk of grade ≥ 3 neutropenia despite an upfront irinotecan dose reduction. Although a precise dose reduction in this patient population has not been established, our practice is consistent with current guidelines suggesting the reduction of irinotecan dose in patients with the UGT1A1*28*28 genotype [15,16,17].
In addition, the median dose reduction we applied was in line with those indicated by previous studies that prospectively investigated dose reductions by nearly 30% when irinotecan was to be administered to homozygous carriers at doses comprised between 150 mg/m2 [18,19] and 180 mg/m2 [20]. Nevertheless, most of those studies investigated only a small number of patients with UGT1A1*28 homozygous polymorphisms, ranging between one and seven [18,19]. As such, these numbers do not allow any conclusions. On the other hand, with a pre-therapeutic median irinotecan dose reduction equal to 40%, we observed among 27 UGT1A1*28 homozygous carriers a 39% rate of grade ≥ 3 neutropenia. These findings are similar to those reported by Iwasa et al. who detected a 46% rate of grade ≥ 3 neutropenia in a population of East-Asian patients with homozygous genotypes (or heterozygous UGT1A1*28 and *6 haplotypes) who received a reduced starting dose of irinotecan equal to 150 mg/m2 [20]. The breakdown according to *28/*28 and the *6/*6 genotypes was not provided, although homozygosity for UGT1A1*6 has been exclusively observed in Asian populations, where the frequency of UGT1A1*6 is higher than UGT1A1*28 [21].
Previous studies revealed that in patients with a UGT1A1*28/*28 genotype, irinotecan doses ≤150 mg⁄m2 carried a risk of hematologic toxicity not statistically different from the risk reported in patients with one or two wild-type alleles [22]. In contrast, a meta-analysis [23] reported an increased risk of neutropenia not only at medium or high doses of irinotecan, but also at lower doses, in patients with a UGT1A1*28/*28 homozygous genotype.
The limited number of UGT1A1*28/*28 carriers in the current study does not allow to draw firm conclusions on irinotecan dose thresholds and risk of severe neutropenia. However, we showed that this risk is inversely related to the upfront irinotecan dose reduction. The discrepancies with the aforementioned data [22] might be due to a certain degree of clinical heterogeneity (such as cancer type, therapeutic line, combination regimens).
In the present study, the risk of grade ≥ 3 neutropenia in *28 homozygous carriers is increased when compared to patients with heterozygous (39%) or wild-type genotypes that received full dose irinotecan (20%), or reduced doses on the basis of clinical judgment (15%). Our findings are in line with a prior report from a Japanese series [18] where a non-significant increase of neutropenia incidence was also observed in patients carrying the UGT1A1*6/*28 heterozygous genotype, despite initially reduced irinotecan doses. Similar to Caucasian patients, a reduced glucuronidation is the molecular underpinning for hematological toxicities associated with this genotype. Consistently, a recent publication by Hulshof et al. again reports on higher rates of grade ≥ 3 neutropenia among *28 and *93 homozygous carriers who receive irinotecan, despite a pre-therapeutic 30% dose reduction [24].
Importantly, all patients who were FP-naïve were further genotyped for DPYD polymorphisms and this suggests that the toxicities could not be related to concomitant FP-containing treatment.
As expected, the concentration of total bilirubin was increased in patients with homozygous polymorphisms in UGT1A1 compared to the heterozygous/wild-type group.
The package insert of irinotecan approved by US Food and Drug Administration indicates that patients with total bilirubin levels between 1.0 and 2.0 mg/dL have a greater likelihood of grade 3–4 neutropenia [25]. In contrast, data for bilirubin levels > 2.0 mg/dL are lacking given that clinical trials of irinotecan excluded all patients with such baseline laboratory values. In theory, evidence for UGT1A1 genotype-guided dosing of irinotecan should be established on the basis of a randomized controlled trial on treatment outcome, such as response rate. However, such a trial is hardly feasible because of the large number of patients needed to achieve sufficient power, which would require at least 6000 patients to be prospectively screened [14].
This study has several limitations, as data on concomitant therapies were not routinely captured in the current series. Besides chemotherapy, a number of drugs often administered to cancer patients may reduce the glucuronidation process, resulting in an increased cellular exposure that eventually determines specific toxicity patterns. Whereas current knowledge on the therapeutic effects of UGT polymorphisms mainly relates to irinotecan and mycophenolate mofetil metabolism, the clinical relevance of UGT polymorphisms other than UGT1A1*28 still needs to be fully explored [26]. Furthermore, additional UGT1 variants might modulate irinotecan-induced toxicities. One of them is the rs11563250G variant allele which is linked to a significantly decreased risk of neutropenia in patients treated with irinotecan-based chemotherapy, thereby suggesting a protective effect [27].

5. Conclusions

Despite the dose reduction we applied, we observed a numerically higher rate of severe neutropenia in patients with homozygous polymorphisms in the UGT1A1 gene (*28/*28) when compared to heterozygous or wild-type genotypes (*1/*1 or *1/*28 allele carriers) with and without dose reduction. Nevertheless, albeit non-significant, our data suggest that such a risk is inversely proportional to the irinotecan dose reduction that is applied. These findings also indicate that a dose reduction does not prevent the onset of severe neutropenia in patients with gastrointestinal malignancies carrying the UGT1A1*28/*28 genotype. However, additional measures should be considered to improve the irinotecan safety in these patients, including the prophylactic use of G-CSF among those felt to yield an increased risk of febrile neutropenia, or further dose reductions, if supported by correlative pharmacokinetic data. In conclusion, our data suggest that there is a relationship between the percentage of drug reduction and neutropenia. Genetic testing is therefore indicated to identify a group of patients at a higher risk of hematological toxicity.

Author Contributions

Conceptualization N.P., R.M., M.T.S. and A.S.; data curation, N.P., A.D., A.M. and L.G. (Laura Giordano); formal analysis L.G. (Laura Giordano); investigation, T.P., S.B., M.G.P., A.C., S.S. and L.G. (Luca Germagnoli); writing—original draft preparation, N.P., L.G. (Laura Giordano), R.M. and L.R.; writing—review and editing, N.P., R.M. and L.R.; supervision, L.G. (Luca Germagnoli) and A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the guidelines of the Declaration of Helsinki and approved by the Institutional Review Board of Humanitas Research Hospital (protocol code ONC-OSS-03.2020, approved 26 May 2020).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study undergoing UGT1A1 and DPYD genotyping. For patients who had died by the time of this analysis or patients who were lost at follow-up, the need for informed consent was waived by the IRB for the purposes of this study.

Data Availability Statement

The data that support the findings of this study are available on request from the corresponding authors L.R. and R.M. The data are not publicly available due to them containing information that could compromise research participant privacy/consent.

Conflicts of Interest

N.P. reports receiving consulting fees from Amgen, Merck Serono, Servier; lectures fees from AbbVie, Gilead, Lilly, Sanofi; travel expenses from Amgen, ArQule; and institutional research funding from Basilea, Merck Serono, Servier. L.R. reports receiving consulting fees from Amgen, ArQule, AstraZeneca, Basilea, Bayer, BMS, Celgene, Eisai, Exelixis, Genenta, Hengrui, Incyte, Ipsen, IQVIA, Lilly, MSD, Nerviano Medical Sciences, Roche, Sanofi, Servier, Taiho Oncology, and Zymeworks; lecture fees from AbbVie, Amgen, Bayer, Eisai, Gilead, Incyte, Ipsen, Lilly, Merck Serono, Roche, and Sanofi; travel expenses from Ipsen; and institutional research funding from Agios, ARMO BioSciences, AstraZeneca, BeiGene, Eisai, Exelixis, Fibrogen, Incyte, Ipsen, Lilly, MSD, Nerviano Medical Sciences, Roche, and Zymeworks.

References

  1. Cremolini, C.; Schirripa, M.; Antoniotti, C.; Moretto, R.; Salvatore, L.; Masi, G.; Falcone, A.; Loupakis, F. First-line chemotherapy for mCRC—A review and evidence-based algorithm. Nat. Rev. Clin. Oncol. 2015, 12, 607–619. [Google Scholar] [CrossRef] [PubMed]
  2. Falcone, A.; Ricci, S.; Brunetti, I.; Pfanner, E.; Allegrini, G.; Barbara, C.; Crinò, L.; Benedetti, G.; Evangelista, W.; Fanchini, L.; et al. Phase III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal cancer: The Gruppo Oncologico Nord Ovest. J. Clin. Oncol. 2007, 25, 1670–1676. [Google Scholar] [PubMed]
  3. 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 1995, 75, 11–17. [Google Scholar] [CrossRef]
  4. Marcuello, E.; Altés, A.; Menoyo, A.; Del Rio, E.; Gómez-Pardo, M.; Baiget, M. UGT1A1 gene variations and irinotecan treatment in patients with metastatic colorectal cancer. Br. J. Cancer 2004, 91, 678–682. [Google Scholar] [CrossRef]
  5. Palomaki, G.E.; Bradley, L.A.; Douglas, M.P.; Kolor, K.; Dotson, W.D. Can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? An evidence-based review. Genet. Med. 2009, 11, 21–34. [Google Scholar] [CrossRef] [Green Version]
  6. Yong, W.P.; Innocenti, F.; Ratain, M.J. The role of pharmacogenetics in cancer therapeutics. Br. J. Clin. Pharmacol. 2006, 62, 35–46. [Google Scholar] [CrossRef] [Green Version]
  7. O’Dwyer, P.J.; Catalano, R.B. Uridine diphosphate glucuronosyltransferase (UGT) 1A1 and irinotecan: Practical pharmacogenomics arrives in cancer therapy. J. Clin. Oncol. 2006, 24, 4534–4538. [Google Scholar] [CrossRef]
  8. Satoh, T.; Ura, T.; Yamada, Y.; Yamazaki, K.; Tsujinaka, T.; Munakata, M.; Nishina, T.; Okamura, S.; Esaki, T.; Sasaki, Y.; et al. Genotype-directed, dose-finding study of irinotecan in cancer patients with UGT1A1*28 and/or UGT1A1*6 polymorphisms. Cancer Sci. 2011, 102, 1868–1873. [Google Scholar] [CrossRef]
  9. Lankisch, T.O.; Schulz, C.; Zwingers, T.; Erichsen, T.J.; Manns, M.P.; Heinemann, V.; Strassburg, C.P. Gilbert’s Syndrome and irinotecan toxicity: Combination with UDP-glucuronosyltransferase 1A7 variants increases risk. Cancer Epidemiol. Biomark. Prev. 2008, 17, 695–701. [Google Scholar] [CrossRef] [Green Version]
  10. Cecchin, E.; Innocenti, F.; D’Andrea, M.; Corona, G.; De Mattia, E.; Biason, P.; Buonadonna, A.; Toffoli, G. Predictive role of the UGT1A1, UGT1A7, and UGT1A9 genetic variants and their haplotypes on the outcome of metastatic colorectal cancer patients treated with fluorouracil, leucovorin, and irinotecan. J. Clin. Oncol. 2009, 27, 2457–2465. [Google Scholar] [CrossRef]
  11. Innocenti, F.; Kroetz, D.L.; Schuetz, E.; Dolan, M.E.; Ramírez, J.; Relling, M.; Chen, P.; Das, S.; Rosner, G.L.; Ratain, M.J. Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J. Clin. Oncol. 2009, 27, 2604–2614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kweekel, D.; Guchelaar, H.J.; Gelderblom, H. Clinical and pharmacogenetic factors associated with irinotecan toxicity. Cancer Treat. Rev. 2008, 34, 656–669. [Google Scholar] [CrossRef] [PubMed]
  13. Tejpar, S.; Yan, P.; Piessevaux, H.; Dietrich, D.; Brauchli, P.; Klingbiel, D.; Fiocca, R.; Delorenzi, M.; Bosman, F.; Roth, A.D. Clinical and pharmacogenetic determinants of 5-fluorouracyl/leucovorin/irinotecan toxicity: Results of the PETACC-3 trial. Eur. J. Cancer 2018, 99, 66–77. [Google Scholar] [CrossRef]
  14. Hulshof, E.C.; Deenen, M.J.; Guchelaar, H.J.; Gelderblom, H. Pre-therapeutic UGT1A1 genotyping to reduce the risk of irinotecan-induced severe toxicity: Ready for prime time. Eur. J. Cancer 2020, 141, 9–20. [Google Scholar] [CrossRef] [PubMed]
  15. KNMP-DPWG. UGT1A1: Irinotecan. 2018. Available online: https://www.g-standaard.nl/risicoanalyse/B0001694.PDF (accessed on 29 November 2021).
  16. Raccomandazioni Per Analisi Farmacogenetiche. Available online: https://www.aiom.it/wp-content/uploads/2019/10/2019_Racc-analisi-farmacogenetiche_v26.3.2020.pdf (accessed on 29 November 2021).
  17. Etienne-Grimaldi, M.C.; Boyer, J.C.; Thomas, F.; Quaranta, S.; Picard, N.; Loriot, M.A.; Narjoz, C.; Poncet, D.; Gagnieu, M.C.; Ged, C.; et al. UGT1A1 genotype and irinotecan therapy: General review and implementation in routine practice. Fundam. Clin. Pharmacol. 2015, 29, 219–237. [Google Scholar] [CrossRef] [PubMed]
  18. Fujii, H.; Yamada, Y.; Watanabe, D.; Matsuhashi, N.; Takahashi, T.; Yoshida, K.; Suzuki, A. Dose adjustment of irinotecan based on UGT1A1 polymorphisms in patients with colorectal cancer. Cancer Chemother. Pharmacol. 2019, 83, 123–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Boisdron-Celle, M.; Metges, J.P.; Capitain, O.; Adenis, A.; Raoul, J.L.; Lecomte, T.; Lam, Y.H.; Faroux, R.; Masliah, C.; Poirier, A.L.; et al. A multicenter phase II study of personalized FOLFIRI-cetuximab for safe dose intensification. Semin. Oncol. 2017, 44, 24–33. [Google Scholar] [CrossRef]
  20. Iwasa, S.; Muro, K.; Morita, S.; Park, Y.S.; Nakamura, M.; Kotaka, M.; Nishina, T.; Matsuoka, H.; Ahn, J.B.; Lee, K.W.; et al. Impact of UGT1A1 genotype on the efficacy and safety of irinotecan-based chemotherapy in metastatic colorectal cancer. Cancer Sci. 2021, 112, 4669–4678. [Google Scholar] [CrossRef] [PubMed]
  21. Marques, S.C.; Ikediobi, O.N. The clinical application of UGT1A1 pharmacogenetic testing: Gene-environment interactions. Hum. Genom. 2010, 4, 238–249. [Google Scholar] [CrossRef] [Green Version]
  22. Hu, Z.Y.; Yu, Q.; Pei, Q.; Guo, C. Dose-dependent association between UGT1A1*28 genotype and irinotecan-induced neutropenia: Low doses also increase risk. Clin. Cancer Res. 2010, 16, 3832–3842. [Google Scholar] [CrossRef] [Green Version]
  23. Hoskins, J.M.; Goldberg, R.M.; Qu, P.; Ibrahim, J.G.; McLeod, H.L. UGT1A1*28 genotype and irinotecan-induced neutropenia: Dose matters. J. Natl. Cancer Inst. 2007, 99, 1290–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Hulshof, E.C.; de With, M.; de Man, F.M.; Creemers, G.J.; Deiman, B.; Swen, J.J.; Houterman, S.; Koolen, S.; Bins, S.; Thijs, A.; et al. UGT1A1 genotype-guided dosing of irinotecan: A prospective safety and cost analysis in poor metaboliser patients. Eur. J. Cancer 2022, 162, 148–157. [Google Scholar] [CrossRef] [PubMed]
  25. FDA. Camptosar: Full Prescribing Information. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/020571s048lbl.pdf (accessed on 29 November 2021).
  26. Stingl, J.C.; Bartels, H.; Viviani, R.; Lehmann, M.L.; Brockmöller, J. Relevance of UDP-glucuronosyltransferase polymorphisms for drug dosing: A quantitative systematic review. Pharmacol. Ther. 2014, 141, 92–116. [Google Scholar] [CrossRef] [PubMed]
  27. Chen, S.; Laverdiere, I.; Tourancheau, A.; Jonker, D.; Couture, F.; Cecchin, E.; Villeneuve, L.; Harvey, M.; Court, M.H.; Innocenti, F.; et al. A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients. Pharmacogenomics J. 2015, 15, 513–520. [Google Scholar] [CrossRef]
Table 1. UGT1A1 allele frequencies in the whole cohort.
Table 1. UGT1A1 allele frequencies in the whole cohort.
UGT1A1 AlleleNumber of Patients%
UGT1A1*1/*1 (wild-type)12751.42
UGT1A1*1/*289237.25
UGT1A1*28/*282811.34
Total247100.00
Table 2. DPYD allele frequencies in the whole cohort.
Table 2. DPYD allele frequencies in the whole cohort.
DPYD AlleleNumber of Patients%
DPYD*1/*1 (wild-type)17597.77
Heterozygous DPYD*2A21.12
DPYD c.2846A>T10.56
DPYD c.496A>G + DPYD*5 [1905+1G=;1627A>G]10.56
Total179100.00
Table 3. Patient demographics according to initial irinotecan dose.
Table 3. Patient demographics according to initial irinotecan dose.
Clinical CharacteristicsUpfront Reduced Irinotecan Dose (*28/*28), N = 28Upfront Full Irinotecan Dose (*1/*1, *1/*28), N = 56p
Median age, years (range)66.5 (41.1–80.0)61.7 (36.5–80.6)0.168
Median body surface area (range)1.76 (1.46–2.15)1.81 (1.48–2.35)0.479
Female N (%)12 (42.9%)20 (35.7%)0.525
ECOG Performance Status 0.925
-015 (53.6%)27 (48.2%)
-112 (42.9%)26 (46.4%)
-21 (3.6%)3 (5.4%)
Primary tumor location N (%) 0.016
-colorectal cancer13 (46.4%)39 (69.6%)
-bilio-pancreatic cancer9 (32.1%)15 (26.8%)
-gastric cancer6 (21.4%)2 (3.6%)
Median baseline total bilirubin (mg/dL) (range)1.16 (0.3–2.0)0.6 (0.24–1.6)<0.001
Median baseline neutrophil count (range)4.25 × 103/mm3 (2.2–23.3)4.45 × 103/mm3 (1.9–14)0.755
Planned median dose of irinotecan to be administered (mg/m2; range)180 (60–200)180 (80–200)0.489
Combination therapy #23 (82.1%)51 (91.1%)0.234
ECOG: Eastern Cooperative Oncology Group; # Includes the following chemotherapy regimens: Folinic Acid-Fluorouracil-Irinotecan-Oxaliplatin (Folfirinox); Folinic Acid-Fluorouracil-Irinotecan (Folfiri); Folfiri plus aflibercept; Folfiri plus bevacizumab; Folfiri plus anti-Epidermal Growth Factor Receptor antibodies.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Personeni, N.; Giordano, L.; Michelini, A.; D’Alessio, A.; Cammarota, A.; Bozzarelli, S.; Pressiani, T.; Prete, M.G.; Sandri, M.T.; Stioui, S.; et al. Implementing Pre-Therapeutic UGT1A1 Genotyping in Clinical Practice: A Real-Life Study. J. Pers. Med. 2022, 12, 204. https://doi.org/10.3390/jpm12020204

AMA Style

Personeni N, Giordano L, Michelini A, D’Alessio A, Cammarota A, Bozzarelli S, Pressiani T, Prete MG, Sandri MT, Stioui S, et al. Implementing Pre-Therapeutic UGT1A1 Genotyping in Clinical Practice: A Real-Life Study. Journal of Personalized Medicine. 2022; 12(2):204. https://doi.org/10.3390/jpm12020204

Chicago/Turabian Style

Personeni, Nicola, Laura Giordano, Angelica Michelini, Antonio D’Alessio, Antonella Cammarota, Silvia Bozzarelli, Tiziana Pressiani, Maria Giuseppina Prete, Maria Teresa Sandri, Sabine Stioui, and et al. 2022. "Implementing Pre-Therapeutic UGT1A1 Genotyping in Clinical Practice: A Real-Life Study" Journal of Personalized Medicine 12, no. 2: 204. https://doi.org/10.3390/jpm12020204

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop