U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Pratt VM, Scott SA, Pirmohamed M, et al., editors. Medical Genetics Summaries [Internet]. Bethesda (MD): National Center for Biotechnology Information (US); 2012-.

Cover of Medical Genetics Summaries

Medical Genetics Summaries [Internet].

Show details

Irinotecan Therapy and UGT1A1 Genotype

, MD.

Author Information and Affiliations

Created: ; Last Update: April 4, 2018.

Estimated reading time: 21 minutes

Introduction

Irinotecan (brand name Camptosar) is a topoisomerase I inhibitor widely used in the treatment of cancer. It is most frequently used in combination with other drugs to treat advanced or metastatic colorectal cancer. However, irinotecan therapy is associated with a high incidence of toxicity, including severe neutropenia and diarrhea (1).

Irinotecan is converted in the body to an active metabolite known as SN-38, which is then inactivated and detoxified by a UDP-glucuronosyltransferase (UGT) enzyme encoded by the UGT1A1 gene. The UGT enzymes are responsible for glucuronidation, a process that transforms lipophilic metabolites into water-soluble metabolites that can be excreted from the body.

The risk of irinotecan toxicity increases with genetic variants associated with reduced UGT enzyme activity, such as UGT1A1*28. The presence of this variant results in reduced excretion of irinotecan metabolites, which leads to increased active irinotecan metabolites in the blood. Approximately 10% of North Americans carry 2 copies of the UGT1A1*28 allele (homozygous, UGT1A1 *28/*28), and are more likely to develop neutropenia following irinotecan therapy (1, 2, 3).

The FDA-approved drug label for irinotecan states that “when administered as a single-agent, a reduction in the starting dose by at least one level of irinotecan hydrochloride injection should be considered for patients known to be homozygous for the UGT1A1*28 allele. However, the precise dose reduction in this patient population is not known and subsequent dose modifications should be considered based on individual patient tolerance to treatment” (Table 1) (1).

The Dutch Pharmacogenetics Working Group (DPWG) of the Royal Dutch Association for the Advancement of Pharmacy (KNMP) recommends starting with 70% of the standard dose for homozygous carriers of the UGT1A1*28 allele. If the patient tolerates this initial dose, the dose can be increased guided by the neutrophil count. They state that no action is needed for heterozygous carriers of the UGT1A1*28 allele (e.g., UGT1A1 *1/*28) (Table 2) (4). In addition, the French National Network of Pharmacogenetics (RNPGx) has proposed a decision tree for guiding irinotecan prescribing based on the UGT1A1 genotype and irinotecan dose (Table 3) (5).

Table 1.

FDA (2017) Drug Label for Irinotecan. Therapeutic Recommendations based on UGT1A1 Genotype. Dosage and Administration.

GenotypeRecommendations
UGT1A1 *28/*28 When administered as a single-agent, a reduction in the starting dose by at least one level of irinotecan hydrochloride injection, USP1 should be considered for patients known to be homozygous for the UGT1A1*28 allele. However, the precise dose reduction in this patient population is not known and subsequent dose modifications should be considered based on individual patient tolerance to treatment.

Please see Therapeutic Recommendations based on Genotype for more information from the FDA. Table adapted from (1).

Table 2.

DPWG (2014) Recommendations for Irinotecan and UGT1A1 Genotype.

Phenotype / genotypeRecommendations
UGT1A1 intermediate metabolizer (IM)NO action is needed for this gene-drug interaction.
UGT1A1 poor metabolizer (PM)Start with 70% of the standard dose
If the patient tolerates this initial dose, the dose can be increased, guided by the neutrophil count.
UGT1A1 *1/*28 NO action is needed for this gene-drug interaction.
UGT1A1 *28/*28 Start with 70% of the standard dose
If the patient tolerates this initial dose, the dose can be increased, guided by the neutrophil count.

Please see Therapeutic Recommendations based on Genotype for more information from the DPWG. Table is adapted from (4).

Table 3.

RNPGx (2017) Recommendations for Irinotecan and UGT1A1 Genotype.

Dose of irinotecanRecommendation
low doses
(<180 mg/m2/week)
Presence of the UGT1A1*28 allele is not a major risk factor (little difference in risk of hematological or digestive toxicity irrespective of the genotype)
180—230 mg/m2 spaced by 2—3-week intervalsPatients who are homozygous for the UGT1A1*28 allele have a higher risk of hematological and/or digestive toxicity than patients who are heterozygous or non-carriers.
For these *28/*28 patients, a 25-30% dose reduction is recommended, especially if the patient presents other risk factors for toxicity.
Dose can be adjusted for subsequent cycles depending on the tolerance.
240 mg/m2 or higher spaced by 2—3-week intervalsHomozygous UGT1A1*28 patients have a greatly increased risk of hematological toxicity (neutropenia) compared with other genotypes, contraindicating administration at this higher dose and leading to discussion of a standard dose depending on the associated risk factors.
Administration of an intensive dose (240 mg/m2) is recommended only for *1/*1 patients, or for *1/*28 patients who have no other risk factors and who benefit from intensive surveillance.

Please see Therapeutic Recommendations based on Genotype for more information from the RNPGx. Table is adapted from (5).

Drug: Irinotecan

Irinotecan is used to treat colorectal cancer, which is the third most common cancer worldwide (6). It is often used in combination with other drugs to treat patients with advanced or metastatic colorectal cancer, when the cancer has recurred, or has progressed following initial treatment. A common irinotecan-based combination therapy is referred to as FOLFIRI (FOLinic acid [also known as leucovorin], Fluorouracil, IRInotecan).

Irinotecan is a semisynthetic derivative of the antineoplastic agent camptothecin, which derives its name from the Camptotheca tree where it was first isolated. Like camptothecin, irinotecan is an inhibitor of the nuclear enzyme, topoisomerase I. This enzyme catalyzes a number of nuclear processes; regulation of DNA supercoiling, replication, recombination, and repair.

Topoisomerase I decreases the torsional strain in the helical strands of DNA by making single strand breaks in the DNA. Single strands of DNA pass through the breaks and bind to the topoisomerase to form a cleavable complex. Once the DNA is sufficiently relaxed and the passage of strands has been completed, topoisomerase re-ligates the broken DNA strands and allows for transcription to proceed (7, 8).

Irinotecan is a pro-drug, and is converted by carboxylesterase enzymes to the active metabolite SN-38, which is 100–1000 times more potent that its parent drug, after administration by intravenous injection (9). The SN-38 metabolite exerts its cytotoxic effects by binding to the cleavable complex to form a ternary drug-topoisomerase-DNA complex. This complex is thought to prevent the re-ligation of the single strand breaks, which interrupts the moving DNA replication fork. The arrest of replication and the interaction between replication enzymes and the ternary complex introduces lethal double-stranded breaks in DNA causing irreparable DNA damage and subsequent cell apoptosis. (10, 11).

SN-38 is lipophilic, and it needs to be inactivated by undergoing phase II metabolism (glucuronidation). The resulting conjugated SN-38 glucuronide is water-soluble, and is mainly excreted through the bile, with about 30% excreted by the kidneys (12).

Irinotecan-based combination therapy has been found to be superior in overall response and survival when compared with the use of 5-fluorouracil/leucovorin therapy alone for patients with metastatic colorectal cancer (3). However, the use of irinotecan is limited by a high incidence of unpredictable and severe dose-limiting toxicity, including severe neutropenia, fever, and diarrhea (13). The rate of grade 3 or 4 adverse events is around 20-25% (14). Approximately 7% of patients who present with severe neutropenia and fever following treatment with irinotecan will die from these complications (3, 15, 16, 17, 18).

Gene: UGT1A1

The UGT enzymes (uridine diphosphate-glucuronosyltransferase, or UDP-glucuronosyltransferase) are a superfamily of enzymes that metabolize a wide range of lipophilic molecules such as bilirubin, steroids, toxins, and drugs—including irinotecan’s active metabolite, SN-38. These enzymes mediate the process of glucuronidation, which is a phase II metabolic pathway during which glucuronic acid is conjugated to specific targets to convert them to water-soluble metabolites that can then be eliminated from the body.

The UGT genes are polymorphic, and genomic processes, such as copy-number variations, variant splicing, and epigenetic factors, likely contribute to their diversity. As a result, the substrates that the UGT enzymes catalyze are particularly variable (19).

The UGT superfamily contains at least 117 enzymes divided into 4 families, of which UGT1A is a member (20). The UGT1A gene locus, located on chromosome 2q37, is complex—it encodes multiple genes and pseudogenes, and alternatively spliced isoforms also exist (21).

The UGT1A locus contains multiple alternative first coding exons, each of which has its own promoter site, enabling the transcription of 9 unique UGT1A enzymes (22). One of these transcripts is UGT1A1, which encodes UGT1A1, the bilirubin-UGT enzyme. Whereas many UGT enzymes overlap in the substrates they glucuronidate, UGT1A1 is the only enzyme that glucuronidates bilirubin (23).

Bilirubin is a yellow waste product produced during the catabolism of heme, a constituent of hemoglobin. When old or damaged red blood cells are broken down in the spleen, their hemoglobin is broken down to heme, which is then converted into bilirubin. The UGT1A1 enzyme converts this toxic, insoluble form of bilirubin (unconjugated bilirubin) to its nontoxic form (conjugated bilirubin). Because conjugated bilirubin is water-soluble, it can be dissolved in bile and eliminated with solid waste. If bilirubin is not eliminated and instead accumulates to high levels (hyperbilirubinemia), it can cause a yellowish discoloration of the skin and eyes, a condition known as jaundice.

Variants of the UGT1A1 gene that decrease UGT1A1 enzyme activity can lead to jaundice. The data suggests that one copy of *28 allele results in about a 35% decrease in transcriptional activity, and 2 copies (*28/*28, homozygous) results in about a 70% decrease (24, 25).

The jaundice may be mild, as seen in Gilbert syndrome, or severe, as seen in Crigler-Najjar syndrome. Crigler-Najjar syndrome presents in 2 forms called type 1 and type 2. Type 1 is the extremely severe form where affected individuals can die in childhood due to kernicterus (bilirubin-induced brain injury), although they may survive for longer with treatment. Type 2 is less severe; the affected individuals are less likely to develop kernicterus and most survive into adulthood.

Currently, over 135 genetic variants of UGT1A1 have been reported (23, 26). UGT1A1*1 is the wild-type allele associated with normal enzyme activity. The most common variant allele is UGT1A1*28, which is commonly found in African-Americans (0.42–0.45 allele frequency) and Caucasians (0.26–0.31), and is less common in Asian populations (0.09–0.16) (27, 28). Within Caucasian and African American populations, the UGT1A1*28 variant is a common cause of Gilbert syndrome and is also a cause of Crigler-Najjar syndrome types 1 and 2 (19, 27).

The UGT1A1*28 [(TA)7TAA] variant contains an extra thymine-adenine (TA) repeat within the TATA box promoter region (7 TA repeats compared with 6 in the wild-type allele) (29). This extra (TA) repeat decreases the rate of transcription initiation of the UGT1A1 gene, leading to decreased enzyme activity and decreased glucuronidation of bilirubin to about 30% of wild-type levels (30).

Another variant allele, UGT1A1*37 [(TA)8TAA], has 8 TA repeats at this site, and results in reduced promoter activity of the gene to levels lower than the UGT1A1*28 allele. In contrast, the UGT1A1*36 [(TA)5TAA] allele only has 5 repeats and is associated with increased promoter activity and a reduced risk of neonatal hyperbilirubinemia (a common, and typically benign condition). Both UGT1A1*36 and UGT1A1*37 occur almost exclusively in populations of African origin, with estimated allele frequencies of 0.03–0.10 and 0.02–0.07, respectively.

The UGT1A1*28 variant is also associated with drug toxicity. Approximately 10% of the North American population is homozygous for the *28 allele (*28/*28 genotype, also known as UGT1A1 7/7 genotype) and are at an increased risk of neutropenia following intravenous irinotecan therapy (28). The rate of severe neutropenia in *28/*28 homozygous patients is as high as 36%, and is strongly associated with a higher hospitalization rate (7, 31, 32).

There is less evidence to support a link between UGT1A1 genotype and irinotecan treatment-related diarrhea, and there is conflicting data on whether an individual’s UGT1A1 genotype influences their response to irinotecan therapy (8, 33).

Another variant allele, UGT1A1*6, is more prevalent in Asian populations, with an allele frequency of around 15–30% in Chinese, Korean, and Japanese populations (24, 34, 35). In this variant, there is a switch of amino acids, from a glycine to an arginine at position 71 within a coding region (p.Arg71Gly). Individuals who are homozygous for this allele have reduced UGT1A1 enzyme activity, which can cause Gilbert syndrome and prolonged neonatal jaundice (36, 37, 38, 39). This variant also appears to be an important predictor of severe toxicity to irinotecan therapy in Asian populations (35, 40, 41, 42, 43, 44, 45, 46, 47).

In addition to genetic variations in the UGT1A1 gene, several other genetic markers may influence the risk of irinotecan toxicity. These include genetic variation in the adenosine triphosphate (ATP)-binding cassette (ABC) transporter genes, ABCC1 and ABCB2 (43, 48, 49), the solute carrier (SLC) transporter genes (48, 50, 51), the transforming growth factor (TGFB) gene (52), and the xenobiotic-sensing receptor, NR1I2 (53).

The emerging data suggests that other variant alleles may have a protective effect. The newly discovered marker rs11563250 (NM_001287395.1:c.-1068A>G), located in the 3′-flanking region of UGT1A1, has a major A allele (rs11563250A) and a relatively common variant G allele (rs11563250G, found in 12% of the population). Carriers of the G allele have a lower risk of irinotecan-induced neutropenia. They also tend to have lower total plasma bilirubin levels, suggesting that this variant is associated with an enhanced capacity for glucuronidation. Evidence suggests that carriers of rs11563250G could tolerate a higher dose of irinotecan, especially if they also have the UGT1A1*1/*1 genotype (54).

Genetic Testing

The NIH’s Genetic Testing Registry provides examples of the genetic tests currently available for irinotecan response and for the UGT1A1 gene.

Genetic testing can be used to optimize irinotecan dosing. For example, the use of genotyping in selective cases may make the following patient choices possible:

• If the patient prefers aggressive treatment: genotyping might allow higher dosing for *1/*1 and *1/*28 genotypes (55, 56, 57, 58, 59).

  • If the patient prefers maximizing quality of life: genotyping might allow lower dosing for *28/*28 genotype (7, 31, 32).

Genotyping may also enable irinotecan to be added to the treatment of other gastrointestinal tumors without the risk of hematologic toxicity (60). Genotyping may also be used as part of the management of Gilbert syndrome (15).

In the USA, the common *1 and *28 UGT1A1 alleles comprise 98–99% of genotypes (61). Routine genotyping typically tests for UGT1A1 *1/*1, *1/*28, and *28/*28 genotypes (also known as 6/6, 6/7, and 7/7, respectively).

Routine screening does not rule out other UGT1A1 polymorphisms that are more common in specific populations (7). For example, the UGT1A1*6 allele is common in Asian populations, and in Japan, a reduced dose of irinotecan is recommended for individuals with UGT1A1 *6/*6, *6/*28, and *28/*28 genotypes (62). In addition, routine screening does not identify patients who are being under-dosed and could potentially tolerate a much higher dose of irinotecan.

The adoption of preemptive UGT1A1*28 genotyping to increase irinotecan safety and efficacy in clinical practice is still limited and often not covered by health insurance, despite the significant costs of treating irinotecan-related toxicities (63, 64).

Part of the reason that healthcare providers forgo testing may be because the standard dose of irinotecan used in FOLFIRI is low (180 mg/m2). A phase II trial is currently determining whether dosing irinotecan based on genotype, as part of a FOLFIRI treatment regime, is effective and safe. The standard irinotecan dose of 180 mg/m2 is being used for patients with the 28/*28 genotype, a dose of 260 mg/m2 is being used for patients with the *1/*28 genotype, and a dose of 310 mg/m2 is being used for patients with the *1/*1 genotype (65).

Therapeutic Recommendations based on Genotype

This section contains excerpted2 information on gene-based dosing recommendations. Neither this section nor other parts of this review contain the complete recommendations from the sources.

2017 Statement from the US Food and Drug Administration (FDA)

Individuals who are homozygous for the UGT1A1*28 allele (UGT1A1 7/7 genotype) are at increased risk for neutropenia following initiation of Irinotecan Hydrochloride Injection, USP treatment.

In a study of 66 patients who received single-agent Irinotecan Hydrochloride Injection, USP (350 mg/m2 once-every-3-weeks), the incidence of grade 4 neutropenia in patients homozygous for the UGT1A1*28 allele was 50%, and in patients heterozygous for this allele (UGT1A1 6/7 genotype) the incidence was 12.5%. No grade 4 neutropenia was observed in patients homozygous for the wild-type allele (UGT1A1 6/6 genotype).

When administered as a single-agent, a reduction in the starting dose by at least one level of Irinotecan Hydrochloride Injection, USP should be considered for patients known to be homozygous for the UGT1A1*28 allele. However, the precise dose reduction in this patient population is not known and subsequent dose modifications should be considered based on individual patient tolerance to treatment.

UGT1A1 Testing

A laboratory test is available to determine the UGT1A1 status of patients. Testing can detect the UGT1A1 6/6, 6/7 and 7/7 genotypes.

Please review the complete therapeutic recommendations that are located here: (1).

2017 Recommendations from the French National Network of Pharmacogenetics (RNPGx)

Interpreting Results

The RNPGx has proposed a decision tree for guiding irinotecan prescription based on the UGT1A1 genotype and the protocol’s theoretical dose:

  • for low doses (< 180 mg/m2 /week), presence of the UGT1A1*28 allele is not a major risk factor (little difference in risk of hematological or digestive toxicity irrespective of the genotype);
  • for doses in the 180—230mg/m2 spaced by 2—3-week intervals, patients who are homozygous for the UGT1A1*28 allele have a higher risk of hematological and/or digestive toxicity than patients who are heterozygous or non-carriers. For these *28/*28 patients, a 25% to 30% dose reduction is recommended, especially if the patient presents other risk factors for toxicity. Dose can be adjusted for subsequent cycles depending on the tolerance;
  • for doses of 240mg/m2 or higher spaced by 2—3 weeks intervals, homozygous UGT1A1*28 patients have a greatly increased risk of hematological toxicity (neutropenia) compared with other genotypes, contraindicating administration at this higher dose and leading to discussion of a standard dose depending on the associated risk factors. Administration of an intensive dose (240 mg/m2) is recommended only for *1/*1 patients, or for *1/*28 patients who have no other risk factors and who benefit from intensive surveillance.

[...]

The first-intention of this strategy for analysis of UGT1A1 status is to detect the *28 variant, the most common deficiency variant observed in the Caucasian population, to be performed before initiating treatment. Referring to the level of evidence classification for RNPGx recommendations detailed in the article by Picard et al. in this issue, UGT1A1 genotyping is advisable for a standard dose (180—230mg/m2) and essential for intensified dose (> 240 mg/m2).

Thus, individualized treatment can be proposed based on the UGT1A1 genotype, with either a dose reduction for *28/*28 homozygous patients, or possibly dose intensification for non-carriers of the *28 allele.

For the other UGT1A1 alleles, genotyping is performed by a limited number of laboratories and is considered a second- intention test.

Moreover, the RNPGx suggests that this analysis could be performed concomitantly with other genetic explorations for colorectal cancer patients (search for KRAS, BRAF mutations. . .) and constitutional (search for DYPD variants) in order to guarantee optimal irinotecan therapy within adequate delay for optimal hospital practices.

Please review the complete therapeutic recommendations that are located here: (5).

2014 Recommendations from the Dutch Pharmacogenetics Working Group (DPWG) of the Royal Dutch Association for the Advancement of Pharmacy (KNMP)

UGT1A1 Intermediate Metabolizers (IM)
NO action is needed for this gene-drug interaction.
This genetic variation (IM) is more common in Western populations than the wild-type (*1/*1). This means that treatment is largely geared to patients with this genetic variation. Adjustment of the treatment is therefore not useful.

UGT1A1 Poor Metabolizers (PM)
Genetic variation reduces conversion of irinotecan to inactive metabolites. This increases the risk of serious, life-threatening adverse events.
Recommendation:
1. Start with 70% of the standard dose
If the patient tolerates this initial dose, the dose can be increased, guided by the neutrophil count.

UGT1A1 *1/*28
NO action is needed for this gene-drug interaction.
This genetic variation (*1/*28) is more common in Western populations than the wild-type (*1/*1). This means that treatment is largely geared to patients with this genetic variation. Adjustment of the treatment is therefore not useful.

UGT1A1 *28/*28
Genetic variation reduces conversion of irinotecan to inactive metabolites. This increases the risk of serious, life-threatening adverse events.
Recommendation:
1. Start with 70% of the standard dose
If the patient tolerates this initial dose, the dose can be increased, guided by the neutrophil count.

Please review the complete therapeutic recommendations that are located here: (4).

Nomenclature of selected UGT1A1 variants

Common allele nameAlternative namesHGVS reference sequencedbSNP reference identifier for allele location
CodingProtein
UGT1A1*1 (TA)6TAANM_000463.2:c.-53_-52TA[7]Not applicable—variant occurs in a non-coding (TATA box promoter) region rs8175347
UGT1A1*6 211G>A
Gly71Arg
NM_000463.2:c.211G>A NP_000454.1:p.Gly71Arg rs4148323
UGT1A1*36 (TA)5TAANM_000463.2:c.-53_-52TA[6]Not applicable—variant occurs in a non-coding (TATA box promoter) region rs8175347
UGT1A1*28 (TA)7TAA NM_000463.2:c.-53_-52[8] Not applicable—variant occurs in a non-coding (TATA box promoter) region rs8175347
UGT1A1*37 (TA)8TAANM_000463.2:c.-53_-52TA[9]Not applicable—variant occurs in a non-coding (TATA box promoter) region rs8175347

UGT1A1*1 is the wild-type allele and is associated with normal enzyme activity.
Note: The UGT1A1*28 variant has 8 TA repeats, as shown by the “[8]” in the official HGVS term, “NM_000463.2:c.-53_-52[8]”. In the medical literature, the term “(TA)7TAA” is commonly used. Here, 7 of the TA repeats are shown in parentheses “(TA)7”, followed by the 8th repeat “(TAA)”.

For an overview of the haplotypes for UGT1A1, please see the PharmGKB’s Haplotype Translation Table.

Guidelines for the description and nomenclature of gene variations are available from the Human Genome Variation Society (HGVS).

Acknowledgments

The author would like to thank Ben Kong, PharmD, BCPS, Clinical Pharmacist, Oregon Health & Science University, Oregon, USA; Otito F. Iwuchukwu, RPh, PhD, Assistant Professor of Pharmaceutical Sciences, Fairleigh Dickinson University School of Pharmacy, New Jersey, USA; Man Yee Merl, PharmD, BCOP, Senior Clinical Pharmacy Specialist, Smilow Cancer Hospital at Yale-New Haven Hospital, New Haven, Connecticut, USA; Jai N. Patel, PharmD, BCOP, Chief, Pharmacology Research, and Associate Professor, Division of Hematology/Oncology, Department of Cancer Pharmacology, Levine Cancer Institute, Charlotte, North Carolina, USA; Ryouichi Tsunedomi, PhD, Assistant Professor, Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Japan; and Ji-Ye Yin, Department of Clinical Pharmacology, Xiangya Hospital, Central South University, China; for reviewing this summary.

First Edition (2015)

The author would like to thank Victoria M. Pratt, PhD, FACMG, Director, Pharmacogenomics Diagnostic Laboratory, Indiana University School of Medicine, Indiana, USA; Mia Wadelius, Senior Lecturer, Uppsala University, Uppsala, Sweden; and Wafaa M. Rashed, Head of Clinical Trials Unit, GCT Research Specialist, and IRB Coordinator, at the Children’s Cancer Hospital, Cairo, Egypt, for reviewing this summary.

Version History

To view earlier versions of this summary, please see:

Update: June 3, 2015

Created: May 27, 2015

References

1.
IRINOTECAN HYDROCHLORIDE- irinotecan hydrochloride injection [package insert]. Orlando, FL, USA: Ingenus Pharmaceuticals; 2017. Available from: https://dailymed​.nlm​.nih.gov/dailymed/drugInfo​.cfm?setid=d04f2471-3085-4fc8-a657-bb3918d48e6eu
2.
Fujita, K. and A. Sparreboom, Pharmacogenetics of irinotecan disposition and toxicity: a review. Curr Clin Pharmacol, 2010. 5(3): p. 209-17. [PubMed: 20406168]
3.
Liu, X., D. Cheng, Q. Kuang, G. Liu, et al., Association of UGT1A1*28 polymorphisms with irinotecan-induced toxicities in colorectal cancer: a meta-analysis in Caucasians. Pharmacogenomics J, 2014. 14(2): p. 120-9. [PMC free article: PMC3992871] [PubMed: 23529007]
4.
Irinotecan – UGT1A1, Netherlands, [Cited May 2017]. Available from: http://kennisbank​.knmp.nl [Access is restricted to KNMP membership.]
5.
Quaranta, S. and F. Thomas, Pharmacogenetics of anti-cancer drugs: State of the art and implementation - recommendations of the French National Network of Pharmacogenetics. Therapie, 2017. 72(2): p. 205-215. [PubMed: 28262261]
6.
Shike, M., S.J. Winawer, P.H. Greenwald, A. Bloch, et al., Primary prevention of colorectal cancer. The WHO Collaborating Centre for the Prevention of Colorectal Cancer. Bull World Health Organ, 1990. 68(3): p. 377-85. [PMC free article: PMC2393072] [PubMed: 2203551]
7.
Should UGT1A1 Genotyping Be Used to Predict Response to Irinotecan Chemotherapy? EGAPP™ Recommendation Statement. Public Health Genomics 2009 May 5, 2015; Available from: https://archive​.cdc.gov​/#/details?q=UGT1A1&start​=0&rows​=10&url=https://www​.cdc​.gov/genomics/gtesting​/egapp/recommend/ugt1a1.htm.
8.
Dias, M.M., R.A. McKinnon and M.J. Sorich, Impact of the UGT1A1*28 allele on response to irinotecan: a systematic review and meta-analysis. Pharmacogenomics, 2012. 13(8): p. 889-99. [PubMed: 22676194]
9.
Chabot, G.G., Clinical pharmacokinetics of irinotecan. Clin Pharmacokinet, 1997. 33(4): p. 245-59. [PubMed: 9342501]
10.
Pommier, Y., P. Pourquier, Y. Fan and D. Strumberg, Mechanism of action of eukaryotic DNA topoisomerase I and drugs targeted to the enzyme. Biochim Biophys Acta, 1998. 1400(1-3): p. 83-105. [PubMed: 9748515]
11.
Di Paolo, A., G. Bocci, M. Polillo, M. Del Re, et al., Pharmacokinetic and pharmacogenetic predictive markers of irinotecan activity and toxicity. Curr Drug Metab, 2011. 12(10): p. 932-43. [PubMed: 21787264]
12.
Slatter, J.G., L.J. Schaaf, J.P. Sams, K.L. Feenstra, et al., Pharmacokinetics, metabolism, and excretion of irinotecan (CPT-11) following I.V. infusion of [(14)C]CPT-11 in cancer patients. Drug Metab Dispos, 2000. 28(4): p. 423-33. [PubMed: 10725311]
13.
Hoskins, J.M., R.M. Goldberg, P. Qu, J.G. Ibrahim, et al., UGT1A1*28 genotype and irinotecan-induced neutropenia: dose matters. J Natl Cancer Inst, 2007. 99(17): p. 1290-5. [PubMed: 17728214]
14.
Tam, V.C., S. Rask, T. Koru-Sengul and S. Dhesy-Thind, Generalizability of toxicity data from oncology clinical trials to clinical practice: toxicity of irinotecan-based regimens in patients with metastatic colorectal cancer. Curr Oncol, 2009. 16(6): p. 13-20. [PMC free article: PMC2794679] [PubMed: 20016742]
15.
Douillard, J.Y., D. Cunningham, A.D. Roth, M. Navarro, et al., Irinotecan combined with fluorouracil compared with fluorouracil alone as first-line treatment for metastatic colorectal cancer: a multicentre randomised trial. Lancet, 2000. 355(9209): p. 1041-7. [PubMed: 10744089]
16.
Obradovic, M., A. Mrhar and M. Kos, Cost-effectiveness of UGT1A1 genotyping in second-line, high-dose, once every 3 weeks irinotecan monotherapy treatment of colorectal cancer. Pharmacogenomics, 2008. 9(5): p. 539-49. [PubMed: 18466101]
17.
Lankisch, T.O., C. Schulz, T. Zwingers, T.J. Erichsen, et al., Gilbert's Syndrome and irinotecan toxicity: combination with UDP-glucuronosyltransferase 1A7 variants increases risk. Cancer Epidemiol Biomarkers Prev, 2008. 17(3): p. 695-701. [PubMed: 18349289]
18.
Ratain, M.J., Irinotecan dosing: does the CPT in CPT-11 stand for "Can't Predict Toxicity"? J Clin Oncol, 2002. 20(1): p. 7-8. [PubMed: 11773147]
19.
Guillemette, C., Pharmacogenomics of human UDP-glucuronosyltransferase enzymes. Pharmacogenomics J, 2003. 3(3): p. 136-58. [PubMed: 12815363]
20.
Mackenzie, P.I., K.W. Bock, B. Burchell, C. Guillemette, et al., Nomenclature update for the mammalian UDP glycosyltransferase (UGT) gene superfamily. Pharmacogenet Genomics, 2005. 15(10): p. 677-85. [PubMed: 16141793]
21.
van Es, H.H., A. Bout, J. Liu, L. Anderson, et al., Assignment of the human UDP glucuronosyltransferase gene (UGT1A1) to chromosome region 2q37. Cytogenet Cell Genet, 1993. 63(2): p. 114-6. [PubMed: 8467709]
22.
Girard, H., E. Levesque, J. Bellemare, K. Journault, et al., Genetic diversity at the UGT1 locus is amplified by a novel 3' alternative splicing mechanism leading to nine additional UGT1A proteins that act as regulators of glucuronidation activity. Pharmacogenet Genomics, 2007. 17(12): p. 1077-89. [PubMed: 18004212]
23.
Strassburg, C.P., Pharmacogenetics of Gilbert's syndrome. Pharmacogenomics, 2008. 9(6): p. 703-15. [PubMed: 18518849]
24.
Chapter 1 - Principles of Pharmacogenomics: Pharmacokinetic, Pharmacodynamic, and Clinical Implications., Y.W. Francis Lam, L.H.C.; [Cited March 2018]. Available from: https://www​.sciencedirect​.com/science/book/9780123919182
25.
Barbarino, J.M., C.E. Haidar, T.E. Klein and R.B. Altman, PharmGKB summary: very important pharmacogene information for UGT1A1. Pharmacogenet Genomics, 2014. 24(3): p. 177-83. [PMC free article: PMC4091838] [PubMed: 24492252]
26.
UGT Official Nomenclature: UGT1A and UGT2B haplotypes and SNPs tables., [Cited March 2018]. Available from: https://www​.pharmacogenomics​.pha.ulaval.ca​/ugt-alleles-nomenclature/
27.
Beutler, E., T. Gelbart and A. Demina, Racial variability in the UDP-glucuronosyltransferase 1 (UGT1A1) promoter: a balanced polymorphism for regulation of bilirubin metabolism? Proc Natl Acad Sci U S A, 1998. 95(14): p. 8170-4. [PMC free article: PMC20948] [PubMed: 9653159]
28.
Hall, D., G. Ybazeta, G. Destro-Bisol, M.L. Petzl-Erler, et al., Variability at the uridine diphosphate glucuronosyltransferase 1A1 promoter in human populations and primates. Pharmacogenetics, 1999. 9(5): p. 591-9. [PubMed: 10591539]
29.
ClinVar: UGT1A1*28, [Cited March 20, 2018]. Available from: https://www​.ncbi.nlm​.nih.gov/clinvar/variation/12275/
30.
Bosma, P.J., J.R. Chowdhury, C. Bakker, S. Gantla, et al., The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in Gilbert's syndrome. N Engl J Med, 1995. 333(18): p. 1171-5. [PubMed: 7565971]
31.
Shulman, K., I. Cohen, O. Barnett-Griness, A. Kuten, et al., Clinical implications of UGT1A1*28 genotype testing in colorectal cancer patients. Cancer, 2011. 117(14): p. 3156-62. [PMC free article: PMC3117027] [PubMed: 21287524]
32.
Etienne-Grimaldi, M.C., J.C. Boyer, F. Thomas, S. Quaranta, et al., UGT1A1 genotype and irinotecan therapy: General review and implementation in routine practice. Fundam Clin Pharmacol, 2015. 29(3): p. 219-37. [PubMed: 25817555]
33.
EGAPP, Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Working Group. Recommendations from the EGAPP Working Group: can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Genet Med, 2009. 11(1): p. 15-20. [PMC free article: PMC2743610] [PubMed: 19125128]
34.
Akaba, K., T. Kimura, A. Sasaki, S. Tanabe, et al., Neonatal hyperbilirubinemia and a common mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene in Japanese. J Hum Genet, 1999. 44(1): p. 22-5. [PubMed: 9929972]
35.
Zhang, X., J.F. Yin, J. Zhang, S.J. Kong, et al., UGT1A1*6 polymorphisms are correlated with irinotecan-induced neutropenia: a systematic review and meta-analysis. Cancer Chemother Pharmacol, 2017. 80(1): p. 135-149. [PubMed: 28585035]
36.
Akaba, K., T. Kimura, A. Sasaki, S. Tanabe, et al., Neonatal hyperbilirubinemia and mutation of the bilirubin uridine diphosphate-glucuronosyltransferase gene: a common missense mutation among Japanese, Koreans and Chinese. Biochem Mol Biol Int, 1998. 46(1): p. 21-6. [PubMed: 9784835]
37.
Yamamoto, K., H. Sato, Y. Fujiyama, Y. Doida, et al., Contribution of two missense mutations (G71R and Y486D) of the bilirubin UDP glycosyltransferase (UGT1A1) gene to phenotypes of Gilbert's syndrome and Crigler-Najjar syndrome type II. Biochim Biophys Acta, 1998. 1406(3): p. 267-73. [PubMed: 9630669]
38.
Maruo, Y., K. Nishizawa, H. Sato, Y. Doida, et al., Association of neonatal hyperbilirubinemia with bilirubin UDP-glucuronosyltransferase polymorphism. Pediatrics, 1999. 103(6 Pt 1): p. 1224-7. [PubMed: 10353933]
39.
Boyd, M.A., P. Srasuebkul, K. Ruxrungtham, P.I. Mackenzie, et al., Relationship between hyperbilirubinaemia and UDP-glucuronosyltransferase 1A1 (UGT1A1) polymorphism in adult HIV-infected Thai patients treated with indinavir. Pharmacogenet Genomics, 2006. 16(5): p. 321-9. [PubMed: 16609363]
40.
Hazama, S., H. Mishima, R. Tsunedomi, Y. Okuyama, et al., UGT1A1*6, 1A7*3, and 1A9*22 genotypes predict severe neutropenia in FOLFIRI-treated metastatic colorectal cancer in two prospective studies in Japan. Cancer Sci, 2013. 104(12): p. 1662-9. [PMC free article: PMC7653527] [PubMed: 24033692]
41.
Atasilp, C., P. Chansriwong, E. Sirachainan, T. Reungwetwattana, et al., Correlation of UGT1A1(*)28 and (*)6 polymorphisms with irinotecan-induced neutropenia in Thai colorectal cancer patients. Drug Metab Pharmacokinet, 2016. 31(1): p. 90-4. [PubMed: 26830078]
42.
Takano, M., K. Yamamoto, T. Tabata, Y. Minegishi, et al., Impact of UGT1A1 genotype upon toxicities of combination with low-dose irinotecan plus platinum. Asia Pac J Clin Oncol, 2016. 12(2): p. 115-24. [PubMed: 26862009]
43.
Yan, L., X.F. Wang, L.M. Wei, Y.L. Nie, et al., Effects of UGT1A1*6, UGT1A1*28, and ABCB1-3435C>T polymorphisms on irinotecan induced toxicity in Chinese cancer patients. Int J Clin Pharmacol Ther, 2016. 54(3): p. 193-9. [PubMed: 26857783]
44.
Liu, X.H., J. Lu, W. Duan, Z.M. Dai, et al., Predictive Value of UGT1A1*28 Polymorphism In Irinotecan-based Chemotherapy. J Cancer, 2017. 8(4): p. 691-703. [PMC free article: PMC5370513] [PubMed: 28367249]
45.
Liu, D., J. Li, J. Gao, Y. Li, et al., Examination of multiple UGT1A and DPYD polymorphisms has limited ability to predict the toxicity and efficacy of metastatic colorectal cancer treated with irinotecan-based chemotherapy: a retrospective analysis. BMC Cancer, 2017. 17(1): p. 437. [PMC free article: PMC5480170] [PubMed: 28637434]
46.
Xu, C., X. Tang, Y. Qu, S. Keyoumu, et al., UGT1A1 gene polymorphism is associated with toxicity and clinical efficacy of irinotecan-based chemotherapy in patients with advanced colorectal cancer. Cancer Chemother Pharmacol, 2016. 78(1): p. 119-30. [PubMed: 27220761]
47.
Cui, C., C. Shu, D. Cao, Y. Yang, et al., UGT1A1*6, UGT1A7*3 and UGT1A9*1b polymorphisms are predictive markers for severe toxicity in patients with metastatic gastrointestinal cancer treated with irinotecan-based regimens. Oncol Lett, 2016. 12(5): p. 4231-4237. [PMC free article: PMC5104260] [PubMed: 27895797]
48.
Chen, S., L. Villeneuve, D. Jonker, F. Couture, et al., ABCC5 and ABCG1 polymorphisms predict irinotecan-induced severe toxicity in metastatic colorectal cancer patients. Pharmacogenet Genomics, 2015. 25(12): p. 573-83. [PubMed: 26352872]
49.
Li, M., E.L. Seiser, R.M. Baldwin, J. Ramirez, et al., ABC transporter polymorphisms are associated with irinotecan pharmacokinetics and neutropenia. Pharmacogenomics J, 2018. 18(1): p. 35-42. [PMC free article: PMC5432414] [PubMed: 27845419]
50.
Crona, D.J., J. Ramirez, W. Qiao, A.J. de Graan, et al., Clinical validity of new genetic biomarkers of irinotecan neutropenia: an independent replication study. Pharmacogenomics J, 2016. 16(1): p. 54-9. [PMC free article: PMC4729652] [PubMed: 25869015]
51.
Toshimoto, K., A. Tomaru, M. Hosokawa and Y. Sugiyama, Virtual Clinical Studies to Examine the Probability Distribution of the AUC at Target Tissues Using Physiologically-Based Pharmacokinetic Modeling: Application to Analyses of the Effect of Genetic Polymorphism of Enzymes and Transporters on Irinotecan Induced Side Effects. Pharm Res, 2017. 34(8): p. 1584-1600. [PMC free article: PMC5498655] [PubMed: 28397089]
52.
Li, J., Q. Yu, S. Fu, M. Xu, et al., A novel genetic score model of UGT1A1 and TGFB pathway as predictor of severe irinotecan-related diarrhea in metastatic colorectal cancer patients. J Cancer Res Clin Oncol, 2016. 142(7): p. 1621-8. [PubMed: 27160286]
53.
Mbatchi, L.C., J. Robert, M. Ychou, J.C. Boyer, et al., Effect of Single Nucleotide Polymorphisms in the Xenobiotic-sensing Receptors NR1I2 and NR1I3 on the Pharmacokinetics and Toxicity of Irinotecan in Colorectal Cancer Patients. Clin Pharmacokinet, 2016. 55(9): p. 1145-57. [PubMed: 27116457]
54.
Chen, S., I. Laverdiere, A. Tourancheau, D. Jonker, et al., A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients. Pharmacogenomics J, 2015. 15(6): p. 513-20. [PMC free article: PMC6174029] [PubMed: 25778466]
55.
Toffoli, G., M.R. Sharma, E. Marangon, B. Posocco, et al., Genotype-Guided Dosing Study of FOLFIRI plus Bevacizumab in Patients with Metastatic Colorectal Cancer. Clin Cancer Res, 2017. 23(4): p. 918-924. [PMC free article: PMC6777349] [PubMed: 27507617]
56.
Phelip, J.M., L. Mineur, C. De la Fouchardiere, E. Chatelut, et al., High Resectability Rate of Initially Unresectable Colorectal Liver Metastases After UGT1A1-Adapted High-Dose Irinotecan Combined with LV5FU2 and Cetuximab: A Multicenter Phase II Study (ERBIFORT). Ann Surg Oncol, 2016. 23(7): p. 2161-6. [PubMed: 26739304]
57.
Toffoli, G., E. Cecchin, G. Gasparini, M. D'Andrea, et al., Genotype-driven phase I study of irinotecan administered in combination with fluorouracil/leucovorin in patients with metastatic colorectal cancer. J Clin Oncol, 2010. 28(5): p. 866-71. [PMC free article: PMC4872310] [PubMed: 20038727]
58.
Marcuello, E., D. Paez, L. Pare, J. Salazar, et al., A genotype-directed phase I-IV dose-finding study of irinotecan in combination with fluorouracil/leucovorin as first-line treatment in advanced colorectal cancer. Br J Cancer, 2011. 105(1): p. 53-7. [PMC free article: PMC3137420] [PubMed: 21654688]
59.
Innocenti, F., R.L. Schilsky, J. Ramirez, L. Janisch, et al., Dose-finding and pharmacokinetic study to optimize the dosing of irinotecan according to the UGT1A1 genotype of patients with cancer. J Clin Oncol, 2014. 32(22): p. 2328-34. [PMC free article: PMC4105486] [PubMed: 24958824]
60.
McWilliams, R.R., N.R. Foster, M.R. Mahoney, T.C. Smyrk, et al., North Central Cancer Treatment Group N0543 (Alliance): A phase 2 trial of pharmacogenetic-based dosing of irinotecan, oxaliplatin, and capecitabine as first-line therapy for patients with advanced small bowel adenocarcinoma. Cancer, 2017. 123(18): p. 3494-3501. [PMC free article: PMC5589489] [PubMed: 28493308]
61.
Evaluation of Genomic Applications in, P. and G. Prevention Working, Recommendations from the EGAPP Working Group: can UGT1A1 genotyping reduce morbidity and mortality in patients with metastatic colorectal cancer treated with irinotecan? Genet Med, 2009. 11(1): p. 15-20. [PMC free article: PMC2743610] [PubMed: 19125128]
62.
Fujita, K., Y. Kubota, H. Ishida and Y. Sasaki, Irinotecan, a key chemotherapeutic drug for metastatic colorectal cancer. World J Gastroenterol, 2015. 21(43): p. 12234-48. [PMC free article: PMC4649109] [PubMed: 26604633]
63.
Roncato, R., E. Cecchin, M. Montico, E. De Mattia, et al., Cost Evaluation of Irinotecan-Related Toxicities Associated With the UGT1A1*28 Patient Genotype. Clin Pharmacol Ther, 2017. [PubMed: 28074472]
64.
Butzke, B., F.S. Oduncu, F. Severin, A. Pfeufer, et al., The cost-effectiveness of UGT1A1 genotyping before colorectal cancer treatment with irinotecan from the perspective of the German statutory health insurance. Acta Oncol, 2016. 55(3): p. 318-28. [PubMed: 26098842]
65.
Genotype-Directed Study Of Irinotecan Dosing In FOLFIRI + BevacizumabTreated Metastatic Colorectal Cancer, [Cited March 2018]. Available from: https://clinicaltrials.gov/show/NCT02138617

Footnotes

1

USP stands for the United States Pharmacopeia. The USP establishes standards that promote safe medication use (e.g., procurement, prescribing, transcribing, order entry, preparation, dispensing, administration, and monitoring of medications).

2

The FDA labels specific drug formulations. We have substituted the generic names for any drug labels in this excerpt. The FDA may not have labeled all formulations containing the generic drug.

Copyright Notice

All Medical Genetics Summaries content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 International (CC BY 4.0) license which permits copying, distribution, and adaptation of the work, provided the original work is properly cited and any changes from the original work are properly indicated. Any altered, transformed, or adapted form of the work may only be distributed under the same or similar license to this one.

Bookshelf ID: NBK294473PMID: 28520360

Views

Related Summaries by Gene

Tests in GTR by Condition

Tests in GTR by Gene

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...