Abstract
The clinical application of Pharmacogenomics (PGx) has improved patient safety. However, comprehensive PGx testing has not been widely adopted in clinical practice, and significant opportunities exist to further optimize PGx in cancer care. This systematic review and meta‐analysis aim to evaluate the safety outcomes of reported PGx‐guided strategies (Analysis 1) and identify well‐studied emerging pharmacogenomic variants that predict severe toxicity and symptom burden (Analysis 2) in patients with cancer. We searched MEDLINE, EMBASE, CENTRAL, clinicaltrials.gov, and International Clinical Trials Registry Platform from inception to January 2023 for clinical trials or comparative studies evaluating PGx strategies or unconfirmed pharmacogenomic variants. The primary outcomes were severe adverse events (SAE; ≥ grade 3) or symptom burden with pain and vomiting as defined by trial protocols and assessed by trial investigators. We calculated pooled overall relative risk (RR) and 95% confidence interval (95%CI) using random effects models. PROSPERO, registration number CRD42023421277. Of 6811 records screened, six studies were included for Analysis 1, 55 studies for Analysis 2. Meta‐analysis 1 (five trials, 1892 participants) showed a lower absolute incidence of SAEs with PGx‐guided strategies compared to usual therapy, 16.1% versus 34.0% (RR = 0.72, 95%CI 0.57–0.91, p = 0.006, I 2 = 34%). Meta‐analyses 2 identified nine medicine(class)‐variant pairs of interest across the TYMS, ABCB1, UGT1A1, HLA‐DRB1, and OPRM1 genes. Application of PGx significantly reduced rates of SAEs in patients with cancer. Emergent medicine‐variant pairs herald further research into the expansion and optimization of PGx to improve systemic anti‐cancer and supportive care medicine safety and efficacy.
INTRODUCTION
Clinical pharmacogenomics (PGx) plays an increasingly important role in patient safety. There is abundant evidence demonstrating that comprehensive germline PGx‐guided medicine optimization strategies reduce severe adverse events (SAEs) in general medicine patients. 1 , 2 Such strategies, supported by guidelines such as those published by the Dutch Pharmacogenetics Working Group (DPWG) 3 or Clinical Pharmacogenetics Implementation Consortium (CPIC), 4 have been associated with especially high effect size in cancer care but studies were inadequately powered to show this safety benefit. 2
Patients with cancer frequently receive high‐risk medicines for the treatment of their cancer and management of associated symptoms. Relative to community averages, they are also likely to be older and have other co‐morbidities contributing to polypharmacy (prescribed five or more medicines). Therefore, this patient group has a high frequency of adverse medicine events and symptom‐driven complaints, leading to unplanned emergency room presentations and unplanned hospital admissions. 5 As the global burden of cancer increases, 6 pre‐emptive and preventative strategies such as those offered through the promise of PGx become increasingly important to reduce patient morbidity and improve the efficiency of health care resource utilization. Although established PGx strategies deliver significantly improved outcomes, 1 , 2 there remain considerable opportunities in cancer care to build on current evidence to improve the specificity and sensitivity of germline pharmacogenomic testing and reduce further the overall toxicity and symptom burden associated with anti‐cancer and supportive care medicines. At the time of this review, there were 77 medicines associated with a CPIC/DPWG recommendation, of which six are systemic anti‐cancer therapies (irinotecan, tamoxifen and classes fluoropyrimidines, and thiopurines) and another 20 are supportive care medicines frequently initiated within a cancer care setting. 7
The primary objectives of this meta‐analysis are to (1) evaluate in patients with cancer the safety outcomes of CPIC‐ or DPWG‐endorsed PGx‐guided strategies compared to usual therapy, and (2) synthesize the evidence for other clinically relevant pharmacogenomic variants that may predict severe toxicity and symptom burden that are not included among current CPIC or DPWG recommendations. A secondary objective is to evaluate the impact of PGx‐guided strategies on radiologically confirmed objective disease response to treatment within studies.
METHODS
Data sources and search strategy
Advanced literature searches were completed in April 2023 using the following electronic databases: Medline (Ovid), Embase (Ovid), and Cochrane CENTRAL (Wiley), as well as the clinical trials registers clinicaltrials.gov and International Clinical Trials Registry Platform (ICTRP). Articles were limited to English language only with no publication year restrictions. The search strategy was developed using the PICO framework specific to each database with oversight from a medical librarian (SLa). In Medline, the search strategy consisted of a combination of exploded subject headings (MESH) and various keywords to best identify the target literature (Table S1). Subject headings applied in Medline included: “Pharmacogenetics,” “Neoplasms,” as well as “randomized controlled trials,” “clinical trials,” and “comparative studies.” The subjects were combined in their associated cluster groups with keywords where all word variations were searched. The adjacency operator was applied in some instances that linked words in proximity. Filters were applied to ensure studies included adults. The “AND” operator was then applied to combine all separate concepts and yield the final relevant publications. The search in Embase followed a similar strategy with variations according to the subject thesaurus (Emtree). In Cochrane CENTRAL, ClinicalTrials.gov and ICTRP, keyword combinations were used. All published literature until January 31, 2023 was captured including any errata.
Data extraction and quality of assessment
All publications were downloaded, and then screened using Covidence. Non‐English abstracts and abstracts grossly out of scope were excluded. Full‐text review applying inclusion/exclusion criteria was undertaken by two independent reviewers (SL and MS). Disagreement was resolved by consensus with a third author, CK. Study selection, data checking, and reporting were undertaken according to the Preferred Reporting Items for Systematic Review and Meta‐Analyses (PRISMA). 8
Eligibility criteria and study selection
Two separate analyses were performed, first to evaluate the impact of PGx strategies on safety (Analysis 1) using interventional data, and second to explore evidence for a clinically relevant association between safety outcomes and other studied variants not yet associated with PGx recommendations (Analysis 2) using observational data.
Analysis 1: eligible studies were randomized or non‐randomized controlled trials (including cluster randomized trials and secondary analyses of clinical trials) and comparative studies for comparing PGx versus usual therapy. Only studies that conformed to CPIC or DPWG recommendations for actionable medicine‐gene pairs were included, with comparisons made between equivalent groups, either variant and wild‐type carriers or variant carriers alone.
Analysis 2: Where no PGx guideline is established, eligible studies compared safety outcomes for variant carriers versus wild‐type carriers. Any clinical trials were included.
Other inclusion criteria were that participants must have received active treatment with systemic anti‐cancer therapy, radiotherapy, or surgery, participants were required to be aged ≥15 years to include adolescents and adults, at least one cohort must have utilized germline pharmacogenomic testing with genotyping performed on non‐tumor tissue (such as peripheral blood or saliva), and studies must have reported at least one of the primary outcomes; severe toxicity defined as Grade ≥3 according to the Common Terminology Criteria for Adverse Events (CTCAE) definitions and grades, or as otherwise classified within individual studies, or symptom burden defined according to validated visual analog scales such as Brief Pain Inventory (BPI) or rates of complete emesis control. Where there were multiple reports from a single trial, only a single report – one with the highest number of total cases or events relevant to the primary outcome comparison was included.
Studies were excluded if there was no full text (abstracts only/conference proceedings), if they were small with a sample size <25, the mean age of patients was <15 years, or if patients received an allogeneic transplant or other donor tissue, or had no diagnosis of cancer, or if only tumor tissue was genotyped, and if outcomes relating to severe toxicity or symptom burden were not reported. PGx discovery‐only studies where putative associations were not subsequently replicated or validated prospectively were also excluded. Reports of cancer prognostic genes (e.g., BRCA1), and tumor or somatic genes (e.g., BRAF) comparisons were excluded.
Assessment of risk of bias in included studies
Risk of bias (RoB) was assessed using validated tools applicable to the design of the study. The Risk Of Bias In Non‐randomized Studies of Interventions (ROBINS‐I) tool 9 and revised tool to assess RoB in randomized trials (RoB 2) 10 were utilized to assess the studies considered in Analysis 1 (comparison of PGx‐guided dosing vs. usual therapy for CPIC/DPWG‐endorsed medicine‐gene pair recommendations). For Analysis 2 (comparison of variant carriers vs. wild‐type carriers) the Newcastle‐Ottawa Quality Assessment Scale (NOQAS) 11 was used to assess cohort and case–control studies within clinical trials, with overall scores classified as low (8–9), moderate (7–6) or critical (≤5). NOQAS like ROBINS‐I is recommended by the Cochrane Scientific Committee for non‐randomized studies and is more relevant for non‐interventional studies. Three independent reviewers (SL, CK, JN) performed the quality assessment. Disagreement was assessed by additional reviewers (co‐authors) for consensus. Certainty assessment for findings of pooled analyses was independently conducted by two authors (SL, CK) with an additional reviewer (JN) mediating any disagreements according to the GRADE approach and summarized using GRADE Evidence Profiles. 12 Any differences in anti‐cancer therapy intensity or dose–response gradients were considered as part of the indirectness assessments for GRADE.
Outcome measures
Two primary analyses were conducted: PGx versus Usual Therapy (Analysis 1), and variant carriers versus wild‐type carriers (Analysis 2). The two primary outcomes were the proportion of patients with severe adverse events (SAEs; Analysis 1 and Analysis 2) and symptom burden assessments (Analysis 2) occurring during study follow‐up. The proportion of study participants who demonstrated an at‐risk genotype was calculated if all studies within the subset analysis confirmed conformance with Hardy–Weinberg equilibrium. For studies that reported more events than patients, an event rate per patient years was calculated based on the duration of patient follow‐up reported for the study. Sensitivity analyses were conducted to determine any impact of this variation on calculated risk ratios. For symptom burden, brief pain inventory scores were used for pain, and rates of complete emesis control were used for vomiting. A secondary analysis was conducted for studies that did not provide adequate information for case or event rate data to be aggregated. A secondary outcome measure for Analysis 1 was the objective disease response rate based on the Response Evaluation Criteria in Solid Tumors (RECIST) Version 1.1 in studies reporting safety.
Statistical analysis
Descriptive statistics relating to outcomes of toxicity or symptom burden for variant carriers versus wild‐type carriers and outcomes of toxicity and overall survival for PGx versus Usual therapy were summarized. Continuous variables were described as mean and standard deviation; dichotomous variables were described as counts and percentages. Meta‐analyses were performed for outcome comparisons between (a) PGx versus Usual Therapy and (b) variant carriers versus wild‐type carriers. Data was pooled and analyzed using Review Manager 5.4 with random effects methods applied to ensure conservative estimates of statistical significance given the expected heterogeneity between studies. Effect measures for all outcomes were reported as Risk Ratio (RR) with 95% confidence intervals (95% CI). Heterogeneity was quantified using the homogeneity test (χ 2 with k‐1 df; p = 0.1 for significance) and I 2statistic. Due to the small number of trials, and expected clinical heterogeneity based on the patient cohorts, the I 2 statistic has been emphasized.
RESULTS
Studies identification and characteristics
The search strategy identified 6811 studies from EMBASE (n = 2439), Cochrane Library (n = 1508), and MEDLINE (n = 2243) (Figure 1). A total of 1676 duplicates were excluded. A further 4585 studies were excluded as irrelevant based on a review of the title and abstract. Following full‐text review, a further 438 studies were excluded due to different outcome measures (n = 164), other study design (n = 57), somatic genes or prognostic genes (n = 52), non‐full‐text (n = 32), non‐implementation studies for CPIC/DPWG‐endorsed PGx‐guided strategies (n = 26), other intervention (n = 24) and discovery only associations without validation or replication (n = 24), sample size <25 (n = 10). Two studies were excluded from Analysis 1 as these reported results from another included trial. 13 , 14
FIGURE 1.
PRISMA flow diagram. Preferred reporting items for systematic reviews and meta‐analyses (PRISMA) flow diagram. CPIC, clinical pharmacogenetics implementation consortium; DPWG, Dutch pharmacogenetics working group; FP, fluoropyrimidines (e.g., 5‐fluorouracil, capecitabine); 5HT3A, 5HT3‐antagonists (e.g., ondansetron, granisetron, palonosetron); MTX, methotrexate; R‐CHOP = rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone.
The research strategy and application of inclusion criteria yielded 73 eligible studies; six studies for Analysis 1 and 40 studies as subset analyses for Analysis 2. There were another 15 studies for secondary analysis for Analysis 2 where only RR was reported and authors did not provide adequate information to the calculate number of events and patients. The remaining 12 studies were single studies evaluating separate medicine‐gene variant pairs (see Figure 1).
Analysis 1 – PGx (guideline established) versus usual therapy
Of the six studies included in Analysis 1, three were randomized studies 2 , 15 , 16 and three were non‐randomized studies with either concurrent 17 or historical controls. 18 , 19 Characteristics of the 5 eligible clinical trials for which data could be aggregated are summarized in Table 1. The sixth study was evaluated within the secondary analysis (Table S5). 2 Three studies evaluated PGx‐guided dose reduction of fluoropyrimidines to reduce toxicity, two studies evaluated PGx‐guided dose escalation of irinotecan to increase efficacy and reported on toxicities, and one randomized cross‐over cluster trial evaluated optimization of medicine or dose selection for multiple medicines, both anti‐cancer therapies and supportive care medicines, within an oncology cluster to reduce toxicity. There were no other studies that specifically evaluated PGx interventions compared to usual therapy for supportive care medicines such as systemic analgesic opioids or antiemetics.
TABLE 1.
Characteristics of studies included in meta‐analysis for Analysis 1: PGx versus usual therapy.
First author's last name, Year | Study design | Total N recruited | Cancer diagnosis | Summary of PGx intervention and goal | Age range | Geographical regions (countries) |
---|---|---|---|---|---|---|
Deenen, 2016 a NCT00838370 | Prospective cohort + historical comparator | 66 | Gastrointestinal, gynecological, and other cancers | 1 CPIC/DPWG‐endorsed DPYD variant guided dose reduction or avoidance of fluoropyrimidines to reduce toxicity | 44–76 | Europe (Netherlands only) |
Henricks, 2018 NCT02324452 | Prospective cohort + historical comparator | 424 | Breast, gastrointestinal, and other cancers | 4 CPIC/DPWG‐endorsed DPYD variant guided dose reduction or avoidance of fluoropyrimidines to reduce toxicity | 21–91 | Europe (Netherlands only) |
Boisdron‐Celle, 2017 NCT01547923 | Prospective cohort + comparator | 1116 | Locally advanced and metastatic colorectal cancer | 4 CPIC/DPWG‐endorsed DPYD variant guided dose reduction or avoidance of fluoropyrimidines to reduce toxicity | 24–88 | Europe (France only) |
Tsai, 2020 NCT02256800 | Randomized controlled, parallel arm trial | 213 | Metastatic colorectal cancer | 1 CPIC/DPWG‐endorsed UGT1A1 variant guided dose escalation of irinotecan to improve treatment efficacy | 25–83 | East Asia (Taiwan only) |
Paez, 2019 NCT01639326 | Randomized controlled, parallel arm trial | 82 | Metastatic colorectal cancer | 1 CPIC/DPWG‐endorsed UGT1A1 variant guided dose escalation of irinotecan to improve treatment efficacy | 40–77 | Europe (Spain only) |
Lunenburg 2018 and Henricks 2019 excluded given the same trial data used and outcomes collected retrospectively.
Analysis 1: Risk of bias for included studies
In general, the quality of included studies was moderate to high, as summarized in Table S2. Authors recognized bias associated with open‐label design of all trials and use of historical controls within some trials with endpoints mostly assessed and graded by unblinded investigators. However, review authors considered it unlikely that primary endpoints of SAEs would be either under‐ or over‐reported in any arm. Consequently, lack of blinding was deemed to be associated with low RoB. RoB scores were allocated for each domain and aggregated as per methods. The overall evidence was qualified as moderate using GRADE (Table 3).
TABLE 3.
Grade Evidence Profile: (Analysis 1, Outcome 1) severe adverse events and (Analysis 1, Outcome 2) treatment efficacy for PGx compared to Usual Therapy, and (Analysis 2, Outcomes 3–11) severe adverse events for emergent pharmacogenomic variant allele carriers compared to wild‐type (WT) carriers in patients receiving treatment for cancer.
Quality assessment | Number of events/patients (%) | Effect | Certainty of the evidence (GRADE) | Importance | ||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
№ of studies | Study design | Risk of bias | Inconsistency | Indirectness | Imprecision | Other considerations | PGx or Variant | Usual Therapy or WT | Relative (95% CI) | Absolute | ||
Outcome 1: CPGx reduces severe adverse events compared to usual therapy | ||||||||||||
5 | Clinical trials | Moderate a | Not serious b | Not serious c | Not serious d | Consistent association (with secondary analysis study) e | 156/968 (16.1%) | 314/924 (34.0%) | RR 0.72 (0.57 to 0.91) | 179 fewer events per 1000 patients | ⨁⨁⨁◯ Moderate | Critical |
Outcome 2: CPGx improves irinotecan efficacy compared to usual therapy | ||||||||||||
2 | Clinical trials | Low f | Not serious g | Serious h | Not serious i | None | 104/147 (70.7%) | 61/145 (42.1%) | OR 3.33 (2.05–5.41) | 286 more objective responses per 1000 patients | ⨁⨁◯◯ Low | Limited importance |
Outcome 3: TYMS 5′UTR 28base pair repeat (rs45445694) homozygous and heterozygous carriers are predisposed to increased risk of fluoropyrimidine‐associated severe adverse events. | ||||||||||||
5 | Observational studies within clinical trials | Moderate j | Not serious k | Not serious c | Not serious l | Understated effect m | 326/711 (45.9%) | 189/865 (21.8%) | RR 1.37 (1.15–1.63) | 241 more events per 1000 patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 4: ABCB1 3435C>T (rs1045642) homozygous carriers are predisposed to increased risk of irinotecan‐associated severe adverse events | ||||||||||||
3 | Observational studies within clinical trials | Low n | Not serious o | Not serious c | Not serious p | Strong association q | 33/66 (50.0%) | 12/67 (17.9%) | RR 2.70 (1.56, 4.67) | 321 more per 1000 patients | ⨁⨁⨁⨁ High | Important |
Outcome 5: UGT1A1*93 (rs10929302) homozygous and heterozygous carriers are predisposed to increased risk of irinotecan‐associated severe adverse events | ||||||||||||
3 | Observational studies within clinical trials | Moderate r | Not serious s | Not serious c | Not serious p | None | 29/49 (59.2%) | 137/498 (27.5%) | RR 2.08 (1.31, 3.31) | 317 more events per 1000 patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 6: UGT1A1*28 (rs3064744) homozygous carriers are predisposed to an increased risk of severe adverse events from treatment with sacituzumab govitecan | ||||||||||||
2 | Observational studies within clinical trials | Moderate r | Not serious | Not serious | Somewhat t | Moderate association u | 85/97 (87.6%) | 185/347 (53.3%) | RR 1.65 (1.46, 1.87) | 343 more events per 1000patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 7: HLA‐DRB1*07:01 homozygous and heterozygous carriers are predisposed to increased risk of lapatinib‐associated severe adverse events | ||||||||||||
2 | Observational studies within clinical trials | Moderate r | Not serious | Not serious | Not serious | Strong association v | 49/433 (11.3%) | 16/1481 (1.1%) | RR 10.36 (5.94, 18.06) | 102 more events per 1000patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 8: UGT1A1*28 homozygous carriers are predisposed to increased risk of nilotinib‐associated severe adverse events | ||||||||||||
2 | Observational studies within clinical trials | Low | Not serious | Not serious | Somewhat w | Strong association x | 7/18 (38.9%) | 4/49 (8.2%) | RR 4.50 (1.59, 12.74) | 307 more events per 1000 patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 9: ABCB1 c1236T>C (rs1045642) homozygous and heterozygous carriers are predisposed to an increased risk of palbociclib‐associated severe adverse events | ||||||||||||
1 | Observational studies within clinical trials | Low r | Not serious | Not serious c | Not serious | None | 58/152 (38.2%) | 45/195 (23.1%) | RR 1.65 (1.19, 2.29) | 151 more events per 1000 patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 10: ABCB1 1199G>A rs2229109 homozygous and heterozygous carriers are predisposed to an increased risk of severe adverse events from R‐CHOP/AVCBP like chemotherapy regimens | ||||||||||||
1 | Observational studies within clinical trials | Low r | Not serious s | Not serious c | Somewhat y | Strong association z | 13/57 (22.8%) | 45/676 (6.7%) | RR 3.53 (2.03, 6.15) | 161 more events per 1000patients | ⨁⨁⨁◯ Moderate | Important |
Outcome 11: OPRM1 118 A>G homozygous and heterozygous carriers are predisposed to greater pain following the commencement of opioid analgesics | ||||||||||||
4 | Observational studies | Serious aa | Serious ab | Not serious | Not serious | None | N/A | N/A | Mean Difference 1.61 (0.77, 2.45) | N/A | ⨁⨁◯◯ Low | Important |
Abbreviations: CI, confidence interval; OR, odds ratio; RR, risk ratio.
RoB: three studies scored moderate and three studies scored low‐risk in the RoB Assessment.
Inconsistency: I 2 = 34%, 95% Cl of all five individual studies are overlapping; heterogeneity of studies explained with differing aims (treatment efficacy vs. toxicity) and sensitivity analysis conducted.
Indirectness: The evidence directly answers the health care question asked.
Imprecision: The 95% Cl does not cross one, and criteria for optimal information size is met, that is, total number of patients included is more than the number of patients generated by a conventional sample size calculation for a single adequately powered trial.
Other considerations include publication bias, large effect, plausible confounding, and dose‐response gradient. Publication bias has no impact. Effect is consistent across participants with unfavorable genotype.
RoB: Two studies recorded low risk in the RoB assessment.
Inconsistency: p‐value <0.0001 and I 2 = 0%, 95% Cl of both studies are overlapping.
Indirectness: Objective response is a surrogate marker of treatment efficacy; overall survival results are conflicting. High‐dose irinotecan is not routinely used in clinical practice.
Imprecision: The 95% Cl does not cross one, and criteria for optimal information size is met.
RoB: Three studies were low risk; two studies were moderate risk in the RoB assessment.
Inconsistency: p‐value <0.01 and I 2 = 26%, 95% Cl of all five individual studies are overlapping.
Imprecision: the 95% Cl does not cross one and criteria for optimal information size is met.
Other considerations include plausible confounding as two studies that showed lower effect size included heterozygous variant carriers in the control arm.
RoB: two studies recorded as low‐risk, one study recorded as moderate risk in the RoB assessment.
Inconsistency: p‐value <0.001; I 2 = 0%. 95% Cl of all three individual studies are overlapping. Although sample size is less than 200, tau square is zero indicating low variability.
Imprecision: The 95% Cl does not cross one, and criteria for optimal information size is met.
Other considerations include large effect assessed as RR = 2.7 and strength of effects associated with homozygosity. This results in certainty of evidence being upgraded by one level. Despite small sample size (less than 200 patients), funnel plot analysis suggests no publication bias.
RoB: all studies recorded as moderate‐risk in the RoB assessment.
Inconsistency: p‐value <0.0001; I 2 = 0%; 95% Cl of individual studies do not cross 1 and both are overlapping.
Imprecision: The 95% Cl does not cross one. Although the total number of patients included in the systematic review is more than the number of patients generated by a conventional sample size calculation for a single adequately powered trial, the variant carrier arm in the systematic review has less patients than would be calculated for single trial (97 vs. 123). This results in certainty of evidence being downgraded by one level.
Other considerations. RR = 1.65 (RR between 0.5 and 2.0) showing the effect was moderate.
Other considerations include large effect assessed as RR = 10.36. This results in certainty of evidence being upgraded by one level.
Imprecision: The 95% Cl does not cross one. Although total number of patients included in the systematic review is more than the number of patients generated by a conventional sample size calculation for a single adequately powered trial, the variant carrier arm in the systematic review has less patients than would be calculated for a single trial (18 vs. 29). This results in certainty of evidence being downgraded by one level.
Other considerations include large effect assessed as RR = 4.5. This results in certainty of evidence being upgraded by one level.
Imprecision: The 95% Cl does not cross one. Although the total number of patients included in the systematic review is more than the number of patients generated by a conventional sample size calculation for a single adequately powered trial, the variant carrier arm in the systematic review has less patients than would be calculated for a single trial (57 vs. 75). This results in certainty of evidence being downgraded by one level.
Other considerations include large effect assessed as RR = 3.4. This results in certainty of evidence being upgraded by one level.
All studies were moderate to high risk in the RoB assessment as these were mostly non‐clinical trial datasets.
Inconsistency: I 2 = 91%, 95% Cl of all individual studies do not overlap.
Analysis 1: Effect of intervention on severe adverse events
The meta‐analysis (5 trials, 1892 participants) found lower absolute incidence of SAEs in patients treated with PGx‐guided strategies compared to usual therapy, 16.1% versus 34.0% (RR = 0.72, 95%CI 0.57–0.91, Z = 2.76, I 2 = 34%, p = 0.006), Figure 2. Studies were considered homogenous with I 2 < 40%. In secondary analysis, the study by Swen et al. showed a strong effect size for the Italy cohort which comprised of oncology patients; OR = 0.24 (95%CI 0.06–1.01) but this study was underpowered and did not achieve statistical significance. 2
FIGURE 2.
Forest plot for Analysis 1: Severe adverse events during the study period for Pharmacogenomics (PGx) guided strategy versus usual therapy.
Three studies reported deaths, with lower incidences of treatment‐related death among DPYD variant allele carriers in the PGx arm compared to Usual Therapy arm (0% vs. 2.5% 17 ; 0% vs. 10% 18 ; 0% vs. 8% 19 ).
Analysis 1; secondary outcome: Effect of intervention on cancer treatment efficacy
Only two of the six studies reporting on safety outcomes also evaluated efficacy outcomes (total 292 participants). The objective disease response rate in the PGx arm versus Usual Therapy arm was 71% versus 42% (RR = 3.33, 95%CI 2.05–5.41, I 2 = 0%) indicating a strong association for higher treatment response to irinotecan with UGT1A1‐guided dose escalation strategies. One study showed an overall survival benefit (median 30.0 vs. 22.0 months; p = 0.02) while the other did not demonstrate a statistically significant benefit (26 vs. 17.6 months, p = 0.74).
Analysis 2 (no PGx guideline established): Severe adverse events or symptom burden
Overall, 67 studies reporting SAEs assessed 21 pharmacogenes and 19 variants that are not yet associated with CPIC/DPWG recommendations for medicine optimization. Data from 40 studies were pooled for primary Analysis 2 to evaluate 21 medicine‐gene variant pairs. Twenty‐three studies (6781 patients) were conducted in countries with large European‐ancestry populations; 4 studies (6485 patients) included mixed populations across countries with representation from European, Asian, and some Latin American and African ancestries; 10 studies (819 patients) included patients with East Asian ancestry only, and three studies (398 patients) included patients with Latin American ancestry (182 patients) or Middle Eastern ancestry (216 patients) only. The most studied disease streams were gastrointestinal cancers (19 studies, 4533 patients); breast cancers (8 studies, 8107 patients); mixed solid tumors (7 studies, 1237 patients); and hematological cancers (3 studies, 905 patients). Characteristics of the included clinical trials for which data could be aggregated are summarized in Table 2, and severe adverse events for these studies are summarized in Table S4. The most frequently studied genes were MTHFR (n = 7) for fluoropyrimidines and methotrexate, UGT1A1 (n = 8) for irinotecan (variants other than UGT1A1*28 and UGT1A1*6), nilotinib, sacituzumab govitecan, and etoposide, ABCB1 (n = 9) for irinotecan, palbociclib, and anthracycline‐vinca alkaloid‐cyclophosphamide based therapies, TYMS (n = 5) for fluoropyrimidines, UGT1A6/UGT1A7/UGT1A9 (n = 5) for irinotecan, and HLA‐DRB1 (n = 2) for lapatinib. Ten studies reported on symptom burden changes for pain or vomiting assessed six pharmacogenes: ABCB1, ABCG2, COMT, OPRM1, CYP3A4, and SLC6A4.
TABLE 2.
Characteristics of studies included in meta‐analysis for Analysis 2: SNP carriers versus wild‐type carriers (2 or more studies for individual medicine: pharmacogenomic variant pairs).
First author last name, Year; Trial no. of original clinical study (if known) | Study design | Total N | Medicine Class: Gene pairs of interest | SNPs evaluated (rsID) | Tissue type genotyped | Cancer diagnosis | Treatment regimens | Age (range or mean +/− SD) | Geographical Regions (countries) | Hardy‐Weinberg equilibrium reported and in equilibrium? |
---|---|---|---|---|---|---|---|---|---|---|
Boige, 2010; NCT00126256 | Cohort study from Phase III RCT | 346 |
Fluoropyrimidines: TYMS, MTHFR, DPYD Irinotecan: UGT1A1 |
TYMS 3′UTR 9 bp‐indel (rs11280056) TYMS 5′UTR 28 bp‐repeat (rs45445694) MTHFR c.1298A>C (rs1801131) MTHFR c.677C>T (rs1801133) UGT1A1*93; c.3156G>A (rs10929302) |
Peripheral blood | Metastatic colorectal cancer | LV5FU2, FOLFOX6, FOLFIRI | 34–83 | UK/Europe (France) | Yes |
Carlini, 2005 | Cohort study from Phase II single‐arm trial | 66 | Irinotecan: UGT1A9 | UGT1A9*22; 118(T)9 > 10 (rs3832043) | Peripheral blood | Metastatic colorectal cancer | Capecitabine/Irinotecan | 40–81 | North America (US) | Yes |
Cote, 2007 | Cohort study from Phase III trial | 93 |
Irinotecan: UGT1A1 Irinotecan: ABCB1 |
UGT1A1*93; c.3156G>A (rs10929302) ABCB1/rs1045642 (3435C>T) |
Resected non‐tumor tissue | Stage III colorectal cancer | LV5FU2‐irinotecan | N/A | UK/Europe (France) | Yes |
Castro‐rojas, 2017 | Single‐arm clinical trial | 63 | Fluoropyrimidines: TYMS |
TYMS 5′UTR 28 bp‐repeat (rs45445694) TYMS 3′UTR 9 bp‐indel (rs11280056); not reported |
Peripheral blood | Stage III‐IV colorectal cancer | FOLFOX, XELOX | N/A | Latin America (Mexico) | Yes |
Glimelius, 2011 | Cohort study from RCT | 140 |
Fluoropyrimidines: MTHFR Irinotecan: ABCB1 |
MTHFR c.677C>T rs1801133 ABCB1/rs1045642 (3435C>T) |
Resected non‐tumor tissue | Stage IV colorectal cancer | FLIRI, LV5FU2‐irinotecan | 42–76 | UK/Europe (Sweden/Europe) | Yes |
Hazama, 2013 UMIN000002388 UMIN000002476 |
Cohort study from two Phase II RCTs | 75 |
Irinotecan: UGT1A1 Irinotecan: UGT1A9 |
UGT1A1*60 UGT1A1*93 UGT1A9*1b UGT1A7*3 |
Peripheral blood | Metatstatic or refractory colorectal cancer | Modified FOLFIRI | N/A | East Asia | Yes |
Han, 2009 a | Cohort study from two Phase II RCTs | 107 |
Irinotecan: ABCB1 Irinotecan: UGT1A9 Irinotecan: ABCC2 Irinotecan: SLCO1B1 Irinotecan: CYP3A5 |
ABCB1 3435C>T (rs1045642) UGT1A9*22; 118(T)9 > 10 (rs3832043) ABCC2 − 24C>T (rs717620), ABCC2 – 1249G>A (rs2273697), and 3972C>T (rs3740066), SLCO1B1 − 11187G>A, 388A > G (rs2306283), and 521T > C (rs4149056), CYP3A5 22893G>A (*3, rs776746) |
Peripheral blood | Stage IIIb‐IV non‐small lung cancer | Irinotecan/cisplatin | 29–76 | East Asia | Yes |
Jennings, 2013 | Single‐arm clinical trial | 254 | Fluoropyrimidines: MTHFR Fluoropyrimidines: TYMS |
MTHFR c.1298A>C (rs1801131) MTHFR c.677C>T (rs1801133) TYMS 5′UTR 28 bp‐repeat (rs45445694) |
Peripheral blood | Colorectal cancer | 5‐FU or capecitabine monotherapy or FOLFIRI, CAPIRI, FOLFOX, CAPOX | 23–88 | UK/Europe | Yes |
Kim, 2013 |
Phase II single‐arm clinical trial | 43 |
Irinotecan: UGT1A6 Irinotecan: UGT1A7 |
UGT1A6*2a UGT1A7*3 |
Peripheral blood | Previously untreated metastatic colorectal cancer | TIROX | 30–67 | East Asia (Korea) | Not reported |
Levi, 2017 |
Single‐arm clinical trial | 52 | Irinotecan: ABCB1 | ABCB1 3435C>T (rs1045642) | Peripheral blood | Colorectal cancer with unresectable liver metastases | Hepatic artery infusion of irinotecan, 5‐fluorouracil and oxaliplatin and IV cetuximab | 33–76 | UK/Europe (multiple) | Yes |
Martinez‐Balibrea, 2010 | Cohort study of Phase III RCT | 149 |
Irinotecan: UGT1A9 Irinotecan: UGT1A7 |
UGT1A9*22; 118(T)9 > 10 (rs3832043) UGT1A7*3 |
Peripheral blood | Metastatic colorectal cancer | FUIRI; FOLFIRI | N/A | UK/Europe | Yes |
McLeod, 2010 |
Cohort study of Phase III RCT | 520 |
Irinotecan: UGT1A1 Irinotecan: ABCB1 |
UGT1A1*93; c.3156G>A (rs10929302) ABCB1; 3435C>T (rs1045642) |
Peripheral blood | Metastatic colorectal cancer | IFL (fluorouracil ± irinotecan), FOLFOX, and IROX | 26–85 | North America (US) | Yes |
Park, 2011 | Phase II single‐arm clinical trial | 44 |
Irinotecan: UGT1A6 Irinotecan: UGT1A7 |
UGT1A6*2 UGT1A7*3 |
Peripheral blood | Metastatic gastric cancer | TIROX | 27–66 | East Asia | Yes |
Ruzzo, 2017 |
Cohort study of Phase III RCT | 534 | Fluoropyrimidines: DPYD |
DPYD*6; p.V732I; c.2194G>A (rs1801160) DPYD; c.496 A > G (rs2297595) |
Peripheral blood |
Surgically resected stage III and high‐risk stage II colorectal cancer |
FOLFOX‐4 or XELOX adjuvant chemotherapy | 57–70 (IQR) | UK/Europe (Italy) | Yes |
Schwab, 2008 | Single‐arm clinical trial | 683 | Fluoropyrimidines: MTHFR Fluoropyrimidines: TYMS |
MTHFR c.1298A>C (rs1801131) MTHFR c.677C>T (rs1801133) TYMS 5′UTR 28 bp‐repeat (rs45445694) |
Peripheral blood | Gastrointestinal cancers, cancer of unknown primary and breast cancer | Varied 5FU regimens | N/A | UK/Europe |
Yes (MTHFR) Moderate deviation for TYMS |
Seo, 2009 | Single‐arm clinical trial | 94 | Fluoropyrimidines: TYMS | TYMS 5′UTR 28 bp‐repeat (rs45445694) | Peripheral blood | Metastatic or relapsed gastric cancer | Modified FOLFOX, Modified FOLFIRI | N/A | East Asia (Korea) | Not reported |
Smyth, 2017 | Cohort study of Phase III RCT | 124 | Fluoropyrimidines: TYMS | TYMS 5′UTR 28 bp‐repeat (rs45445694) | Resected non‐tumor tissue | Resectable gastroesophageal cancer | Perioperative chemotherapy with ECF | 29–85 | UK/Europe (UK) | Yes |
Thomas, 2011 | Single‐arm clinical trial | 131 | Fluoropyrimidines: MTHFR |
MTHFR c.1298A>C (rs1801131) MTHFR c.677C>T (rs1801133) |
Peripheral blood | Advanced or metastatic rectal cancer | 5‐FU ± irinotecan + radiotherapy | N/A | North America (US) | Yes |
Boige, 2016 |
Cohort study of Phase III RCT | 1545 | Fluoropyrimidines: DPYD | DPYD*6; c.2194G>A (rs1801160) | Peripheral blood | Stage III Colon Cancer | FOLFOX4 +/− cetuximab | 19–75 | UK/Europe (France) | Not reported |
Bardia, 2021 b |
Cohort study of Phase I/II RCT | 403 | Sacituzumab Govitecan: UGT1A6 | UGT1A1*28 (rs3064744) | Peripheral blood | Multiple metastatic cancers including breast, gastrointestinal, lung | Sacituzumab govitecan 8 mg/kg or 10 mg/kg D1, D8 q21d | 31–90 | North America (US) | Not reported |
Rugo, 2022 |
Cohort study of Phase III RCT | 243 | Sacituzumab Govitecan: UGT1A6 | UGT1A1*28 (rs3064744) | Peripheral blood | Refractory/relapse metastatic triple negative breast cancer | Sacituzumab govitecan 10 mg/kg D1, D8 q21d | 27–82 | UK/Europe, North America | Not reported |
Schaid, 2014 |
Cohort study of Phase III RCT | 1104 | Lapatinib: HLA‐DRB1 | HLA‐DRB1*07:01 | Peripheral blood | Early‐stage breast cancer | Lapatinib 1500 mg orally daily | 25–87 | UK/Europe, Americas, Africa, Australasia | Not reported |
Spraggs, 2018 |
Cohort study of Phase III RCT | 4618 | Lapatinib: HLA‐DRB1 | HLA‐DRB1*07:01 | Peripheral blood | Early‐stage breast cancer | Lapatinib monotherapy, and in combination or sequence with trastuzumab, in addition to concurrent taxane | N/A | Asia, Australasia, Americas, Africa, UK/Europe. | Not reported |
Abumiya, 2014 | Single‐arm clinical trial | 34 | Nilotinib: UGT1A1 |
UGT1A1*28 (rs3064744) UGT1A1*6 (rs4148323) |
Peripheral blood | Chronic myeloid leukemia | Nilotinib | 31–83 | East Asia (Japan) | Not reported |
Singer, 2007 |
Cohort study of Phase I/II parallel arm NRCT | 111 | Nilotinib: UGT1A1 | UGT1A1*28 (rs3064744) | Peripheral blood | Chronic myeloid leukemia | Nilotinib | 55.1 ± 15.7 | Asia, Australasia, North America, UK/Europe | Not reported |
Toffoli, 2003 | Cohort study of single‐arm clinical trial | 43 | Methotrexate: MTHFR | MTHFR 677TT (rs1801133) | Peripheral blood | Ovarian cancer | Oral MTX 1.25 mg (one‐half tablet of 2.5 mg) every 12 hr for 21 days. | 24–77 | UK/Europe | Not reported |
Choi, 2017 | Single‐arm clinical trial | 111 | Methotrexate: MTHFR | MTHFR 677TT (rs1801133) | Peripheral blood | Primary central nervous system lymphoma | MTX 3500 mg/m2 for 2 hours on day 1, q2w. | 17–86 | East Asia | Yes |
Goey, 2016 |
Cohort study of single‐arm clinical trial | 25 | Belinostat: UGT1A1 |
UGT1A1*28 (rs3064744) UGT1A1*6 (rs4148323) UGT1A1*60 (rs4124874) |
Peripheral blood | Advanced cancer | Belinostat + cisplatin 80 mg/m2 D1 + etoposide 100 mg/m2 D1, D3 and D4 | 40–78 | North America (US) | Yes |
Iwata, 2021 |
Cohort study of Phase III RCTs | 652 | Palbociclib: ABCB1 | ABCB1 1236T > C (rs1128503) | Peripheral blood | Advanced breast cancer | Palbociclib D1‐21 q 28 days | N/A | Asia, Australasia, North America, UK/Europe | Yes |
Metharom, 2011 [35] | Cohort study of Phase II RCT | 33 | Gemcitabine: CDA | CDA c.79 A>C (rs2072671) | Peripheral blood | Pancreatic cancer, NCLC | Gemcitabine weekly for 4 weeks | N/A | Australasia | Not reported |
Joerger, 2012 | Cohort study of single‐arm clinical trial | 146 | Gemcitabine:CDA | CDA c.79 A>C (rs2072671) | Peripheral blood | NSCLC | Gemcitabine D1, D8 followed by cisplatin or carboplatin D1 | 37–79 | UK/Europe | Yes |
Jordheim, 2015 NCT00140660, NCT00140595, NCT00144807, NCT00169143, NCT00144755, and NCT00135499 |
Cohort study of RCTs | 760 | Rituximab‐doxorubicin‐vinca alkaloid‐cyclophosphamide based chemotherapy: ABCB1 | ABCB1 1199G>A (rs2229109) | Peripheral blood | DLBCL, other NHL | R‐CHOP/R‐ACVBP | 18–93 | UK/Europe (Belgium France, Switzerland) | Yes |
Reyes‐Gibby, 2007 c | Cohort study | 207 |
Morphine: OPRM1 Morphine: COMT |
OPRM1 118A > G (rs1799971) COMT Val158Met (rs4680) |
Peripheral blood | Multiple cancers: Urology, Breast, Lung, other | Post‐surgery | 29–89 | UK/Europe (Norway) | Yes |
Rakvag, 2008 d | Cohort study | 197 | Morphine: COMT | COMT Val158Met (rs4680) | Peripheral blood | Multiple cancers: Urology, Lung, Breast, Hematology | Post‐surgery | 64 +/−13 | UK/Europe (Norway) | Yes |
Liu, 2012 | Cohort study | 96 | Tramadol: OPRM1 | OPRM1 118A > G (rs1799971) | Peripheral blood | Gastrointestinal cancers; oxaliplatin‐associated neuropathy | Oxaliplatin related neuropathy | N/A | East Asia (China) | Not reported |
Pu, 2019 ChiCTR‐PR‐16007775 |
Cohort study of RCT | 59 |
Oxycodone: OPRM1 Sufentanil: CYP3A4 |
OPRM1 118A > G (rs1799971) Sufentanil: CYP3A4*1G |
Peripheral blood | Gastrointestinal cancers | Post‐surgery | N/A | East Asia (China) | Yes |
Babaoglu, 2005 | Cohort study | 216 | Granisetron: ABCB1 | ABCB1 3435C>T (rs1045642) | Peripheral blood | Multiple cancers: breast cancer, lymphoma, and lung cancer | Moderately or highly emetogenic chemotherapy | 46 +/− 10.7 | Middle East (Turkey) | Yes |
Tsuji, 2017 e UMIN000009335 | Cohort study of Phase III RCT | 156 |
Granisetron: ABCB1 Palonosetron: ABCG2 |
ABCB1 3435C>T (rs1045642) ABCB1 2677GT > A (rs2032582) ABCG2 421C > A (rs2231142) |
Peripheral blood | Advanced cancer | Cisplatin ≥50 mg/m2 | 31–76 | East Asia (Japan) | Yes |
Lavanderos, 2019 | Cohort study | 119 | 5HT3antagonist: ABCB1 | ABCB1 3435C>T (rs1045642) | Peripheral blood | Germinal (seminoma or non‐seminoma) testicular cancer | Bleomycin, cisplatin, etoposide | 16–56 | Latin America (Chile) | Not assessed |
Note: LVF5FU2 = leucovorin 200 mg/m2 and 5‐fluorouracil 400 mg/m 2 intravenously on day 1, followed by a 46‐h protracted infusion of 5‐fluorouracil 2400 mg/m2.
TIROX = S‐1/irinotecan/oxaliplatin: S‐1 40 mg/m2 bd. on days 1–14; irinotecan 150 mg/m2 plus oxaliplatin 85 mg/m2 on Day 1 every 3 weeks.
IROX = irinotecan 200 mg/m2 and oxaliplatin 85 mg/m2 on Day 1 every 3 week.
FOLFOX = oxaliplatin 85 mg/m2 and leucovorin 1, 200 mg/m2 and 5‐fluorouracil bolus 400 mg/m2 on day 1, followed by 22‐h infusion of 5‐fluorouracil 600 mg/m2.
XELOX = oxaliplatin 135 mg/m2 on day 1 and capecitabine 1000 mg/m2 twice daily for 14 days.
FOLFIRI = irinotecan 180 mg/m2 and leucovorin 400 mg/m2, followed by bolus 400–500 mg/m2 5‐fluorouracil, followed by 46‐h infusion of 5‐fluorouracil 2400–3000 mg/m2.
FLIRI = irinotecan 180 mg/m2 Day 1, bolus 5‐Fluorouracil 500 mg/m2 and Leucovorin 60 mg/m2 on day 1 and 2.
R‐CHOP = rituximab 37,550 mg/m2, doxorubicin 50 mg/m2, vincristine 1.4 mg/m2 (cap 2 mg), cyclophosphamide 750 mg/m2 D1 and prednisolone 100 mg/m2 D1‐5.
R‐ACVBP = rituximab 375 mg/m2, doxorubicin 75 mg/m2 and cyclophosphamide 1200 mg/m2 D1, vindesine 2 mg/m2 D1&5, bleomycin 10 mg D1&5, prednisone 60 mg/m2 D1‐5.
Han, 2006 excluded given same trial.
Ocean, 2017 excluded given same trial.
Klepstad, 2004 excluded given duplicate cohort.
Rakvag, 2005 excluded given duplicate cohort.
Yokoi, 2018 excluded given same trial.
Analysis 2 (no PGx guideline established): Risk of bias for included studies
The included studies were mostly low to moderate RoB, as summarized in Table S3. Authors recognized bias associated with comparator arms including variant allele carriers (e.g., heterozygous) or where the comparative study did not report Hardy–Weinberg equilibrium. RoB scores were allocated for each domain and aggregated as per methods. The Overall evidence was qualified using GRADE (Table 3).
Fluoropyrimidines: TYMS 5′UTR 28base pair repeat (rs45445694) homozygous and heterozygous
Seven studies 20 , 21 , 22 , 23 , 24 , 25 , 26 reported on SAEs across 1747 participants, and another study was excluded as authors reported tissue samples were frequently too degraded for TYMS genotyping. 27 Pooled analysis showed a higher incidence of fluoropyrimidine‐associated SAEs in patients homozygous or heterozygous for variant rs45445694 compared to wild‐type, 46.4% versus 24.6% (RR = 1.31, 95%CI 1.12–1.54, I 2 = 26%), see Figure 3a. Two studies 24 , 25 were rated as high RoB as the control arm included heterozygotes of variant rs45445694, potentially underestimating the effect size. Pooled data for studies reporting homozygous carriers only was associated with less heterogeneity and higher RR (24.9% vs. 14.3%; RR = 1.8, 95%CI 1.33, 2.44, I 2 = 0%). In secondary analysis, Ruzzo et al. 28 reported that despite a low number of SAEs, the risk of vomiting was higher (OR = 8.83; p = 0.049) in carriers of rs45445694, see Table S6.
FIGURE 3.
Forest plots for Analysis 2: Severe adverse events for variant carriers vs. wild‐type carriers. (a) TYMS 5′UTR (rs45445694) homozygous and heterozygous carriers versus wild‐type in patients treated with fluoropyrimidine‐based chemotherapy, (b) DPYD*6 (rs1801160) homozygous and heterozygous carriers versus wild‐type in patients treated with fluoropyrimidine‐based chemotherapy (c) ABCB1 3435C>T (rs1045642) homozygous carriers versus wild‐type in patients treated with irinotecan‐based chemotherapy, (d) UGT1A1*93 (rs10929302) carriers versus wild‐type in patients treated with irinotecan‐based chemotherapy, (e) UGT1A1*28 (rs3064744) carriers versusversus wild‐type in all patients treated with sacituzumab govitecan, (f) HLA‐DRB1*07:01 carriers versus wild‐type in all patients treated with lapatinib, (g) UGT1A1*28 carriers versus wild‐type in all patients treated with nilotinib, (h) Pain scores (mean ± standard deviation) in OPRM1 118A>G carriers versus wild‐type in patients initiated on systemic opioid medication.
Note: (a) Seo, 2009 excluded from the RR calculation due to the high risk of bias (heterozygous carriers were included in the “wild‐type” arm). With the inclusion of Seo 2009, RR = 1.37 (1.12, 1.67), p = 0.002. (c) Hh = Homozygous and heterozygous carriers (Han 2009; McLeod 2010) excluded from RR calculation. (d) Event rate per patient year used for McLeod 2010; RR not significantly different in sensitivity analysis. (e) Event rate per patient year used for Rugo 2022; RR not significantly different in sensitivity analysis.
Fluoropyrimidines: DPYD*6 (rs1801160) homozygous and heterozygous
Two studies 29 , 30 reported on SAEs across 2042 participants. Pooled analysis showed that both homozygous and heterozygous carriers of DPYD*6 allele were at higher risk of fluoropyrimidine‐associated SAEs, 60.0% vs. 44.6% (RR = 1.42; 95%CI = 1.10–1.84, I 2 = 73%), see Figure 3b.
Irinotecan: ABCB1 3435C>T (rs1045642) homozygous
Five studies 23 , 27 , 31 , 32 , 33 reported on SAEs across 343 participants. Overall, only 3 studies reported outcomes for homozygotes versus wild‐type, 27 , 31 , 33 wherein pooled analysis showed a significantly higher incidence of irinotecan‐associated SAEs in patients homozygous for variant rs1045642 compared to wild‐type, 50% versus 18% (2.70, 95%CI 1.56–4.67), (I 2 = 0%), see Figure 3c, with homozygous carriers accounting for 23% of participants. In secondary analysis, Lara et al reported a similar summary effect size. 34 Statistical significance was lost when heterozygous carriers were included in the pooled analysis.
Irinotecan: UGT1A1 *93 (rs10929302) homozygous
Three studies 20 , 23 , 31 (394 participants) in aggregate demonstrated higher incidence of irinotecan‐associated SAEs in homozygous carriers of the UGT1A1*93 (−3156G>A; rs10929302) variant compared to wild‐type, 59.1% versus 27.5% (RR = 2.08, 95%CI 1.31–3.31, I 2 = 64%), see Figure 3d. Approximately 8% of participants were homozygous carriers. Mcleod 23 reported a greater number of events than patients. Accordingly, data for this study was converted to an event rate per patient year for pooled analysis. In secondary analysis, Innocenti et al. also found that ‐3156G>A was a significant predictor of severe neutropenia and distinguished different phenotypes within TA indel genotypes. 35
Sacituzumab govitecan: UGT1A1 *28 (rs3064744) homozygous
Two studies 36 , 37 (370 participants) demonstrated higher incidence of SAEs in homozygous carriers of UGT1A1*28 (rs3064744) variant compared to wild‐type, 87.6% versus 53.3% (RR = 1.55 95%CI 1.46–1.87, I 2 = 0%), Figure 3e. As Rugo 37 reported a greater number of events than patients, data was converted to an event rate per patient years for pooled analysis.
Lapatinib: HLA‐DRB1 *07:01 homozygous and heterozygous
Two studies 38 , 39 (1914 participants) demonstrated higher incidence of lapatinib‐associated SAEs, in particular, hepatotoxicity among carriers of HLA‐DRB1*07:01 variant compared to wild‐type, 11.3% versus 1.1% (RR = 10.36, 95%CI 5.94–18.06, I 2 = 0%), Figure 3f.
Nilotinib: UGT1A1 *28 (rs3064744) homozygous
Two small studies 40 , 41 (67 participants) demonstrated higher incidence of nilotinib‐associated SAEs, among homozygous carriers of UGT1A1*28 variant compared to wild‐type, 39% versus 8.2% (RR = 4.5, 95%CI 1.59–12.74, I 2 = 0%), Figure 3g.
Opioids: OPRM1 118 A>G
Four studies 42 , 43 , 44 , 45 (423 participants) demonstrated higher pain scores among carriers of the OPRM1 118A>G variant compared to wild‐type, with a mean difference of 1.61 (95%CI 0.77–2.45, p = 0.0002, I 2 = 91%), Figure 3h. Heterogeneity was considerable as indications for opioid use varied from procedure‐related pain to cancer pain. In secondary analysis, a validation cohort of approximately 620 patients receiving opioids for moderate to severe cancer pain, unrelated to a procedure, showed no association between this variant and pain intensity. 46
Palbociclib: ABCB1 rs1128503 homozygous and heterozygous
One study 47 (347 participants) reported on the interactions between palbociclib and variants of the ABCB1 gene, c1236C>T (rs1128503) and c3435C>T (rs1045642). A higher incidence of palbociclib‐associated SAEs occurred among homozygous and heterozygous carriers of the c1236C>T variant compared to wild‐type, 38% versus 23% (RR = 1.65 95%CI 1.19–2.29, p = 0.003) and 32% versus 23% (RR = 1.37 95%CI 1.03–1.84, p = 0.03) respectively but not c3435C>T (rs1045642) variant. Of the participants studied, 23% were homozygous carriers of c1236C>T and 70% were homozygous or heterozygous carriers. The interaction of c1236C>T variant on palbociclib was significant in non‐Asian patients (32% vs. 19%) but not in Asians (50% vs. 67%).
R‐CHOP/AVCBP: ABCB1 rs2229109 homozygous and heterozygous
In one study 48 of 760 participants, a higher incidence of SAEs for non‐Hodgkin lymphoma patients receiving chemotherapy with regimen R‐CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone) or R‐AVCBP (rituximab, doxorubicin, cyclophosphamide, vindesine, bleomycin, prednisone) occurred among homozygous and heterozygous carriers of the ABCB1 1199G>A (rs2229109) variant compared to wild‐type, 22.8% versus 6.5% (RR = 3.53; 95%CI 2.03–6.15, p < 0.0001). Approximately, 8% of participants carried this variant allele. In secondary analysis, Rossi et al 49 reported a higher risk of severe cardiac toxicity in heterozygous carriers compared to wild‐type; OR = 1.89 (95%CI 1.15–3.12, p = 0.01).
Other medicine: variant pairs
Meta‐analysis from two studies 50 , 51 totaling 166 participants showed higher incidence of gemcitabine‐associated SAEs among homozygous and heterozygous carriers of the CDA c79 A>C (rs2072671) variant compared to wild‐type, 36.4% versus 16.7% (RR = 2.25, 95%CI 1.13–4.47, I 2 = 16%). However, secondary analysis demonstrated no correlation between this CDA genotype and occurrence of toxicities, 52 and a conflicting effect with this variant associated with a lower risk of severe toxicities. 53
Pairs that did not show a significant effect on incidence of SAEs included fluoropyrimidines: MTHFR rs1801133 (5 studies, 20 , 22 , 24 , 27 , 54 754 participants), RR = 0.89, 95%CI 0.6–1.3, I 2 = 0%; fluoropyrimidines: MTHFR rs1801131 (4 studies, 20 , 22 , 24 , 54 625 participants), RR = 1.05, 95%CI 0.65–1.70, I 2 = 0% (and secondary analysis 55 ); irinotecan: UGT1A7*3 (3 studies, 56 , 57 , 58 190 participants), RR = 1.15, 95%CI 0.77–1.74, I 2 = 0%; irinotecan: UGT1A6*2 (2 studies, 56 , 58 86 participants), RR = 1.8, 95%CI 0.26–12.48, I 2 = 53%; irinotecan: UGT1A9*1b (4 studies, 32 , 57 , 59 , 60 394 participants), RR = 0.91, 95%CI 0.57–1.43, I 2 = 61%, and pazopanib: HLA‐B*57:01 (1 study, 61 1002 participants, RR = 1.14, 95%CI 0.55–2.36).
Two studies 62 , 63 that evaluated MTHFR rs1801133 in patients treated with methotrexate (362 participants), each utilizing very different dosing regimens, were associated with a higher incidence of SAEs compared to wild‐type, 39% versus 24.3%, but this was not a statistically significant result in pooled analysis (RR = 1.8 95%CI 0.81–3.99; I 2 = 24%). For 5‐HT3 antagonists used as antiemetics, there was no statistically significant effect on loss of emesis control associated with ABCB1 rs1045642 (40% variant carriers vs. 29.5% wild‐type; RR = 1.37, 95% CI 0.46–4.07, I 2 = 65%) in pooled analysis of 3 studies 64 , 65 , 66 (407 participants) or ABCG2 rs2231142 66 , 67 (20.0% variant carriers vs. 24.3% wild‐type; RR = 1.17, 95% CI 0.08–16.93). Secondary analysis included assessments of association of taxane‐related toxicities with ARHGEF10, 68 FGD4 68 , 69 ; and everolimus with CYP3A4/5 70 Data could not be pooled to evaluate medicine: variant pairs that were associated with single and inadequately powered studies including imatinib: ABCB1 1236T>C rs1128503, 71 high dose etoposide: UGT1A1*28, 72 sorafenib: CYP3A5, 73 sorafenib: ABCB1 3435C>T rs1045642, 74 belinostat: UGT1A1*28, 75 sunitinib: ABCG2 421 C>A rs2231142 76 (including secondary analysis 77 ), taxanes: ATP‐binding cassette family of genes, 78 , 79 (including secondary analysis 80 , 81 ); taxanes: CYP3A4/5, 82 (including secondary analysis 83 , 84 , 85 , 86 ); pralatrexate: ATIC, 87 morphine: SLC6A4, 88 vincristine: CEP72. 89 These results are shown in Tables S4 and S5.
DISCUSSION
This meta‐analysis demonstrates that for the medicine‐gene pairs that have been subject to study in adult patients with cancer, adoption of evidence‐based pharmacogenomic‐guided prescribing significantly reduces the incidence of clinically relevant, adverse medicine‐related events including chemotherapy‐related deaths. Our secondary analysis suggests the potential for greater impact when comprehensive and multigene testing is employed as part of the PGx strategy, in support of large‐scale PGx implementation approaches across multiple jurisdictions, including but not limited to cancer care. These studies have also demonstrated feasibility and acceptance in clinical practice. 2 , 90 , 91 , 92 , 93 Here, we recommend, for priority clinical validation within a cancer care context, eight pharmacogenomic variants that are not currently associated with a recommendation for medicines optimization from CPIC or DPWG guidelines but have been found to be predictive of a higher risk of SAEs compared to wild‐type allele carriers. These will be studied in a prospective clinical trial ACTRN12624000079549.
The clinical applicability for these emergent variants is important, as assessed by GRADE, given adverse events were prospectively collected and limited to SAEs only, analysis was generally restricted to clinical trial datasets where pharmacovigilance requirements ensure robust follow‐up of SAEs, and the effect sizes were generally moderate or large. The same regimens were used across intervention/at‐risk arms and comparator arms with standard dose adjustments applied depending on toxicities; and differences in dose–response gradients were considered as part of the generalisability assessments for GRADE. Only studies that prospectively replicated or validated a PGx association were included, and not PGx discovery‐only cohorts or studies that may utilize proximal surrogate outcomes such as pharmacokinetic changes or pharmacodynamic biochemical responses. It is well‐reported that false positive findings are typically associated with discovery studies that have design limitations or test many potential associations without proper statistical correction for multiple comparisons. 94 Finally, studies that genotyped tumor tissue were excluded given that somatic variants may be acquired during carcinogenesis and prior reports have shown conflicting data when tumor tissue was utilized for pharmacogenomic analysis. 95 Although most included studies utilized peripheral blood for genotyping, a minority of studies utilized fresh frozen normal tissue, and where authors 27 reported low quality of sampled tissue for variant analysis, these data were excluded. Notably, in limiting our included studies to robust clinical datasets, less studied and more novel variants with important associations may not have been identified through this approach.
The present review demonstrated that DPYD‐guided dosing of fluoropyrimidines reduced SAEs and treatment‐related deaths as shown in previously published reviews of diverse study design. 96 Although the included studies did not assess the impact on treatment efficacy, a newer study has shown no detrimental impact on survival for many variants 97 and further research is underway to optimize the accuracy of fluoropyrimidine dosing. 98 , 99 We highlight a small effect size on the incidence of severe toxicity among homozygous and heterozygous carriers of DPYD*6, which supports findings from a recent meta‐analysis of any grade fluoropyrimidine‐associated toxicity. 100 The DPWG and CPIC have regarded this variant as having a negligible effect on clinically relevant enzymatic degradation of fluoropyrimidines based on studies conducted largely in European countries. However, the minor allele frequency of this variant is highest in some South Asian populations (9%) where increased toxicity has been suggested in small studies, 101 , 102 and in African populations where it has been associated with reduced enzymatic activity conditional on the presence of a linked variant p.Y186C. 103 This review also supports findings from a previously published meta‐analysis associating TYMS 5′UTR with an increased risk of adverse events. 104 Combined with DPYD, concurrent TYMS genotyping may enhance the predictability of patients at elevated risk for adverse events. 105 This is particularly important where target concentration intervention modeling cannot be utilized to guide dosing as in the case of the oral prodrug capecitabine where intracellular concentrations of 5‐fluoroucail cannot be routinely measured nor have been correlated with systemic concentrations of capecitabine. 99
This meta‐analysis also showed that in addition to patients who are carriers of UGT1A1*28 (rs3064744) and UGT1A1*6 (rs4148323) variants, patients who have the UGT1A1*93 (rs10929302), and ABCB1 3435 C>T (rs1045642) variant alleles are at significantly increased risk of severe toxicity from irinotecan‐based therapies. SN‐38, a DNA topoisomerase I inhibitor, is the active metabolite of irinotecan. It is inactivated by intracellular glucuronidation via the uridine diphosphate glucuronosyltransferase (UGT) family of enzymes including UGT1A1. Both UGT1A1*28 and UGT1A1*6 variants, recognized by the CPIC and DPWG, diminish glucuronidation and increase exposure to SN‐38 with patients who are homozygous for either or both variants requiring dose reduction of irinotecan to avoid toxicities. UGT1A1*93 has been reported to be in linkage disequilibrium with *28 in populations of European‐ancestry 106 and therefore may not have an independent effect on irinotecan toxicity, however, these findings have not been confirmed across multiple ethnicities. The ABCB1 encoded transporter, ABCB1 or P‐glycoprotein actively expels irinotecan, SN‐38, and its glucuronide out of hepatocytes into the bile canaliculus lumen and intestinal lumen. Homozygous carriers of ABCB1 3435C>T (rs1045642) may be predisposed to increased risk of toxicity through reduced efflux ability of ABCB1 and reduced clearance of SN‐38. 107 The minor allele frequency is highest in African (78%) and Asian (62%) ancestries and lowest in European (48%) and South Asian (39%) ancestries. 101 While other variants such as ABCB1 1236C>T (rs1128503) have previously been correlated with increased exposure to irinotecan and its active metabolite SN‐38, 108 our analysis did not uncover an association of severe toxicities within a prospective clinical trial dataset. The influence of ABCB1 variants on irinotecan‐induced severe toxicity may be better elucidated through haplotype analysis. 109 These medicine‐variant interactions may also be relevant for the third‐generation antibody‐drug conjugate, sacituzumab govitecan which is comprised of monoclonal antibody hRS7 conjugated to SN38 via a hydrolyzable linker. Although available data suggest homozygous UGT1A1*28 carriers are predisposed to increased toxicity from sacituzumab govitecan (65.0% vs. 46.6%; RR = 3.57, 95%CI 1.83–6.95) similar to irinotecan, further analysis is warranted to determine if other variants of relevance to irinotecan, namely UGT1A1*6, UGT1A1*93 and ABCB1 3435 C>T are equally important for toxicities related to sacituzumab govitecan.
The ABCB1 gene was shown in this review to be an important predictor of toxicity across a range of anti‐cancer regimens including the more novel Cyclin‐dependent kinases 4 and 6 inhibitors. Despite evidence of improved overall survival from CDK 4/6 inhibitors in hormone‐receptor positive, human epidermal growth factor receptor 2 (HER2)–negative advanced breast cancer, wide inter‐individual variability in therapeutic benefit and toxicity have been reported. The ABCB1 1236C>T variant was associated with higher rates of toxicity from palbociclib in non‐Asians (32% vs. 19%) but not in Asians. While an exposure‐safety relationship for palbociclib has previously been reported with higher palbociclib exposure predisposing to a significantly higher risk of neutropenia and thrombocytopenia, there is no evidence that an exposure‐response relationship at the standard fixed dose exists. 110 , 111 Inter‐ethnic differences in tolerability of small molecule anti‐cancer agents have previously been correlated to ethnic differences in the expression and activity of metabolic enzymes and transporters including for the P‐glycoprotein. 112 Ethnicity was not well‐reported across included studies and if reported, it was race that was generally self‐identified by participants. Most studies were conducted in European‐ancestry population‐rich geographical regions, followed by East Asian countries. The landscape for pharmacogenomic mapping across geographical regions has improved and enabled a more accurate determination of genomic variation across other ethnic populations including for Indigenous peoples. 113 , 114 , 115 , 116 Investigation of ancestry‐specific differences in linkage disequilibrium will elucidate lesser‐known variants that are of greater impact in poorly studied populations and highlight the differential impact on drug response by ancestry. 117
There are a few limitations of this meta‐analysis. First, there is heterogeneity in the types of studies evaluated under Analysis 1 (PGx vs. Usual Therapy) where the included studies aimed to either enhance cancer treatment efficacy or reduce cancer treatment toxicity, and different strategies, both single variant testing and broad gene panel testing were evaluated. Regardless of the treatment goal of the individual studies, overall, PGx is demonstrated to improve safety, and where maintaining dose intensity of chemotherapy is important for disease control, avoidance of toxicities will minimize chemotherapy dose delays or interruption and may have an overall impact on treatment outcomes. Furthermore, a subset analysis was conducted to clarify the impact on clinically relevant safety outcomes. Second, criteria were not concordant for Analysis 2 (no PGx guideline recommendation) where CTCAE criteria were used to define adverse events but symptom burden was defined by prespecified measures of brief pain inventory and emesis control rates. Accordingly, Analysis 2 comprised of subset analyses for individual medicine‐variant pairs where there was a limited number of good quality studies that could be aggregated for assessment of individual medicine: variant effects. Where genotype and outcomes data were not available for aggregation, those studies provided a narrative synthesis to either corroborate or contest the effect estimates from pooled analyses.
CONCLUSION
In conclusion, this comprehensive analysis of all available English language studies confirms the clinical validity of CPIC/DPWG‐endorsed pharmacogenomics‐guided approaches for medicines optimization to reduce unwarranted and preventable SAEs in patients with cancer. We advocate for future studies evaluating the pharmacogenomics of anti‐cancer therapies to include efficacy endpoints and not just safety endpoints. Additionally, eight emerging pharmacogenomic variants are identified as priorities for future research, prior to consideration for implementation into clinical practice.
AUTHOR CONTRIBUTIONS
All authors wrote the manuscript. S.Li. and C.M.J.K. designed the research; S.Li., S.La., M.S., C.M.J.K., and J.N. performed the research. All authors analyzed the data.
FUNDING INFORMATION
No funding was received for this work.
CONFLICT OF INTEREST STATEMENT
SLo receives research funding from institutions from Novartis, Bristol Myers Squibb, MSD, Puma Biotechnology, Eli Lilly, Nektar Therapeutics, Astra Zeneca, and Seattle Genetics. She has acted as a consultant (not compensated) to Seattle Genetics, Novartis, Bristol Myers Squibb, MSD, AstraZeneca, Eli Lilly, Pfizer, Gilead Therapeutics, and Roche‐Genentech. She has acted as a consultant (paid to institution) to Aduro Biotech, Novartis, GlaxoSmithKline, Roche‐Genentech, Astra Zeneca, Silverback Therapeutics, G1 Therapeutics, PUMA Biotechnologies, Pfizer, Gilead Therapeutics, Seattle Genetics, Daiichi Sankyo, MSD, Amunix, Tallac Therapeutics, Eli Lilly and Bristol Myers Squibb. JFS receives an honorarium from AbbVie, Astra Zeneca, Beigene, BMS, Genor Bio, Gilead, Janssen, Roche, and TG Therapeutics for participation on advisory boards, and research funding from Abbvie, BMS, Janssen, and Roche. None of these Sponsors were involved in the review. All other authors declared no competing interests for this work.
CONSENT FOR PUBLICATION
All authors have reviewed the final manuscript and provided consent for publication.
PROTOCOL REGISTRATION
The study methods were specified in advance and documented in a study protocol. The study protocol was registered with PROSPERO, registration number CRD42023421277. Prospero protocol is publicly accessible. Minor amendments to the protocol were made after the commencement of the review justified by preliminary searches, screening, and data review, to refine the inclusion criteria.
REPORTING GUIDELINES
Systematic reviews and meta‐analyses (PRISMA) checklist provided in Supplementary Tables.
Supporting information
Table S1.
Lingaratnam S, Shah M, Nicolazzo J, et al. A systematic review and meta‐analysis of the impacts of germline pharmacogenomics on severe toxicity and symptom burden in adult patients with cancer. Clin Transl Sci. 2024;17:e13781. doi: 10.1111/cts.13781
DATA AVAILABILITY STATEMENT
All data generated or analyzed during this study are included in this published article [and its supplementary information files].
REFERENCES
- 1. David V, Fylan B, Bryant E, Smith H, Sagoo GS, Rattray M. An analysis of pharmacogenomic‐guided pathways and their effect on medication changes and hospital admissions: a systematic review and meta‐analysis. Front Genet. 2021;12:698148. doi: 10.3389/fgene.2021.698148 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Swen JJ, van der Wouden CH, Manson LE, et al. A 12‐gene pharmacogenetic panel to prevent adverse drug reactions: an open‐label, multicentre, controlled, cluster‐randomised crossover implementation study. Lancet. 2023;401(10374):347‐356. doi: 10.1016/S0140-6736(22)01841-4 [DOI] [PubMed] [Google Scholar]
- 3. Swen JJ, Nijenhuis M, van Rhenen M, et al. Pharmacogenetic information in clinical guidelines: the European perspective. Clin Pharmacol Ther. 2018;103(5):795‐801. doi: 10.1002/cpt.1049 [DOI] [PubMed] [Google Scholar]
- 4. Relling MV, Klein TE. CPIC: clinical Pharmacogenetics implementation consortium of the pharmacogenomics research network. Clin Pharmacol Ther. 2011;89(3):464‐467. doi: 10.1038/clpt.2010.279 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Caterino JM, Adler D, Durham DD, et al. Analysis of diagnoses, symptoms, medications, and admissions among patients with cancer presenting to emergency departments. JAMA Netw Open. 2019;2(3):e190979. doi: 10.1001/jamanetworkopen.2019.0979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Slomski A. Global cancer burden grew from 2010 to 2019. JAMA. 2022;327(9):804. doi: 10.1001/jama.2022.2188 [DOI] [PubMed] [Google Scholar]
- 7. Whirl‐Carrillo M, Huddart R, Gong L, et al. An evidence‐based framework for evaluating pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2021;110(3):563‐572. doi: 10.1002/cpt.2350 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Page MJ, McKenzie JE, Bossuyt PM, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372:n71. doi: 10.1136/bmj.n71 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Sterne JA, Hernan MA, Reeves BC, et al. ROBINS‐I: a tool for assessing risk of bias in non‐randomised studies of interventions. BMJ. 2016;355:i4919. doi: 10.1136/bmj.i4919 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Sterne JAC, Savovic J, Page MJ, et al. RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ. 2019;366:l4898. doi: 10.1136/bmj.l4898 [DOI] [PubMed] [Google Scholar]
- 11. Wells GSB, O'Connell D, Peterson J, Welch V, Losos M, Tugwell P. The Newcastle‐Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta‐Analyses. 2013.
- 12. Schünemann H BJ, Guyatt G, Oxman A. The GRADE working group . GRADE Handbook for Grading Quality of Evidence and Strength of Recommendations;2013. Cochrane. [Google Scholar]
- 13. Henricks LM, van Merendonk LN, Meulendijks D, et al. Effectiveness and safety of reduced‐dose fluoropyrimidine therapy in patients carrying the DPYD*2A variant: a matched pair analysis. Int J Cancer. 2019;144(9):2347‐2354. doi: 10.1002/ijc.32022 [DOI] [PubMed] [Google Scholar]
- 14. Lunenburg C, Henricks LM, Dreussi E, et al. Standard fluoropyrimidine dosages in chemoradiation therapy result in an increased risk of severe toxicity in DPYD variant allele carriers. Eur J Cancer. 2018;104:210‐218. doi: 10.1016/j.ejca.2018.07.138 [DOI] [PubMed] [Google Scholar]
- 15. Paez D, Tobena M, Fernandez‐Plana J, et al. Pharmacogenetic clinical randomised phase II trial to evaluate the efficacy and safety of FOLFIRI with high‐dose irinotecan (HD‐FOLFIRI) in metastatic colorectal cancer patients according to their UGT1A 1 genotype. Br J Cancer. 2019;120(2):190‐195. doi: 10.1038/s41416-018-0348-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Tsai HL, Huang CW, Lin YW, et al. Determination of the UGT1A1 polymorphism as guidance for irinotecan dose escalation in metastatic colorectal cancer treated with first‐line bevacizumab and FOLFIRI (PURE FIST). Eur J Cancer. 2020;138:19‐29. doi: 10.1016/j.ejca.2020.05.031 [DOI] [PubMed] [Google Scholar]
- 17. Boisdron‐Celle M, Capitain O, Faroux R, et al. Prevention of 5‐fluorouracil‐induced early severe toxicity by pre‐therapeutic dihydropyrimidine dehydrogenase deficiency screening: assessment of a multiparametric approach. Semin Oncol. 2017;44(1):13‐23. doi: 10.1053/j.seminoncol.2017.02.008 [DOI] [PubMed] [Google Scholar]
- 18. Deenen MJ, Meulendijks D, Cats A, et al. Upfront genotyping of DPYD*2A to individualize Fluoropyrimidine therapy: a safety and cost analysis. J Clin Oncol. 2016;34(3):227‐234. doi: 10.1200/JCO.2015.63.1325 [DOI] [PubMed] [Google Scholar]
- 19. Henricks LM, Lunenburg C, de Man FM, et al. DPYD genotype‐guided dose individualisation of fluoropyrimidine therapy in patients with cancer: a prospective safety analysis. Lancet Oncol. 2018;19(11):1459‐1467. doi: 10.1016/S1470-2045(18)30686-7 [DOI] [PubMed] [Google Scholar]
- 20. Boige V, Mendiboure J, Pignon JP, et al. Pharmacogenetic assessment of toxicity and outcome in patients with metastatic colorectal cancer treated with LV5FU2, FOLFOX, and FOLFIRI: FFCD 2000‐05. J Clin Oncol. 2010;28(15):2556‐2564. doi: 10.1200/JCO.2009.25.2106 [DOI] [PubMed] [Google Scholar]
- 21. Castro‐Rojas CA, Esparza‐Mota AR, Hernandez‐Cabrera F, et al. Thymidylate synthase gene variants as predictors of clinical response and toxicity to fluoropyrimidine‐based chemotherapy for colorectal cancer. Drug Metab Pers Ther. 2017;32(4):209‐218. doi: 10.1515/dmpt-2017-0028 [DOI] [PubMed] [Google Scholar]
- 22. Jennings BA, Loke YK, Skinner J, et al. Evaluating predictive pharmacogenetic signatures of adverse events in colorectal cancer patients treated with fluoropyrimidines. PLoS One. 2013;8(10):e78053. doi: 10.1371/journal.pone.0078053 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. McLeod HL, Sargent DJ, Marsh S, et al. Pharmacogenetic predictors of adverse events and response to chemotherapy in metastatic colorectal cancer: results from north American gastrointestinal intergroup trial N9741. J Clin Oncol. 2010;28(20):3227‐3233. doi: 10.1200/JCO.2009.21.7943 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Schwab M, Zanger UM, Marx C, et al. Role of genetic and nongenetic factors for fluorouracil treatment‐related severe toxicity: a prospective clinical trial by the German 5‐FU toxicity study group. J Clin Oncol. 2008;26(13):2131‐2138. doi: 10.1200/JCO.2006.10.4182 [DOI] [PubMed] [Google Scholar]
- 25. Seo BG, Kwon HC, Oh SY, et al. Comprehensive analysis of excision repair complementation group 1, glutathione S‐transferase, thymidylate synthase and uridine diphosphate glucuronosyl transferase 1A1 polymorphisms predictive for treatment outcome in patients with advanced gastric cancer treated with FOLFOX or FOLFIRI. Oncol Rep. 2009;22(1):127‐136. [PubMed] [Google Scholar]
- 26. Smyth E, Zhang S, Cunningham D, et al. Pharmacogenetic analysis of the UK MRC (Medical Research Council) MAGIC trial: Association of Polymorphisms with toxicity and survival in patients treated with perioperative Epirubicin, cisplatin, and 5‐fluorouracil (ECF) chemotherapy. Clin Cancer Res. 2017;23(24):7543‐7549. doi: 10.1158/1078-0432.CCR-16-3142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Glimelius B, Garmo H, Berglund A, et al. Prediction of irinotecan and 5‐fluorouracil toxicity and response in patients with advanced colorectal cancer. Pharmacogenomics J. 2011;11(1):61‐71. doi: 10.1038/tpj.2010.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Ruzzo A, Graziano F, Galli F, et al. Genetic markers for toxicity of adjuvant oxaliplatin and fluoropyrimidines in the phase III TOSCA trial in high‐risk colon cancer patients. Sci Rep. 2014;4:6828. doi: 10.1038/srep06828 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Boige V, Vincent M, Alexandre P, et al. DPYD genotyping to predict adverse events following treatment with fluorouracil‐based adjuvant chemotherapy in patients with stage III colon cancer: a secondary analysis of the PETACC‐8 randomized clinical trial. JAMA Oncol. 2016;2(5):655‐662. doi: 10.1001/jamaoncol.2015.5392 [DOI] [PubMed] [Google Scholar]
- 30. Ruzzo A, Graziano F, Galli F, et al. Dihydropyrimidine dehydrogenase pharmacogenetics for predicting fluoropyrimidine‐related toxicity in the randomised, phase III adjuvant TOSCA trial in high‐risk colon cancer patients. Br J Cancer. 2017;117(9):1269‐1277. doi: 10.1038/bjc.2017.289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Cote JF, Kirzin S, Kramar A, et al. UGT1A1 polymorphism can predict hematologic toxicity in patients treated with irinotecan. Clin Cancer Res. 2007;13(11):3269‐3275. doi: 10.1158/1078-0432.CCR-06-2290 [DOI] [PubMed] [Google Scholar]
- 32. Han JY, Lim HS, Park YH, Lee SY, Lee JS. Integrated pharmacogenetic prediction of irinotecan pharmacokinetics and toxicity in patients with advanced non‐small cell lung cancer. Lung Cancer. 2009;63(1):115‐120. doi: 10.1016/j.lungcan.2007.12.003 [DOI] [PubMed] [Google Scholar]
- 33. Levi F, Karaboue A, Etienne‐Grimaldi MC, et al. Pharmacokinetics of Irinotecan, Oxaliplatin and 5‐fluorouracil during hepatic artery Chronomodulated infusion: a translational European OPTILIV study. Clin Pharmacokinet. 2017;56(2):165‐177. doi: 10.1007/s40262-016-0431-2 [DOI] [PubMed] [Google Scholar]
- 34. Lara PN Jr, Natale R, Crowley J, et al. Phase III trial of irinotecan/cisplatin compared with etoposide/cisplatin in extensive‐stage small‐cell lung cancer: clinical and pharmacogenomic results from SWOG S0124. J Clin Oncol. 2009;27(15):2530‐2535. doi: 10.1200/JCO.2008.20.1061 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Innocenti F, Undevia SD, Iyer L, et al. Genetic variants in the UDP‐glucuronosyltransferase 1A1 gene predict the risk of severe neutropenia of irinotecan. J Clin Oncol. 2004;22(8):1382‐1388. doi: 10.1200/JCO.2004.07.173 [DOI] [PubMed] [Google Scholar]
- 36. Bardia A, Messersmith WA, Kio EA, et al. Sacituzumab govitecan, a Trop‐2‐directed antibody‐drug conjugate, for patients with epithelial cancer: final safety and efficacy results from the phase I/II IMMU‐132‐01 basket trial. Ann Oncol. 2021;32(6):746‐756. doi: 10.1016/j.annonc.2021.03.005 [DOI] [PubMed] [Google Scholar]
- 37. Rugo HS, Tolaney SM, Loirat D, et al. Safety analyses from the phase 3 ASCENT trial of sacituzumab govitecan in metastatic triple‐negative breast cancer. NPJ Breast Cancer. 2022;8(1):98. doi: 10.1038/s41523-022-00467-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Schaid DJ, Spraggs CF, McDonnell SK, et al. Prospective validation of HLA‐DRB1*07:01 allele carriage as a predictive risk factor for lapatinib‐induced liver injury. J Clin Oncol. 2014;32(22):2296‐2303. doi: 10.1200/JCO.2013.52.9867 [DOI] [PubMed] [Google Scholar]
- 39. Spraggs CF, Parham LR, Briley LP, et al. Characterisation of the HLA‐DRB1*07:01 biomarker for lapatinib‐induced liver toxicity during treatment of early‐stage breast cancer patients with lapatinib in combination with trastuzumab and/or taxanes. Pharmacogenomics J. 2018;18(3):480‐486. doi: 10.1038/tpj.2017.39 [DOI] [PubMed] [Google Scholar]
- 40. Abumiya M, Takahashi N, Niioka T, et al. Influence of UGT1A1 6, 27, and 28 polymorphisms on nilotinib‐induced hyperbilirubinemia in Japanese patients with chronic myeloid leukemia. Drug Metab Pharmacokinet. 2014;29(6):449‐454. doi: 10.2133/dmpk.DMPK-14-RG-031 [DOI] [PubMed] [Google Scholar]
- 41. Singer JB, Shou Y, Giles F, et al. UGT1A1 promoter polymorphism increases risk of nilotinib‐induced hyperbilirubinemia. Leukemia. 2007;21(11):2311‐2315. doi: 10.1038/sj.leu.2404827 [DOI] [PubMed] [Google Scholar]
- 42. Klepstad P, Rakvag TT, Kaasa S, et al. The 118 a > G polymorphism in the human mu‐opioid receptor gene may increase morphine requirements in patients with pain caused by malignant disease. Acta Anaesthesiol Scand. 2004;48(10):1232‐1239. doi: 10.1111/j.1399-6576.2004.00517.x [DOI] [PubMed] [Google Scholar]
- 43. Liu YC, Wang WS. Human mu‐opioid receptor gene A118G polymorphism predicts the efficacy of tramadol/acetaminophen combination tablets (ultracet) in oxaliplatin‐induced painful neuropathy. Cancer. 2012;118(6):1718‐1725. doi: 10.1002/cncr.26430 [DOI] [PubMed] [Google Scholar]
- 44. Pu J, Wang N, Huang ZK, He XY, Yuan HB. Correlation between gene polymorphism and opioid efficacy in patients with gastric or intestinal cancer. Eur Rev Med Pharmacol Sci. 2019;23(21):9393‐9410. doi: 10.26355/eurrev_201911_19432 [DOI] [PubMed] [Google Scholar]
- 45. Reyes‐Gibby CC, Shete S, Rakvag T, et al. Exploring joint effects of genes and the clinical efficacy of morphine for cancer pain: OPRM1 and COMT gene. Pain. 2007;130(1–2):25‐30. doi: 10.1016/j.pain.2006.10.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Fladvad T, Fayers P, Skorpen F, Kaasa S, Klepstad P. Lack of association between genetic variability and multiple pain‐related outcomes in a large cohort of patients with advanced cancer: the European Pharmacogenetic opioid study (EPOS). BMJ Support Palliat Care. 2012;2(4):351‐355. doi: 10.1136/bmjspcare-2012-000212 [DOI] [PubMed] [Google Scholar]
- 47. Iwata H, Umeyama Y, Liu Y, et al. Evaluation of the association of polymorphisms with palbociclib‐induced neutropenia: pharmacogenetic analysis of PALOMA‐2/‐3. Oncologist. 2021;26(7):e1143‐55. doi: 10.1002/onco.13811 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Jordheim LP, Ribrag V, Ghesquieres H, et al. Single nucleotide polymorphisms in ABCB1 and CBR1 can predict toxicity to R‐CHOP type regimens in patients with diffuse non‐Hodgkin lymphoma. Haematologica. 2015;100(5):e204‐6. doi: 10.3324/haematol.2014.120113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Rossi D, Rasi S, Franceschetti S, et al. Analysis of the host pharmacogenetic background for prediction of outcome and toxicity in diffuse large B‐cell lymphoma treated with R‐CHOP21. Leukemia. 2009;23(6):1118‐1126. doi: 10.1038/leu.2008.398 [DOI] [PubMed] [Google Scholar]
- 50. Joerger M, Burgers SA, Baas P, et al. Germline polymorphisms in patients with advanced nonsmall cell lung cancer receiving first‐line platinum‐gemcitabine chemotherapy: a prospective clinical study. Cancer. 2012;118(9):2466‐2475. doi: 10.1002/cncr.26562 [DOI] [PubMed] [Google Scholar]
- 51. Metharom E, Galettis P, Manners S, et al. The pharmacological advantage of prolonged dose rate gemcitabine is restricted to patients with variant alleles of cytidine deaminase c.79A>C. Asia Pac. J Clin Oncol. 2011;7(1):65‐74. doi: 10.1111/j.1743-7563.2010.01354.x [DOI] [PubMed] [Google Scholar]
- 52. Serdjebi C, Gagniere J, Desrame J, et al. FFCD‐1004 clinical trial: impact of cytidine deaminase activity on clinical outcome in gemcitabine‐monotherapy treated patients. PLoS One. 2015;10(8):e0135907. doi: 10.1371/journal.pone.0135907 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Innocenti F, Jiang C, Sibley AB, et al. An initial genetic analysis of gemcitabine‐induced high‐grade neutropenia in pancreatic cancer patients in CALGB 80303 (Alliance). Pharmacogenet Genomics. 2019;29(6):123‐131. doi: 10.1097/FPC.0000000000000375 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Thomas F, Motsinger‐Reif AA, Hoskins JM, et al. Methylenetetrahydrofolate reductase genetic polymorphisms and toxicity to 5‐FU‐based chemoradiation in rectal cancer. Br J Cancer. 2011;105(11):1654‐1662. doi: 10.1038/bjc.2011.442 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Zarate R, Rodriguez J, Bandres E, et al. Oxaliplatin, irinotecan and capecitabine as first‐line therapy in metastatic colorectal cancer (mCRC): a dose‐finding study and pharmacogenomic analysis. Br J Cancer. 2010;102(6):987‐994. doi: 10.1038/sj.bjc.6605595 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Kim SY, Kong SY, Shin A, et al. S‐1 plus irinotecan and oxaliplatin for the first‐line treatment of patients with metastatic colorectal cancer: a prospective phase II study and pharmacogenetic analysis. Br J Cancer. 2013;109(6):1420‐1427. doi: 10.1038/bjc.2013.479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Martinez‐Balibrea E, Abad A, Martinez‐Cardus A, et al. UGT1A and TYMS genetic variants predict toxicity and response of colorectal cancer patients treated with first‐line irinotecan and fluorouracil combination therapy. Br J Cancer. 2010;103(4):581‐589. doi: 10.1038/sj.bjc.6605776 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Park SR, Kong SY, Rhee J, et al. Phase II study of a triplet regimen of S‐1 combined with irinotecan and oxaliplatin in patients with metastatic gastric cancer: clinical and pharmacogenetic results. Ann Oncol. 2011;22(4):890‐896. doi: 10.1093/annonc/mdq435 [DOI] [PubMed] [Google Scholar]
- 59. Carlini LE, Meropol NJ, Bever J, et al. UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res. 2005;11(3):1226‐1236. [PubMed] [Google Scholar]
- 60. Hazama S, Mishima H, Tsunedomi R, 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):1662‐1669. doi: 10.1111/cas.12283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Xu CF, Johnson T, Wang X, et al. HLA‐B*57:01 confers susceptibility to Pazopanib‐associated liver injury in patients with cancer. Clin Cancer Res. 2016;22(6):1371‐1377. doi: 10.1158/1078-0432.CCR-15-2044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Choi YJ, Park H, Lee JS, et al. Methotrexate elimination and toxicity: MTHFR 677C>T polymorphism in patients with primary CNS lymphoma treated with high‐dose methotrexate. Hematol Oncol. 2017;35(4):504‐509. doi: 10.1002/hon.2363 [DOI] [PubMed] [Google Scholar]
- 63. Toffoli G, Russo A, Innocenti F, et al. Effect of methylenetetrahydrofolate reductase 677C>T polymorphism on toxicity and homocysteine plasma level after chronic methotrexate treatment of ovarian cancer patients. Int J Cancer. 2003;103(3):294‐299. doi: 10.1002/ijc.10847 [DOI] [PubMed] [Google Scholar]
- 64. Babaoglu MO, Bayar B, Aynacioglu AS, et al. Association of the ABCB1 3435C>T polymorphism with antiemetic efficacy of 5‐hydroxytryptamine type 3 antagonists. Clin Pharmacol Ther. 2005;78(6):619‐626. doi: 10.1016/j.clpt.2005.08.015 [DOI] [PubMed] [Google Scholar]
- 65. Lavanderos MA, Cayun JP, Roco A, et al. Association study among candidate genetic polymorphisms and chemotherapy‐related severe toxicity in testicular cancer patients. Front Pharmacol. 2019;10:206. doi: 10.3389/fphar.2019.00206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Tsuji D, Yokoi M, Suzuki K, et al. Influence of ABCB1 and ABCG2 polymorphisms on the antiemetic efficacy in patients with cancer receiving cisplatin‐based chemotherapy: a TRIPLE pharmacogenomics study. Pharmacogenomics J. 2017;17(5):435‐440. doi: 10.1038/tpj.2016.38 [DOI] [PubMed] [Google Scholar]
- 67. Yokoi M, Tsuji D, Suzuki K, et al. Genetic risk factors for chemotherapy‐induced nausea and vomiting in patients with cancer receiving cisplatin‐based chemotherapy. Support Care Cancer. 2018;26(5):1505‐1513. doi: 10.1007/s00520-017-3974-3 [DOI] [PubMed] [Google Scholar]
- 68. Boora GK, Kulkarni AA, Kanwar R, et al. Association of the Charcot‐Marie‐Tooth disease gene ARHGEF10 with paclitaxel induced peripheral neuropathy in NCCTG N08CA (alliance). J Neurol Sci. 2015;357(1–2):35‐40. doi: 10.1016/j.jns.2015.06.056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Baldwin RM, Owzar K, Zembutsu H, et al. A genome‐wide association study identifies novel loci for paclitaxel‐induced sensory peripheral neuropathy in CALGB 40101. Clin Cancer Res. 2012;18(18):5099‐5109. doi: 10.1158/1078-0432.CCR-12-1590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Pascual T, Apellaniz‐Ruiz M, Pernaut C, et al. Polymorphisms associated with everolimus pharmacokinetics, toxicity and survival in metastatic breast cancer. PLoS One. 2017;12(7):e0180192. doi: 10.1371/journal.pone.0180192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Au A, Aziz Baba A, Goh AS, et al. Association of genotypes and haplotypes of multi‐drug transporter genes ABCB1 and ABCG2 with clinical response to imatinib mesylate in chronic myeloid leukemia patients. Biomed Pharmacother. 2014;68(3):343‐349. doi: 10.1016/j.biopha.2014.01.009 [DOI] [PubMed] [Google Scholar]
- 72. Moeung S, Chevreau C, Marsili S, et al. Pharmacokinetic and Pharmacogenetic study of etoposide in high‐dose protocol (TI‐CE) for advanced germ cell tumors. Pharm Res. 2020;37(7):147. doi: 10.1007/s11095-020-02861-5 [DOI] [PubMed] [Google Scholar]
- 73. Boudou‐Rouquette P, Narjoz C, Golmard JL, et al. Early sorafenib‐induced toxicity is associated with drug exposure and UGTIA9 genetic polymorphism in patients with solid tumors: a preliminary study. PLoS One. 2012;7(8):e42875. doi: 10.1371/journal.pone.0042875 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Qin C, Cao Q, Li P, et al. The influence of genetic variants of sorafenib on clinical outcomes and toxic effects in patients with advanced renal cell carcinoma. Sci Rep. 2016;6:20089. doi: 10.1038/srep20089 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Goey AK, Sissung TM, Peer CJ, et al. Effects of UGT1A1 genotype on the pharmacokinetics, pharmacodynamics, and toxicities of belinostat administered by 48‐hour continuous infusion in patients with cancer. J Clin Pharmacol. 2016;56(4):461‐473. doi: 10.1002/jcph.625 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Low SK, Fukunaga K, Takahashi A, et al. Association study of a functional variant on ABCG2 gene with Sunitinib‐induced severe adverse drug reaction. PLoS One. 2016;11(2):e0148177. doi: 10.1371/journal.pone.0148177 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Kim HR, Park HS, Kwon WS, et al. Pharmacogenetic determinants associated with sunitinib‐induced toxicity and ethnic difference in Korean metastatic renal cell carcinoma patients. Cancer Chemother Pharmacol. 2013;72(4):825‐835. doi: 10.1007/s00280-013-2258-y [DOI] [PubMed] [Google Scholar]
- 78. Kim HS, Kim MK, Chung HH, et al. Genetic polymorphisms affecting clinical outcomes in epithelial ovarian cancer patients treated with taxanes and platinum compounds: a Korean population‐based study. Gynecol Oncol. 2009;113(2):264‐269. doi: 10.1016/j.ygyno.2009.01.002 [DOI] [PubMed] [Google Scholar]
- 79. Narita S, Tsuchiya N, Yuasa T, et al. Outcome, clinical prognostic factors and genetic predictors of adverse reactions of intermittent combination chemotherapy with docetaxel, estramustine phosphate and carboplatin for castration‐resistant prostate cancer. Int J Clin Oncol. 2012;17(3):204‐211. doi: 10.1007/s10147-011-0275-6 [DOI] [PubMed] [Google Scholar]
- 80. De Marchi P, Melendez ME, Laus AC, et al. The role of single‐nucleotide polymorphism (SNPs) in toxicity of induction chemotherapy based on cisplatin and paclitaxel in patients with advanced head and neck cancer. Oral Oncol. 2019;98:48‐52. doi: 10.1016/j.oraloncology.2019.09.013 [DOI] [PubMed] [Google Scholar]
- 81. Sissung TM, Rajan A, Blumenthal GM, et al. Reproducibility of pharmacogenetics findings for paclitaxel in a heterogeneous population of patients with lung cancer. PLoS One. 2019;14(2):e0212097. doi: 10.1371/journal.pone.0212097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Sim S, Bergh J, Hellstrom M, Hatschek T, Xie H. Pharmacogenetic impact of docetaxel on neoadjuvant treatment of breast cancer patients. Pharmacogenomics. 2018;19(16):1259‐1268. doi: 10.2217/pgs-2018-0080 [DOI] [PubMed] [Google Scholar]
- 83. Gandara DR, Kawaguchi T, Crowley J, et al. Japanese‐US common‐arm analysis of paclitaxel plus carboplatin in advanced non‐small‐cell lung cancer: a model for assessing population‐related pharmacogenomics. J Clin Oncol. 2009;27(21):3540‐3546. doi: 10.1200/JCO.2008.20.8793 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Marsh S, Paul J, King CR, Gifford G, McLeod HL, Brown R. Pharmacogenetic assessment of toxicity and outcome after platinum plus taxane chemotherapy in ovarian cancer: the Scottish randomised trial in ovarian cancer. J Clin Oncol. 2007;25(29):4528‐4535. doi: 10.1200/JCO.2006.10.4752 [DOI] [PubMed] [Google Scholar]
- 85. Mysona D, Dorr K, Ward A, Shaver E, Rungruang B, Ghamande S. Pharmacogenetics as a predictor chemotherapy induced peripheral neuropathy in gynecologic cancer patients treated with Taxane‐based chemotherapy. Gynecol Oncol. 2023;168:114‐118. doi: 10.1016/j.ygyno.2022.10.021 [DOI] [PubMed] [Google Scholar]
- 86. Petty WJ, Knight SN, Mosley L, et al. A pharmacogenomic study of docetaxel and gemcitabine for the initial treatment of advanced non‐small cell lung cancer. J Thorac Oncol. 2007;2(3):197‐202. doi: 10.1097/JTO.0b013e318031cd89 [DOI] [PubMed] [Google Scholar]
- 87. Malhotra U, Mukherjee S, Fountzilas C, et al. Pralatrexate in combination with Oxaliplatin in advanced Esophagogastric cancer: a phase II trial with predictive molecular correlates. Mol Cancer Ther. 2020;19(1):304‐311. doi: 10.1158/1535-7163.MCT-19-0240 [DOI] [PubMed] [Google Scholar]
- 88. Zhu XL, Han X, Xin XF, Song HJ, Guo H. Correlations of analgesic dosage of morphine with SLC6A4 gene polymorphisms in patients with lung cancer. Eur Rev Med Pharmacol Sci. 2020;24(9):5046‐5052. doi: 10.26355/eurrev_202005_21197 [DOI] [PubMed] [Google Scholar]
- 89. Stock W, Diouf B, Crews KR, et al. An inherited genetic variant in CEP72 promoter predisposes to vincristine‐induced peripheral neuropathy in adults with acute lymphoblastic leukemia. Clin Pharmacol Ther. 2017;101(3):391‐395. doi: 10.1002/cpt.506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Eadon MT, Desta Z, Levy KD, et al. Implementation of a pharmacogenomics consult service to support the INGENIOUS trial. Clin Pharmacol Ther. 2016;100(1):63‐66. doi: 10.1002/cpt.347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Gottesman O, Scott SA, Ellis SB, et al. The CLIPMERGE PGx program: clinical implementation of personalized medicine through electronic health records and genomics‐pharmacogenomics. Clin Pharmacol Ther. 2013;94(2):214‐217. doi: 10.1038/clpt.2013.72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Pulley JM, Denny JC, Peterson JF, et al. Operational implementation of prospective genotyping for personalized medicine: the design of the Vanderbilt PREDICT project. Clin Pharmacol Ther. 2012;92(1):87‐95. doi: 10.1038/clpt.2011.371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Weitzel KW, Alexander M, Bernhardt BA, et al. The IGNITE network: a model for genomic medicine implementation and research. BMC Med Genet. 2016;9:1. doi: 10.1186/s12920-015-0162-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Xiao R, Boehnke M. Quantifying and correcting for the winner's curse in genetic association studies. Genet Epidemiol. 2009;33(5):453‐462. doi: 10.1002/gepi.20398 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Goetz MP, Rae JM, Suman VJ, et al. Pharmacogenetics of tamoxifen biotransformation is associated with clinical outcomes of efficacy and hot flashes. J Clin Oncol. 2005;23(36):9312‐9318. doi: 10.1200/JCO.2005.03.3266 [DOI] [PubMed] [Google Scholar]
- 96. Glewis S, Alexander M, Khabib MNH, et al. A systematic review and meta‐analysis of toxicity and treatment outcomes with pharmacogenetic‐guided dosing compared to standard of care BSA‐based fluoropyrimidine dosing. Br J Cancer. 2022;127(1):126‐136. doi: 10.1038/s41416-022-01779-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Knikman JE, Wilting TA, Lopez‐Yurda M, et al. Survival of patients with cancer with DPYD variant alleles and dose‐individualized Fluoropyrimidine therapy‐a matched‐pair analysis. J Clin Oncol. 2023;41:5421. doi: 10.1200/JCO.22.02780 [DOI] [PubMed] [Google Scholar]
- 98. Glewis S, Alexander M, Lingaratnam S, et al. Pharmacogenomics guided dosing for fluoropyrimidine and irinotecan chemotherapies for patients with cancer (PACIFIC‐PGx): study protocol of a multicentre clinical trial. Acta Oncol. 2022;61(9):1136‐1139. doi: 10.1080/0284186X.2022.2109423 [DOI] [PubMed] [Google Scholar]
- 99. Hodroj K, Barthelemy D, Lega JC, et al. Issues and limitations of available biomarkers for fluoropyrimidine‐based chemotherapy toxicity, a narrative review of the literature. ESMO Open. 2021;6(3):100125. doi: 10.1016/j.esmoop.2021.100125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Kim W, Cho YA, Kim DC, Lee KE. Elevated risk of Fluoropyrimidine‐associated toxicity in European patients with DPYD genetic polymorphism: a systematic review and meta‐analysis. J Pers Med. 2022;12(2):225. doi: 10.3390/jpm12020225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. National Center for Biotechnology Information: dbSNP:rs<enter number>. Accessed May 31, 2023. https://www.ncbi.nlm.nih.gov/snp/
- 102. Patil VM, Noronha V, Joshi A, et al. Dihydropyrimidine dehydrogenase mutation in neoadjuvant chemotherapy in head and neck cancers: myth or reality? South Asian J Cancer. 2016;5(4):182‐185. doi: 10.4103/2278-330X.195338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Offer SM, Lee AM, Mattison LK, Fossum C, Wegner NJ, Diasio RB. A DPYD variant (Y186C) in individuals of African ancestry is associated with reduced DPD enzyme activity. Clin Pharmacol Ther. 2013;94(1):158‐166. doi: 10.1038/clpt.2013.69 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Jennings BA, Kwok CS, Willis G, Matthews V, Wawruch P, Loke YK. Functional polymorphisms of folate metabolism and response to chemotherapy for colorectal cancer, a systematic review and meta‐analysis. Pharmacogenet Genomics. 2012;22(4):290‐304. doi: 10.1097/FPC.0b013e328351875d [DOI] [PubMed] [Google Scholar]
- 105. Saif MW, Hachem H, Purvey S, et al. Pharmacogenetic variants in the DPYD and TYMS genes are clinically significant predictors of Fluoropyrimidine toxicity: are we ready for use in our clinical practice. Arch Pharmacol Ther. 2020;2(1):6‐8. [PMC free article] [PubMed] [Google Scholar]
- 106. Cecchin E, Innocenti F, D'Andrea M, et al. 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(15):2457‐2465. doi: 10.1200/JCO.2008.19.0314 [DOI] [PubMed] [Google Scholar]
- 107. Tagen M, Zhuang Y, Zhang F, et al. P‐glycoprotein, but not multidrug resistance protein 4, plays a role in the systemic clearance of irinotecan and SN‐38 in mice. Drug Metab Lett. 2010;4(4):195‐201. doi: 10.2174/187231210792928251 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Mathijssen RH, Marsh S, Karlsson MO, et al. Irinotecan pathway genotype analysis to predict pharmacokinetics. Clin Cancer Res. 2003;9(9):3246‐3253. [PubMed] [Google Scholar]
- 109. Salvador‐Martin S, Garcia‐Gonzalez X, Garcia MI, et al. Clinical utility of ABCB1 genotyping for preventing toxicity in treatment with irinotecan. Pharmacol Res. 2018;136:133‐139. doi: 10.1016/j.phrs.2018.08.026 [DOI] [PubMed] [Google Scholar]
- 110. Flaherty KT, Lorusso PM, Demichele A, et al. Phase I, dose‐escalation trial of the oral cyclin‐dependent kinase 4/6 inhibitor PD 0332991, administered using a 21‐day schedule in patients with advanced cancer. Clin Cancer Res. 2012;18(2):568‐576. doi: 10.1158/1078-0432.CCR-11-0509 [DOI] [PubMed] [Google Scholar]
- 111. Verheijen RB, Yu H, Schellens JHM, Beijnen JH, Steeghs N, Huitema ADR. Practical recommendations for therapeutic drug monitoring of kinase inhibitors in oncology. Clin Pharmacol Ther. 2017;102(5):765‐776. doi: 10.1002/cpt.787 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Touma JA, McLachlan AJ, Gross A. The role of ethnicity in personalized dosing of small molecule tyrosine kinase inhibitors used in oncology. Translat Cancer Res. 2017;6:S1558‐S1591. doi: 10.21037/tcr.2017.09.09 [DOI] [Google Scholar]
- 113. Dandara C, Masimirembwa C, Haffani YZ, et al. African pharmacogenomics consortium: consolidating pharmacogenomics knowledge, capacity development and translation in Africa: consolidating pharmacogenomics knowledge, capacity development and translation in Africa. AAS Open Res. 2019;2:19. doi: 10.12688/aasopenres.12965.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Lopez‐Cortes A, Esperon P, Martinez MF, et al. Editorial: Pharmacogenetics and pharmacogenomics in Latin America: ethnic variability, new insights in advances and perspectives: a RELIVAF‐CYTED initiative, Volume II. Front Pharmacol. 2023;14:1211712. doi: 10.3389/fphar.2023.1211712 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Runcharoen C, Fukunaga K, Sensorn I, et al. Prevalence of pharmacogenomic variants in 100 pharmacogenes among southeast Asian populations under the collaboration of the southeast Asian pharmacogenomics research network (SEAPharm). Hum Genome Var. 2021;8(1):7. doi: 10.1038/s41439-021-00135-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Samarasinghe SR, Hoy W, Jadhao S, McMorran BJ, Guchelaar HJ, Nagaraj SH. The pharmacogenomic landscape of an indigenous Australian population. Front Pharmacol. 2023;14:1180640. doi: 10.3389/fphar.2023.1180640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Davis BH, Limdi NA. Translational pharmacogenomics: discovery, evidence synthesis and delivery of race‐conscious medicine. Clin Pharmacol Ther. 2021;110(4):909‐925. doi: 10.1002/cpt.2357 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1.
Data Availability Statement
All data generated or analyzed during this study are included in this published article [and its supplementary information files].