Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Colin F. Spraggs is active.

Publication


Featured researches published by Colin F. Spraggs.


Journal of Clinical Oncology | 2011

HLA-DQA1*02:01 Is a Major Risk Factor for Lapatinib-Induced Hepatotoxicity in Women With Advanced Breast Cancer

Colin F. Spraggs; Laura R. Budde; Linda P. Briley; Nan Bing; Charles J. Cox; Karen S. King; John C. Whittaker; Vincent Mooser; Alaknanda J. Preston; S. Stein; Lon R. Cardon

PURPOSE Hepatobiliary adverse events (AEs) have been observed in a small proportion of patients with metastatic breast cancer (MBC) treated with lapatinib. This study sought to identify gene variants associated with lapatinib-induced ALT elevation and hepatobiliary AEs. PATIENTS AND METHODS A two-stage pharmacogenetic investigation of ALT elevation was conducted in lapatinib-treated patients with MBC. Exploratory marker identification evaluated classical HLA alleles, candidate genes, and genome-wide screening in 37 cases with ALT greater than 3 times the upper limit of normal (ULN) and 286 controls with ALT ≤ 1× ULN, selected from 901 lapatinib-treated patients in 12 trials. Markers achieving prespecified association thresholds were progressed to an independent confirmatory data set of 24 ALT cases and 155 controls selected from a subsequent trial of 374 lapatinib-treated patients. RESULTS Of 58 variants associated with ALT elevation in the exploratory data set, four exceeded the prespecified significance threshold in the confirmatory analysis. These variants reside in the same MHC genomic locus and include HLA-DQA1*02:01. In the confirmatory study, DQA1*02:01 allele carriage was present in 71% of ALT cases and in 21% of controls (P < .001; odds ratio, 9.0; 95% CI, 3.2 to 27.4). As a predictor of liver safety risk in ALT cases versus noncases, DQA1*02:01 had negative and positive predictive values of 0.97 (95% CI, 0.95 to 0.99) and 0.17 (95% CI 0.10 to 0.26), respectively. CONCLUSION These results support a role for immune mechanisms in lapatinib-induced hepatotoxicity. Further work is required to determine whether testing for DQA1*02:01 allele carriage is clinically useful in managing liver safety risk during lapatinib treatment.


Journal of Clinical Oncology | 2011

Pazopanib Efficacy in Renal Cell Carcinoma: Evidence for Predictive Genetic Markers in Angiogenesis-Related and Exposure-Related Genes

C. Xu; N. Bing; H. A. Ball; Dilip Rajagopalan; Cora N. Sternberg; Thomas E. Hutson; Paul de Souza; Zhengyu G. Xue; Lauren McCann; Karen S. King; Leigh J. Ragone; John C. Whittaker; Colin F. Spraggs; Lon R. Cardon; Vincent Mooser; Lini Pandite

PURPOSE Pazopanib, an oral angiogenesis inhibitor, is approved for the treatment of advanced renal cell carcinoma (RCC). Response to pazopanib monotherapy varies between patients, and no validated biomarkers predictive of treatment outcome have been identified. We tested the hypothesis that this variability is partially dependent on germline genetic variants that may affect pazopanib exposure or angiogenesis pathways. PATIENTS AND METHODS Twenty-seven functional polymorphisms within 13 genes were evaluated in 397 patients with RCC. Genetic association with progression-free survival (PFS) and objective response rate (RR) was analyzed using the Cox proportional hazards model and proportional odds model, respectively. RESULTS Three polymorphisms in IL8 and HIF1A and five polymorphisms in HIF1A, NR1I2, and VEGFA showed nominally significant association (P ≤ .05) with PFS and RR, respectively. Compared with the wild-type AA genotype (median PFS, 48 weeks), the IL8 2767TT variant genotype showed inferior PFS (27 weeks, P = .009). The HIF1A 1790AG genotype was associated with inferior PFS and reduced RR, compared with the wild-type GG genotype (median PFS, 20 v 44 weeks; P = .03; RR, 30% v 43%, P = .02). Reductions in RR were detected for the NR1I2 -25385TT genotype, compared with the wild-type CC genotype (37% v 50%, P = .03), and for the VEGFA -1498CC genotype compared with the TT genotypes (33% v 51%). CONCLUSION Germline variants in angiogenesis- and exposure-related genes may predict treatment response to pazopanib monotherapy in patients with RCC. If validated, these markers may explain why certain patients fail antiangiogenesis therapy and they may support the use of alternative strategies to circumvent this issue.


British Journal of Cancer | 2010

Pazopanib-induced hyperbilirubinemia is associated with Gilbert's syndrome UGT1A1 polymorphism

Xu Cf; Reck Bh; Zhengyu Xue; Huang L; Baker Kl; Chen M; Chen Ep; Ellens He; Mooser Ve; Cardon Lr; Colin F. Spraggs; Lini Pandite

Background:Pazopanib has shown clinical activity against multiple tumour types and is generally well tolerated. However, isolated elevations in transaminases and bilirubin have been observed. This study examined polymorphisms in molecules involved in pharmacokinetic and pharmacodynamic pathways of pazopanib and their association with hepatic dysfunction.Methods:Twenty-eight polymorphisms in 11 genes were evaluated in pazopanib-treated renal cell carcinoma patients. An exploratory analysis was conducted in 116 patients from a phase II study; a replication study was conducted in 130 patients from a phase III study.Results:No polymorphisms were associated with alanine aminotransferase elevation. The Gilberts uridine-diphosphoglucuronate glucuronosyltransferase 1A1 (UGT1A1) TA-repeat polymorphism was significantly associated with pazopanib-induced hyperbilirubinemia in the phase II study. This association was replicated in the phase III study (P<0.01). Patients with TA6/TA6, TA6/TA7, and TA7/TA7 genotypes experienced median bilirubin increases of 0.31, 0.37, and 0.71 × upper limit of the normal range (ULN), respectively. Of the 38 patients with hyperbilirubinemia (⩾1.5 × ULN), 32 (84%) were either TA7 homozygotes (n=18) or TA7 heterozygotes (n=14). For TA7 homozygotes, the odds ratio (95% CI) for developing hyperbilirubinemia was 13.1 (5.3–32.2) compared with other genotypes.Conclusions:The UGT1A1 polymorphism is frequently associated with pazopanib-induced hyperbilirubinemia. These data suggest that some instances of isolated hyperbilirubinemia in pazopanib-treated patients are benign manifestations of Gilberts syndrome, thus supporting continuation of pazopanib monotherapy in this setting.


European Urology | 2012

Potential impact of adding genetic markers to clinical parameters in predicting prostate biopsy outcomes in men following an initial negative biopsy: findings from the REDUCE trial.

A. Karim Kader; Jielin Sun; Brian H. Reck; Paul Newcombe; Seong Tae Kim; Fang-Chi Hsu; Ralph B. D’Agostino; Sha Tao; Zheng Zhang; Aubrey R. Turner; Greg T. Platek; Colin F. Spraggs; John C. Whittaker; Brian R. Lane; William B. Isaacs; Deborah A. Meyers; Eugene R. Bleecker; Frank M. Torti; Jeffery M. Trent; John D. McConnell; S. Lilly Zheng; Lynn D. Condreay; Roger S. Rittmaster; Jianfeng Xu

BACKGROUND Several germline single nucleotide polymorphisms (SNPs) have been consistently associated with prostate cancer (PCa) risk. OBJECTIVE To determine whether there is an improvement in PCa risk prediction by adding these SNPs to existing predictors of PCa. DESIGN, SETTING, AND PARTICIPANTS Subjects included men in the placebo arm of the randomized Reduction by Dutasteride of Prostate Cancer Events (REDUCE) trial in whom germline DNA was available. All men had an initial negative prostate biopsy and underwent study-mandated biopsies at 2 yr and 4 yr. Predictive performance of baseline clinical parameters and/or a genetic score based on 33 established PCa risk-associated SNPs was evaluated. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Area under the receiver operating characteristic curves (AUC) were used to compare different models with different predictors. Net reclassification improvement (NRI) and decision curve analysis (DCA) were used to assess changes in risk prediction by adding genetic markers. RESULTS AND LIMITATIONS Among 1654 men, genetic score was a significant predictor of positive biopsy, even after adjusting for known clinical variables and family history (p = 3.41 × 10(-8)). The AUC for the genetic score exceeded that of any other PCa predictor at 0.59. Adding the genetic score to the best clinical model improved the AUC from 0.62 to 0.66 (p<0.001), reclassified PCa risk in 33% of men (NRI: 0.10; p=0.002), resulted in higher net benefit from DCA, and decreased the number of biopsies needed to detect the same number of PCa instances. The benefit of adding the genetic score was greatest among men at intermediate risk (25th percentile to 75th percentile). Similar results were found for high-grade (Gleason score ≥ 7) PCa. A major limitation of this study was its focus on white patients only. CONCLUSIONS Adding genetic markers to current clinical parameters may improve PCa risk prediction. The improvement is modest but may be helpful for better determining the need for repeat prostate biopsy. The clinical impact of these results requires further study.


Journal of Clinical Oncology | 2014

Prospective Validation of HLA-DRB1*07:01 Allele Carriage As a Predictive Risk Factor for Lapatinib-Induced Liver Injury

Daniel J. Schaid; Colin F. Spraggs; Shannon K. McDonnell; Laura Rae Parham; Charles J. Cox; Bent Ejlertsen; Dianne M. Finkelstein; Erica Rappold; Joan Curran; Lon R. Cardon; Paul E. Goss

PURPOSE Liver injury is a serious adverse event leading to permanent discontinuation of lapatinib in affected patients. This study aimed to validate previously associated major histocompatibility complex (MHC) variants as predictors of risk of liver injury by using a large, randomized, placebo-controlled trial of lapatinib in human epidermal growth factor receptor 2-positive, early-stage breast cancer (Tykerb Evaluation After Chemotherapy [TEACH]: Lapatinib Versus Placebo In Women With Early-Stage Breast Cancer). PATIENTS AND METHODS The frequency of ALT elevation cases was compared among four MHC variants in 1,194 patients randomly assigned to lapatinib. Cumulative ALT elevation time courses during treatment were also compared between carriers and noncarriers of specified MHC variants. RESULTS In lapatinib-treated patients, there was a significant difference in ALT case incidence between HLA carriers and noncarriers. The highly correlated alleles HLA-DRB1*07:01 and HLA-DQA1*02:01 (study frequency, 22.4%) were associated with ALT elevation (odds ratio, 14) between cases (n = 37) and controls (n = 1,071). These associations strengthened at higher ALT elevation thresholds and in Hys Law cases. In lapatinib-treated patients, the overall risk for National Cancer Institute-Common Terminology Criteria for Adverse Events grade 3 ALT elevation (> 5× upper limit of normal) was 2.1%; HLA allele carriers had an increased risk of 7.7%; in noncarriers, risk was reduced to 0.5%, comparable to ALT elevation for all patients receiving placebo. The increase in ALT case incidence in the lapatinib arm showed no evidence of plateau during 1 year of lapatinib treatment. CONCLUSION These results validate HLA-DRB1*07:01 allele carriage as a predictor of increased risk of lapatinib-induced liver injury and implicate an immune pathology. The HLA association could support clinical management of patients experiencing hepatotoxicity during lapatinib treatment.


Journal of Hepatology | 2011

Association of the hemochromatosis gene with pazopanib-induced transaminase elevation in renal cell carcinoma

C. Xu; Brian H. Reck; Vicki L. Goodman; Zhengyu Xue; Lingkang Huang; Michael R. Barnes; Beena T. Koshy; Colin F. Spraggs; Vincent Mooser; Lon R. Cardon; Lini Pandite

BACKGROUND & AIMS Pazopanib has demonstrated clinical benefit in patients with advanced renal cell carcinoma (RCC) and is generally well tolerated. However, transaminase elevations have commonly been observed. This 2-stage study sought to identify genetic determinants of alanine transaminase (ALT) elevations in pazopanib-treated white patients with RCC. METHODS Data from two separate clinical studies were used to examine the association of genetic polymorphisms with maximum on-treatment ALT levels. RESULTS Of 6852 polymorphisms in 282 candidate genes examined in an exploratory dataset of 115 patients, 92 polymorphisms in 40 genes were significantly associated with ALT elevation (p<0.01). Two markers (rs2858996 and rs707889) in the HFE gene, which are not yet known to be associated with hemochromatosis, showed evidence for replication. Because of multiple comparisons, there was a 12% likelihood the replication occurred by chance. These two markers demonstrated strong linkage disequilibrium (r(2)=0.99). In the combined dataset, median (25-75th percentile) maximum ALT values were 1.2 (0.7-1.9), 1.1 (0.8-2.5), and 5.4 (1.9-7.6)×ULN for rs2858996 GG (n=148), GT (n=82), and TT (n=1 2) genotypes, respectively. All 12 TT patients had a maximum ALT>ULN, and 8 (67%) had ALT≥3×ULN. The odds ratio (95% CI) for ALT≥3×ULN for TT genotype was 39.7 (2.2-703.7) compared with other genotypes. As a predictor of ALT≥3×ULN, the TT genotype had a negative predictive value of 0.83 and positive predictive value of 0.67. No TT patients developed liver failure. CONCLUSIONS The rs2858996/rs707889 polymorphisms in the HFE gene may be associated with reversible ALT elevation in pazo-panib-treated patients with RCC.


Clinical Pharmacology & Therapeutics | 2012

Lapatinib-Induced Liver Injury Characterized by Class II HLA and Gilbert's Syndrome Genotypes

Colin F. Spraggs; L R Parham; Christine M. Hunt; C T Dollery

Lapatinib is a clinically important component of the treatment for HER2‐positive metastatic breast cancer and has an acceptable safety profile. Lapatinib‐associated Hys Law cases have been characterized using human leukocyte antigen (HLA) DQA1*02:01/DRB1*07:01 and Gilberts syndrome UGT1A1*28/*28 genotypes. The HLA‐positive cases had higher alanine aminotransferase (ALT) elevation, whereas the HLA‐negative cases had a higher incidence of Gilberts syndrome. The findings of our study, which extend this HLA association to lapatinib‐associated serious liver injury, emphasize the importance of Gilberts syndrome in the interpretation of Hys Law and may lead to methods for enhancing patient safety.


Pharmacogenomics | 2013

Genetic characterization to improve interpretation and clinical management of hepatotoxicity caused by tyrosine kinase inhibitors

Colin F. Spraggs; C. Xu; Christine M. Hunt

Tyrosine kinase inhibitors (TKIs) represent important therapeutic alternatives to, or combinations with, traditional cytotoxic chemotherapy. Despite their selective molecular targeting and demonstrated clinical benefit, TKIs produce a range of serious adverse events, including drug-induced liver injury, that require careful patient management to maintain treatment benefit without harm. Genetic characterization of serious adverse events can identify mechanisms of injury and improve safety risk management. This review presents pharmacogenetic comparisons of two approved TKIs, lapatinib and pazopanib, which reveal different mechanisms of injury and inform the characteristics and risk of serious liver injury in treated patients. The data presented demonstrate the utility of genetic studies to investigate drug-induced liver injury and potentially support its management in patients.


British Journal of Cancer | 2015

IL8 polymorphisms and overall survival in pazopanib- or sunitinib-treated patients with renal cell carcinoma

Chun-Feng Xu; Toby Johnson; Jesús García-Donas; Toni K. Choueiri; Cora N. Sternberg; Ian D. Davis; N. Bing; Keith C. Deen; Zhengyu Xue; Lauren McCann; Emilio Esteban; John C. Whittaker; Colin F. Spraggs; Cristina Rodríguez-Antona; Lini Pandite; Robert J. Motzer

Background:We evaluated germline single nucleotide polymorphisms (SNPs) for association with overall survival (OS) in pazopanib- or sunitinib-treated patients with advanced renal cell carcinoma (aRCC).Methods:The discovery analysis tested 27 SNPs within 13 genes from a phase III pazopanib trial (N=241, study 1). Suggestive associations were then pursued in two independent datasets: a phase III trial (COMPARZ) comparing pazopanib vs sunitinib (N=729, study 2) and an observational study of sunitinib-treated patients (N=89, study 3).Results:In study 1, four SNPs showed nominally significant association (P≤0.05) with OS; two of these SNPs (rs1126647, rs4073) in IL8 were associated (P≤0.05) with OS in study 2. Because rs1126647 and rs4073 were highly correlated, only rs1126647 was evaluated in study 3, which also showed association (P≤0.05). In the combined data, rs1126647 was associated with OS after conservative multiple-test adjustment (P=8.8 × 10−5; variant vs reference allele hazard ratio 1.32, 95% confidence interval: 1.15–1.52), without evidence for heterogeneity of effects between studies or between pazopanib- and sunitinib-treated patients.Conclusions:Variant alleles of IL8 polymorphisms are associated with poorer survival outcomes in pazopanib- or sunitinib-treated patients with aRCC. These findings provide insight in aRCC prognosis and may advance our thinking in development of new therapies.


Annals of Oncology | 2012

Concomitant use of pazopanib and simvastatin increases the risk of transaminase elevations in patients with cancer

C. Xu; Z. Xue; N. Bing; Karen S. King; Lauren McCann; P. de Souza; V. L. Goodman; Colin F. Spraggs; Vincent Mooser; Lini Pandite

Alanine aminotransferase (ALT) elevations were reported in oncology trials of the oral angiogenesis inhibitor pazopanib [1]. Statins, which are widely used to treat hypercholesterolemia [2], are also associated with ALT elevations. As pazopanib and statins are substrates for the same key metabolizing enzymes [e.g. cytochrome P450 3A4 (CYP3A4)] and drug transporters [e.g. P-glycoprotein (Pgp) and breast cancer resistance protein (BCRP)] [2–3], it is plausible that concomitant administration of pazopanib and statins may alter their systemic and/or hepatic exposures, leading to increased toxicities such as liver injury. Using individual patient data from 11 pazopanib clinical studies for the treatment of cancer (n = 976), we evaluated the effects of concomitant pazopanib and statin use on the incidence of ALT elevation [≥3 × upper limit of normal (ULN)]. The incidence of ALT≥ 3 × ULN was 21% in patients who received both pazopanib and any statin and 14% in patients who did not receive statins (P = 0.10; Table 1). Simvastatin and atorvastatin were the two most commonly used statins in this patient population. The incidence of ALT≥ 3 × ULN (27%) was significantly higher in simvastatin users than in patients who did not receive statins (P = 0.04; Table 1). Although the incidence of ALT≥ 3 × ULN was also higher in atorvastatin users (17%) than in patients who did not receive statins (14%), this difference did not reach statistical significance (P = 0.59). Among patients with ALT≥ 3 × ULN who received both pazopanib and simvastatin, ALT recovery to <2.5 × ULN (grade ≤1 by common toxicity criteria v3) was documented for 10/11 patients (91%) after either (i) no alteration for pazopanib and simvastatin therapy (n = 2); (ii) discontinuation of simvastatin only (n = 2); (iii) discontinuation of pazopanib only (n = 4); or (iv) discontinuation of both simvastatin and pazopanib (n = 2). There were insufficient follow-up ALT data to assess recovery for the one remaining patient after pazopanib was discontinued. The ALT recovery rate (to <2.5 × ULN) for patients receiving concomitant pazopanib and simvastatin was comparable with that seen in the larger group of pazopanib-treated study patients with renal cell carcinoma who had ALT≥ 3 × ULN (96/106, 91%) [4]. Exploratory pharmacogenetic analysis revealed that the ABCG2 (BCRP) 421C>A polymorphism may be associated with ALT elevation in patients taking pazopanib and simvastatin. Those with the variant 421C>A allele had a higher incidence of ALT≥ 3 × ULN (5/7, 71%) compared with patients with wild-type genotype (2/21, 10%; odds ratio = 19.6, 95% confidence interval 1.9–231.6; P = 0.004). This polymorphism was not associated with ALT elevations in pazopanib-treated patients without concurrent use of statins. This study showed that concomitant use of pazopanib and simvastatin increased the risk of transaminase elevations in patients with cancer. In addition to implementing the recommended dose modification guidelines for pazopanib, discontinuation of simvastatin should be considered to manage the risk of liver injury in cancer patients receiving both medications. Furthermore, hepatotoxicity/transaminase elevations have been reported in patients receiving other multi-tyrosine kinase inhibitors (e.g. imatinib, erlotinib, lapatinib, nilotinib, and sunitinib) that are also metabolized primarily by CYP3A4 [5]. The effect of concomitant administration of these tyrosine kinase inhibitors and statins on potential hepatotoxicity remains to be determined.

Collaboration


Dive into the Colin F. Spraggs's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge