Skip to main content
The Journal of Molecular Diagnostics : JMD logoLink to The Journal of Molecular Diagnostics : JMD
. 2019 May;21(3):491–502. doi: 10.1016/j.jmoldx.2019.01.009

Analytical Validation of Variants to Aid in Genotype-Guided Therapy for Oncology

Marelize Swart , Wesley M Stansberry , Victoria M Pratt †,, Elizabeth B Medeiros , Patrick J Kiel , Fei Shen , Bryan P Schneider , Todd C Skaar
PMCID: PMC6504676  PMID: 30794985

Abstract

The Clinical Laboratory Improvement Amendments of 1988 require that pharmacogenetic genotyping methods need to be established according to technical standards and laboratory practice guidelines before testing can be offered to patients. Testing methods for variants in ABCB1, CBR3, COMT, CYP3A7, C8ORF34, FCGR2A, FCGR3A, HAS3, NT5C2, NUDT15, SBF2, SEMA3C, SLC16A5, SLC28A3, SOD2, TLR4, and TPMT were validated in a Clinical Laboratory Improvement Amendments–accredited laboratory. Because no known reference materials were available, existing DNA samples were used for the analytical validation studies. Pharmacogenetic testing methods developed here were shown to be accurate and 100% analytically sensitive and specific. Other Clinical Laboratory Improvement Amendments–accredited laboratories interested in offering pharmacogenetic testing for these genetic variants, related to genotype-guided therapy for oncology, could use these publicly available samples as reference materials when developing and validating new genetic tests or refining current assays.


Genetic variants exist in genes coding for enzymes that are targets of oncology medications or responsible for their metabolism and transport. Pharmacogenetic tests are used to assess whether an individual has the variant allele for these known genetic changes and provide information on risk of toxicity or inefficacy to assist a patient's medical care team in developing therapeutic strategies. For example, the cytotoxic agent 5-fluorouracil undergoes fluoropyrimidine catabolism facilitated by dihydropyrimidine dehydrogenase. Individuals with reduced-function or no-function variants in the DPYD gene (codes for dihydropyrimidine dehydrogenase) have reduced activity of dihydropyrimidine dehydrogenase, reduced 5-fluorouracil clearance, increased half-life, and profound dose-related toxicities (eg, mucositis, diarrhea, neutropenia, and neurotoxicity). Treatment outcomes can be improved by testing for the DPYD variants and then following recommendations according to the Clinical Pharmacogenetics Implementation Consortium (https://cpicpgx.org, last accessed October 29, 2018) guidelines, which suggest a reduction in the dose by 25% to 50% or avoiding 5-fluorouracil depending on the specific DPYD genetic variants present.1 Pharmacogenetic tests can be used to improve patient care only if the test is analytically and clinically valid.

The U.S. Food and Drug Administration includes pharmacogenetic test information in drug labels for several approved oncology medications, including belinostat (UGT1A1), irinotecan (UGT1A1), nilotinib (UGT1A1), pazopanib (UGT1A1 and HLA-B), capecitabine (DPYD), cisplatin (TPMT), mercaptopurine (TPMT), and thioguanine (TPMT). The suggestion that patients should be tested for genetic variants in the genes included in drug labels results in the need for more clinical laboratories with the ability to validate and perform pharmacogenetic testing. The Clinical Laboratory Improvement Amendments of 1988 were developed to regulate all facilities or sites in the United States that test human specimens for health assessment or to diagnose, prevent, or treat disease. Pharmacogenetic tests need to be established according to the technical standards and laboratory practice guidelines required by the Clinical Laboratory Improvement Amendments before testing can be offered to patients.

To achieve regulatory requirements and meet best practice standards, the testing laboratories often will use reference materials for assay development and validation, quality control, and proficiency testing. Genomic DNA samples from cell lines or remaining de-identified patient material are used regularly to develop and validate assays. The CDC established the Genetic Testing Reference Material Coordination Program in 2010 to address the need for characterized genomic DNA reference materials. Several DNA samples from the Coriell Cell Repository (Camden, NJ) have been tested for genetic variants in five commonly tested genes: CYP2D6, CYP2C19, CYP2C9, VKORC1, and UGT1A1.2, 3 As more genetic variants are established as markers of toxicity or inefficacy to cytotoxic agents, more pharmacogenetic testing methods can be validated to offer testing to patients with cancer. This study provides the rationale for chosen genes and variants as well as the analytical validation of genotyping methods for pharmacogenetic variants. For analytical validation, approximately 200 Coriell DNA samples for the variants of which methods were being validated were screened and Sanger sequencing was used as an orthogonal method on a subset of samples both positive and negative for the variants of interest. All of the genes included in the analytical validation are involved in metabolism and transport of medicines (ABCB1, CBR3, CYP3A7, SLC16A5, SLC28A3, TPMT, NT5C2, and NUDT15), are targets of medications (FCGR2A and FCGR3A), or have an unclear role (COMT, C8ORF34, HAS3, SBF2, SEMA3C, SOD2, and TLR4), in a Clinical Laboratory Improvement Amendments–accredited laboratory, related to genotype-guided therapy for oncology.

Materials and Methods

Selection of Variants

Oncology pharmacogenetic literature was reviewed to select 27 clinically relevant genetic variants in 17 genes that have been associated with interindividual variability in efficacy or toxicity of cytotoxic agents. Genetic variants in ABCB1 were selected because ABCB1 codes for the drug transporter P-glycoprotein and these variants are associated with variability in achieving complete control of chemotherapy-induced nausea and vomiting when using ondansetron.4, 5, 6 The CBR3 gene encodes for a carbonyl reductase that is involved in metabolism of anthracyclines. The selected variant is associated with an increased risk of anthracycline-induced cardiomyopathy in pediatric patients.7, 8, 9 Similarly, the HAS3 and SLC28A3 variants were included for their role in the risk of anthracycline-induced cardiomyopathy.10, 11, 12, 13, 14, 15, 16 Although TPMT is involved in metabolism of thiopurines, the particular variant included in this study is associated with an increased risk of cisplatin-induced hearing impairment. Similarly, the COMT genetic variants were included in this study based on studies reporting that the variants play a role in cisplatin-induced hearing impairment in pediatric patients with medulloblastoma, neuroblastoma, or osteosarcoma.17, 18, 19, 20, 21, 22 Similar to SLC28A3, SLC16A5 codes for a protein that is a member of the solute carrier transporter superfamily. In a recent in vitro study, cisplatin induced expression of SLC16A5 in a dose-dependent manner. The selected SLC16A5 variant was identified as a marker of hearing loss in a group of testicular cancer patients treated with cisplatin-containing chemotherapy.23 Many clinically used medications are metabolized by CYP3A enzymes including CYP3A7, which is expressed in a fraction of adult human livers. The CYP3A7*1C allele is associated with lower urinary unconjugated estrogen metabolite levels and an increased risk of mortality among individuals with breast cancer treated with medicines that are CYP3A substrates.24, 25 A genome-wide association study among Korean individuals with advanced non–small-cell lung cancer receiving irinotecan plus cisplatin reported associations between the SEMA3C variants and an increased risk of grade 4 neutropenia and the C8orf34 variant and increased risk of grade 3 diarrhea.26 Genetic variants in FCGR2A and FCGR3A were included because these genes code for fragment C–receptor subtypes that are targets for trastuzumab or rituximab binding. The variants are associated with an altered risk of disease progression and progression-free survival.27, 28, 29, 30, 31, 32, 33, 34, 35 The NT5C2 gene codes for an enzyme involved in the dephosphorylation of monophosphorylated gemcitabine and the particular variant included is associated with decreased clearance of intravenous gemcitabine.36, 37 NUDT15 was selected because it is important in the metabolism of thiopurines and variants in NUDT15 are associated with an increased risk of thiopurine-induced toxicity. The relationship between the function of SBF2 and taxanes is unknown, however, five variants have been associated with an increased risk of taxane-induced peripheral neuropathy in a group of African Americans.38 The protein coded for by SOD2 is a manganese superoxide dismutase that acts as a mitochondrial antioxidant enzyme by endogenously converting superoxide into oxygen and hydrogen peroxide. The SOD2 variant selected was reported to affect enzyme function and increase the risk of asparaginase-induced hepatotoxicity. TLR4 is a member of the Toll-like receptor family, which plays a role in activation of innate immunity. Individuals with the selected TLR4 variant were more likely to experience methotrexate-induced gastrointestinal, liver, pneumonitis, and skin and mucosal adverse events.39 Table 1 summarizes the risk genotypes for each genetic variant related to a specific medication. Currently, no clinical guidelines are available to recommend dose adjustment for these selected genetic variants.

Table 1.

Selected Germline Genetic Variants and Genotypes Related to Oncology Therapy

Gene and rs number Genotypes Description and references Relevant population PharmGKB level of evidence CPIC level of evidence Medications with suggested testing in FDA label
ABCB1 rs1045642 T/T More likely to experience efficacy and achieve complete control with ondansetron treatment in postoperative or chemotherapy-induced nausea and vomiting4, 5, 6 2A C/D
ABCB1 rs1128503 T/T
ABCB1 rs2032582 T/T 2A
CBR3 rs1056892 G/G Increased risk of anthracycline-induced cardiomyopathy or reduced ejection fraction if cumulative anthracycline exposure is 1 to 250 mg/m27, 8, 9 Pediatric: Caucasian/European 2B D
COMT rs4646316 and rs9332377 rs4646316T/T and rs9332377C/C Decreased risk of cisplatin-induced hearing impairment17, 18, 19, 20, 21, 22 Pediatric: Caucasian/European 3 C/D
CYP3A7 rs45446698 (*1C) *1/*1 and *1/*1C Lower urinary unconjugated estrogen metabolite levels and increased risk of mortality if treated with CYP3A substrates24, 25 Caucasian/European breast and lung cancer patients
C8ORF34 rs1517114 G/C and C/C Increased risk of irinotecan-induced grade 3 diarrhea Asian advanced non–small-cell lung cancer patients 2B D
FCGR2A rs1801274 G/G and A/G Increased risk of stable or progressive disease and more likely to have shorter progression-free survival after treatment of HER2 + breast cancer with trastuzumab-based therapy27, 28, 29
Increased risk of stable or progressive disease and more likely to have shorter progression-free survival after treatment of lymphoma with rituximab-based therapy30
2B
FCGR3A rs396991 C/C Decreased risk of stable or progressive disease and more likely to have longer progression-free survival after treatment of HER2+ metastatic breast cancer with trastuzumab-based therapy27, 28, 29
Decreased risk of stable or progressive disease and more likely to have longer progression-free survival after treatment of lymphoma with rituximab-based therapy30, 31, 32, 33, 34, 35
2B D
HAS3 rs2232228 A/A and A/G Increased risk of anthracycline-induced cardiomyopathy or reduced ejection fraction if cumulative anthracycline exposure is 1 to 450 mg/m210 Pediatric: Caucasian/European 2B D
NT5C2 rs11598702 A/A Decreased clearance of intravenous gemcitabine36, 37 Caucasian solid-tumor patients 2B D
NUDT15 rs116855232 and rs186364861 (*3 and *5) *1/*3, *1/*5, *3/*3, *5/*5, and *3/*5 Increased risk of thiopurine-induced toxicity, including early leukopenia, neutropenia, alopecia totalis, pancytopenia, and treatment discontinuation 1B A/B Mercaptopurine and thioguanine
SBF2 rs7102464 T/T and C/T Increased risk of taxane-induced peripheral neuropathy38 African American
SBF2 rs146987383 C/C and G/C
SBF2 rs141368249 A/A and G/A
SBF2 rs117957652 C/C and G/C
SBF2 rs149501654 G/G and C/G
SEMA3C rs7779029 C/C and T/C Increased risk of irinotecan-induced severe neutropenia26 Advanced non–small-cell lung cancer patients 2B D
SEMA3C rs11979430 T/T and C/T
SLC16A5 rs4788863 T/T and T/C Decreased risk of cisplatin-induced hearing impairment Caucasian/European testicular cancer patients 3
SLC28A3 rs885004 A/A and G/A Decreased risk of anthracycline-induced cardiomyopathy or reduced ejection fraction11, 12, 13, 14, 15, 16 Pediatric: Caucasian/European 2B D
SLC28A3 rs7853758 A/A and G/A
SOD2 rs4880 C/C Increased risk of asparaginase-induced hepatotoxicity Hispanic or Caucasian/European acute lymphoblastic leukemia patients 3 D
TLR4 rs4986790 G/G and A/G Increased risk of methotrexate-induced gastrointestinal (nausea, vomiting, diarrhea, and constipation), liver (increased liver enzyme levels), pneumonitis, and skin and mucosal adverse events39 3
TPMT rs12201199 T/A and A/A Increased risk of cisplatin-induced hearing impairment Pediatric: Caucasian/European 3 Cisplatin

CPIC, Clinical Pharmacogenetics Implementation Consortium; FDA, U.S. Food and Drug Administration; HER2+, human epidermal growth factor receptor 2 positive/erb-b2 receptor tyrosine kinase 2 positive.

Several of these genetic variants have not been reviewed by PharmGKB or CPIC and have not been assigned a level of evidence.

Samples

A total of 189 reference DNA samples obtained from Coriell Cell Repository2, 3 were used for analytical validation (Supplemental Table S1).

TaqMan Genotyping for Selected Variants

Commercially available genotyping assays and reagents were used for each variant. DNA was amplified by real-time PCR on the LifeTech QuantStudio 12K Flex software version 1.2.2 (Thermo Fisher Scientific, Waltham, MA) and subjected to TaqMan allelic discrimination using commercially available LifeTech reagents in a custom-designed open array. The assay identification numbers are shown in Table 2.

Table 2.

Assay Results: Intra-assay and Interassay Concordance, Accuracy, Precision, Sensitivity, and Specificity

Gene rs number TaqMan assay ID Intra-assay concordance Interassay concordance Verified by Sanger sequencing Accuracy Robustness Analytical sensitivity Analytical specificity
ABCB1 rs1045642 C___7586657_20 100% (12 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (12 samples) Yes 100% (95% CI, 93–100) 100% (95% CI, 83–100)
rs1128503 C___7586662_10
rs2032582 C_11711720C_30
CBR3 rs1056892 C___9483603_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (11 samples) Yes 100% (95% CI, 68–100) 100% (95% CI, 78–100)
COMT rs4646316 C__29193982_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (11 samples) Yes 100% (95% CI, 77–100) 100% (95% CI, 89–100)
rs9332377 C__29614343_10
CYP3A7 rs45446698 (*1C) C__30634320_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) No 100% (96 samples) Yes 100% (95% CI, 51–100) 100% (95% CI, 98–100)
C8orf34 rs1517114 C___8341581_20 100% (7 samples in triplicate) 100% (7 samples in triplicate) Yes 100% (16 samples) Yes 100% (95% CI, 86–100) 100% (95% CI, 68–100)
FCGR2A rs1801274 C___9077561_20 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (9 samples) Yes 100% (95% CI, 70–100) 100% (95% CI, 70–100)
FCGR3A rs396991 C__25815666_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (10 samples) Yes 100% (95% CI, 72–100) 100% (95% CI, 90–100)
HAS3 rs2232228 C___3283947_1_ 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (12 samples) Yes 100% (95% CI, 68–100) 100% (95% CI, 81–100)
NT5C2 rs11598702 C__11196884_20 100% (7 samples in triplicate) 100% (7 samples in triplicate) Yes 100% (16 samples) Yes 100% (95% CI, 61–100) 100% (95% CI, 87–100)
NUDT15 rs116855232 (*3) C_154823200_10 100% (7 samples in triplicate) 100% (7 samples in triplicate) Yes 100% (24 samples) Yes 100% (95% CI, 65–100) 100% (95% CI, 96–100)
rs186364861 (*5) C_181955856_10
SBF2 rs7102464 C__29019176_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (12 samples and an additional 9 samples from another laboratory) Yes 100% (95% CI, 81–100) 100% (95% CI, 98–100)
rs146987383 C_161447122_10
rs141368249 C_161190467_10
rs117957652 C_152435684_10
rs149501654 C_161562183_10
SEMA3C rs7779029 C____334680_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (12 samples) Yes 100% (95% CI, 78–100) 100% (95% CI, 89–100)
rs11979430 C___2621121_10
SLC16A5 rs4788863 C____156080_10 100% (7 samples in triplicate) 100% (7 samples in triplicate) Yes 100% (16 samples) Yes 100% (95% CI, 85–100) 100% (95% CI, 72–100)
SLC28A3 rs885004 C___2752627_10 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (11 samples) Yes 100% (95% CI, 70–100) 100% (95% CI, 90–100)
rs7853758 C___1820227_30
SOD2 rs4880 C___8709053_10 100% (7 samples in triplicate) 100% (7 samples in triplicate) Yes 100% (8 samples) Yes 100% (95% CI, 44–100) 100% (95% CI, 77–100)
TLR4 rs4986790 C__11722238_20 100% (14 samples in triplicate) 100% (18 samples in triplicate) Yes 100% (12 samples) Yes 100% (95% CI, 21–100) 100% (95% CI, 86–100)
TPMT rs12201199 C__31923406_10 100% (7 samples in triplicate) 100% (7 samples in triplicate) Yes 100% (16 samples) Yes 100% (95% CI, 70–100) 100% (95% CI, 86–100)

Robustness means obtaining the same genotyping result using two different instruments on different days and using input DNA within a concentration range of 15.4 to 50.8 ng/μL.

Primer Design and Sanger Sequencing of Samples for Accuracy

Primers for each genetic variant were designed specifically for the gene of interest (by aligning the gene sequence with that of genes with similar sequences to select a region; ie, unique to the gene of interest). The following tools were used for primer design: Primer 3 version 2004, which was developed by Rozen and Skaletsky40 in 2000 (http://bioinfo.ut.ee/primer3-0.4.0, last accessed October 29, 2018), NCBI Primer-BLAST (http://www.ncbi.nlm.nih.gov/tools/primer-blast), and the IDT OligoAnalyzer (Integrated DNA Technologies, Inc., Coralville, IA). Integrated DNA Technologies, Inc., performed the primer synthesis. Primer sequences for each genetic variant are provided in Table 3 with PCR amplification conditions.41, 42

Table 3.

Primer Sequences and PCR Amplification Conditions for Validated Genetic Variants before Performing Verification by Sanger Sequencing

Gene rs number HGVS nomenclature Sequence accession number Forward primer sequence Reverse primer sequence PCR annealing temperature, degrees Celsius PCR product/amplicon length, bp
ABCB1 rs1045642 c.3435T>C or p.Ile1145= NM_000927.4 or NP_000918.2 5′-ACTCTTGTTTTCAGCTGCTTG-3′41 5′-AGAGACTTACATTAGGCAGTGACTC-3′41 63 231
rs1128503 c.1236T>C or p.Gly412= 5′-TGTGTCTGTGAATTGCCTTGAAG-3′42 5′-CCTCTGCATCAGCTGGACTGT-3′42 63 149
rs2032582 c.2677T>G/A or p.Ser893Ala/Thr 5′-ATGGTTGGCAACTAACACTGTTA-3′42 5′-AGCAGTAGGGAGTAACAAAATAACA-3′42 63 208
CBR3 rs1056892 c.730G>A or p.Val244Met NM_001236.3 or NP_001227.1 5′-CCAGGACCAGTGAAGACAGA-3′ 5′-CCGAAGCAGACGTTTACCAG-3′ 63 166
COMT rs4646316 c.615+310C>T NM_000754.3 5′-ACACGCTTCTCTTGGAGGTG-3′ 5′-CTGTCTAGCCTCACTCGGG-3′ 63 519
rs9332377 c.616-367C>A/T 5′-GCTTGTTGATGGGAGGTCTG-3′ 5′-TCCCTTAGAACAGCATGTGG-3′ 61 217
C8ORF34 rs1517114 c.736+8162C>G/T/A NM_052958.2 5′-CTGTGCTTTCTCGTCTTCAG-3′ 5′-CAGCCTGGAACCTACCCTTG-3′ 58 238
FCGR2A rs1801274 c.500A>G or p.His167Arg NM_001136219.1 or NP_001129691.1 5′-CAAGCCTCTGGTCAAGGTCA-3′ 5′-AAGGATTCCCCTTAGCCCCT-3′ 58 663
FCGR3A rs396991 c.841T>C/G or p.Phe281Leu/Val NM_000569.7: or NP_000560.6 5′-CACATATTTACAGAATGGCAAAGG-3′ 5′-GATTCTGGAGGCTGGTGCTACA-3′ 58 969
HAS3 rs2232228 c.279A>G or p.Ala93= NM_001199280.1 or NP_001186209.1 5′-GTGACGGGCTACCAGTTCAT-3′ 5′-CACAACCCAAGGGACCTAGA-3′ 58 654
NT5C2 rs11598702 c.175+1178A>G/C NM_012229.4 5′-GACGGGTTTATAGGTGCAGC-3′ 5′-TCAATGACTTCTTGCCCAGT-3′ 58 222
NUDT15 rs116855232 (*3) c.415C>T or p.Arg139Cys NM_018283.3 or NP_060753.1 5′-GCCTTTGTAAACTGGGCTTC-3′ 5′-CAAATCTTCTCGGCCACCTA-3′ 58 411
rs186364861 (*5) c.52G>A or p.Val18Ile 5′-CATTCCCCAACCTGATAGCC-3′ 5′-CAACCGAGCCTTTCCTCTTC-3′ 58 296
SBF2 rs7102464 c.2035G>A or p.Glu679Lys NM_030962.3 or NP_112224.1 5′-ACAGAAACTTGCCCCTGGAG-3′ 5′-ACCCAAATACACTGGCAGGA-3′ 63 289
rs146987383 c.2050C>G or p.Leu684Val 5′-ACAGAAACTTGCCCCTGGAG-3′ 5′-ACCCAAATACACTGGCAGGA-3′ 63 289
rs141368249 c.2081C>T or p.Ala694Val 5′-ACAGAAACTTGCCCCTGGAG-3′ 5′-ACCCAAATACACTGGCAGGA-3′ 63 289
rs117957652 c.3292C>G/T or p.Leu1098Val/= 5′-CCTGTCTTGGTGTAAGAGTCTTCT-3′ 5′-ACCTCTTTTTGGAGCCCACT-3′ 63 843
rs149501654 c.4111G>C or p.Val1371Leu 5′-TCTTCATCCGCAGAACTTCA-3′ 5′-AGTGTGCCTTTGGTGGGTAG-3′ 63 649
SEMA3C rs7779029 c.103+13883A>G NM_006379.3 5′-GGCTTAGGTCTCTGCCCTTT-3′ 5′-GTTCCCATTTCCAGGCTCCA-3′ 58 200
rs11979430 c.103+36739G>A 5′-GGAAAGGGCAGACTGTGGTA-3′ 5′-ACCAAACCTCTTCAGGGTGA-3′ 58 383
SLC16A5 rs4788863 c.121T>C or p.Leu81= NM_004695.3 or NP_004686.1 5′-AGGTCCCCCTGTTGACTTCT-3′ 5′-TGAAATCTGGTGAAACCTTAGGA-3′ 58 725
SLC28A3 rs885004 c.862-360C>T NM_022127.2 5′-TGTGTCTGCCATCCAGTAGG-3′ 5′-CCTGGTGCTAAAAAGACATGG-3′ 58 161
rs7853758 c.1381C>T or p.Leu461= NM_022127.2 or NP_071410.1 5′-CCCCTGACAACTCCTTGGTA-3′ 5′-CAGGGGCGTGATGTGATTAT-3′ 58 239
SOD2 rs4880 c.47T>C or p.Val16Ala NM_000636.3 or NP_000627.2 5′-CTGTGCTTTCTCGTCTTCAG-3′ 5′-CAGCCTGGAACCTACCCTTG-3′ 58 238
TLR4 rs4986790 c.776A>G or p.Asp299Gly NM_003266.3 or NP_612564.1 5′-AGTCCATCGTTTGGTTCTGG-3′ 5′-TGCCATTGAAAGCAACTCTG-3′ 58 635
TPMT rs12201199 c.419+94T>A NM_000367.3 5′-GTTCTTCGGGGAACATTTCA-3′ 5′-AAGTGATTGAGCCACAAGCC-3′ 58 975

Accession numbers are available at https://www.ncbi.nlm.nih.gov/snp.

HGVS, Human Genome Variation Society.

PCR amplification was performed under the following conditions: initial denaturation at 98°C for 30 seconds, followed by 35 cycles of denaturation at 98°C for 10 seconds, annealing at the specific annealing temperature provided in Table 3 for 10 seconds, primer extension at 72°C for 30 seconds, and final extension at 72°C for 5 minutes. A MyCycler Thermal cycler (Bio-Rad, Hercules, CA) was used and the PCR reaction contained the following reagents: 10 ng genomic DNA, 1× Platinum SuperFi PCR Master Mix (Thermo Fisher Scientific), and 0.112 μmol/L of the forward and reverse primers (Integrated DNA Technologies, Inc.).

Purification of the PCR amplicons was performed using the MinElute PCR Purification Kit (Qiagen, Hilden, Germany). The protocol was adjusted by performing elution twice in 20 μL of DNase-free water. Purified samples were mixed with 0.25 μmol/L of the primer used for sequencing and submitted to ACGT, Inc. (Wheeling, IL) for Sanger sequencing. Analysis of the sequences was performed using the BioEdit biological sequence alignment editor version 7.0.5 (Ibis Therapeutics, Carlsbad, CA).

Results

All of the variants were detected using the TaqMan reagents. Both the amplification traces and allelic discrimination plots showed good allele separation (http://tools.thermofisher.com/content/sfs/manuals/cms_042798.pdf, last accessed November 11, 2018). The sequencing results were compared with the genotyping results and were 100% concordant (Table 2). The number of variant alleles and nonvariant (ie, wild type) alleles detected by sequencing were evaluated to calculate the analytical sensitivity and specificity. The analytical sensitivity was 100% for the detection of variant alleles, with no reported false negatives. The analytical specificity was 100% for detection of nonvariant alleles, with no false-positive results reported (confidence intervals varied based on samples tested and allele frequency) (Table 2).

DNA samples obtained from Coriell Cell Repository were used to assess intra-assay and interassay variation. Intra-assay (within-assay) variation studies showed that all three replicates of the samples analyzed in the same run had concordant results. The interassay (between-assay) variation study showed that the samples consistently had the same result across three separate runs. All assays were robust, and consistent genotyping results were obtained using two different instruments on different days and using input DNA within a concentration range of 15.4 to 50.8 ng/μL.

A total of 189 DNA samples from Coriell Cell Repository were genotyped successfully, with the results provided in Supplemental Table S1. Sanger sequencing was used as an orthogonal method to confirm the accuracy of the array genotyping results for a majority of the 27 variants, except for SBF2 rs146987383 and SBF2 rs149501654. All 189 Coriell reference materials were negative for both the SBF2 rs146987383 and rs149501654 variants and known reference materials were obtained from a research laboratory. The number of samples from the Coriell sample set that carried a variant allele for each of the 27 genetic variants is summarized in Table 4 to show how many samples in this data set had a variant allele. The variant alleles for CYP3A7 rs45446698, NUDT15 rs116855232, NUDT15 rs186364891, SBF2 rs141368249, and SBF2 rs117957652 are rare and were observed in only five, six, one, five, and three samples, respectively (Table 4).

Table 4.

Genotype Frequencies for Validated Genetic Variants

Gene and rs number HGVS nomenclature Genotype n Genotype frequencies
ABCB1 rs1045642 c.3435T>C or p.Ile1145= T/T 34 0.18
T/C 84 0.44
C/C 71 0.38
ABCB1 rs1128503 c.1236T>C or p.Gly412= T/T 28 0.15
T/C 88 0.47
C/C 73 0.39
ABCB1 rs2032582 c.2677T>G/A or p.Ser893Ala/Thr T/T 33 0.17
T/G 70 0.37
G/G 86 0.46
CBR3 rs1056892 c.730G>A or p.Val244Met G/G 65 0.34
G/A 101 0.53
A/A 23 0.12
COMT rs4646316 c.615+310C>T C/C 117 0.62
C/T 57 0.30
T/T 15 0.08
COMT rs9332377 c.616-367C>A/T C/C 125 0.66
C/T 59 0.31
T/T 5 0.03
CYP3A7 rs45446698 (*1C) c.-232A>C A/A 184 0.97
A/C 4 0.02
C/C 1 0.01
C8orf34 rs1517114 c.736+8162C>G/T/A C/C 22 0.12
C/G 81 0.43
G/G 86 0.46
FCGR2A rs1801274 c.500A>G or p.His167Arg A/A 59 0.31
A/G 94 0.50
G/G 36 0.19
FCGR3A rs396991 c.841T>C/G or p.Phe281Leu/Val T/T 78 0.41
T/G 94 0.50
G/G 17 0.09
HAS3 rs2232228 c.279A>G or p.Ala93 A/A 81 0.43
A/G 87 0.46
G/G 21 0.11
NT5C2 rs11598702 c.175+1178A>G/C A/A 102 0.54
A/G 75 0.40
G/G 12 0.06
NUDT15 rs116855232 (*3) c.415C>T or p.Arg139Cys C/C 183 0.97
C/T 6 0.03
T/T 0 0.00
NUDT15 rs186364861 (*5) c.52G>A or p.Val18Ile G/G 188 0.99
G/A 1 0.01
A/A 0 0.00
SBF2 rs7102464 c.2035G>A or p.Glu679Lys G/G 168 0.89
G/A 18 0.10
A/A 3 0.01
SBF2 rs146987383 c.2050C>G or p.Leu684Val C/C 0 0.00
C/G 0 0.00
G/G 189 1.00
SBF2 rs141368249 c.2081C>T or p.Ala694Val C/C 4 0.02
C/T 1 0.01
T/T 184 0.97
SBF2 rs117957652 c.3292C>G/T or p.Leu1098Val/= C/C 0 0.00
C/G 3 0.02
G/G 186 0.98
SBF2 rs149501654 c.4111G>C or p.Val1371Leu G/G 0 0.00
G/C 0 0.00
C/C 189 1.00
SEMA3C rs7779029 c.103+13883A>G A/A 134 0.71
A/G 44 0.23
G/G 11 0.06
SEMA3C rs11979430 c.103+36739G>A G/G 143 0.76
G/A 39 0.21
A/A 7 0.03
SLC16A5 rs4788863 c.121T>C or p.Leu81= T/T 24 0.13
T/C 71 0.38
C/C 94 0.49
SLC28A3 rs885004 c.862-360C>T C/C 143 0.76
C/T 44 0.23
T/T 2 0.01
SLC28A3 rs7853758 c.1381C>T or p.Leu461= C/C 123 0.65
C/T 56 0.30
T/T 10 0.05
SOD2 rs4880 c.47T>C or p.Val16Ala T/T 86 0.46
T/C 62 0.33
C/C 41 0.21
TLR4 rs4986790 c.776A>G or p.Asp299Gly A/A 176 0.93
A/G 13 0.07
G/G 0 0.00
TPMT rs12201199 c.419+94T>A T/T 18 0.10
T/A 26 0.14
A/A 143 0.76

HGVS, Human Genome Variation Society.

Discussion

Pharmacogenomic testing methods can be complex to generate and are complicated by gene sequence similarity between members of the same gene family. The benefit of publishing our validated genotyping methods is that other Clinical Laboratory Improvement Amendments–accredited laboratories can access this information and confidently establish these methods knowing that the assays are robust, accurate, and had 100% analytical sensitivity and specificity. Furthermore, identification of samples with the variant allele among genomic DNA samples from the Coriell Cell Repository is useful for other clinical laboratories. These publicly available DNA samples and associated data can be used when developing and validating new genetic tests or refining current assays. The 1000 Genomes Project43 or the Exome Aggregation Consortium Project44 may be used as another resource for identifying reference materials. Having validated methods and positive samples will improve standardization of pharmacogenomic testing across clinical laboratories.

The goal of analytical validation in clinical laboratories is to determine how well the test system can detect what it is designed to detect (ie, defined genetic variants from genomic DNA). Analytical validation can be challenging when there is a lack of reference materials for the defined variants, or lack of truth. We chose to screen approximately 200 Coriell DNA samples for the variants that were being validated and to use Sanger sequencing as an orthogonal method on a subset of both positive and negative samples to determine correct genotype. This approach works well when variants are rather common, and have greater than 0.01 frequency. For rarer alleles, a different approach was chosen. A research laboratory was contacted and DNA samples were requested for validation studies.

A novel discovery during the validation studies was that several of the variants were in cis. Several DNA samples were positive for more than two variants in the same gene (eg, ABCB1, COMT, SBF2, SEMA3C, and SLC28A3). In clinical testing, some examples, including CFTR (p.R117H and c.1210−12[5][7][9]),45 CYP2D6,46 and MTHFR47 cis variants, have been well documented as both impacting clinical phenotype and as a confounder for clinical interpretation. If any of these markers are used in risk models for toxicity, these risk models may need to be revised.

Establishing standardized methods is challenging, but once these genotyping methods are validated another difficulty is the interpretation and implementation of the test results. Interpretation of pharmacogenomic test results for patients with cancer is particularly complex because both germline and somatic DNA alterations could inform therapy: somatic mutations can be used to select a targeted therapeutic agent whereas germline genetic variation can highlight the possible risk of toxicity or inefficacy of therapy. An approach for clinical pharmacogenomic testing in oncology is to perform testing on cancer patients as part of precision genomics initiatives/clinics and then discuss test results during molecular tumor boards. Molecular tumor boards are forums through which interprofessional teams discuss and interpret genomic test results and make treatment recommendations. If a clinical genetics testing laboratory with the ability to perform pharmacogenomic tests is aligned with a molecular tumor board, testing can be performed and results can be used to assess a cancer patient's risk of toxicity or treatment inefficacy when decisions are made about which cytotoxic agents are preferred. If a clinical testing laboratory is in proximity and its services are integrated into a molecular tumor board, there may be added benefits such as shorter turnaround time and, in some cases, genotyping before therapy selection instead of reactive genotyping. This approach along with the provided pharmacogenetic testing methods for genetic variants in ABCB1, CBR3, COMT, CYP3A7, C8ORF34, FCGR2A, FCGR3A, HAS3, NT5C2, NUDT15, SBF2, SEMA3C, SLC16A5, SLC28A3, SOD2, TLR4, and TPMT have the potential to better understand a patient's risk of toxicity or treatment inefficacy for oncology medications such as taxanes, anthracyclines, platinum agents, trastuzumab, rituximab, and 5-hydroxytriptamine-3–receptor antagonists.

Acknowledgments

M.S., W.M.S., V.M.P., and T.C.S wrote the manuscript; M.S., W.M.S., V.M.P., E.B.M., P.J.K., B.P.S., and T.C.S. designed the research; M.S., W.M.S., V.M.P., and E.B.M. performed the research and analyzed the data; and F.S. and B.P.S. provided additional control DNA samples.

Footnotes

Supported by the NIH-funded National Human Genome Research Institute—Implementing Genomics in Practice (NHGRI-IGNITE) network project grant U01HG007762 (T.C.S. and V.M.P.); the Indiana University Health–Indiana University School of Medicine Strategic Research Initiative (V.M.P. and E.B.M.); the Vera Bradley Foundation for Breast Cancer (M.S. and T.C.S.); the Indiana University Grand Challenge Precision Health Initiative (B.P.S. and T.C.S.); and the Indiana Institute for Personalized Medicine (B.P.S. and T.C.S.).

Disclosures: The Indiana University School of Medicine Pharmacogenomics Laboratory is a fee-for-service clinical laboratory that offers clinical pharmacogenetic testing.

Supplemental material for this article can be found at https://doi.org/10.1016/j.jmoldx.2019.01.009.

Supplemental Data

Supplemental Table S1
mmc1.xlsx (33.2KB, xlsx)

References

  • 1.Amstutz U., Henricks L.M., Offer S.M., Barbarino J., Schellens J.H.M., Swen J.J., Klein T.E., McLeod H.L., Caudle K.E., Diasio R.B., Schwab M. Clinical Pharmacogenetics Implementation Consortium (CPIC) guideline for dihydropyrimidine dehydrogenase genotype and fluoropyrimidine dosing: 2017 update. Clin Pharmacol Ther. 2018;103:210–216. doi: 10.1002/cpt.911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pratt V.M., Zehnbauer B., Wilson J.A., Baak R., Babic N., Bettinotti M., Buller A., Butz K., Campbell M., Civalier C., El-Badry A., Farkas D.H., Lyon E., Mandal S., McKinney J., Muralidharan K., Noll L., Sander T., Shabbeer J., Smith C., Telatar M., Toji L., Vairavan A., Vance C., Weck K.E., Wu A.H., Yeo K.T., Zeller M., Kalman L. Characterization of 107 genomic DNA reference materials for CYP2D6, CYP2C19, CYP2C9, VKORC1, and UGT1A1: a GeT-RM and Association for Molecular Pathology collaborative project. J Mol Diagn. 2010;12:835–846. doi: 10.2353/jmoldx.2010.100090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pratt V.M., Everts R.E., Aggarwal P., Beyer B.N., Broeckel U., Epstein-Baak R., Hujsak P., Kornreich R., Liao J., Lorier R., Scott S.A., Smith C.H., Toji L.H., Turner A., Kalman L.V. Characterization of 137 genomic DNA reference materials for 28 pharmacogenetic genes: a GeT-RM collaborative project. J Mol Diagn. 2016;18:109–123. doi: 10.1016/j.jmoldx.2015.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.He H., Yin J.Y., Xu Y.J., Li X., Zhang Y., Liu Z.G., Zhou F., Zhai M., Li Y., Li X.P., Wang Y., Zhou H.H., Liu Z.Q. Association of ABCB1 polymorphisms with the efficacy of ondansetron in chemotherapy-induced nausea and vomiting. Clin Ther. 2014;36:1242–1252.e1242. doi: 10.1016/j.clinthera.2014.06.016. [DOI] [PubMed] [Google Scholar]
  • 5.Babaoglu M.O., Bayar B., Aynacioglu A.S., Kerb R., Abali H., Celik I., Bozkurt A. Association of the ABCB1 3435C>T polymorphism with antiemetic efficacy of 5-hydroxytryptamine type 3 antagonists. Clin Pharmacol Ther. 2005;78:619–626. doi: 10.1016/j.clpt.2005.08.015. [DOI] [PubMed] [Google Scholar]
  • 6.Zoto T., Kilickap S., Yasar U., Celik I., Bozkurt A., Babaoglu M.O. Improved anti-emetic efficacy of 5-HT3 receptor antagonists in cancer patients with genetic polymorphisms of ABCB1 (MDR1) drug transporter. Basic Clin Pharmacol Toxicol. 2015;116:354–360. doi: 10.1111/bcpt.12334. [DOI] [PubMed] [Google Scholar]
  • 7.Blanco J.G., Sun C.L., Landier W., Chen L., Esparza-Duran D., Leisenring W., Mays A., Friedman D.L., Ginsberg J.P., Hudson M.M., Neglia J.P., Oeffinger K.C., Ritchey A.K., Villaluna D., Relling M.V., Bhatia S. Anthracycline-related cardiomyopathy after childhood cancer: role of polymorphisms in carbonyl reductase genes--a report from the Children's Oncology Group. J Clin Oncol. 2012;30:1415–1421. doi: 10.1200/JCO.2011.34.8987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Blanco J.G., Leisenring W.M., Gonzalez-Covarrubias V.M., Kawashima T.I., Davies S.M., Relling M.V., Robison L.L., Sklar C.A., Stovall M., Bhatia S. Genetic polymorphisms in the carbonyl reductase 3 gene CBR3 and the NAD(P)H:quinone oxidoreductase 1 gene NQO1 in patients who developed anthracycline-related congestive heart failure after childhood cancer. Cancer. 2008;112:2789–2795. doi: 10.1002/cncr.23534. [DOI] [PubMed] [Google Scholar]
  • 9.Visscher H., Rassekh S.R., Sandor G.S., Caron H.N., van Dalen E.C., Kremer L.C., van der Pal H.J., Rogers P.C., Rieder M.J., Carleton B.C., Hayden M.R., Ross C.J., CPNDS Consortium Genetic variants in SLC22A17 and SLC22A7 are associated with anthracycline-induced cardiotoxicity in children. Pharmacogenomics. 2015;16:1065–1076. doi: 10.2217/pgs.15.61. [DOI] [PubMed] [Google Scholar]
  • 10.Wang X., Liu W., Sun C.L., Armenian S.H., Hakonarson H., Hageman L., Ding Y., Landier W., Blanco J.G., Chen L., Quinones A., Ferguson D., Winick N., Ginsberg J.P., Keller F., Neglia J.P., Desai S., Sklar C.A., Castellino S.M., Cherrick I., Dreyer Z.E., Hudson M.M., Robison L.L., Yasui Y., Relling M.V., Bhatia S. Hyaluronan synthase 3 variant and anthracycline-related cardiomyopathy: a report from the children's oncology group. J Clin Oncol. 2014;32:647–653. doi: 10.1200/JCO.2013.50.3557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Visscher H., Ross C.J., Rassekh S.R., Sandor G.S., Caron H.N., van Dalen E.C., Kremer L.C., van der Pal H.J., Rogers P.C., Rieder M.J., Carleton B.C., Hayden M.R., CPNDS Consortium Validation of variants in SLC28A3 and UGT1A6 as genetic markers predictive of anthracycline-induced cardiotoxicity in children. Pediatr Blood Cancer. 2013;60:1375–1381. doi: 10.1002/pbc.24505. [DOI] [PubMed] [Google Scholar]
  • 12.Visscher H., Ross C.J., Rassekh S.R., Barhdadi A., Dube M.P., Al-Saloos H., Sandor G.S., Caron H.N., van Dalen E.C., Kremer L.C., van der Pal H.J., Brown A.M., Rogers P.C., Phillips M.S., Rieder M.J., Carleton B.C., Hayden M.R., Canadian Pharmacogenomics Network for Drug Safety Consortium Pharmacogenomic prediction of anthracycline-induced cardiotoxicity in children. J Clin Oncol. 2012;30:1422–1428. doi: 10.1200/JCO.2010.34.3467. [DOI] [PubMed] [Google Scholar]
  • 13.Tanaka M., Javle M., Dong X., Eng C., Abbruzzese J.L., Li D. Gemcitabine metabolic and transporter gene polymorphisms are associated with drug toxicity and efficacy in patients with locally advanced pancreatic cancer. Cancer. 2010;116:5325–5335. doi: 10.1002/cncr.25282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Reichwagen A., Ziepert M., Kreuz M., Godtel-Armbrust U., Rixecker T., Poeschel V., Reza Toliat M., Nurnberg P., Tzvetkov M., Deng S., Trumper L., Hasenfuss G., Pfreundschuh M., Wojnowski L. Association of NADPH oxidase polymorphisms with anthracycline-induced cardiotoxicity in the RICOVER-60 trial of patients with aggressive CD20(+) B-cell lymphoma. Pharmacogenomics. 2015;16:361–372. doi: 10.2217/pgs.14.179. [DOI] [PubMed] [Google Scholar]
  • 15.Hertz D.L., Caram M.V., Kidwell K.M., Thibert J.N., Gersch C., Seewald N.J., Smerage J., Rubenfire M., Henry N.L., Cooney K.A., Leja M., Griggs J.J., Rae J.M. Evidence for association of SNPs in ABCB1 and CBR3, but not RAC2, NCF4, SLC28A3 or TOP2B, with chronic cardiotoxicity in a cohort of breast cancer patients treated with anthracyclines. Pharmacogenomics. 2016;17:231–240. doi: 10.2217/pgs.15.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Serie D.J., Crook J.E., Necela B.M., Dockter T.J., Wang X., Asmann Y.W., Fairweather D., Bruno K.A., Colon-Otero G., Perez E.A., Thompson E.A., Norton N. Genome-wide association study of cardiotoxicity in the NCCTG N9831 (Alliance) adjuvant trastuzumab trial. Pharmacogenet Genomics. 2017;27:378–385. doi: 10.1097/FPC.0000000000000302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Ross C.J., Katzov-Eckert H., Dube M.P., Brooks B., Rassekh S.R., Barhdadi A., Feroz-Zada Y., Visscher H., Brown A.M., Rieder M.J., Rogers P.C., Phillips M.S., Carleton B.C., Hayden M.R., CPNDS Consortium Genetic variants in TPMT and COMT are associated with hearing loss in children receiving cisplatin chemotherapy. Nat Genet. 2009;41:1345–1349. doi: 10.1038/ng.478. [DOI] [PubMed] [Google Scholar]
  • 18.Pussegoda K., Ross C.J., Visscher H., Yazdanpanah M., Brooks B., Rassekh S.R., Zada Y.F., Dube M.P., Carleton B.C., Hayden M.R., CPNDS Consortium Replication of TPMT and ABCC3 genetic variants highly associated with cisplatin-induced hearing loss in children. Clin Pharmacol Ther. 2013;94:243–251. doi: 10.1038/clpt.2013.80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Yang J.J., Lim J.Y., Huang J., Bass J., Wu J., Wang C., Fang J., Stewart E., Harstead E.H., E S., Robinson G.W., Evans W.E., Pappo A., Zuo J., Relling M.V., Onar-Thomas A., Gajjar A., Stewart C.F. The role of inherited TPMT and COMT genetic variation in cisplatin-induced ototoxicity in children with cancer. Clin Pharmacol Ther. 2013;94:252–259. doi: 10.1038/clpt.2013.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Thiesen S., Yin P., Jorgensen A.L., Zhang J.E., Manzo V., McEvoy L., Barton C., Picton S., Bailey S., Brock P., Vyas H., Walker D., Makin G., Bandi S., Pizer B., Hawcutt D.B., Pirmohamed M. TPMT, COMT and ACYP2 genetic variants in paediatric cancer patients with cisplatin-induced ototoxicity. Pharmacogenet Genomics. 2017;27:213–222. doi: 10.1097/FPC.0000000000000281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lanvers-Kaminsky C., Malath I., Deuster D., Ciarimboli G., Boos J., Am Zehnhoff-Dinnesen A.G. Evaluation of pharmacogenetic markers to predict the risk of cisplatin-induced ototoxicity. Clin Pharmacol Ther. 2014;96:156–157. doi: 10.1038/clpt.2014.67. [DOI] [PubMed] [Google Scholar]
  • 22.Hagleitner M.M., Coenen M.J., Patino-Garcia A., de Bont E.S., Gonzalez-Neira A., Vos H.I., van Leeuwen F.N., Gelderblom H., Hoogerbrugge P.M., Guchelaar H.J., Te Loo M.W. Influence of genetic variants in TPMT and COMT associated with cisplatin induced hearing loss in patients with cancer: two new cohorts and a meta-analysis reveal significant heterogeneity between cohorts. PLoS One. 2014;9:e115869. doi: 10.1371/journal.pone.0115869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Drogemoller B.I., Monzon J.G., Bhavsar A.P., Borrie A.E., Brooks B., Wright G.E.B., Liu G., Renouf D.J., Kollmannsberger C.K., Bedard P.L., Aminkeng F., Amstutz U., Hildebrand C.A., Gunaretnam E.P., Critchley C., Chen Z., Brunham L.R., Hayden M.R., Ross C.J.D., Gelmon K.A., Carleton B.C. Association between SLC16A5 genetic variation and cisplatin-induced ototoxic effects in adult patients with testicular cancer. JAMA Oncol. 2017;3:1558–1562. doi: 10.1001/jamaoncol.2017.0502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Johnson N., De Ieso P., Migliorini G., Orr N., Broderick P., Catovsky D., Matakidou A., Eisen T., Goldsmith C., Dudbridge F., Peto J., Dos-Santos-Silva I., Ashworth A., Ross G., Houlston R.S., Fletcher O. Cytochrome P450 allele CYP3A7*1C associates with adverse outcomes in chronic lymphocytic leukemia, breast, and lung cancer. Cancer Res. 2016;76:1485–1493. doi: 10.1158/0008-5472.CAN-15-1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sood D., Johnson N., Jain P., Siskos A.P., Bennett M., Gilham C., Busana M.C., Peto J., Dos-Santos-Silva I., Keun H.C., Fletcher O. CYP3A7*1C allele is associated with reduced levels of 2-hydroxylation pathway oestrogen metabolites. Br J Cancer. 2017;116:382–388. doi: 10.1038/bjc.2016.432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Han J.Y., Shin E.S., Lee Y.S., Ghang H.Y., Kim S.Y., Hwang J.A., Kim J.Y., Lee J.S. A genome-wide association study for irinotecan-related severe toxicities in patients with advanced non-small-cell lung cancer. Pharmacogenomics J. 2013;13:417–422. doi: 10.1038/tpj.2012.24. [DOI] [PubMed] [Google Scholar]
  • 27.Musolino A., Naldi N., Bortesi B., Pezzuolo D., Capelletti M., Missale G., Laccabue D., Zerbini A., Camisa R., Bisagni G., Neri T.M., Ardizzoni A. Immunoglobulin G fragment C receptor polymorphisms and clinical efficacy of trastuzumab-based therapy in patients with HER-2/neu-positive metastatic breast cancer. J Clin Oncol. 2008;26:1789–1796. doi: 10.1200/JCO.2007.14.8957. [DOI] [PubMed] [Google Scholar]
  • 28.Tamura K., Shimizu C., Hojo T., Akashi-Tanaka S., Kinoshita T., Yonemori K., Kouno T., Katsumata N., Ando M., Aogi K., Koizumi F., Nishio K., Fujiwara Y. FcgammaR2A and 3A polymorphisms predict clinical outcome of trastuzumab in both neoadjuvant and metastatic settings in patients with HER2-positive breast cancer. Ann Oncol. 2011;22:1302–1307. doi: 10.1093/annonc/mdq585. [DOI] [PubMed] [Google Scholar]
  • 29.Gavin P.G., Song N., Kim S.R., Lipchik C., Johnson N.L., Bandos H., Finnigan M., Rastogi P., Fehrenbacher L., Mamounas E.P., Swain S.M., Wickerham D.L., Geyer C.E., Jr., Jeong J.H., Costantino J.P., Wolmark N., Paik S., Pogue-Geile K.L. Association of polymorphisms in FCGR2A and FCGR3A with degree of trastuzumab benefit in the adjuvant treatment of ERBB2/HER2-positive breast cancer: analysis of the NSABP B-31 trial. JAMA Oncol. 2017;3:335–341. doi: 10.1001/jamaoncol.2016.4884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Weng W.K., Levy R. Two immunoglobulin G fragment C receptor polymorphisms independently predict response to rituximab in patients with follicular lymphoma. J Clin Oncol. 2003;21:3940–3947. doi: 10.1200/JCO.2003.05.013. [DOI] [PubMed] [Google Scholar]
  • 31.Nishio M., Endo T., Fujimoto K., Yamamoto S., Obara M., Yamaguchi K., Takeda Y., Goto H., Kasahara I., Sato N., Koike T. FCGR3A-158V/F polymorphism may correlate with the levels of immunoglobulin in patients with non-Hodgkin's lymphoma after rituximab treatment as an adjuvant to autologous stem cell transplantation. Eur J Haematol. 2009;82:143–147. doi: 10.1111/j.1600-0609.2008.01174.x. [DOI] [PubMed] [Google Scholar]
  • 32.Kim D.H., Jung H.D., Kim J.G., Lee J.J., Yang D.H., Park Y.H., Do Y.R., Shin H.J., Kim M.K., Hyun M.S., Sohn S.K. FCGR3A gene polymorphisms may correlate with response to frontline R-CHOP therapy for diffuse large B-cell lymphoma. Blood. 2006;108:2720–2725. doi: 10.1182/blood-2006-01-009480. [DOI] [PubMed] [Google Scholar]
  • 33.Ahlgrimm M., Pfreundschuh M., Kreuz M., Regitz E., Preuss K.D., Bittenbring J. The impact of Fc-gamma receptor polymorphisms in elderly patients with diffuse large B-cell lymphoma treated with CHOP with or without rituximab. Blood. 2011;118:4657–4662. doi: 10.1182/blood-2011-04-346411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Quartuccio L., Fabris M., Pontarini E., Salvin S., Zabotti A., Benucci M., Manfredi M., Biasi D., Ravagnani V., Atzeni F., Sarzi-Puttini P., Morassi P., Fischetti F., Tomietto P., Bazzichi L., Saracco M., Pellerito R., Cimmino M., Schiavon F., Carraro V., Semeraro A., Caporali R., Cavagna L., Bortolotti R., Paolazzi G., Govoni M., Bombardieri S., De Vita S. The 158VV Fcgamma receptor 3A genotype is associated with response to rituximab in rheumatoid arthritis: results of an Italian multicentre study. Ann Rheum Dis. 2014;73:716–721. doi: 10.1136/annrheumdis-2012-202435. [DOI] [PubMed] [Google Scholar]
  • 35.Kim S.H., Jeong I.H., Hyun J.W., Joung A., Jo H.J., Hwang S.H., Yun S., Joo J., Kim H.J. Treatment outcomes with rituximab in 100 patients with neuromyelitis optica: influence of FCGR3A polymorphisms on the therapeutic response to rituximab. JAMA Neurol. 2015;72:989–995. doi: 10.1001/jamaneurol.2015.1276. [DOI] [PubMed] [Google Scholar]
  • 36.Mitra A.K., Kirstein M.N., Khatri A., Skubitz K.M., Dudek A.Z., Greeno E.W., Kratzke R.A., Lamba J.K. Pathway-based pharmacogenomics of gemcitabine pharmacokinetics in patients with solid tumors. Pharmacogenomics. 2012;13:1009–1021. doi: 10.2217/pgs.12.81. [DOI] [PubMed] [Google Scholar]
  • 37.Khatri A., Williams B.W., Fisher J., Brundage R.C., Gurvich V.J., Lis L.G., Skubitz K.M., Dudek A.Z., Greeno E.W., Kratzke R.A., Lamba J.K., Kirstein M.N. SLC28A3 genotype and gemcitabine rate of infusion affect dFdCTP metabolite disposition in patients with solid tumours. Br J Cancer. 2014;110:304–312. doi: 10.1038/bjc.2013.738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schneider B.P., Lai D., Shen F., Jiang G., Radovich M., Li L., Gardner L., Miller K.D., O'Neill A., Sparano J.A., Xue G., Foroud T., Sledge G.W., Jr. Charcot-Marie-Tooth gene, SBF2, associated with taxane-induced peripheral neuropathy in African Americans. Oncotarget. 2016;7:82244–82253. doi: 10.18632/oncotarget.12545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kooloos W.M., Wessels J.A., van der Straaten T., Allaart C.F., Huizinga T.W., Guchelaar H.J. Functional polymorphisms and methotrexate treatment outcome in recent-onset rheumatoid arthritis. Pharmacogenomics. 2010;11:163–175. doi: 10.2217/pgs.09.139. [DOI] [PubMed] [Google Scholar]
  • 40.Rozen S., Skaletsky H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 2000;132:365–386. doi: 10.1385/1-59259-192-2:365. [DOI] [PubMed] [Google Scholar]
  • 41.Swart M., Ren Y., Smith P., Dandara C. ABCB1 4036A>G and 1236C>T polymorphisms affect plasma efavirenz levels in South African HIV/AIDS patients. Front Genet. 2012;3:236. doi: 10.3389/fgene.2012.00236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Rhodes K.E., Zhang W., Yang D., Press O.A., Gordon M., Vallbohmer D., Schultheis A.M., Lurje G., Ladner R.D., Fazzone W., Iqbal S., Lenz H.J. ABCB1, SLCO1B1 and UGT1A1 gene polymorphisms are associated with toxicity in metastatic colorectal cancer patients treated with first-line irinotecan. Drug Metab Lett. 2007;1:23–30. doi: 10.2174/187231207779814328. [DOI] [PubMed] [Google Scholar]
  • 43.Genomes Project C., Auton A., Brooks L.D., Durbin R.M., Garrison E.P., Kang H.M., Korbel J.O., Marchini J.L., McCarthy S., McVean G.A., Abecasis G.R. A global reference for human genetic variation. Nature. 2015;526:68–74. doi: 10.1038/nature15393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lek M., Karczewski K.J., Minikel E.V., Samocha K.E., Banks E., Fennell T. Analysis of protein-coding genetic variation in 60,706 humans. Nature. 2016;536:285–291. doi: 10.1038/nature19057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Watson M.S., Cutting G.R., Desnick R.J., Driscoll D.A., Klinger K., Mennuti M., Palomaki G.E., Popovich B.W., Pratt V.M., Rohlfs E.M., Strom C.M., Richards C.S., Witt D.R., Grody W.W. Cystic fibrosis population carrier screening: 2004 revision of American College of Medical Genetics mutation panel. Genet Med. 2004;6:387–391. doi: 10.1097/01.GIM.0000139506.11694.7C. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yang Y., Botton M.R., Scott E.R., Scott S.A. Sequencing the CYP2D6 gene: from variant allele discovery to clinical pharmacogenetic testing. Pharmacogenomics. 2017;18:673–685. doi: 10.2217/pgs-2017-0033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Brown N.M., Pratt V.M., Buller A., Pike-Buchanan L., Redman J.B., Sun W., Chen R., Crossley B., McGinniss M.J., Quan F., Strom C.M. Detection of 677CT/1298AC “double variant” chromosomes: implications for interpretation of MTHFR genotyping results. Genet Med. 2005;7:278–282. doi: 10.1097/01.gim.0000159904.92850.d5. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Table S1
mmc1.xlsx (33.2KB, xlsx)

Articles from The Journal of Molecular Diagnostics : JMD are provided here courtesy of American Society for Investigative Pathology

RESOURCES