Pharmaco-Genomics: Methods and Protocols
Pharmaco-Genomics: Methods and Protocols
Federico Innocenti
Ron H. N. van Schaik Editors
Pharmaco-
genomics
Methods and Protocols
Second Edition
METHODS IN M O L E C U L A R B I O LO G Y ™
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Second Edition
Edited by
Federico Innocenti
Division of Pharmacotherapy and Experimental Therapeutics
Lineberger Comprehensive Cancer Center, Institute for Pharmacogenomics and
Individualized Therapy, Eshelman School of Pharmacy
University of North Carolina, Chapel Hill, NC, USA
Based upon the success of its first edition, the second edition of Pharmacogenomics: Methods
and Protocols aims to continue to provide readers with high-quality content on the most
innovative and commonly adopted technologies in the field of pharmacogenomics. Many
contributors to this book are leading experts in this field.
Pharmacogenomics: Methods and Protocols has become an established guide for investi-
gators in the selection and the experimental application of pharmacogenomic technolo-
gies. Using the extensive information in the materials and methods sections, investigators
will be able to easily perform each technique in their laboratories. This book is unique in
that it identifies and highlights problems that might be encountered in performing a spe-
cific technique and how to overcome these. Each procedure is described in a stepwise
fashion, providing detailed information from leading experts that is usually not found in
research articles.
Pharmacogenomics aims to study the genetic basis of interpatient variability in response
to drug therapy. Understanding an individual’s genetic makeup is the key to creating per-
sonalized drugs with greater efficacy and safety. Various technologies are currently available,
and this book aids the researchers’ decision on the most suitable method to apply.
In this updated edition, an introductory chapter describes the history of pharmacoge-
nomics and its current status. It is followed by Part II, which includes a variety of tech-
niques that are currently available to interrogate a patient’s genome. Readers will find
detailed information on eight technologies for SNP detection, plus three in-depth chapters
on recent technological developments in epigenetic techniques, sequencing, and quality
control. Relative to the first edition, newer methods such as SmartAmp, GoldenGate, and
Luminex X MAP have now been included.
Part III describes six methodologies and tools to assess and infer the functional sig-
nificance of allele variation in humans, including more innovative in vitro models (assays
to detect allelic imbalance or the effects of nonsynonymous variants and to guide iden-
tification of candidate genes) and in vivo assays in mice (use of genomically characterized
inbred mice and the hydrodynamic tail vein assay for human promoters and
enhancers).
Part IV describes current tools for supporting the translation and implementation of
pharmacogenomic markers in the clinic. Here, readers will find five completely new
chapters on the latest repositories of pharmacogenomic information, a summary guide
to the most recent Web-based resources of interest to pharmacogenomic researchers,
and two key examples of algorithms and guidelines for treatment personalization based
upon genetics.
Pharmacologists, geneticists, molecular biologists, and physicians in academic institu-
tions, in biotechnology, and in pharmaceutical industries will find Pharmacogenomics:
Methods and Protocols, second edition an essential reference and a valuable source on the
latest information in this field.
v
vi Preface
We are extremely grateful to all the authors for their excellent contributions making this
book a comprehensive and up-to-date resource for investigators in pharmacogenomics.
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 355
Contributors
xi
xii Contributors
Introduction to Pharmacogenomics
Chapter 1
Abstract
Pharmacogenomics and its predecessor pharmacogenetics study the contribution of genetic factors to the
interindividual variability in drug efficacy and safety. One of the major goals of pharmacogenomics is to
tailor drugs to individuals based on their genetic makeup and molecular profile. From early findings in the
1950s uncovering inherited deficiencies in drug metabolism that explained drug-related adverse events, to
nowadays genome-wide approaches assessing genetic variation in multiple genes, pharmacogenomics has
come a long way. The evolution of pharmacogenomics has paralleled the evolution of genotyping tech-
nologies, the completion of the human genome sequencing and the HapMap project. Despite these
advances, the implementation of pharmacogenomics in clinical practice has yet been limited. Here we
present an overview of the history and current applications of pharmacogenomics in patient selection, dos-
ing, and drug development with illustrative examples of these categories. Some of the challenges in the
field and future perspectives are also presented.
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_1, © Springer Science+Business Media, LLC 2013
3
4 Rosane Charlab and Lei Zhang
Clinical
Age Practice
DDI
Fig. 1 The drug response “puzzle.” Genetic makeup is just one of many factors that contribute to interindividual
variability in drug response. The contribution of each factor can be different for different patient–drug associa-
tions. DDI drug–drug interactions
2 Pharmacogenetics Is Born
Table 1
Pharmacogenetics is born: seminal findings
Variable Associated
Drug clinical effect Gene mechanisma References
Phenylthiourea Inability to taste “PTU nontaster Coding SNPs in [11, 12]
(PTU) phenylthiourea trait” taste receptor
TAS2R1
Primaquine Primaquine-related G6PD G6PD deficiency [92]
(antimalarial) hemolytic anemia
Succinilcholine Prolonged muscle Pseudocholinesterase Pseudocholinesterase [93]
(muscle relaxant) paralysis and deficiency
apnea
Isoniazid Isoniazid-induced NAT2 Reduced function [94, 95]
(antituberculosis) peripheral NAT2 variants
neuropathy in
“slow-acetylators”
Debrisoquine Adverse response to CYP2D6 Reduced function [96, 97]
(antihypertensive) debriquisone in CYP2D6 variants
“poor
metabolizers”
Thiopurines Increased risk of TPMT Reduced function [98]
myelossupression TPMT variants
in “poor
metabolizers”
Adapted from refs. 15, 27
Genes: TAS2R1 taste receptor, type 2, member 1; G6PD glucose-6-phosphate dehydrogenase; NAT2 N-acetyltransferase
2; CYP2D6 cytochrome P450, family 2, subfamily D, polypeptide 6; TPMT thiopurine S-methyltransferase
a
Associated mechanisms at molecular level were in some instances identified years apart of the original findings
Table 2
Genetic variation among individuals can influence pharmacokinetics and/or pharmacodynamics
and affect drug benefit/risk profile
Gene (and
associated
Therapeutic agent Clinical effect variants) Problem affecting efficacy/safety
Metabolism
Warfarin Anticoagulant CYP2C9 PK: Increased risk of bleeding in PMs
Codeine Analgesic CYP2D6 PK: Lack of analgesia in CYP2D6 PMs,
(pro-drug) toxicity in UMs
Nortriptyline Antidepressant CYP2D6 PK: Increased adverse event risk in CYP2D6
PMs, decreased efficacy in UMs
Clopidogrel Anti-thrombotic CYP2C19 PK: Decreased bioactivation and reduced
(pro-drug) response to clopidogrel in PMs
Mercaptopurine Antineoplastic TPMT PK: Increased myelosupression risk in PMs
Irinotecan Antineoplastic UGT1A1 PK: Increased hematological toxicity risk in
the reduced function UGT1A1*28
carriers
Transport
Simvastatin Lipid-lowering drug SLCO1B1 PK: SNP associated with reduced hepatic
uptake and increased risk of statin-
induced myopathy
Drug target
Warfarin Anticoagulant VKORC1 PD: Decreased dose requirement associated
to VKORC1 SNP
Beta blockers Treat high blood ADRB1 PD: SNPs associated to variability in
pressure/decrease response to beta blockers
heart rate
Trastuzumab Antineoplastic ERBB2 PD: Effective in patients overexpressing
(HER2) HER2 receptor on tumor cells
Other
Abacavir Anti-HIV HLA-B*5701 PD/mechanistic: High risk of severe
hypersensitivity reaction in HLA-B*5701
allele carriers
Carbamezapine Anticonvulsant HLA-B*1502 PD/mechanistic: High risk of SJS/TEN
with HLA-B*1502 allele in Han Chinese
Pegylated Antiviral for IL28B PD/mechanistic: SNP near to IL28B gene
interferon hepatitis C is associated to response to PegIFN and
(PegINF) and RBV for patients with chronic genotype
ribovarin (RBV) 1 HCV infection [99]
Modified from refs. 22, 27, 100, 101
PK pharmacokinetics, PD pharmacodynamics, SNP single nucleotide polymorphism, PM poor metabolizer, UM ultrarapid
metabolizer, SJS/TEN Stevens Johnson syndrome/Toxic epidermal necrolysis, HIV human immunodeficiency virus, HCV
Hepatitis C virus
Genes: CYP2C9 cytochrome P450, family 2, subfamily C, polypeptide 9; CYP2D6 cytochrome P450, family 2, subfam-
ily D, polypeptide 6; CYP2C19 cytochrome P450, family 2, subfamily C, polypeptide 19; TPMT thiopurine
S-methyltransferase; UGT1A1 UDP glucuronosyltransferase 1 family, polypeptide A1; SLCO1B1 solute carrier organic
anion transporter family, member 1B1; VKORC1 vitamin K epoxide reductase complex, subunit 1; ADRB1 adrenergic,
beta-1-, receptor; ERBB2 (HER2) v-erb-b2 erythroblastic leukemia viral oncogene homolog 2, neuro/glioblastoma
derived oncogene homolog (avian); HLA-B major histocompatibility complex, class I, B; IL28B interleukin 28B (inter-
feron, lambda 3)
8 Rosane Charlab and Lei Zhang
5 Pharmacogenomics Applications
5.1 Pharmacoge- Toxicity effects and serious adverse drug reactions (SADRs) are
nomics and Adverse responsible for a large amount of deaths and hospitalizations per
Drug Reactions: year in the US, for the preclusion of potential drugs for entering the
Who Is at Risk? market and for postmarketing withdrawal of approved drugs [36].
SADRs known as type A are predictable events based on the drug
pharmacokinetic and pharmacodynamic properties, and are typi-
cally dose-related. SADRs can also be unpredictable (of idiosyn-
cratic nature; known as type B) and not related to dose, and thus
more worrisome. Some type B SADRs have an underlying immu-
noallergenic mechanism not yet completely understood [36, 40].
Pharmacogenomic approaches can be used to uncover potential
gene(s) associated to the adverse events and develop biomarkers for
Pharmacogenomics: Historical Perspective and Current Status 11
5.2 Predictors of Individualized drug therapy is particularly needed for agents with
Efficacy: Who Benefits narrow therapeutic index and when the consequences of drug tox-
the Most? icity or lack of efficacy are severe and potentially fatal such as for
antineoplastic agents or anticoagulants [60].
5.2.1 Anticancer Drugs Only a small proportion of cancer patients respond to available
and Tumor Molecular anticancer therapies. Furthermore, patients can develop resistance
Landscape to therapy in a short period of time. Tumor genomes present sev-
eral genetic or genomic structural changes and are highly hetero-
geneous. This heterogeneity can be observed both among different
patients with the same tumor type, and within the same patient
when different tumor sites are compared at molecular level. This
striking variability allied to narrow therapeutic index of many anti-
cancer agents make cancer pharmacogenomics a field rich in oppor-
tunities for tailoring therapy to individual patients. Recent examples
with several cancers [61–63] suggest that tumor types classically
subdivided and classified by histopathology can be further subdi-
vided in molecular subsets. These subsets are characterized by spe-
cific alterations at molecular level that “drive” the tumor (the
so-called “oncogenic drivers”) and are critical for its survival (e.g.,
point mutations, translocations, amplifications). Most of these
genetic alterations are acquired or somatic, i.e., they are only pres-
ent in the tumor tissue.
The increased understanding of the tumor molecular land-
scape and signaling has driven the development of antineoplastic
agents against these specific tumor molecular alterations (mostly in
the drug target or associated pathway). Impressive response rates
have been observed with this approach in the molecularly defined
subset of patients positive for the alteration and/or negative for
alterations associated with resistance to therapy. This targeting
approach is also expected to have a better safety profile than most
chemotherapy drugs—nonselective and designed to fit all.
However, in this scenario, traditional histologies may be subdi-
vided in smaller and smaller molecular subsets adding complexity
and challenge to drug development. One of the earlier examples is
the use of tamoxifen in estrogen receptor-positive metastatic breast
cancer. Other examples (Table 3) include imatinib, a tyrosine
kinase inhibitor indicated for BCR-ABL translocation positive
chronic myelogenous leukemia (CML), trastuzumab, a monoclo-
nal antibody with clinical efficacy in patients with breast cancer
positive for v-erb-b2 erythroblastic leukemia viral oncogene homo-
log 2 (HER2) gene amplification or overexpression of the HER2
protein, erlotinib, an epidermal growth factor receptor (EGFR)
tyrosine kinase inhibitor most effective in non-small-cell lung can-
cer (NSCLC) positive for EGFR activating mutations,
14 Rosane Charlab and Lei Zhang
Table 3
Examples of targeted oncology agents and associated tumor molecular alterations
Labeling
Fig. 2 Pharmacogenomic maneuvers in drug development. Original Published Source: Figure 4 from reference
[86]. PGx pharmacogenomic, FIH first in human, DDI drug–drug interactions, HV healthy volunteers, ADME
absorption, distribution, metabolism, excretion, D/R dose–response, C/R concentration–response
6 Future Perspectives
Disclaimer
The views expressed in this article are those of the authors and may
not necessarily represent FDA policy. No official endorsement is
intended, nor should be inferred.
Acknowledgment
The authors acknowledge Dr. Issam Zineh for critical review of the
manuscript.
Pharmacogenomics: Historical Perspective and Current Status 19
References
ceptibility to the effects of chemotherapy. bench. Nat Rev Gastroenterol Hepatol 8(4):
Clin Cancer Res 15(15):4806–4814 202–211
33. Lee W et al (2005) Cancer pharmacogenom- 48. Pirmohamed M (2010) Pharmacogenetics of
ics: powerful tools in cancer chemotherapy idiosyncratic adverse drug reactions. Handb
and drug development. Oncologist Exp Pharmacol 196:477–491
10(2):104–111 49. Becquemont L (2010) HLA: a pharmacoge-
34. Judson R et al (2000) The predictive power nomics success story. Pharmacogenomics
of haplotypes in clinical response. 11(3):277–281
Pharmacogenomics 1(1):15–26 50. Mallal S et al (2008) HLA-B*5701 screening
35. Fujiwara Y, Minami H (2010) An overview of for hypersensitivity to abacavir. N Engl J Med
the recent progress in irinotecan pharmacoge- 358(6):568–579
netics. Pharmacogenomics 11(3):391–406 51. Hung SI et al (2006) Genetic susceptibility to
36. Becquemont L (2009) Pharmacogenomics of carbamazepine-induced cutaneous adverse
adverse drug reactions: practical applications drug reactions. Pharmacogenet Genomics
and perspectives. Pharmacogenomics 10(6): 16(4):297–306
961–969 52. TEGRETOL(Carbamazepine) Labeling
37. Wu X et al (2009) Strategies to identify phar- [Online]. http://www.accessdata.fda.gov/
macogenomic biomarkers: candidate gene, drugsatfda_docs/label/2011/
pathway-based, and genome-wide approaches. 016608s100s102 s.s., 018927s041s042,020
In: Innocenti F (ed) Genomics and pharma- 234s031s033lbl.pdf. Accessed 31 Oct 2011
cogenomics in anticancer drug development 53. McCormack M et al (2011) HLA-A*3101
and clinical response. Humana Press, Totowa and carbamazepine-induced hypersensitivity
NJ, pp 353–370 reactions in Europeans. N Engl J Med
38. Feero WG et al (2010) Genomic medicine— 364(12):1134–1143
an updated primer. N Engl J Med 362(21): 54. Ozeki T et al (2011) Genome-wide association
2001–2011 study identifies HLA-A*3101 allele as a genetic
39. Metzker ML (2010) Sequencing risk factor for carbamazepine-induced cutane-
technologies—the next generation. Nat Rev ous adverse drug reactions in Japanese popula-
Genet 11(1):31–46 tion. Hum Mol Genet 20(5):1034–1041
40. Wilke RA et al (2007) Identifying genetic risk 55. Somkrua R et al (2011) Association of HLA-
factors for serious adverse drug reactions: cur- B*5801 allele and Allopurinol-induced
rent progress and challenges. Nat Rev Drug Stevens Johnson syndrome and toxic epider-
Discov 6(11):904–916 mal necrolysis: a systematic review and meta-
41. Andrade RJ et al (2009) Drug-induced liver analysis. BMC Med Genet 12(1):118
injury: insights from genetic studies. 56. Niemi M (2010) Transporter pharmacoge-
Pharmacogenomics 10(9):1467–1487 netics and statin toxicity. Clin Pharmacol
42. Huang YS (2010) Tailored drug therapy for Ther 87(1):130–133
mitigating drug-induced liver injury: is this 57. Link E et al (2008) SLCO1B1 variants and
the era of genetic screening? Pers Med statin-induced myopathy—a genomewide
7(1):5–8 study. N Engl J Med 359(8):789–799
43. Wang L et al (2011) Genomics and drug 58. Walko CM, McLeod H (2009)
response. N Engl J Med 364(12):1144–1153 Pharmacogenomic progress in individualized
44. Daly AK et al (2009) HLA-B*5701 genotype dosing of key drugs for cancer patients. Nat
is a major determinant of drug-induced liver Clin Pract Oncol 6(3):153–162
injury due to flucloxacillin. Nat Genet 59. Innocenti F, Ratain MJ (2006)
41(7):816–819 Pharmacogenetics of irinotecan: clinical per-
45. Kindmark A et al (2008) Genome-wide phar- spectives on the utility of genotyping.
macogenetic investigation of a hepatic adverse Pharmacogenomics 7(8):1211–1221
event without clinical signs of immunopathol- 60. Wilke RA, Dolan ME (2011) Genetics and vari-
ogy suggests an underlying immune patho- able drug response. JAMA 306(3):306–307
genesis. Pharmacogenomics J 8(3):186–195 61. Carlquist JF, Anderson JL (2011)
46. Singer JB et al (2010) A genome-wide study Pharmacogenetic mechanisms underlying
identifies HLA alleles associated with unanticipated drug responses. Discov Med
lumiracoxib-related liver injury. Nat Genet 11(60):469–478
42(8):711–714 62. Lovly CM, Carbone DP (2011) Lung cancer
47. Tujios S, Fontana RJ (2011) Mechanisms of in 2010: one size does not fit all. Nat Rev Clin
drug-induced liver injury: from bedside to Oncol 8(2):68–70
Pharmacogenomics: Historical Perspective and Current Status 21
63. Biankin AV, Hudson TJ (2011) Somatic vari- 77. Yuan HY et al (2005) A novel functional
ation and cancer: therapies lost in the mix. VKORC1 promoter polymorphism is associ-
Hum Genet 130(1):79–91 ated with inter-individual and inter-ethnic dif-
64. Chapman PB et al (2011) Improved survival ferences in warfarin sensitivity. Hum Mol
with vemurafenib in melanoma with BRAF Genet 14(13):1745–1751
V600E mutation. N Engl J Med 364(26): 78. Gage BF, Lesko LJ (2008) Pharmacogenetics
2507–2516 of warfarin: regulatory, scientific, and clinical
65. Shaw AT et al (2011) Effect of crizotinib on issues. J Thromb Thrombolysis 25(1):45–51
overall survival in patients with advanced non- 79. Caraco Y et al (2008) CYP2C9 genotype-
small-cell lung cancer harbouring ALK gene guided warfarin prescribing enhances the effi-
rearrangement: a retrospective analysis. Lancet cacy and safety of anticoagulation: a
Oncol 12(11):1004–1012 prospective randomized controlled study.
66. Hutchinson L (2010) Targeted therapies: Clin Pharmacol Ther 83(3):460–470
activated PI3K/AKT confers resistance to 80. Gage BF et al (2008) Use of pharmacogenetic
trastuzumab but not lapatinib. Nat Rev Clin and clinical factors to predict the therapeutic
Oncol 7(8):424 dose of warfarin. Clin Pharmacol Ther 84(3):
67. Ellis LM, Hicklin DJ (2009) Resistance to 326–331
targeted therapies: refining anticancer therapy 81. Lenzini P et al (2010) Integration of genetic,
in the era of molecular oncology. Clin Cancer clinical, and INR data to refine warfarin
Res 15(24):7471–7478 dosing. Clin Pharmacol Ther 87(5):
68. PLAVIX (Clopidogrel) Labeling [Online]. 572–578
http://www.accessdata.fda.gov/drugsatfda_ 82. COUMADIN (Warfarin) prescribing informa-
docs/label/2011/020839s051lbl.pdf . tion. [Online]. www.accessdata.fda.gov/drug-
Accessed 28 Aug satfda_docs/label/2010/009218s108lbl.pdf.
69. Mega JL et al (2009) Cytochrome p-450 Accessed 28 Aug
polymorphisms and response to clopidogrel. 83. Tetrabenazine Clinical Pharmacology Review.
N Engl J Med 360(4):354–362 [Online]. http://www.accessdata.fda.gov/
70. Mega JL et al (2010) Reduced-function drugsatfda_docs/nda/2008/
CYP2C19 genotype and risk of adverse clini- 021894s000TOC.cfm. Accessed 28 Aug
cal outcomes among patients treated with 84. XENAZINE (Tetrabenazine) Labeling
clopidogrel predominantly for PCI: a meta- [Online]. http://www.accessdata.fda.gov/
analysis. JAMA 304(16):1821–1830 drugsatfda_docs/label/2011/021894s004lbl.
71. Scott SA et al (2011) Clinical pharmacoge- pdf. Accessed 28 Aug
netics implementation consortium guidelines 85. Stingl Kirchheiner JC, Brockmoller J (2011)
for cytochrome P450-2C19 (CYP2C19) gen- Why, when, and how should pharmacogenet-
otype and Clopidogrel therapy. Clin ics be applied in clinical studies? Current and
Pharmacol Ther 90(2):328–332 future approaches to study designs. Clin
72. Daly AK, King BP (2003) Pharmacogenetics Pharmacol Ther 89(2):198–209
of oral anticoagulants. Pharmacogenetics 86. Zineh I, Pacanowski MA (2011)
13(5):247–252 Pharmacogenomics in the assessment of ther-
73. Kim MJ et al (2009) A regulatory science per- apeutic risks versus benefits: inside the United
spective on warfarin therapy: a pharmacoge- states food and drug administration.
netic opportunity. J Clin Pharmacol 49(2): Pharmacotherapy 31(8):729–735
138–146 87. Diamandis M et al (2010) Personalized medi-
74. Scordo MG et al (2002) Influence of CYP2C9 cine: marking a new epoch in cancer patient
and CYP2C19 genetic polymorphisms on war- management. Mol Cancer Res 8(9):
farin maintenance dose and metabolic clear- 1175–1187
ance. Clin Pharmacol Ther 72(6):702–710 88. Houle D et al (2010) Phenomics: the next
75. Sconce EA et al (2005) The impact of challenge. Nat Rev Genet 11(12):855–866
CYP2C9 and VKORC1 genetic polymor- 89. Pirmohamed M et al (2011) The phenotype
phism and patient characteristics upon warfa- standardization project: improving pharma-
rin dose requirements: proposal for a new cogenetic studies of serious adverse drug
dosing regimen. Blood 106(7):2329–2333 reactions. Clin Pharmacol Ther 89(6):
76. Marsh S et al (2006) Population variation in 784–785
VKORC1 haplotype structure. J Thromb 90. Lanktree MB et al (2010) Phenomics:
Haemost 4(2):473–474 expanding the role of clinical evaluation in
22 Rosane Charlab and Lei Zhang
Abstract
Denaturing high-performance liquid chromatography (DHPLC) is an accurate and efficient screening
technique used for detecting DNA sequence changes by heteroduplex analysis. It can also be used for
genotyping of single nucleotide polymorphisms (SNPs). The high sensitivity of DHPLC has made this
technique one of the most reliable approaches to mutation analysis and, therefore, used in various areas of
genetics, both in the research and clinical arena. This chapter describes the methods used for mutation
detection analysis and the genotyping of SNPs by DHPLC on the WAVE™ system from Transgenomic
Inc. (“WAVE” and “DNASep” are registered trademarks, and “Navigator” is a trademark, of Transgenomic,
used with permission. All other trademarks are property of the respective owners).
1 Introduction
1.1 Mutation The basis of mutation detection by DHPLC is the formation and
Detection by DHPLC discrimination of homoduplex and heteroduplex DNA molecules
that can be created when a DNA sequence change is present on
one allele [1]. The DHPLC cartridge (DNASep™) contains a non-
porous matrix consisting of polystyrene–divinylbenzene copolymer
beads. The beads are alkylated with C-18 chains which form single
C–C bonds, are electrostatically neutral and do not interact with
nucleic acids [2]. DNA binds to the cartridge matrix by the use of
triethylammonium acetate (TEAA) that serves as an ion-pairing
reagent between nucleic acids and the beads in the cartridge. The
positively charged triethylammonium ion bonds to the negatively
charged phosphate group on the DNA backbone and the hydro-
phobic groups of triethylammonium acetate interact with the
hydrophobic C-18 chains on the copolymer beads. DNA is eluted
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_2, © Springer Science+Business Media, LLC 2013
25
26 Donna Lee Fackenthal et al.
denature
and cool
A C G T A T GC
A T GC
1.2 Genotyping The DHPLC instrument can also be used for genotyping of SNPs.
by DHPLC DNA sequence fragments that differ at a single base pair position
can be distinguished on the DHPLC due to the differing hydro-
phobicities of different base pairs that can cause a change in their
elution profile [6]. This characteristic is taken advantage of in geno-
type applications used on the DHPLC and one that has been used
successfully is single-base extension (SBE) genotyping [7]. Single-
base extension on the DHPLC (SBE-DHPLC) is performed by an
initial polymerase chain reaction of an amplicon with a single-base
change to be genotyped followed by an extension reaction using an
oligonucleotide that acts as a single-base extension primer. The
SBE primer is annealed downstream or upstream immediately
adjacent to the SNP to be genotyped in the 5′ to 3′ direction.
Thermosequenase extends the 3′ end of the extension primer with the
appropriate ddNTP. The primer extends one base only because the
ddNTP terminates further extension. Extended products are sepa-
rated on the DHPLC based on the hydrophobicity of the last base,
so although the lengths of the extended products are the same for
different alleles, the hydrophobicity of the extended products of
each allele will be different.
Another variation of single-base extension genotyping on the
DHPLC is primer extension genotyping where a combination of
dNTPs and ddNTPs are added to the reaction so that depending
on the allele present, either extension beyond the single-base or
just single-base extension occurs [8]. Separation of the extended
products then becomes a function of the differing lengths of the
two extended products. This review will focus on the protocol for
single-base extension genotyping.
The utility of the DHPLC for genotyping is not as widespread as
its mutation detection application. However, the utility and effective-
ness of SBE-DHPLC for genotyping purposes has been clearly
demonstrated [7]. In our experience genotyping by SBE-DHPLC is
a very robust technique and has often worked where other methods
of genotyping have failed. It is a very useful methodology for medium
scale genotyping projects of approximately 500–1,000 samples.
2 Materials
2.1 Instrumentation 1. The WAVE system from Transgenomic Inc., Omaha, NE, is the
most widely used system for DHPLC analysis. The methods
described in this chapter pertain specifically to the WAVE™
Nucleic Acid Fragment Analysis System 3500HT, although
applicable to other WAVE™ model types (see Note 1). The
3500HT system is a high throughput system that allows for
analysis of hundreds of samples. It consists of six major compo-
nents (Fig. 2): degasser, in which the two ion-pairing buffers
Sample Introduction
Degasser
Waste Container
Computer
Buffers A, B, C, D
Fig. 2 System flowpath of the WAVE nucleic acid fragment analysis system 3500HT
Denaturing High-Performance Liquid Chromatography for Mutation Detection… 29
2.2 Software At the time of printing, Navigator™ Software Version 3.0 is the
operating software for the WAVE™ platform (Transgenomic, Inc.,
Omaha, NE).
3 Methods
3.1 Mutation The following considerations should be taken into account when
Detection by choosing amplicons to be analyzed by DHPLC for mutation
Heteroduplex Analysis detection:
3.1.1 PCR 1. The optimal size of the amplicons should be 180–700 bp.
Amplicon Design 2. The melting temperature range of the amplicon should be
between 52 and 75 °C.
3. Ideally choose one amplicon with one melting domain as
opposed to multiple melting domains. Figure 3a shows an
amplicon with a single melting domain and Fig. 3b demon-
strates an amplicon with multiple melting domains. When the
amplicon of interest has multiple melting domains it may be
necessary to break the fragment into smaller amplicons with
one melting domain each or incorporate GC clamps (see Note 4)
to PCR primers to even out melting domains within the ampli-
con, thereby obtaining one melting temperature.
a
1.00
Helical Fraction %
0.75
0.50
0.25
0.00
0 50 100 150 200 250
Base Position
Domain 1
b
1.00
0.75 Domain 2
Helical Fraction %
0.50
0.25
0.00
0 50 100 150 200 250 300
Base Position
Fig. 3 (a) Amplicon with single melting domain; (b) Amplicon with two melting
domains
32 Donna Lee Fackenthal et al.
3.1.2 Preparation of PCR 1. PCRs for subsequent DHPLC analysis are performed using
Samples for Mutation regular touchdown PCR protocols and in 50 μL volumes to
Detection allow for sufficient volume injections for DHPLC analysis at
various temperatures.
2. Negative control DNA samples should be included for every
amplicon being analyzed and positive control DNA samples
should be included when available (see Note 5). A negative
control is a sample with no sequence change in the amplicon
and a positive control is a sample with a known sequence
change in the amplicon being analyzed. A blank (H2O) control
should also be included to check for PCR contamination.
3. After the PCR, samples are denatured and gradually allowed
to reanneal to create homoduplex and heteroduplex products.
To do this, samples are briefly spun down and denatured and
slowly reannealed over 60 min with the following cycling
profile: [95 °C for 5 min, ramp 95 °C → 45 °C over 60 min,
45 °C for 30 s, hold at 4 °C] (see Note 6).
3.1.3 Instrument 1. The Cartridge for sample injections should be installed in the
and Cartridge Preparation WAVE oven as per manufacturer’s instructions and the oven
temperature should be set to 50 °C for sizing PCR products
(see Note 7).
2. The volumes of Buffer A, Buffer B, Solution D and the Syringe
Wash Solution (Buffer C) should be checked to make sure that
sufficient buffer exists for the number of injections to be per-
formed (see Note 3). Check waste receptacle, exchange if
receptacle is almost full to capacity.
3. Wash the cartridge with 100 % Buffer D at flow rate 1.5 mL/
min for 10 min.
This is performed by entering 100 for %D and 0 for %B
and %C and changing the flow rate to 1.5 mL/min on the
pump keypad.
4. Wash the syringe five times by pushing the WASH button on
the instrument’s autosampler.
5. Purge the pumps by setting Buffer A, Buffer B, and Solution D
to 33 % each, flip purge valve in the pump chamber to the
“open” position, press “purge.” Purge for 2 min. Press “purge”
again and close the purge valve. Purging the pump helps to
eliminate air bubbles.
Denaturing High-Performance Liquid Chromatography for Mutation Detection… 33
3.1.4 Set-up of Project Certain criteria are important to set up as default settings as
Defaults they pertain to all mutation detection runs. Once these settings are
created they need not be entered each time prior to each run.
1. On the Menu Bar, choose Setup then Project Defaults.
2. In the Equilibrate Cartridge area: check the Before first Injection
box and enter 3 min. This is necessary for equilibrating the
cartridge prior to the first injection. Check the After Temperature
Change box and enter 5 min. Again, it is necessary to equilibrate
the cartridge (see Note 9). Check the After Gradient Change
box and enter 5 min. This allows for a 5 min equilibration of
the cartridge in between changing of the buffer gradient.
3. In the Injection Ordering area: check the Run in Temperature
Order Ascending. This allows for samples to be injected in
ascending temperature order thereby minimizing the number
of times the oven needs to change temperature.
4. In the Clean Options area: check Normal Clean (see Note 10).
5. In the Injection area: Select Injection Type ALL. This injection
type gives better intensity. In the Default Injection Volume
enter 7 μL. In the Feed Volume, that is, the volume of syringe
wash solution injected into the flow path, enter 25 μL when
the Injection Type is ALL.
6. Disable Tray Change Request is optional. If this is checked,
the tray change prompt will not appear when a run is started.
This is especially useful when two trays are used for one run.
3.1.6 Create Sample A sample sheet is a table that specifies the injection order and method
Sheet (Injection Table) type to use for a series of samples to be analyzed. In a sample sheet,
information including such variables as sample name, sample loca-
tion in the tray (vial), method to be used (that links to information
such as application type, volume to be injected for each sample, oven
temperature, clean type, and flow rate) is listed. This needs to be set
up for every set of samples to be analyzed and prior to each run.
1. The sample sheet should be set up following the Navigator™
Software Manual that lists detailed step-by-step instructions.
2. It is recommended that each sample be injected once for sizing
(see Note 7). This is performed using a Sizing application type,
that is, DS (double-stranded) Single Fragment (see Note 11).
It is recommended that each sample be injected three times for
mutation detection analysis, using the three different tempera-
tures calculated for optimal detection of sequence change for
the particular amplicon in question (see Note 12).
3.1.7 Running Samples 1. After all the daily maintenance has been performed
(Subheading 3.1.3, steps 2–6) and the sample sheet created,
run the samples by highlighting specific injections then press-
ing the run injection button indicated by the green triangle or
simply pressing the run injection button when all injections in
the sample sheet are to be run.
2. The first 3 min of the run is an equilibration. The equilibration
line should be flat at 0 mV. A slight deviation in the line is nor-
mal. If the line is not flat at 0 mV the run must be discontinued
and the cartridge equilibrated for an additional 10 min.
3.1.8 Analyzing Results 1. As previously mentioned, for all amplicons being analyzed for
mutation detection, a normal control (a sample with no
sequence change in the amplicon) should be included, and a
positive control (a sample with a known sequence change in
the amplicon), if available, should be included.
2. Compare the chromatogram of the normal control with the
experimental samples for analysis. An absence of a change in
the chromatogram between the experimental sample and the
normal control indicates no sequence change present in the
amplicon of the experimental sample. If a sequence change is
present in the amplicon of the experimental sample this will be
36 Donna Lee Fackenthal et al.
Experimental Sample
Fig. 4 Normal and experimental samples indicating homoduplex and heteroduplex peaks
3.2.5 Instrument 1. The cartridge for sample injections should be installed in the
and Cartridge Preparation WAVE oven as per manufacturer’s instructions and oven tem-
perature should be set to 70 °C to keep extension products
denatured.
2. The volumes of Buffer A, Buffer B, Solution D and the Syringe
Wash Solution (Buffer C) should be checked to make sure that
sufficient buffer exists for the number of injections to be per-
formed (see Note 3). Check waste receptacle, exchange if
receptacle is almost full to capacity.
3. Wash the cartridge with 100 % Solution D at flow rate 1.5 mL/
min for 10 min.
This is performed by entering 100 for %D and 0 for %B
and %C and changing the flow rate to 1.5 mL/min on the
pump keypad.
4. Wash the syringe five times by pushing the WASH button on
the instrument’s autosampler.
5. Purge the pumps by setting Buffer A, Buffer B, and Solution D
to 33 % each, flip purge valve in the pump chamber to the
“open” position, press “purge.” Purge for 2 min. Press “purge”
again and close the purge valve. Purging the pump helps to
eliminate air bubbles.
6. Equilibrate the cartridge at 65 % Buffer A and 35 % Buffer B at
flow 1.5 mL/min for 20 min. This is performed by entering
35 for %B and 0 for %C and %D and changing the flow rate to
1.5 mL/min on the pump keypad.
Note: Steps 2–6 should be performed once daily prior to run-
ning samples on the instrument. This helps to keep the cartridge
and flow path clean and free from impurities. In addition, for opti-
mal instrument and cartridge performance, regular quality control
procedures should be performed at regular intervals (see Note 8).
3.2.6 Set-up Certain criteria are important to set up as default settings as they
of Project Defaults pertain to all single-base extension runs. Once these settings are
created they need not be entered each time prior to each run.
1. On the Menu Bar, choose Setup then Project Defaults.
2. In the Equilibrate Cartridge area: Check the Before first
Injection box and enter 3 min. This is necessary for equilibrating
the cartridge prior to the first injection.
Denaturing High-Performance Liquid Chromatography for Mutation Detection… 39
(g) Click Save after entering the parameters but first carefully
check all the values of the parameters as sometimes when
one parameter is entered or modified, it might also change
one or some of the other parameters. If this happens the
original values need to be re-entered. This is a result of the
software application.
3. Time shift (optional)—The time shift is an adjustment in min-
utes that moves the elution of the fragment of interest either
earlier or later in the gradient. The value of the time shift can be
negative (earlier elution) or positive (later elution). The time
shift actually offsets the gradient by the formula: value × slope.
For example, a slope of 2.5 %/min and a time shift value of
+1.0 min decreases all values for %B (not including clean-off)
by 2.5 %. The lower percentage of Buffer B results in peaks
increasing in retention time. A time shift is recommended if, for
example:
1. The peak of interest is eluting too late which would result
in an absence of the peak on the chromatogram. Change
time shift default to a negative value such as −0.5 which
results in earlier elution.
2. The peak of interest elutes too early, then change the value
to +0.5 min.
Essentially, the slope of the gradient changes as a result of a
time shift.
3.2.8 Create Sample A sample sheet is a table that specifies the injection order and method
Sheet (Injection Table) type to use for a series of samples to be analyzed. In a sample sheet,
information including such variables as sample name, sample location
in the tray (vial), method to be used (that links to information such
as application type, volume to be injected for each sample, oven
temperature, clean type and flow rate) is listed. This needs to be set
up for every set of samples to be analyzed and prior to each run.
The sample sheet should be set up following the Navigator™
Software Manual that lists detailed step-by-step instructions.
3.2.9 Running Samples The primers should be initially injected individually to check the
elution time. This is critical when performing multiplex SBE as the
peaks need to be separated by at least 30 s.
Denaturing High-Performance Liquid Chromatography for Mutation Detection… 41
3.2.10 Analyzing 1. On the Injection Page, select the appropriate Tray Name for
Single-Base Extension the run which is to be analyzed and click on the Results tab.
Results Two charts with x and y-axis will be displayed. The x-axis indi-
cates minutes and the y-axis represents the absorbance. In the
Injection Table, under Chart 1 (for graph 1), highlight the
blank control as well as the known genotyped control samples.
The results of the experimental samples will be compared to
these controls and the genotypes determined. Under Chart 2,
highlight each sample individually to read the genotype.
2. Based on the increasing hydrophobicity of the four bases on the
extension products, the elution order is C<G<A<T, i.e., the C
extension product elutes first and the T extension product
elutes last [7]. Figure 5 displays the extension products for a
single reaction whereas Figs. 6 and 7 show extension products
for duplex and triplex reactions, respectively. It should be noted
that the elution order of T and A may sometimes be reversed
(see Note 26).
Fig. 5 Demonstration of single base extension products in three samples and a blank control sample. In this
example the extension products are G and A nucleotides. The genotype of each sample is determined based
on the extension products obtained. u.p. unextended primer
42 Donna Lee Fackenthal et al.
Fig. 6 Demonstration of single base extension products in a duplex reaction where two SNP regions are being
genotyped simultaneously. The extension products are G, T and C, T respectively. The genotype of each sample
is determined based on the extension products obtained. This example demonstrates the results in four experimental
samples and one blank control sample. u.p. unextended primer
Fig. 7 Demonstration of single base extension products in a triplex reaction where three SNP regions are being
genotyped simultaneously. The extension products for all three SNPs are C and A. The genotype of each sample
is determined based on the extension products obtained. This example demonstrates the results in two experi-
mental samples and one blank control sample. U.P. unextended primer. Note—For the first SNP, a reverse
extension primer has been used necessitating reverse complementing of the extended bases for genotype call.
For the second two SNPs forward extension primers have been used
3.3 Mutation 1. This section provides general instructions for the detection of
Detection and mutations using the SURVEYOR® PLUS Mutation Detection
Genotyping Using Kit for WAVE and WAVE HS Systems. In general, processing
SURVEYOR® Nuclease of samples should be carried out from start to finish as
described in the Transgenomic User Guide. If processing of a
sample is stopped before completion of all steps, the DNA
should be stored at −20 °C until the next step is carried out.
However, exposure of any frozen sample to repeated freeze-thaw
Denaturing High-Performance Liquid Chromatography for Mutation Detection… 43
4 Notes
1. There are four WAVE™ system models. The 3500 is the base
model without high throughput capacity. It uses the DNASep™
Cartridge, has a larger mixer than the 3500HT and does not
44 Donna Lee Fackenthal et al.
Fig. 9 Heteroduplex/homoduplex digestion peaks shown overlaid on the homoduplex digestion profile.
Fluorescent signal produced by SURVEYOR® Nuclease digestion of heteroduplex DNA present in 1:64 and
1:128 ratio of homoduplex DNA. Products were analyzed using the WAVE™ System run under non-denaturing
conditions at 50 °C and equipped with a Fluorescence Detector and a High-Sensitivity Accessory for post-
column DNA intercalation with fluorescent dye
3′ End Up cgggacgc
5′ End Up gcgtcccg
25 bp GCT cgcccgccgccgcccgccgcccgtc
30 bp GCT cgcccgccgccgcccgccgcccgtcccgcc
40 bp GCT cgcccgccgcgccccgcgcccgtcccgccgcccccgcccg
10 bp GC cgcccgccgc
15 bp GC cgcccgccgccgccc
20 bp GC cgcccgccgccgcccgccgc
3′ GC 10 bp cgggcggggg
3′ GC 20 bp cgggcgggggcggcgggccg
5′ GC 10 bp gcccccgccg
5′ GC 20 bp gcggcccgccgcccccgccg
20. The extension primer is designed such that its 3′ end lies imme-
diately adjacent to the SNP to be genotyped. The extension
primer can be designed to lie either upstream or downstream
of the SNP. The sequence surrounding the SNP often times
determines which direction to design the extension primer
(i.e., upstream/forward or downstream/reverse). The length
of the extension primer can range between 18 and 24 bp with
an optimal length of 20 bp and aim for a TM of ~60 °C with
~50 % AT and 50 % GC content. When designing the primer
avoid areas of repeats and check primers for hairpin loops and
primer dimer formation especially at the 3′ end of the primer.
Single base changes can be introduced into the primer (usually
towards the 5′ end) if needed to stabilize the structure, break
hairpin loops etc.
21. Dilute the stock enzyme [32 U/μL] 1:12.8 with Thermo
Sequenase™ Enzyme Dilution Buffer. Final concentration should
be 2.5 U/μL. Enzyme solution should be prepared fresh.
22. The following considerations must be taken into account when
designing extension primers for multiplex single-base exten-
sion. There must be sufficient separation between primers, that
is, there must be a minimum of 30 s elution time separating
the primers. This is generally obtained by having primers differ
in length by 2 bp or more. GC content as well as the sequence
content must be considered. GC-rich primers elute earlier than
AT-rich primers. Hydrophobicity of the primers is a factor; for
example, elution times are shorter for cytosine and guanine.
Elution times may also be adjusted with the addition of GC or
AT clamps onto the 5′ end of the extension primer.
23. Prior to pooling reactions one needs to verify that different
extension primers and their extension products can be separated
on the DHPLC by sizing aliquots of the extension primers to
check their separation. Ideally there should be a separation of
a minimum of 30 s between extension primers to allow for
clear separation of the corresponding extension products.
Pooling can be performed when it is difficult to perform mul-
tiplex reactions. Reactions are performed separately and
pooled prior to running on the DHPLC. Pooling (as with
multiplex reactions) saves on DHPLC run times, DHPLC
reagents as well as increases the life span of the DHPLC
cartridge.
24. A “Normal” clean is the 100 % B clean-off step containing
25 % acetonitrile which prevents cartridge gradient fluctuation
for this application, as the gradient conditions for SBE-DHPLC
require a low percentage acetonitrile and small increases over
time. The “Fast” or “Active” clean is the 100 % D clean-off
step containing 75 % acetonitrile.
Denaturing High-Performance Liquid Chromatography for Mutation Detection… 51
25. The gradient duration is modified to 2 min that cuts down the
run time. The gradient duration may be increased to 2.5 min
or more as needed when multiplex SBE is performed.
26. The elution order of T and A may sometimes be switched at
70 °C. Devaney et al. [7] reported an elution order of C<G<A<T
at 70 °C. This elution order has also been observed when a set
of four 16-mer heterooligonucleotides differ in a single base at
the 3′ end [6]. We have also observed an elution order of
C<G<T<A. Our experimental samples are run at a 1.5 mL/min
flow rate using the high-throughput (DNASep™HT) cartridge.
It is possible that the faster run time as well as the dimensions of
the DNASep™HT Cartridge versus the DNASep™ Cartridge
(as used by Devaney et al. [7]) have a subtle effect on the elution
order of T and A. In addition it is known that retention may be
governed not only by the substituted base but also by the
immediate sequence context. For this reason the inclusion of
positive controls with known genotypes (as determined by an
independent method) is very important.
27. Factors Affecting the Quality of SURVEYOR® Nuclease Results:
(a) The quality of the genomic DNA to be amplified: High
quality DNA (from fresh or frozen cells or tissue) should
be used. The DNA should have a concentration of >5 ng/
μL as determined by absorbance at 260 nm, have an
absorbance ratio at 260/280 nm of >1.7 and be >90 %
DNA (i.e., free of most tRNA and rRNA contamination
as judged by appearance on an agarose gel). Store DNA
samples at −20 °C. If the DNA template is extracted from
paraffin-embedded tissue, several additional precautions
can be taken. The extracted DNA can be treated with uracil
DNA glycosylase to prevent amplification of DNA frag-
ments containing deaminated C residues. Often a high
percentage of the A260 adsorbing material extracted from
paraffin-embedded tissue is not amplified well during
PCR. Using a larger amount of starting DNA, e.g., ~50
versus 10 ng, will often help to produce a reasonable
amplification product.
(b) The quality of the PCR amplified DNA: PCR should pro-
duce a sufficiently high yield (>15 ng/μL) of a SINGLE
amplified species of the correct size. We strongly
recommend the use of a proofreading DNA polymerase
(such as Transgenomic Optimase® Polymerase) to reduce
the amount of base misincorporation during PCR (which
leads to the generation of “false” mutations and spurious
SURVEYOR® Nuclease cleavage fragments). Similarly,
nonspecific PCR fragments can be interpreted as muta-
tions and can mask SURVEYOR® Nuclease mismatch
52 Donna Lee Fackenthal et al.
Fig. 10 SURVEYOR Nuclease digestion products of self-annealed Control C homoduplex (HMD) and Control G/C
homoduplex/heteroduplex (HTD). The 633-bp amplicons were PCR amplified with Optimase polymerase from
2 μL of Control G and Control C. Control G/C homoduplex/heteroduplex was formed by hybridizing equal
amounts of Control G and Control C homoduplex PCR product and contains homoduplexes and C/C and G/G
mismatched heteroduplexes. DNA (300, 600, and 1,200 ng) was digested with 1 μL of SURVEYOR Nuclease W,
1 μL of SURVEYOR Enhancer W2, 1/10th volume of 0.15 M MgCl2 and 1/10th volume of Enhancer Cofactor for
60 min at 42 °C. SURVEYOR Nuclease digestion products [180 (6 μL), 400 (12 μL), and 850 ng (24 μL)] were
analyzed using the WAVE system run under non-denaturing conditions at 50 °C and equipped with a
Fluorescence Detector and a High-Sensitivity Accessory for post-column DNA intercalation with fluorescent
dye. Transgenomic 100-bp ladder DNA was run as a marker. The 217- and 416-cleavage products expected
from the Control G/C heteroduplex are clearly visible. Also visible in all three chromatograms is the full-length
633-bp homoduplex. Flat-top peaks were produced by injection of amounts of DNA that saturated the instrument
detector
References
1. Oefner PJ, Underhill PA (1995) Comparative 4. O’Donovan MC et al (1998) Blind analysis of
DNA sequencing by denaturing high- denaturing high-performance liquid chroma-
performance liquid chromatography tography as a tool for mutation detection.
(DHPLC). In: ASHG Annual meeting A2666. Genomics 52:44–49
University of Chicago Press 5. Frueh FW, Noyer-Weidner M (2003) The use
2. Navigator™ Software Manual, Version 3, © of denaturing high performance liquid chro-
(2002–2009) Transgenomic, Inc., used with matography (DHPLC) for the analysis of
permission genetic variations: impact for diagnostics and
3. Cotton RG (1997) Slowly but surely towards pharmacogenetics. Clin Chem Lab Med
better scanning for mutations. Trends Genet 41:452–461
13:43–46
54 Donna Lee Fackenthal et al.
Abstract
For advancing personalized medicine, it is important to incorporate pharmacogenomics data into routine
clinical practice. The SmartAmp method enables us to detect genetic polymorphisms or mutations in tar-
get genes within 30–40 min without DNA isolation and PCR amplification. The SmartAmp method has
been developed based on the concept that DNA amplification per se is the signal for the presence of a
specific target sequence. Differing from the widely used PCR, the SmartAmp reaction is an isothermal
DNA amplification, where the initial step of copying a target sequence from the template DNA is critically
important. For clinical applications, we have created SmartAmp primers and clinical device that detect
genetic polymorphisms of human genes involved in drug-induced toxicity or disease risk. This chapter
addresses both the basic molecular mechanism underlying the SmartAmp method and its practical applica-
tions to detect clinically important single nucleotide polymorphisms (SNPs).
Key words Personalized medicine, Point of care, Adverse reaction, Warfarin, Irinotecan, ABCB1
(MDR1), ABCC11, VKORC1, CYP2C9, UGT1A1
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_3, © Springer Science+Business Media, LLC 2013
55
56 Toshihisa Ishikawa and Yoshihide Hayashizaki
2 Materials
2.1 Enzyme The SmartAmp method utilizes Aac polymerase as a DNA poly-
merase with strand-displacement activity. Aac polymerase is from
the thermophilic bacteria Alicyclobacillus acidocaldarius. This DNA
polymerase is highly resistant to cellular contaminants and hence
works directly on blood samples, just after a simple heat treatment
(98 °C, 3 min) to degrade/denature RNA and proteins. This is a
great advantage of the SmartAmp method over the commonly used
PCR-based techniques. The enzymatic activity of Taq DNA poly-
merase is easily inhibited by impurities in the PCR reaction.
1. Aac polymerase used for the SmartAmp method is a cloned
large fragment of 610 amino acids (69.5 kDa) that carries
DNA strand displacement activity. The optimal temperature
and pH for this enzyme are 60–65 °C and 8.0–8.2,
respectively.
Clinical SNP Detection by the SmartAmp Method 57
2.2 Primers In the SmartAmp method, the entire DNA amplification process
requires five primers: turnback primer (TP), boost primer (BP),
folding primer (FP), and two outer primers (OP1 and OP2)
(Fig. 1). Primers are selected based on those algorithms consider-
ing the free energy, probability of base-pairing, product size range,
optimal melting temperature, and product size range. The design
of these primers contributes to the specificity of SmartAmp. In
particular two primers (TP and FP) are critically important for the
amplification process. The genomic sequence between the anneal-
ing sites of the TP and FP primers is the target region that will be
amplified by the SmartAmp reaction. The other primers (BP, OP1,
and OP2) are additionally employed to accelerate the process and
enhance specificity. Those primers can be synthesized and obtained
from any commercial source, such as Invitrogen.
1. In isothermal DNA amplification by the SmartAmp method,
the initial step of copying a target sequence from the genomic
DNA is a prerequisite. FP and TP hybridize the template
genomic DNA. Next, both products primed for the FP and
TP are detached from template genomic DNA by strand-
displacing DNA polymerase, whose extensions are primed by
OP1 and OP2. Single-stranded DNA products, thus displaced,
become templates in the second step for the opposing FP and
TP. These single stranded DNA products are generated by the
strand-displacement activity of the DNA polymerase, being
primed from the flanking region of OP primers adjacent to the
target sequence. The resulting DNA products are referred to
as “intermediate products” which play key roles in the subse-
quent amplification steps (Figs. 1 and 2).
2. The formation of those intermediate products is the rate-
limiting step in SmartAmp-based isothermal DNA
amplification. Intermediate 1 (IM1) has the TP sequence at
the 5′ end and the FP complementary sequence at the 3′ end.
BP FP OP2
5’ 3’
Genomic DNA
3’ 5’
OP1 TP
turn back 20 b
Fig. 1 Schematic illustration of the SmartAmp reaction using five primers: turnback primer (TP), boost primer
(BP), forward primer (FP), and two outer primers (OP1 and OP2)
58 Toshihisa Ishikawa and Yoshihide Hayashizaki
Self-primed
DNA synthesis
(Concatenation)
FP
3’ Intermediate 1 (IM1)
3’
OP1 TP turn back 3’
5’ 5’
FP OP2
5’ 3’
Genomic DNA
3’ 5’
OP1 TP turn back
3’ 5’ FP OP2
3’
Intermediate 2 (IM2) 3’
5’
TP
Self-primed
DNA synthesis
(Concatenation)
Fig. 2 Formation of intermediate products in the initial step of the SmartAmp reaction. As an initial step,
primer’s priming and DNA polymerase reaction generate two intermediates (IM1 and IM2). The inner primer set
with FP and TP initiates the reaction by hybridizing to opposite strands of a target region. Linear primer exten-
sion products from the FP and TP primers are then released from their templates in a second primer extension
reaction driven by a set of outer primers (OP1 and OP2) that hybridize downstream of the FP and TP primers.
Due to the special features of the FP and TP primers, single-stranded primer extension products from those
primers will refold at their 3′ and 5′ ends to form new priming sites that maintain self-amplification in a cau-
tious process driven by the DNA strand displacement activity of Aac DNA polymerase
Pathway A Pathway B
3’ 3’ 5’
5’
IM1 IM2
5’ 3’
3’ 5’
Fig. 3 Formation of concatenated DNA products in the SmartAmp reaction. Self-priming DNA synthesis from
each of IM1 and IM2 creates hairpin molecules via pathways A and B. These structures lead to further self-
primed DNA synthesis to create dimeric amplicons and then subsequently concatenated DNA products
16q12
5’
ABCC11
1 2 3 4 5 6 7 8
9 10 11 12 13 14 15 16 17 18
19 20 21 22 23 24 25 26
27 28 29 30
3’
SNP(538G or A)
BP FP OP2
Genomic DNA 3’ 5’ 3’ 5’
5’ 3’
-36 -18 -9 +1 +6 +18 +25 +41
-77 -64 -55 -34 Intron
3’ 5’
5’ 3’ 3’
OP1 TP 3’-NH2- 5’
Turn-back CP
5’ 0
Exon 4
Fig. 4 Schematic illustration of the SmartAmp method-based SNP detection. The SNP 538G>A resides in exon
4 of the ABCC11 gene on chromosome 16q12. The lower panel shows the strategy for the SmartAmp method-
based detection of SNP 538G>A in ABCC11 gene
2.5 Samples 1. The blood sample can be obtained with a finger prick.
2. The minimal volume of blood sample to be tested: 2 μl.
3. The blood sample is subsequently pretreated and then applied
to the reaction of SmartAmp-based SNP detection.
3 Methods
3.1 Sample Before applied to the SmartAmp reaction, each blood sample
Pretreatment should be diluted fourfold with 40 mM NaOH and then heated at
98 °C for 3 min. During this pretreatment process, proteins and
RNA are denatured and degraded under alkaline conditions.
Genomic DNA remains intact at concentrations of 5–10 ng/μl in
the sample after the pretreatment.
1. Take 2 μl of the blood sample and mix with 6 μl of 40 mM
NaOH to make a fourfold dilution.
2. Heat the mixed sample solution at 98 °C for 3 min.
3. Chill the sample solution on ice until use.
3.2 SmartAmp The standard reaction mixture (total volume of 25 μl) contains:
Reaction 2.0 μM FP, 2.0 μM TP, 1.0 μM BP, 0.25 μM OP1, and 0.25 μM
OP2, 1.4 mM dNTPs, 5 % DMSO, 20 mM Tris–HCl (pH 8.0),
10 mM KCl, 10 mM (NH4)2SO4, 8 mM MgSO4, 0.1 % (v/v)
Tween®20, 1/100,000-diluted SYBR® Green I, 20 units of Aac
DNA polymerase.
1. Add 1 μl of the pretreated sample to 25 ml of the SmartAmp
reaction mixture.
2. Incubate the SmartAmp reaction mixture at 60 °C for
30–60 min in a real-time PCR instrument.
3. Measure the fluorescence of SYBR® Green I that indicates for-
mation of concatenated DNA products.
TP /Bc FP OP2
CATTGCCAGTGTACTCGGGCCAGTAAGTGGCAGACTTGGTGAGGTTTGGGGGACTCTAGGCTTCAGAGGT-3’
CP (538G)
TP/Bc FP OP2
CATTGCCAGTGTACTCAGGCCAGTAAGTGGCAGACTTGGTGAGGTTTGGGGGACTCTAGGCTTCAGAGGT-3’
CP (538A)
Fig. 5 Partial genomic DNA sequences of the ABCC11 gene carrying WT (538G) and SNP (538A) alleles as well
as the sequences of the primers used for the SmartAmp assay. Arrows indicated the sequence difference
between the WT and SNP alleles
Clinical SNP Detection by the SmartAmp Method 63
Fluorescence (dR)
Fluorescence (dR)
4 Notes
4.1 Primer Design To design primer sets for the reaction, we have developed algo-
rithms specific to SmartAmp primer design [9]. Primer candidates
can be selected based on those algorithms considering the free
energy, probability of base-pairing, product size range, optimal
melting temperature, and product size range. The design of these
primers contributes to the specificity of SmartAmp. In particular
two primers (TP and FP) are critically important for the amplifica-
tion process. For SmartAmp primer design, it is convenient to use
the software program available on the Web site at http://www.
smapDNA.com. Initial candidate primer sets can be generated
with this program.
Primer extension-based SNP detection systems usually require
the SNP detection nucleotide to be engineered precisely at the
3′-end of a specific primer [2]. However, SmartAmp does not have
this limitation and thus there is a far greater versatility in its ability
to detect SNPs. The design options are numerous and the primer
design flexibility is unrivaled. However, the best primer set should
be selected by experimental screening among numerous possible
combinations of primer candidates. The criteria of the screening
are: no mis-amplification, high fidelity and selectivity to the target
SNP, and high sensitivity to the target site. For clinical SNP detec-
tion, the minimal detection limit of SmartAmp-based SNP detec-
tion is 5 ng genomic DNA per 1 μl of sample or even lower.
4.2 Variations There are several applications of the SmartAmp method to detec-
of SmartAmp-Based tion of clinically important genetic polymorphisms of drug metab-
SNP Detection olizing enzymes and transporters.
1. Detection of CYP2C9*2, CYP2C9*3 and vitamin K oxide
reductase VKORC1.
Warfarin is the most widely prescribed anticoagulant for the
treatment of thromboembolic disorders. Because of its narrow
therapeutic index and the large individual variability observed
between warfarin dosage and its anticoagulant effect [10–12], it
is essential to carefully adjust the dosage based on the prothrom-
bin time (PT) expressed as the international normalized ratio.
The genetic polymorphisms of CYP2C9*2 and CYP2C9*3 and
in the vitamin K oxide reductase (VKORC1) promoter
−1639G>A have a great impact on the pharmacokinetic profile
and pharmacological efficacy of warfarin. Genetic testing of a
patient for these SNPs prior to prescription of the drug is of great
importance in warfarin-based individualized pharmacotherapy
that will minimize the risks of adverse reactions and reoccurrence
of thromboembolic episodes. In 2007, the US FDA updated the
labeling for warfarin (http://www.fda.gov/bbs/topics/
NEWS/2007/NEW01684.html) such that genetic testing is
Clinical SNP Detection by the SmartAmp Method 65
4.3 Positive and As the positive control, we use three types of isolated genomic
Negative Controls for DNA with homozygous WT/WT, heterozygous WT/SNP, or
SmartAmp-Based SNP homozygous SNP/SNP in the gene of interest. The sequence of
Detection those genetic polymorphisms should be analyzed and confirmed
by conventional DNA sequence analysis. The concentration of the
genomic DNA in each control is adjusted to be about 10 ng/μl.
The negative control is distilled water. For control experiments,
1 μl of the positive or negative control is taken and added to the
SmartAmp reaction mixture (final volume 25 μl). The negative
control should not lead to any DNA amplification during the
SmartAmp reaction over time up to 60 min.
4.4 Sensitivity Check By using the positive control, the sensitivity of SmartAmp-based
SNP detection can be performed. We gain insight into the minimal
detection limit of SmartAmp-based SNP detection by diluting the
positive controls (WT/WT, WT/SNP, and SNP/SNP) in a step-
wise manner. As described above, the minimal detection limit of
SmartAmp-based SNP detection should be 5 ng genomic DNA
per 1 μl of test sample or even lower. Detection of a minimal detec-
tion limit is a prerequisite before clinical applications, since we use
heat-pretreated blood samples that contain varying numbers of
white blood cells. Precaution is needed for SmartAmp-based SNP
detection, in particular when we use blood samples from patients
with leucopenia.
CCD
Camera
PC
Lens & filter
Dispensing samples
into reaction wells SmartAmp reaction SNP
at 60°C for 30 -40 min digital pattern
&
End-point measurement
Fig. 7 Schematic illustration for end-point detection of SmartAmp-based SNP typing with a CCD camera-linked
digital processor
Acknowledgments
References
1. Mitani Y et al (2007) Rapid SNP diagnostics 2. Mitani Y et al (2009) A rapid and cost-effective
using asymmetric isothermal amplification and SNP detection method: application of
a new mismatch-suppression technology. Nat SmartAmp2 to pharmacogenomics research.
Methods 4:257–262 Pharmacogenomics 10:1187–1197
Clinical SNP Detection by the SmartAmp Method 69
3. Watanabe J et al (2007) Use of a competitive study of 75 patients. Clin Cancer Res 10:
probe in assay design for genotyping of the 5151–5159
UGT1A1*28 microsatellite polymorphism by 17. Iyer L et al (2002) UGT1A1*28 polymor-
the smart amplification process. Biotechniques phism as a determinant of irinotecan disposi-
43:479–484 tion and adverse reactions. Pharmacogenomics
4. Toyoda Y et al (2009) Earwax, osmidrosis, J 2:43–47
and breast cancer: why does one SNP 18. Innocenti F et al (2004) Genetic variants in
(538G>A) in the human ABC transporter the UDP-glucuronosyltransferase 1A1 gene
ABCC11 gene determine earwax type? FASEB predict the risk of severe neutropenia of irino-
J 23: 2001–2013 tecan. J Clin Oncol 22:1382–1388
5. Yoshiura K et al (2006) A SNP in the ABCC11 19. Marcuello E et al (2004) UGT1A1 gene varia-
gene is the determinant of human earwax type. tions and irinotecan treatment in patients with
Nat Genet 38:324–330 metastatic colorectal cancer. Br J Cancer
6. Miura K et al (2007) A strong association 91:678–682
between human earwax-type and apocrine 20. Kitagawa C et al (2005) Genetic polymorphism
colostrum secretion from the mammary gland. in the phenobarbital-responsive enhancer mod-
Hum Genet 121:631–633 ule of the UDPglucuronosyltransferase 1A1
7. Ota I et al (2010) Association between breast gene and irinotecan toxicity. Pharmacogenet
cancer risk and the wild-type allele of human Genomics 15:35–41
ABC transporter ABCC11. Anticancer Res 21. Hasegawa Y et al (2006) Pharmacogenetic
30:5189–5194 approach for cancer treatment-tailored medicine
8. Toyoda Y, Ishikawa T (2010) in practice. Ann N Y Acad Sci 1086:223–232
Pharmacogenomics of human ABC trans- 22. Innocenti F et al (2002) Haplotype structure
porter ABCC11 (MRP8): potential risk of of the UDP-glucuronosyltransferase 1A1 pro-
breast cancer and chemotherapy failure. moter in different ethnic groups.
Anticancer Agents Med Chem 10:617–623 Pharmacogenetics 12:725–733
9. Kimura Y et al (2011) Optimization of turn- 23. Watanabe J et al (2007) Complete suppression
back primers in isothermal amplification. of background amplification using competi-
Nucleic Acids Res 39:e59 tive probe in a SMart-Amplification process assay
10. Kaminsky LS, Zhang ZY (1997) Human P450 for microsatellite polymorphism genotyping of
metabolism of warfarin. Pharmacol Ther UGT1A1*28. Biotechniques 43:479–484
73:67–74 24. Leschzinger GD et al (2007) ABCB1 geno-
11. Cannegieter SC et al (1995) Optimal oral anti- type and PGP expression, function and thera-
coagulant therapy in patients with mechanical peutic drug response: a critical review and
heart valves. N Engl J Med 333:11–17 recommendations for future research.
12. Fihn SD et al (1993) Risk factors for complica- Pharmacogenomics J 7:154–179
tions of chronic anticoagulation. Ann Intern 25. Sakurai A et al (2007) Quantitative SAR
Med 118:511–520 analysis and molecular dynamic simulation to
13. Aomori T et al (2009) Rapid SNP detection of functionally validate nonsynonymous poly-
the cytochrome P-450 (CYP) 2C9 and the vita- morphisms of human ABC transporter
min K oxide reductase (VKORC1) gene for the ABCB1. Biochemistry 46:7678–7693
warfarin dose adjustment by Smart-Amplification 26. Hüebner C et al (2007) Triallelic single
process version 2. Clin Chem 55:804–812 nucleotide polymorphisms and genotyping error
14. Ando Y et al (2000) Polymorphisms of in genetic epidemiology studies: MDR1 (ABCB1)
UDPglucuronosyltransferase gene and irinote- G2677/T/A as an example. Cancer Epidemiol
can adverse reactions: a pharmacogenetic anal- Biomarkers Prev 16: 1185–1192
ysis. Cancer Res 60:6921–6929 27. Ishikawa T et al (2010) Emerging new tech-
15. Ando Y, Hasegawa Y (2005) Clinical pharma- nologies in pharmacogenomics: rapid SNP
cogenetics of irinotecan (CPT-11). Drug detection, molecular dynamic simulation,
Metab Rev 37:565–574 and QSAR analysis methods to validate clini-
16. Rouits E et al (2004) Relevance of different cally important genetic variants of human
UGT1A1 polymorphisms in irinotecan- ABC transporter ABCB1 (P-gp/MDR1).
induced toxicity: a molecular and clinical Pharmacol Ther 126:69–81
Chapter 4
Abstract
Major strengths of mass spectrometry analysis include the accuracy of the detection principle, automatic
data storage as well as simplicity and flexibility of assay design making it a premier choice for targeted
genotyping of sequence variations. We explain the assay principle in detail and give step-by-step laboratory
instructions. Finally, references point toward further use of mass spectrometry analysis for molecular hap-
lotyping, re-sequencing, and quantitative analysis for copy number variations and gene expression studies
are given.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_4, © Springer Science+Business Media, LLC 2013
71
72 Dirk van den Boom et al.
1.1 High-Throughput Despite the plethora of available technologies, not all genotyping
Genotyping methods are suitable for HT genotyping. A major requirement for
HT genotyping is automation—from sample preparation to auto-
mated readout of the genotype. Another requirement is the avail-
ability of sufficient DNA template—the reason why nearly all
methods are based on PCR amplification. Timing, throughput and
accuracy are also critical. Missing or incorrect genotypes, even in a
minor number of samples, may double the time for genotyping.
Either individual samples need to be re-arrayed in a second step
from original plates or repeated from the same source. Average
setup, implementation and process time for an assay are therefore
important factors to consider. Finally, accuracy is extremely impor-
tant, as running all assays in duplicate or triplicate would not be
cost efficient.
Current methods for targeted genotyping combine at least one
of four different principles of allelic discrimination (hybridization,
primer extension, ligation, or restriction) with one of four different
detection techniques (chemi-luminescence/fluorescence, fluores-
cence polarization, resonance energy transfer, and mass spectrom-
etry). Assay formats still range from gel electrophoresis, plates,
particles, fiber arrays and microchip arrays to semi- and homoge-
nous assays that do not require any further sample separation or
purification.
The major strengths of mass spectrometric analysis are the
inherent accuracy of this detection principle, the automatic data
accumulation and interpretation, the high-throughput capacity
and the ability to analyze not only SNPs but also more complex
MALDI-TOF Mass Spectrometry 73
a b
A C GT A C GT
T T
I EP AG I EP ddA dGddC
m/z m/z
Fig. 1 Schematic representation of primer extension reactions and analysis by mass spectrometry for genotyp-
ing of sequence variants/single nucleotide polymorphisms. (a) depicts an example of an extension reaction
where only terminating nucleotides are used in the extension reaction. Alleles are identified by the correspond-
ing molecular mass and the difference in the molecular mass of extension products is driven by the mass dif-
ference between the terminator nucleotides. (b) depicts an example of a multibase extension reaction. Here a
mixture of one elongator with three terminator nucleotides is modelled. The extension products differ in molec-
ular mass by a nucleotide. While single base extension design usually allow for higher multiplexing, mixtures of
elongators/terminators are occasionally the more appropriate choice for genotyping insertion/deletions
rs72549353
I
Dirk van den Boom et al.
...P.rs41279188
...P.rs28371706
...P.rs35350960
...P.rs11572080
...P.rs72549353
...P.rs11568626
...P.rs41303343
...P.rs55785340
...P.rs56296335
...P.rs28399447
...P.rs72552713
...P.rs72558187
...P.rs28399433
...P.rs28399444
...P.rs10509681
...P.rs56318881
...P.rs72558186
D
D
D
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
T
A
A
A
A
A
A
A
A
A
A
A
A
A
A
A
A
A
A
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
G
...EP.rs5030865
...EP.rs7662029
...EP.rs1135836
...EP.rs1801266
...EP.rs5030862
...EP.rs4646278
...EP.rs8177508
...EP.rs4149056
...EP.rs1801267
...EP.rs3740066
...EP.rs2032582
...EP.rs8177517
...EP.rs1138272
...EP.rs9332239
...EP.rs2282143
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
C
UEP.rs628031
UEP.rs316019
UEP.rs776746
UEP.rs1695
40
30
20
Intensity
10
0
4500 5000 5500 6000 6500 7000 7500 8000 8500 9000
Mass
Fig. 2 Depicted is a representative mass spectrum of a 36-plexed SNP genotyping reaction covering multiple sequence variants influencing drug metabolism and
transport (for details please see Everts R. et al., Application Note, Development and Validation of the iPLEX ADME PGx Panel on the MassARRAY System). The inten-
sity of mass signals (y-axis) is plotted against the molecular mass of extension products (x-axis) of all assays in the multiplex. Assays are color-coded consisting
of the unextended primer (EP) referencing the rs number of the sequence variant, e.g., EP.rs2282143, followed by the alleles, e.g., C and T in the same color
MALDI-TOF Mass Spectrometry 77
2 Materials
3 Methods
3.1 Assay Design For designing highly multiplexed genotyping assays, specific primer
Considerations design software is available that designs PCR and EXTEND prim-
ers for each SNP (or insertion/deletion polymorphism) to be
investigated. It uses a multiplexing algorithm developed to take full
advantage of the available mass range while avoiding overlapping
mass signals in the analyzed mass range. The program is also
78 Dirk van den Boom et al.
3.2 PCR To prepare and process the PCR, perform the following steps:
1. Prepare a PCR cocktail as described in Table 1 (volumes are
provided on a per-well basis)
2. Cycle the PCR in a standard thermal cycler according to the
conditions described in Table 2.
3.3 SAP 1. 0.5 U of shrimp alkaline phosphatase (SAP) provided in a 2 μL
volume of enzyme/buffer are then added to each PCR to
dephosphorylate unincorporated dNTPs from the amplifica-
tion reaction. The reaction is incubated at 37 °C for 40 min
followed by inactivation of the enzyme at 85 °C for 5 min.
Final concentration in
Reagent Volume (μL) 7 mL reaction volume
Nanopure Water (HPLC grade) 1.53 N/A
SAP Buffer 0.17 0.025x
SAP Enzyme 0.3 0.5 U
Total volume 2
MALDI-TOF Mass Spectrometry 79
Table 1
PCR cocktail
Table 2
PCR conditions
Cycles Condition
1 95 °C for 2 min
45 95 °C for 30 s
56 °C for 30 s
72 °C for 1 min
1 72 °C for 5 min
1 4 °C hold
3.4 Adjusting The mass signals in the mass spectrum for a multiplexed reaction
Primer Amount may not have comparable heights. Variations in peak height may
stem from (1) inconsistent oligonucleotide quality, (2) inconsistent
oligonucleotide concentration, or (3) different desorption/ioniza-
tion behavior in MALDI. For best multiplexing results, the con-
centrations of MassEXTEND primers should be adjusted to even
out peak heights (intensities) in the mass spectrum. This adjust-
ment must be done prior to preparing the iPLEX/MassEXTEND
reaction cocktail and processing the iPLEX reaction. The following
steps need to be performed to adjust primer mixes:
1. For each multiplex, prepare a mixture of the required primers.
The final concentration of each primer in the primer mix
should be 9 μM. Consider how much primer mix you will
80 Dirk van den Boom et al.
3.5 iPLEX Reaction, Once the MassEXTEND primer mixes have been adjusted, the
Desalting and iPLEX extend reaction cocktail is prepared (Table 3), added to the
Dispensing SAP-treated PCR product, and thermocycled.
Cycle the reaction as indicated in Table 4.
Dilute with 16 μL Nanopure water and add 6 mg CLEAN resin
to the Extend reaction products for conditioning (see Note 8).
Then incubate for 15 min at room temperature and keep the resin
particles in suspension during incubation. Spin the reaction vessel
at 3,200 × g (2,000 rpm for standard rotor centrifuge) prior to the
next step. Using a nanodispenser, 12–15 nL of the reaction product
is then transferred onto a 384-well SpectroCHIP array.
Table 3
Extension reaction cocktail (per reaction well)
Final concentration
Reagent Volume (μL) in 9 μL reaction volume
Nanopure water (HPLC grade) 0.619 N/A
iPLEX Gold buffer 0.200 0.222×
iPLEX termination mix 0.200 1×
Adjusted primer mix (~9 μM each) a
0.940 1.25×/1.875 mM MgCl2
iPLEX enzyme (32 U/μL) 0.041 1.25 U/rxn
Total volume 2.000
a
Note that the primers in an adjusted mix may not be at 9 μM each. Each starts out at 9 μM; however, the addition of
extra amounts of some primers to adjust the mix will change the concentrations
Table 4
Extension conditions
3.7 Other MALDI The focus of this chapter has been genotyping of SNPs using primer
Applications extension methods and MALDI-TOF MS. Within recent years, the
portfolio of applications using MALDI-TOF MS as a detection
platform has expanded significantly. A majority of these new appli-
cations not only rely on the accuracy provided by mass spectrome-
try for qualitative analysis of nucleic acids, but they also have
established measures for quantitative analysis of nucleic acids.
Recent publications describe the use MALDI-TOF MS for relative
quantitation of genetic information in DNA pools and sample mix-
tures [12–15]; re-sequencing methods, which allow the rapid dis-
covery of SNPs, the screening for mutations or signature sequence
based identification of organisms such as pathogens [16–18]; and
also relative and absolute quantitation in gene expression and copy
number variations [19]. A further interesting application is M1-PCR
for haplotyping [20]. Here, multiplex PCR performed on single
DNA molecules generated by dilution is combined with the speci-
ficity of mass spectrometry read-outs to generate up to 25 kB hap-
lotypes. Recent reviews summarize these developments [9].
4 Notes
Table 5
SNPs and suitable termination mixes in addition to SEQUENOM standard
iPLEX termination mix
Acknowledgments
References
12. Bansal A, van den Boom D, Kammerer S, Honisch C, Rodi CP, Bocker S et al (2004)
Honisch C, Adam G, Cantor CR, Kleyn P, High-throughput MALDI-TOF discovery of
Braun A (2002) Association testing by DNA genomic sequence polymorphisms. Genome
pooling: an effective initial screen. Proc Natl Res 14:126–133
Acad Sci USA 99:16871–16874 17. Ehrich M, Böcker S, van den Boom D (2005)
13. Werner M, Sych M, Herbon N, Illig T, König Multiplexed discovery of sequence polymor-
I, Wjst M (2002) Large-scale determination of phisms using base-specific cleavage and
SNP allele frequencies in DNA pools using MALDI-TOF MS. Nucleic Acids Res
MALDI-TOF mass spectrometry. Hum Mutat 33(4):e38
20:57–64 18. Honisch C, Chen Y, Mortimer C, Arnold C,
14. Mohlke KL, Erdos MR, Scott LJ, Fingerlin Schmidt O, van den Boom D, Cantor CR,
TE, Jackson AU, Silander K, Hollstein P, Shah HN, Gharbia SE (2007) Automated
Boehnke M, Collins FS (2002) High- comparative sequence analysis by base-specific
throughput screening for evidence of associa- cleavage and mass spectrometry for nucleic
tion by using mass spectrometry genotyping acid-based microbial typing. Proc Natl Acad
on DNA pools. Proc Natl Acad Sci USA Sci USA 104(25):10649–10654
99:16928–16933 19. Ding C, Cantor CR (2003) A high-throughput
15. Herbon N, Werner M, Braig C, Gohlke H, gene expression analysis technique using com-
Dütsch G, Illig T, Altmüller J, Hampe J, petitive PCR and matrix-assisted laser desorp-
Lantermann A, Schreiber S et al (2003) High- tion ionization time-of-flight MS. Proc Natl
resolution SNP scan of chromosome 6p21 in Acad Sci USA 100:3059–3064
pooled samples from patients with complex 20. Ding C, Cantor CR (2003) Direct molecular
diseases. Genomics 81:510–518 haplotyping of long-range genomic DNA with
16. Stanssens P, Zabeau M, Meersseman G, Remes M1-PCR. Proc Natl Acad Sci USA 100:
G, Gansemans Y, Storm N, Hartmer R, 7449–7453
Chapter 5
Abstract
Polymorphisms associated with genes that code for various drug-metabolizing enzymes (DMEs) and
associated transport proteins can influence the rate of drug metabolism within individuals, thus potentially
affecting drug efficacy and the occurrence of side effects. There are 2,700 unique TaqMan® Drug
Metabolism Genotyping Assays (Life Technologies) for detecting single nucleotide polymorphisms
(SNPs), insertions and deletions (indels), and multinucleotide polymorphisms (MNPs) in both coding and
regulatory regions. These research assays are useful tools for better understanding genetic variation in drug
metabolism. Here we describe the procedure for measuring genetic variation in human DNA using
TaqMan® Drug Metabolism Genotyping Assays. These assays are for research use only and are not intended
for any animal or human therapeutic or diagnostic use.
Key words Single nucleotide polymorphism, SNP, Drug-metabolizing enzymes, DME, Cytochrome
P450, CYP superfamily, TaqMan® Drug Metabolism Genotyping Assays, TaqMan® Genotyper
Software
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_5, © Springer Science+Business Media, LLC 2013
87
88 Toinette Hartshorne
2 Materials
2.1 Assay Each TaqMan® DME assay consists of a single tube containing:
Components ● Two primers for specific amplification of the locus containing
the polymorphism of interest.
TaqMan® Drug Metabolism Genotyping Assays 89
Fig. 1 TaqMan® SNP Genotyping Assay. (1) The four TaqMan® Genotyping Assay components and the target
DNA template with the SNP (in brackets). (2) The denatured DNA target and annealing of the assay compo-
nents. (3) Signal generation leading to specific allele detection
2.3 Selecting a This PCR protocol has been tested using GeneAmp® PCR System
Thermal Cycler or 9700 and the Applied Biosystems® 7900HT Real-Time PCR
Real-Time PCR System System thermal cyclers for PCR amplification, as well as other
Applied Biosystems® instruments, including those listed below (see
Notes 2 and 3).
Instruments:
Thermal Cyclers: GeneAmp® PCR System 9700, Veriti® 384-Well
Thermal Cycler, Veriti® 96-Well Fast Thermal Cycler.
(TaqMan® Drug Metabolism Assays can be performed on Fast
thermal cyclers using standard reagents and standard cycling
protocols.)
Real-Time PCR Systems (These systems allow real-time analysis of
PCR, which is helpful for troubleshooting. QuantStudio™
12K Flex system, ViiA™ 7 system, 7900HT Fast system, 7500
system, 7500 Fast system, StepOnePlus™ system, StepOne™
system (TaqMan® Drug Metabolism Assays can be performed
on Fast real-time PCR systems using standard reagents and
standard cycling protocols).
3 Methods
3.1 PCR During the first step of a TaqMan® Drug Metabolism Genotyping
Amplification Assay, AmpliTaq Gold® DNA Polymerase from the TaqMan®
Genotyping Master Mix (see Note 4) amplifies the target DNA
3.1.1 Overview
using sequence-specific primers. TaqMan® MGB probes from the
Drug Metabolism Genotyping Assay provide a fluorescence read-
out of the amplification of each allele.
3.1.2 General Process PCR amplification requires that you prepare an optical reaction
plate containing the following for each assay:
● No-template controls (NTCs) (at least two are strongly rec-
ommended; see Note 5)
TaqMan® Drug Metabolism Genotyping Assays 91
3.1.3 Reagent ● Keep all TaqMan® Drug Metabolism Genotyping Assays in the
Preparation Guidelines freezer, protected from light, until ready for use. Excessive
exposure to light may affect the fluorescent probes.
92 Toinette Hartshorne
3.1.5 Prepare the The reaction mix contains TaqMan® Drug Metabolism Genotyping
Reaction Mix Assay Mix, TaqMan® Genotyping Master Mix, and DNase-free
water. The recommended final reaction volume, per well, is 5 μL
for a 384-well plate and 25 μL for a 96-well plate. To prepare the
reaction mix:
1. Calculate the number of reactions to be performed for each
assay (see Note 8).
TaqMan® Drug Metabolism Genotyping Assays 93
Dried-down
Wet DNA method DNA method
Volume/well (μL)
3.1.6 Prepare the DNA For each assay and on each reaction plate, run controls to ensure
Reaction Plate optimal analysis and troubleshooting capabilities of TaqMan®
Drug Metabolism Genotyping Assays:
● Two no-template controls (NTCs, DNase-free water) per assay
(strongly recommended)
● Known gDNA controls (optional)
1. Into each well of the DNA reaction plate, pipet the reaction
mix as indicated below (see Note 10). For preparing a 384-
well reaction plate, the volume of reaction mix/well should
be 2.75 μL (wet method) or 5 μL (dried-down DNA
method). For preparing a 96-well reaction plate, the vol-
ume of reaction mix/well should be 13.75 μL (wet method)
or 25 μL (dried-down DNA method).
94 Toinette Hartshorne
3.1.7 Perform PCR The TaqMan® Drug Metabolism Genotyping Assay protocol uses
a 90-s PCR extension time and 50 PCR cycles. These conditions
are chosen for optimal performance because the average amplicon
size of TaqMan® Drug Metabolism Genotyping Assays is longer
than the average amplicon size of most TaqMan® SNP Genotyping
Assays.
To perform PCR:
1. Specify the thermal cycling conditions (see Note 11).
AmpliTaq Gold®
enzyme activation PCR (50 cycles)
3.2 Allelic After PCR amplification, you perform an endpoint plate read using
Discrimination Plate Applied Biosystems® Real-Time PCR System Software. The instru-
Read and Analysis ment software uses the fluorescence measurements made during
the plate read to plot fluorescence (Rn) values based on the signals
from each well. The plotted fluorescence signals indicate which
alleles and genotypes are in each sample.
Refer to the allelic discrimination or genotyping section of the
appropriate instrument user guide for instructions on how to use
the system software to perform the plate read and analysis.
Analyzing data for SNP genotyping requires that you:
1. Create and set up an allelic discrimination plate read document
2. Perform an allelic discrimination plate read on a real-time
PCR instrument system
TaqMan® Drug Metabolism Genotyping Assays 95
4 Notes
Abstract
Despite the influx of high throughput sequencing techniques, there is still a niche for low-medium
throughput genotyping technologies for small-scale screening and validation purposes. Pyrosequencing is
a genotyping assay based on sequencing-by-synthesis. Short runs of sequence around each polymorphism
are generated, allowing for internal controls for each sample. Pyrosequencing can also be utilized to iden-
tify tri-allelic, indel, and short repeat polymorphisms, as well as determining allele percentages for methyla-
tion or pooled sample assessment. This range of applications makes it well-suited to the research laboratory
as a one-stop system.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_6, © Springer Science+Business Media, LLC 2013
97
98 Cristi R. King and Sharon Marsh
2 Materials
2.1 DNA Template DNA from any source can be used in Pyrosequencing assays
(see Note 1). Commonly used kits for manual or machine DNA
extraction, including Gentra, Qiagen, and Oragene do not inhibit
the assay.
2.2 PCR 1. Primer Design Software (custom software is available for a fee
from Qiagen, and is usually included in the purchase of the
Pyrosequencing machine).
2. 1–5 ng DNA template (see Note 2).
3. PCR Mastermix, for example: 30 mM Tris–HCl, 100 mM
potassium chloride, pH 8.05, 400 μM dNTP, and 5 mM mag-
nesium chloride (see Note 3).
4. Hot start Taq Polymerase (see Note 4).
5. DNase- and RNase-free 18.2 mΩ water.
6. DNA Oligonucleotides (primers), one biotinylated.
7. Unskirted 96-well PCR trays.
8. Sealing Film or Silicon Mat for covering PCR plates in a
thermocycler.
9. Thermocycler with 96-well capacity, gradient block, and
heated lid.
3 Methods
Fig. 1 The Pyrosequencing reaction. A modified ATP is used for the nucleotide dispensations to prevent its
direct use by luciferase in the reaction. Modified and published with permission from Biotage AB
3.1 PCR 1. Any primer design software, freely available or custom pur-
Primer Design chased may be used to design PCR primers for Pyrosequencing.
The polymorphism may be in any position of the PCR ampli-
con from one base in from the 3′ end of the PCR primer
sequence to centered between the primers. SNPs, indels,
repeats, etc. do not require specific PCR primer design
modifications.
2. Primers should be between 15 and 30 bases long, with an
optimum size of 20 bases, ideally with a GC:AT ratio around
50 % (although not essential, as you are at the mercy of the
location of the polymorphism).
3. Most amplicon sizes are usable for high quality DNA, how-
ever, amplicon sizes of 100–200 bp are suitable for most tem-
plate sources, including fragmented DNA.
4. Care should be taken to avoid any possible template loops
from primers or the single-stranded amplicon doubling back
on themselves, as these can lead to background problems dur-
ing the Pyrosequencing assay (see Note 6).
5. Optimum primer melting temperature (Tm) is 60 °C, how-
ever, again, the position of the polymorphism determines the
ability to design optimum primers and 50–69 °C will work.
The individual primers should ideally have Tms within 2 °C of
each other to allow effective optimization of the PCR.
6. Primer specificity should be checked by screening the primers
across available human genome sequence using the NCBI Blast
program (http://www.ncbi.nlm.nih.gov/blast/). Extra care
should be taken when designing assays for gene family mem-
bers, e.g., cytochromes, or genes with known pseudogenes,
e.g., DHFR, as cross-hybridization of primers can lead to high
background, reduced signal and/or false positive results.
7. One primer needs to be biotinylated at the 5′ end. Which
primer to be biotinylated is dependent on the Pyrosequencing
primer orientation.
102 Cristi R. King and Sharon Marsh
3.2 Pyrosequencing 1. The entire PCR amplicon sequence, including forward and
Primer Design reverse primer sequences is required to generate the optimum
Pyrosequencing primer. The custom software from Qiagen
(Germany) should be used for optimum results.
2. Unless multiplexing is required (see Note 7), the software
should be defaulted to find both forward and reverse primers
to improve the likelihood of obtaining the optimum primer
sequence. The software will list all possible forward and reverse
primers by score. Often “medium” scores yield usable primers,
as certain scoring parameters are more critical than others (see
Note 8). Template loops likely to cause background will not
affect the overall score can cause problems and should be
avoided (see Note 9).
3. The orientation of the Pyrosequencing primer will determine
the PCR primer to be biotinylated. Forward Pyrosequencing
primers require a biotinylated reverse PCR primer, reverse
Pyrosequencing primers require a biotinylated forward PCR
primer.
72 °C 30 s
Then:
72 °C 5 min
4 °C storage.
2. The gradient PCR should be visualized using a 1 or 2 %
agarose gel. The optimal temperature should give the brightest
single band at the appropriate amplicon size. Care should be
taken to avoid temperatures where a smeared or multiband
product can be seen as these can increase pyrosequencing
background or reduce specificity if co-amplifying a different
DNA region. Where several temperatures of equal band
intensity are available, the highest temperature should be
picked to ensure specificity.
3.4 PCR for 1. Care should be taken to avoid contamination. Ideally a sepa-
Pyrosequencing rate room that does not come in contact with post-PCR ampli-
fied DNA or post-PCR pipettes, reagents and consumables
should be used. The bench area should be swabbed with 70 %
ethanol or 5 % bleach solution before each PCR set-up and
barrier tips should be used for all pipetting steps.
2. 1 μl (1–5 ng) DNA (depending on source, see Note 2) should
be dispensed into an unskirted 96-well PCR tray (see Note
12). At least 1 well should not contain DNA to act as a nega-
tive control (see Note 13).
3. A 20 μl PCR reaction is ideal for Pyrosequencing, however, if
the PCR product is especially strong or wide-peak pyrograms
occur, a 10 μl reaction will work well. For a 20 μl reaction
based on ABI Amplitaq Gold PCR mastermix (Applied
Biosystems, Foster City, CA):
10 μl ABI Amplitaq Gold PCR mix
Forward PCR primer (10 pM final concentration)
Reverse PCR primer (10 pM final concentration)
Up to 19 μl with 18.2 mΩ water
1 μl template
4. The PCR plate should be well sealed using a silicon mat or
adhesive film. The following PCR program should be run (see
Note 14):
93 °C 10 min (or relevant temperature/time for taq activation)
55 cycles of:
95 °C 30 s
X °C 30 s (based on gradient derived annealing temp)
72 °C 30 s
104 Cristi R. King and Sharon Marsh
Then:
72 °C 5 min
4 °C storage.
5. It is possible to directly use the PCR product for
Pyrosequencing, however, it is advisable to check the product
and the negative control on a 1–2 % agarose gel to ensure the
reaction has been performed successfully and no contamina-
tion is present. Contamination is identifiable at the
Pyrosequencing stage, however, it is cheaper and faster to run
an agarose gel than process and run a contaminated/failed
Pyrosequencing plate. 96-well plates should be briefly centri-
fuged and the lid removed with care to prevent sample aerosol
and inadvertent cross-contamination. Typically, 5 μl of the
negative control and 5 μl of five to six wells should give an idea
of the success of the PCR. The Pyrosequencing will not be
affected by the reduction in volume in these wells. Due to the
unusually large number of PCR cycles, some smearing may be
visible on a gel, even if the optimum annealing temperature
has been used. At this stage the smearing typically does not
affect the Pyrosequencing reaction if the PCR primers are spe-
cific and the negative control does not contain product.
6. The PCR product can be stored at 4 °C until needed. PCR
trays should be briefly centrifuged prior as condensation may
occur on the lid, which is a possible source of post PCR
contamination.
3.5 PCR Processing This protocol assumes the use of a streptavidin/sepharose bead
for Pyrosequencing set-up for Pyrosequencing on a 96-well PyroMark system. The
magnetic bead processing method for the PSQ96 or PSQ96MA is
described elsewhere [8].
1. A 96-well Pyrosequencing plate containing Pyrosequencing
primer mix should be set-up as described in Subheading 2.4
(see Note 15):
2. The small volume readily evaporates, if the set-up time is lon-
ger than 10–15 min cover the plate with adhesive film. Primer
plates can be aliquoted in advance and stored at 4 °C. It is
advisable to allow them to reach ambient temperature and
briefly centrifuge them before use after storage.
3. Add 70 μl sepharose bead mix as described in Subheading 2.4
to each well of the PCR product. Replace silicon lid/adhesive
film securely.
4. Shake the 96-well plate for 5 min at room temperature. If
using the eppendorf thermomixer, 1,400 RPM is the opti-
mum speed. This allows the streptavidin coated sepharose
beads to anneal to the biotin tag on the PCR primer. Use the
Pyrosequencing of Clinically Relevant Polymorphisms 105
3.6 Pyrosequencing 1. Open the Pyrosequencing software. A user name and pass-
word is typically required. This is usually set up with instru-
3.6.1 Entering Assay
ment installation. Individual or group-wide passwords can
Details
be used.
106 Cristi R. King and Sharon Marsh
2. If the assay is not already entered into the software, on the left
of the screen click “simplex entry” (see Note 17). In the menu
tree to the right of the simplex entry icon scroll to the top,
right click over “simplex entry” and select “new entry”.
3. The required fields are a unique name for the assay (usually
gene/SNP name) and a sequence to analyze. Usually five to
six bases after the SNP position provides enough information
for the assay. SNPs should be denoted as for example T/C
(tri-allelic or tetra-allelic SNPs can also be entered, e.g.,
G/A/T or G/A/T/C) and indels as for example [GATC].
Short repeats should be entered as a series of indels, e.g., [TA]
[TA][TA]. Clicking “dispensation order” will automatically
generate the least amount of nucleotide dispensations required
for optimum genotype information. The dispensation order
can be manually edited by typing in the dispensation order
field, which is useful for troubleshooting problem assays.
4. Select “show histograms” and the predicted pyrogram pattern
will be displayed on the right. The default screens show both
homozygous patterns and the heterozygous pattern. It is pos-
sible to scroll through histograms on the lower panel, useful if
multiplex of multiple indels are to be analyzed, etc. Selecting
individual or all predicted histograms on the box below the
dispensation order and clicking “export” opens the histograms
in a browser window where they can be printed or saved.
5. Click “save”. At this stage the parameters can no longer be
altered, a duplicate setup with a unique name will need to be
created for any alterations to the assay.
3.6.2 Entering a 1. Select the “SNP run” icon on the far left of the screen.
SNP Run 2. On the menu tree right-click over “SNP run” and select “new
SNP run” (see Note 18).
3. The essential parameters on the setup tab are a unique run
name (e.g., gene/SNP/sample set/date) and the active well
map. The default plate map is for a full 96-well plate. Individual
wells can be selected (hold down control for nonadjacent wells),
clicking the “activate wells” button will grey out unused wells.
In addition, instrument parameters must be selected from the
drop down menu. Usually “instrument parameters” is a default
file, however, care should be taken to ensure the appropriate
parameters are selected for nucleotide or capillary dispensing
tips, as they are not interchangeable. Parameter setup instruc-
tions are found with the dispensing tip packaging.
4. The essential parameters on the setup tab are to select the SNP
assay by clicking on the drop-down menu under “simplex”
and selecting the assay name entered in Subheading 3.6.1, and
to fill the plate map by clicking and dragging over the active
(white) wells (see Note 19).
Pyrosequencing of Clinically Relevant Polymorphisms 107
5. Once the run has been set up, click “save”. This can be edited
post-save, and changes can be re-saved.
6. If multiple plates of the same assay are to be run, on the menu
tree right click over the SNP run you have just entered and
select “duplicate SNP run”. The only parameter necessary is a
unique run name.
3.6.3 Individual Plate 1. On the SNP run setup page described in Subheading 3.6.2,
Run for PyroMark Systems click the “view” tab and select “run”. This will list the appro-
priate volumes of nucleotides, enzyme, and reagent needed
for the individual run.
2. Set up the cartridge holder as shown in Fig. 2. It is essential
that all nucleotide/capillary and reagent tips are clean before
use. To check for blockages in the nucleotide and reagent tips,
fill with 18.2 mΩ water and apply pressure over the top of the
tip. Water should squirt from the bottom of the tip. If this
does not occur, try filling/emptying the tip several times with
water and retry forcing liquid through. If the tip remains
blocked, discard. For nucleotide dispensing tips, do not force
water through them. The hydrophobic disks may dislodge and
prevent the tip from functioning. Rather, ensure the tip has
been rinsed several times in water and has been stored in a
clean, lint-free environment (see Note 20).
3. Nucleotides, enzyme, and substrate are sold as a reagent kit.
Each vial is clearly labeled. Nucleotides come as a solution,
enzyme and reagent are lyophilized and should be resuspended
with 18.2 mΩ water before use; the volumes vary per kit and are
clearly marked on the labels. The enzyme and substrate both
dissolve rapidly and no mixing or shaking is required. Indeed,
this should be avoided as air bubbles in the liquid could cause
tip blockages or inconsistent dispensation. Unused resuspended
enzyme and substrate can be stored at −20 °C for future use.
3.6.4 Batch Runs Using 1. SNP runs should be set up as described in Subheading 3.6.2,
the Automatic Plate Loader saved and closed.
2. Select the “Batch run” icon on the far left of the Pyrosequencing
software, on the menu tree right click over “batch runs” and
select “new batch run”. One to ten plates can be run in each
batch. A unique name for each batch must be provided, and
the instrument parameters must be selected for each batch. If
barcoded plates are not used, uncheck the “barcode” field.
3. On the far left of the software click on the “SNP runs” icon.
From the menu tree, click and drag your SNP runs into the
one to ten slots on the batch window.
4. On the top menu bar select “batch” and “setup information”.
This will open a browser window (may take a few seconds)
with the total amount of nucleotides (which should be doubled
for the capillary dispensing tips), enzyme and reagents needed
for the entire batch.
5. The cartridge should be set up as described in Subheading 3.6.3.
The dispensing tips should be cleaned between every batch
and a test plate should be run prior to every batch.
6. Remove the adhesive film from the Pyrosequencing plates, stack
them (check that the plates can be lifted free without sticking to
the lower plates, occasional warping may occur, causing plates
to stick together, which jams the robotic arm). Place plates in
the robot stacker unit. The correct plate orientation is shown on
the top of the stacker unit. Ensure the plates lie flat on the base
of the stacker unit and are between the grooves. Plate 1 on the
Batch set up should be on the top, plate 10 (or the last plate in
the batch setup) should be on the bottom.
7. Ensure the stacker unit is firmly pushed into place. The nucle-
otides will not dispense if the unit is only partially home.
8. Click the “play” icon. Plates will automatically load and be
discarded throughout the batch.
9. Plates will automatically be analyzed by the software when run
in batch mode. They can be accessed from the batch setup
window or from the individual SNP run files.
3.6.5 Analysis 1. Once the Pyrosequencing run has been analyzed by the soft-
of Pyrosequencing Results ware, the 96-well plate map will be color-coded according to
the result. Blue indicates a well where the pyrogram matches
one of the predicted histograms and a genotype can be accu-
rately called. Orange indicates a possible match with a pre-
dicted histogram, however, human intervention is required to
validate the call. Red indicates a failed well, where no match
with a predicted histogram can be found. Figure 3 shows
pyrograms and associated predicted histograms for the tri-
allelic ABCB1 2677 G>A/T polymorphism.
110 Cristi R. King and Sharon Marsh
Fig. 3 Predicted histograms and actual pyrograms for ABCB1 2677 G>A/T genotypes
Pyrosequencing of Clinically Relevant Polymorphisms 111
4 Notes
References
1. Evans WE, McLeod HM (2003) 6. Saeki M, Saito Y, Jinno H, Tohkin M, Kurose
Pharmacogenomics—drug disposition, drug K, Kaniwa N, Komamura K, Ueno K,
targets, and side effects. N Engl J Med 348: Kamakura S, Kitakaze M, Ozawa S, Sawada J
538–549 (2003) Comprehensive UGT1A1 genotyping
2. Marsh S (2009) Pyrosequencing. In: Patrinos in a Japanese population by pyrosequencing.
GP, Ansorge W (eds) Molecular diagnostics. Clin Chem 49:1182–1185
Elsevier, USA 7. Garsa A, Marsh S, McLeod HL (2005)
3. Freimuth RR, Ameyaw M-M, Pritchard SC, CYP3A4 and CYP3A5 genotyping by
Kwok P-Y, McLeod HL (2004) High- Pyrosequencing. BMC Med Genet 6:19
throughput genotyping methods for pharma- 8. Rose CM, Marsh S, Ameyaw MM, McLeod
cogenomic studies. Curr Pharmacogenomics HL (2003) Pharmacogenetic analysis of clini-
2:21–33 cally relevant genetic polymorphisms. Methods
4. Ahluwalia R, Freimuth R, McLeod HL, Marsh S Mol Med 85:225–237
(2003) Use of pyrosequencing to detect clini- 9. Lee SS, Kim WY, Jang YJ, Shin JG (2008)
cally relevant polymorphisms in dihydropyrimi- Duplex pyrosequencing of the TPMT3C and
dine dehydrogenase. Clin Chem 49:1661–1664 TPMT6 alleles in Korean and Vietnamese pop-
5. Hoskins JM, Marcuello E, Altes A, Marsh S, ulations. Clin Chim Acta 398:82–85
Maxwell T, Van Booven DJ, Pare L, 10. Yu J, Marsh S (2008) SNP and DNA methyla-
Culverhouse R, McLeod HL, Baiget M (2008) tion analysis with Pyrosequencing. In: Wang F
Irinotecan pharmacogenetics: influence of (ed) Biomarker methods in drug discovery and
pharmacodynamic genes. Clin Cancer Res development: methods and protocols.
14:1788–1796 Humana, Totowa, pp 119–140
Chapter 7
Abstract
Sequence variations in the human genome can affect the development of diseases and provide markers for
the identification of genetic diseases and drug susceptibility. Single Nucleotide Polymorphisms (SNPs), the
most abundant sequence variations in the genome, are used in pharmacogenetics as indicators of drug
therapy efficacy in individuals and are important road maps in the route to personalized medicine. This
chapter describes the development of PCR based custom multiplex SNP mutation analysis assays using
Luminex® Multi-Analyte Profiling (xMAP®) Technology. Up to 500 different mutations can be detected
in a single well and up to 384 samples can be analyzed per run.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_7, © Springer Science+Business Media, LLC 2013
115
116 Gonnie Spierings and Sherry A. Dunbar
Table 1
Mutations and polymorphisms detected by the xTAG®
CYP 2D6 Kit v3. All ancillary reagents are included in the kit
Fig. 1 Schematic presentation of the detection modules of Luminex® 200™/FLEXMAP 3D® (a) and MAGPIX® (b)
2 Materials
2. Thermal cycler for 0.2 ml thin wall PCR tubes and 96-well
plates.
3. Microcentrifuge for 1.5 ml and 0.2 ml tubes.
4. Vortex mixer.
5. Mini bath sonicator.
6. Cold block for 1.5 ml and 0.5 ml microcentrifuge tubes.
7. PCR cooler rack for 0.2 ml thin wall PCR tubes (96-well
compatible).
8. Pipettes (P10, P20, P100, P200, P1000).
9. 8 channel pipette (1–10 μl, 5–50 μl, 50–200 μl).
10. Racks for 1.5 ml and 0.5 ml microcentrifuge tubes and for
0.2 ml thin-walled PCR tubes.
11. Dynal MPC®-96S Magnetic Particle Concentrator (see Note 4).
12. 96-well plate magnet compatible with V-bottom plates (see
Notes 5 and 6).
13. Pipette aid.
2.2 Consumables 1. 0.2 ml thin wall polypropylene tubes for PCR (see Note 7).
2. 1.5 ml and 0.5 ml polypropylene microcentrifuge tubes.
3. 25 ml Pipettes.
4. Polypropylene tubes (Falcon® tubes): 15 ml and 50 ml.
5. Aerosol Resistant tips for Pipettes.
6. Corning Costar ® Thermowell® Thin-wall polycarbonate
96-well plate (see Note 8).
7. Bio-Rad Microseal® A.
8. Parafilm M.
9. Reservoir basins.
3 Methods
3.1 Multiplexed The following procedures are for single PCR reactions (see Notes
PCR Reaction 17 and 18). Scale it to analyze up to 96 samples by multiplying the
volumes by the number of samples (see Note 19). PCR should be
performed under optimized conditions. The parameters listed
below are for example purposes only.
1. PCR Set-up
(a) Method A: Asymmetric PCR.
Prepare the following PCR mix per sample: 1× Qiagen
PCR reaction buffer, 1.5 mM MgCl2, 200 μM each dNTP
(-dCTP), 200 μM biotin-dCTP, 0.4–1 μM each TAGged
primer, 0.004–0.1 μM each primer without TAG, 2.5 Units
Qiagen HotStarTaq® polymerase, 50 ng template (see
Notes 20 and 21).
(b) Method B: PCR with Lambda Exonuclease Treatment.
Prepare the following PCR mix per sample: 1× Qiagen
PCR reaction buffer, 1.5 mM MgCl2, 200 µM each dNTP
(-dCTP), 200 µM biotin-dCTP, 0.2 μM each primer,
2.5 Units Qiagen HotStarTaq® polymerase, 50 ng tem-
plate (see Note 21).
(c) Method C: Spacer-modified TAGged Primers.
Prepare the following PCR mix per sample: 1× Qiagen PCR
reaction buffer, 1.5 mM MgCl2, 200 μM each dNTP, 0.2 μM
each primer, 2.5 Units Qiagen HotStarTaq® polymerase,
50 ng Template (see Note 22).
122 Gonnie Spierings and Sherry A. Dunbar
For Method C
1. Select the appropriate MagPlex®-TAG™ microsphere sets and
resuspend according to the instructions described in the
Product Information Sheet provided with your microspheres.
2. Combine 2,500 microspheres of each set per reaction.
3. Dilute/concentrate the MagPlex®-TAG™ microsphere mixture
to 125 of each microsphere set per μl in 1.1× Tm Hybridization
Buffer by vortex and sonication for approximately 20 s.
4. Aliquot 20 μl of the MagPlex®-TAG™ microsphere mixture to
each well.
5. Add 1–5 μl of dH2O to each background well.
6. Add 1–5 μl of each PCR reaction to appropriate wells.
7. Prepare Reporter Mix by diluting SA-PE to 8–10 μg/ml in 1× Tm
Hybridization Buffer.
8. Add 70 μl to each well. Mix gently.
9. Cover the plate to prevent evaporation and hybridize at
37–45 °C for 25–45 min.
10. Analyze 70 μl at hybridization temperature on the Luminex
analyzer according to the system manual.
4 Notes
18. Prior to use, mix all solutions, except enzyme stock solutions,
by short vortex (2–5 s) and settle the reagents to the bottom of
the tube by short centrifugation (2–5 s). Enzyme stock solutions
should be taken from freezer when ready to use and returned
to freezer immediately after use (alternatively it can be kept on
a freezer block). Mix enzyme stock solutions by inverting and
flicking the tube followed by a short centrifugation step (2–5 s)
to settle reagents to the bottom of the tube.
19. When calculating master mix volumes for multiple reactions,
include a minimum of 10 % overage to account for variability
in pipetting. After making the master mix, vortex the solution
(2–5 s) followed by a short centrifugation step (2–5 s) and
then aliquot in the separate tubes.
20. Ratio of TAGged (excess) to non-TAGged (limiting) primer
should be optimized in the 10:1–100:1 range.
21. Ratio of biotinylated to unlabeled dNTPs may require
optimization.
22. During setup, keep master mix and samples on ice or a cold
block. Preheat thermal cycler to 95 °C.
23. If background signals are too high wash steps may need to be
added after the hybridization step and/or after adding the SA-PE.
24. These steps can be performed on a thermal cycler programmed
as follows: Hold at 96 °C, 90 s, Hold at 37 °C, Forever.
25. Optional Wash Procedure after Hybridization Step:
(a) Pellet the MagPlex®-TAG™ microspheres by placing the
plate on a magnetic separator and allow separation to
occur for 30–60 s. Remove the supernatant.
(b) Resuspend the pelleted MagPlex®-TAG™ microspheres in
75 μl of 1× Tm Hybridization Buffer.
(c) Repeat steps (a) and (b). This is a total of two washes.
(d) Pellet the MagPlex®-TAG™ microspheres by placing the
plate on a magnetic separator and allow separation to
occur for 30–60 s. Remove the supernatant.
● Alternatively, wash steps can be performed by centrif-
ugation or vacuum filtration.
– Pellet the MagPlex®-TAG™ microspheres by
centrifugation at ≥2,250 × g for 3 min and remove
the supernatant.
– Pre-wet a 1.2 μm Millipore filter plate with 1×
Tm Hybridization Buffer and filter by vacuum
manifold. Transfer the reactions to the pre-wetted
filter plate and remove the supernatant by vacuum
filtration. Wash twice with 100 μl 1× Tm
Hybridization Buffer.
126 Gonnie Spierings and Sherry A. Dunbar
References
1. Dunbar SA (2006) Applications of Luminex® Methods in molecular biology: Salmonella:
xMAP™ technology for rapid, high-throughput methods and protocols, vol 394. Humana,
multiplexed nucleic acid detection. Clin Chim Totowa, NJ, pp 1–19
Acta 363:71–82 5. Pickering JW et al (2004) Flow cytometric
2. Dunbar SA, Hoffmeyer MR (2013) assay for genotyping cytochrome P450 2C9
Microsphere-based multiplex immunoassays: and 2 C19—comparison with a microelec-
development and applications using Luminex® tronic DNA array. Am J Pharmacogenomics
xMAP® Technology. In: Wild DG (ed) The 4(3):199–207
immunoassay handbook, 4th edition. Elsevier, 6. Bruse E et al (2008) Improvements to bead-
Amsterdam, NL, pp 157–174 based oligonucleotide ligation SNP genotyping
3. Luminex Corporation (2010) Luminex assays. Biotechniques 45:559–571
Corporation launches new FDA cleared phar- 7. Monico CG et al (2007) Comprehensive
macogenetic diagnostic test. xTAG® mutation screening in 55 probands with type 1
CYP2D6Kit can assist physicians in improving primary Hyperoxaluria shows feasibility of a
patient care by helping to determine a person- gene-based diagnosis. J Am Soc Nephrol 18:
alized therapeutic strategy: press release. 1905–1914
PRNewswire via COMTEX. http://www. 8. Tian F et al (2008) A new single nucleotide
prnewswire.com/news-releases/luminex- polymorphism genotyping method based on
corporation- launches-new-fda-clear ed- gap ligase chain reaction and a microsphere
pharmacogenetic-diagnostic-test-108680239. detection assay. Clin Chem Lab Med 46:
html. Accessed 17 Nov 2010 486–489
4. Dunbar SA, Jacobson JW (2007) Quantitative, 9. Deshpande A et al (2010) A rapid multiplex
multiplexed detection of Salmonella and other assay for nucleic acid-based diagnostics.
pathogens by Luminex® xMAP™ suspension J Microbiol Methods 80:155–163
array. In: Schatten H, Eisenbank A (eds)
Chapter 8
Abstract
For the past two decades, linkage analysis and genome-wide analysis have greatly advanced our knowledge
of the human genome. But despite these successes the genetic architecture of diseases remains unknown.
More recently, the availability of next-generation sequencing has dramatically increased our capability for
determining DNA sequences that range from large portions of one individual’s genome to targeted regions
of many genomes in a cohort of interest. In this review, we highlight the successes and shortcomings that
have been achieved using genome-wide association studies (GWAS) to identify the variants contributing to
disease. We further review the methods and use of new technologies, based on next-generation sequencing,
that are becoming increasingly used to expand our knowledge of the causes of genetic disease.
Key words Linkage analysis, Genome-wide association study, Massively parallel sequencing, NGS-
applications, Pharmacogenomics
1 Introduction
Over the last 25 years, exciting progress has been made in identifying
the genetic variants associated with human diseases. During this
time, genes responsible for over 3,000 Mendelian disorders have
been identified (Online Mendelian Inheritance in Man, http://www.
ncbi.nlm.nih.gov/omim); however, identifying variants associated
with complex diseases has proven more difficult. More recently, new
genomic methods have begun to impact this field and currently more
than 1,300 variants have been associated with a variety of complex
diseases (http://www.genome.gov/gwastudies) [1].
Linkage analysis and more recently genome-wide association
studies (GWAS) have been the main tools to identify variants for
Mendelian and complex diseases, respectively. Both methods reveal
genomic regions associated with disease rather than the actual
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_8, © Springer Science+Business Media, LLC 2013
127
128 Eric Londin et al.
each variant alone can yield a minor amount of risk, but when
combined, their effect is substantially increased. In GWAS, these
disease-causing variants may not be identified directly, but rather
genomic locations linked to them may be identified. In fact, statis-
tically significant association is often not found to be within a gene.
Generally, the closest gene associated with the common variant
represents the most likely candidate gene.
The first GWAS performed, in 2005, was for age-related macular
degeneration (AMD). Here, Klein et al. [9], genotyped ~100,000
SNPs throughout the genome in a small sample set of 96 cases and
50 controls resulting in the association of a SNP in the comple-
ment factor H (CFH) gene. The p-value of the associated SNP
surpassed the genome-wide significance threshold; and, the high
effect size (odds-ratio = 4.6) contributed to the highly significant
finding. The success of this study suggested that this would be a
viable approach for other complex disorders. Since the AMD study,
numerous GWAS have been published identifying over 1,300
significant associations (p-value < 5 × 10−8) with over 220 different
traits (http://www.genome.gov/gwastudies) [1].
While thousands of associations have been identified, they have
failed, with few exceptions, to produce results as definitive as the
AMD study. Over time, the number of SNPs assayed has increased
into the millions with tens of thousands of subjects. Even with
these increasingly large studies, only modest associations have been
identified. For example, studies of Crohn’s disease have identified
30 loci, but when combined they only explain ~20 % of the overall
heritability of the disease [10]. Similarly, studies of human height,
which has an estimated heritability of ~80 %, have identified hun-
dreds of loci which explain only 10 % of its overall heritability [11].
These studies are in contrast to AMD in which five common loci
explain over 50 % of the heritability of the disease [12]. These dif-
ferences in study results can be attributed to the fact that some
common diseases, such as AMD, can be explained by a limited
number of common variants of large effect [13]. But, for most
other conditions, this is not the case, and common variants only
account for a small part of the overall heritability of the disease.
In conclusion, even though GWAS has been successful in iden-
tifying many low risk alleles for common disease, the findings do not
explain a large proportion of the heritability of complex diseases [8].
Several possible reasons for this exist. Estimates of heritability, based
on familial aggregation, may be inflated. Genetic and non-genetic
factors contribute to familial aggregation of disease [14], with the
latter not being detected in a GWAS. Second, GWAS tend to iden-
tify loci not genes, and a positive signal is not always in a protein-
coding region making identification of the disease-causing variants
difficult. Therefore, the proportion of heritability explained by the
associated SNPs may be underestimated. Third, not all of the
genome is covered. Finally, the majority of GWAS are underpowered
130 Eric Londin et al.
2.4 NGS Sequencing The newer sequencing technologies can achieve a much higher
Platforms throughput by sequencing a large number of samples in parallel.
Currently, a variety of new platforms are available to perform
massively parallel sequencing, with each platform allowing for dif-
ferences in the scale of the sequence being performed [20–22].
Use of Linkage Analysis, Genome-Wide Association Studies, and Next-Generation… 131
Table 1
Overview of the three main next-generation sequencing platforms
Life Technologies
Roche 454 SOLiD/5500xl Illumina HiSeq
Sequencing Pyrosequencing Sequencing by ligation of Polymerase-mediated
method fluorescently labeled incorporation of terminally
nucleotides labeled fluorescent nucleotides
Library Emulsion PCR Emulsion PCR Enzymatic amplification
amplification
method
Maximum read Up to 1,000 bp Mate-paired: 2 × 50 bp Paired-end: 2 × 150 bp
lengths Paired-end: 75 bp and 35 bp
Fragment: 75 bp
Throughput 700 Mb/run Up to 15 Gb/day for a single Up to 35 Gb/day for a single
flow cell flow cell
Fig. 1 Roche 454 Sequencing. (a) Genomic DNA is fragmented and adapter sequences are ligated onto
fragmented DNA mixed with agarose beads; (b) Emulsion PCR is used to amplify the DNA fragments on the
agarose beads generating millions of amplified sequencing templates on each bead; (c) The beads are deposited
into PicoTiter wells where simultaneous sequencing of the entire genome is performed in thousands of picoliter-
sized wells by Pyrosequencing; (d) Pyrosequencing reactions consist of stepwise elongation of the primer
strand by sequential addition of the individual deoxynucleoside triphosphates in the presence of sulfurylase
and luciferase. Sequence at each elongation step is inferred by measuring light emission as an indicator of
nucleotide incorporation. This method allows for the amplification of up to 1,000 nucleotide size sequences
(with permission from 454 Sequencing© Roche Diagnostics, Branford, CT.)
reactions by two different primers allowing for up to a 99.99 % accuracy to be achieved (with permission from Life Technologies, Carlsbad, CA.)
133
134 Eric Londin et al.
2.5 Applications The increased use of NGS is rapidly expanding our understanding
of NGS of the genetic basis of disease. Perhaps the largest genome sequenc-
ing effort being performed currently is the 1000 Genomes Project
[15]. The project aims to characterize variations in the human
genome by performing both whole-genome and exome sequenc-
ing on roughly 2,500 subjects from diverse population groups.
The pilot phase of this project sequenced 179 individuals and iden-
tified over 15 million new genetic variations. Release of this data
into the public domain will aid additional studies and increase our
knowledge of genetic variation.
Perhaps the greatest success of next-generation sequencing to
date has been in the discovery of variants for rare Mendelian dis-
eases. Instead of sequencing the entire genome, targeted sequenc-
ing of the coding regions as well as whole-exome sequencing has
yielded valuable results in the identification of disease-causing vari-
ants. The initial proof-of-concept that these were viable approaches
came in 2009 with the sequencing of four cases of Freeman–
Sheldon syndrome (FSS) and an additional eight controls [26].
While the cause of the disease was known (MYH3), the authors
were able to identify causative variants within this gene in all four
of their subjects. Since then, whole-exome sequencing has been
used extensively to identify previously unknown causes of a variety
of diseases (Table 2).
Use of Linkage Analysis, Genome-Wide Association Studies, and Next-Generation… 135
Fig. 3 Illumina Sequencing by Synthesis. (a) Genomic DNA is randomly fragmented and adapters are ligated
to both ends; (b)–(f) Bridge amplification is used to create clusters of DNA strands and fluorescently labeled
3′-OH blocked nucleotides are added to the flow cell with DNA polymerase; (g) The strands are extended by
one nucleotide. Following the addition of a single nucleotide, the unused nucleotides and DNA polymerase are
washed away, and the reaction is imaged; (h) and (i) The process is then repeated for another round of nucleo-
tide incorporation (with permission from Illumina, San Diego, CA.)
136 Eric Londin et al.
(continued)
gene
(continued)
139
Table 2
140
(continued)
Saarinen Whole-exome Hodgkin lymphoma A family of four affected Identified mutations in the ataxia-telangiectasia
et al. [67] sequencing cousins (NAPT) gene in the subjects and confirmed
the presence of the mutation in additional
affected subjects
Simpson Whole-exome Hajdu–Cheney Three unrelated affected Identified nonsense mutations in the NOTCH2
et al. [68] sequencing syndrome subjects gene
Snape Whole-exome Mosaic variegated Two affected siblings Identified two mutations in the CEP57 gene,
et al. [69] sequencing aneuploidy syndrome the first is a 2 bp deletion and the second an
(MVA) 11 bp insertion present in both siblings.
Confirmed in an additional 18 affected
individuals
Szperl Whole-exome Celiac disease Two affected subjects from a Identified 12 nonsense mutations with low
et al. [70] sequencing three generation family with frequency present in both individuals. Two of
six affected individuals the variants in CSAG1 and KRT37 genes
were present in all six affected individuals and
two additional variants in the MADD and
GBGT1 genes were also present in 5/6 and
4/6 individuals, respectively
Sundaram Whole-exome Tourette syndrome Ten members of a 3-generation Identified three missense mutations in the
et al. [71] sequencing family MRPL3, DNAJC13 and OFCC1 genes that
segregated with chronic tic disorder
Timmermann Whole-exome Colorectal cancer Tumor and adjacent non- Identified mutations in the intracellular kinase
et al. [72] sequencing affected normal colonic domain of bone morphogenetic protein
tissue from two subjects receptor 1A (BMPR1A)
Method Number of subjects
Study performed Disease studied sequenced Major findings
Tsurusaki Whole-exome X-linked leukodystrophy An affected subject and Identified a nonsense mutation in the MCT8
et al. [73] sequencing unaffected sibling gene
Vissers Whole-exome Mental retardation Ten families with one affected Identified missense de novo mutations in nine
et al. [74] sequencing child genes
Vissers Whole-exome Chondrodysplasia and Three affected individuals Identified a missense mutation in the Golgi-
et al. [75] sequencing abnormal joint resident nucleotide phosphatase (gPAPP)
development gene in all three patients
Wei et al. [76] Whole-exome Melanoma 14 matched normal and Identified 68 genes that have somatic
sequencing metastatic tumors mutations. TRRAP harbored a recurrent
mutation in 4 % of additional patients and a
mutation in GRIN2A was mutated in 33 % of
the melanoma samples
Worthey et al. Whole-exome Intractable inflammatory A single affected subject Identified a single missense mutation in the
[77] sequencing bowel disease X-linked inhibitor of apoptosis gene. The
exome sequencing performed was used to
make a definitive diagnosis of the disease
Yamaguchi et al. Whole-exome Primary failure of tooth Two affected subjects Identified a missense mutation in the
[78] sequencing eruption (PFE) parathyroid hormone 1 receptor gene
(PTH1R) gene
Zhou et al. [79] Whole-exome Hereditary hypotrichosis One affected subject Identified a missense mutation in the ribosomal
sequencing simplex protein L21 (RPL21) gene
Zuchner et al. Whole-exome Retinitis pigmentosa A single affected family Identified a missense mutation in the
[80] sequencing dehydrodolichyl diphosphate synthase
(DHDDS) gene
Use of Linkage Analysis, Genome-Wide Association Studies, and Next-Generation…
141
142 Eric Londin et al.
2.6 Limitations Because most disease-causing variants are located within the
to NGS coding regions of the genome, whole exome sequencing will prove
to be a powerful approach to identify genetic variation, and will
continue to be the method of choice until whole genome sequenc-
ing can be performed more cost effectively. Despite the clear
advantages of exome sequencing, there are drawbacks to this
approach. First, this approach will not detect structural variants
such as copy number changes, which have been implicated in dis-
ease. Second, there is a limitation in the specific exons that are
captured. Additional variants may be located in exons not targeted,
and such, not be identified. Finally, since exome capture is not
sufficiently specific, it now requires sequencing of a much larger
area. This level of sequencing would be equivalent to performing
whole genome sequencing at a low coverage. However, the low
coverage sequencing would likely miss many variants that are
Use of Linkage Analysis, Genome-Wide Association Studies, and Next-Generation… 143
3 Conclusions
Acknowledgments
This work was supported the Kimmel Cancer Center and the
Computational Medicine Center at Thomas Jefferson University
Jefferson Medical College.
References
1. Hindorff LA et al (2009) Potential etiologic strongly influences risk of age-related macular
and functional implications of genome-wide degeneration. Nat Genet 38:1055–1059
association loci for human diseases and traits. 13. Jakobsdottir J et al (2009) Interpretation of
Proc Natl Acad Sci USA 106:9362–9367 genetic association studies: markers with repli-
2. Rommens JM et al (1989) Identification of the cated highly significant odds ratios may be
cystic fibrosis gene: chromosome walking and poor classifiers. PLoS Genet 5:e1000337
jumping. Science 245:1059–1065 14. Rose SP (2006) Commentary: heritability esti-
3. Riordan JR et al (1989) Identification of the mates–long past their sell-by date. Int J
cystic fibrosis gene: cloning and characteriza- Epidemiol 35:525–527
tion of complementary DNA. Science 15. 1000 Genomes Project Consortium, Abecasis
245:1066–1073 GR et al (2010) A map of human genome
4. Kerem B et al (1989) Identification of the cys- variation from population-scale sequencing.
tic fibrosis gene: genetic analysis. Science Nature 467:1061–1073
245:1073–1080 16. Venter JC et al (2001) The sequence of the
5. Risch N, Merikangas K (1996) The future of human genome. Science 291:1304–1351
genetic studies of complex human diseases. 17. Lander ES et al (2001) Initial sequencing and
Science 273:1516–1517 analysis of the human genome. Nature 409:
6. Altmuller J et al (2001) Genomewide scans of 860–921
complex human diseases: true linkage is hard 18. Schloss JA (2008) How to get genomes at one
to find. Am J Hum Genet 69:936–950 ten-thousandth the cost. Nat Biotechnol
7. Consortium IH (2005) A haplotype map of 26:1113–1115
the human genome. Nature 437:1299–1320 19. Hert DG, Fredlake CP, Barron AE (2008)
8. Manolio TA et al (2009) Finding the missing Advantages and limitations of next-generation
heritability of complex diseases. Nature 461: sequencing technologies: a comparison of
747–753 electrophoresis and non-electrophoresis meth-
9. Klein RJ et al (2005) Complement factor H ods. Electrophoresis 29:4618–4626
polymorphism in age-related macular degen- 20. Pareek CS, Smoczynski R, Tretyn A (2011)
eration. Science 308:385–389 Sequencing technologies and genome
10. Barrett JC et al (2008) Genome-wide associa- sequencing. J Appl Genet 52:413–435
tion defines more than 30 distinct susceptibil- 21. Metzker ML (2010) Sequencing technologies—
ity loci for Crohn’s disease. Nat Genet the next generation. Nat Rev Genet 11:31–46
40:955–962 22. Mardis ER (2008) Next-generation DNA
11. Lango AH et al (2010) Hundreds of variants sequencing methods. Annu Rev Genomics
clustered in genomic loci and biological pathways Hum Genet 9:387–402
affect human height. Nature 467:832–838 23. Zheng Z et al (2010) Titration-free massively
12. Maller J et al (2006) Common variation in three parallel pyrosequencing using trace amounts of
genes, including a noncoding variant in CFH, starting material. Nucleic Acids Res 38:e137
Use of Linkage Analysis, Genome-Wide Association Studies, and Next-Generation… 145
24. Margulies M et al (2005) Genome sequencing 42. Blaby-Haas CE, de Crecy-Lagard V (2011)
in microfabricated high-density picolitre reac- Mining high-throughput experimental data to
tors. Nature 437:376–380 link gene and function. Trends Biotechnol
25. Depristo MA et al (2011) A framework for 29:174–182
variation discovery and genotyping using next- 43. Nothnagel M et al (2011) Technology-specific
generation DNA sequencing data. Nat Genet error signatures in the 1000 Genomes Project
43:491–498 data. Hum Genet 130:505–516
26. Ng SB et al (2009) Targeted capture and mas- 44. Al Badr W et al (2011) Exome capture and
sively parallel sequencing of 12 human exomes. massively parallel sequencing identifies a novel
Nature 461:272–276 HPSE2 mutation in a Saudi Arabian child with
27. Ng SB et al (2009) Exome sequencing identi- Ochoa (urofacial) syndrome. J Pediatr Urol
fies the cause of a mendelian disorder. Nat 7:569–573
Genet 42:30–35 45. Alvarado DM et al (2011) Exome sequencing
28. Hoischen A et al (2010) De novo mutations of identifies an MYH3 mutation in a family with
SETBP1 cause Schinzel-Giedion syndrome. distal arthrogryposis type 1. J Bone Joint Surg
Nat Genet 42:483–485 Am 93:1045–1050
29. Bansal V et al (2010) Statistical analysis strate- 46. Barak T et al (2011) Recessive LAMC3 muta-
gies for association studies involving rare vari- tions cause malformations of occipital cortical
ants. Nat Rev Genet 11:773–785 development. Nat Genet 43:590–594
30. Nalls MA et al (2011) Imputation of sequence 47. Becker J et al (2011) Exome sequencing iden-
variants for identification of genetic risks for tifies truncating mutations in human
Parkinson’s disease: a meta-analysis of genome- SERPINF1 in autosomal-recessive osteogene-
wide association studies. Lancet 377:641–649 sis imperfecta. Am J Hum Genet 88:362–371
31. Su Z et al (2011) Next-generation sequencing 48. Bolze A et al (2010) Whole-exome-
and its applications in molecular diagnostics. sequencing-based discovery of human FADD
Expert Rev Mol Diagn 11:333–343 deficiency. Am J Hum Genet 87:873–881
32. Marian AJ (2011) Medical DNA sequencing. 49. Caliskan M et al (2011) Exome sequencing
Curr Opin Cardiol 26:175–180 reveals a novel mutation for autosomal reces-
33. Diamandis EP (2009) Next-generation sive non-syndromic mental retardation in the
sequencing: a new revolution in molecular TECR gene on chromosome 19p13. Hum
diagnostics? Clin Chem 55:2088–2092 Mol Genet 20:1285–1289
34. Choi M et al (2009) Genetic diagnosis by 50. de Greef JC et al (2011) Mutations in ZBTB24
whole exome capture and massively parallel are associated with immunodeficiency, centro-
DNA sequencing. Proc Natl Acad Sci USA meric instability, and facial anomalies syndrome
106:19096–19101 type 2. Am J Hum Genet 88:796–804
35. Maxmen A (2011) Exome sequencing deci- 51. Erlich Y et al (2011) Exome sequencing and
phers rare diseases. Cell 144:635–637 disease-network analysis of a single family
implicate a mutation in KIF1A in hereditary
36. St HC et al (2011) NT5E mutations and spastic paraparesis. Genome Res 21:658–664
arterial calcifications. N Engl J Med 364:
432–442 52. Gilissen C et al (2010) Exome sequencing
identifies WDR35 variants involved in
37. Daly AK (2010) Genome-wide association Sensenbrenner syndrome. Am J Hum Genet
studies in pharmacogenomics. Nat Rev Genet 87:418–423
11:241–246
53. Glazov EA et al (2011) Whole-exome
38. Dewey FE et al (2011) Phased whole-genome re-sequencing in a family quartet identifies
genetic risk in a family quartet using a major POP1 mutations as the cause of a novel skele-
allele reference sequence. PLoS Genet tal dysplasia. PLoS Genet 7:e1002027
7:e1002280
54. Gotz A et al (2011) Exome sequencing identi-
39. Trapnell C, Salzberg SL (2009) How to map fies mitochondrial alanyl-tRNA synthetase
billions of short reads onto genomes. Nat mutations in infantile mitochondrial cardio-
Biotechnol 27:455–457 myopathy. Am J Hum Genet 88:635–642
40. Nielsen R et al (2011) Genotype and SNP call- 55. Greif PA et al (2011) Somatic mutations in acute
ing from next-generation sequencing data. promyelocytic leukemia (APL) identified by
Nat Rev Genet 12:443–451 exome sequencing. Leukemia 25:1519–1522
41. Hinchcliffe M, Webster P (2011) In silico 56. Johnson JO et al (2010) Exome sequencing
analysis of the exome for gene discovery. reveals VCP mutations as a cause of familial
Methods Mol Biol 760:109–128 ALS. Neuron 68:857–864
146 Eric Londin et al.
57. Li Y et al (2010) Resequencing of 200 human disorder of severe and progressive bone loss.
exomes identifies an excess of low-frequency Am J Hum Genet 43:303–305
non-synonymous coding variants. Nat Genet 69. Snape K et al (2011) Mutations in CEP57
42:969–972 cause mosaic variegated aneuploidy syndrome.
58. Liu Y et al (2011) Confirmation by exome Am J Hum Genet 43:527–529
sequencing of the pathogenic role of NCSTN 70. Szperl AM et al (2011) Exome sequencing in a
mutations in acne inversa (hidradenitis suppu- family segregating for celiac disease. Clin
rativa). J Invest Dermatol 131:1570–1572 Genet 80:138–147
59. Mondal K et al (2011) Targeted sequencing of 71. Sundaram SK et al (2011) Exome sequencing
the human X chromosome exome. Genomics of a pedigree with tourette syndrome or
98:260–265 chronic tic disorder. Ann Neurol 69:901–904
60. Montenegro G et al (2011) Exome sequenc- 72. Timmermann B et al (2010) Somatic mutation
ing allows for rapid gene identification in a profiles of MSI and MSS colorectal cancer
Charcot-Marie-Tooth family. Ann Neurol 69: identified by whole exome next generation
464–470 sequencing and bioinformatics analysis. PLoS
61. Ng SB et al (2010) Exome sequencing identi- One 5:e15661
fies MLL2 mutations as a cause of Kabuki syn- 73. Tsurusaki Y et al (2011) Exome sequencing of
drome. Nat Genet 42:790–793 two patients in a family with atypical X-linked
62. O’Roak BJ et al (2011) Exome sequencing in leukodystrophy. Clin Genet 80:161–166
sporadic autism spectrum disorders identifies 74. Vissers LE et al (2010) A de novo paradigm
severe de novo mutations. Nat Genet 43: for mental retardation. Am J Hum Genet
585–589 42:1109–1112
63. O’Sullivan J et al (2011) Whole-exome 75. Vissers LE et al (2011) Chondrodysplasia and
sequencing identifies FAM20A mutations as a abnormal joint development associated with
cause of amelogenesis imperfecta and gingival mutations in IMPAD1, encoding the Golgi-
hyperplasia syndrome. Am J Hum Genet resident nucleotide phosphatase, gPAPP. Am J
88:616–620 Hum Genet 88:608–615
64. Ostergaard P et al (2011) Rapid identification 76. Wei X et al (2011) Exome sequencing identi-
of mutations in GJC2 in primary lymphoe- fies GRIN2A as frequently mutated in mela-
dema using whole exome sequencing com- noma. Nat Genet 43:442–446
bined with linkage analysis with delineation of 77. Worthey EA et al (2011) Making a definitive
the phenotype. J Med Genet 48:251–255 diagnosis: successful clinical application of
65. Puente XS et al (2011) Exome sequencing and whole exome sequencing in a child with intrac-
functional analysis identifies BANF1 mutation table inflammatory bowel disease. Genet Med
as the cause of a hereditary progeroid syn- 13:255–262
drome. Am J Hum Genet 88:650–656 78. Yamaguchi T et al (2011) Exome resequenc-
66. Rios J et al (2010) Identification by whole- ing combined with linkage analysis identifies
genome resequencing of gene defect responsi- novel PTH1R variants in primary failure of
ble for severe hypercholesterolemia. Hum Mol tooth eruption in Japanese. J Bone Miner Res
Genet 19:4313–4318 26(7):1655–1661
67. Saarinen S et al (2011) Exome sequencing 79. Zhou C et al (2011) Mutation in ribosomal
reveals germline NPAT mutation as a candi- protein L21 underlies hereditary hypotrichosis
date risk factor for Hodgkin lymphoma. Blood simplex. Hum Mutat 32:710–714
118:493–498 80. Zuchner S et al (2011) Whole-exome sequenc-
68. Simpson MA et al (2011) Mutations in ing links a variant in DHDDS to retinitis pig-
NOTCH2 cause Hajdu-Cheney syndrome, a mentosa. Am J Hum Genet 88:201–206
Chapter 9
Abstract
The Illumina GoldenGate Assay is a technique that is widely used in molecular genetics to analyze up to
thousands of single nucleotide polymorphism (SNPs) simultaneously, providing data of very high quality
in a fast and efficient manner. This technique allows the user to optimize the number of genetic loci to be
interrogated in a way that best suits their research goals. Here are described in detail all the steps to be
followed in the process of genotyping a custom panel, from panel design through data analysis.
Key words GoldenGate assay, Custom genotyping, Single nucleotide polymorphism, Fluorescence
signal, Cluster analysis
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_9, © Springer Science+Business Media, LLC 2013
147
148 Anna González-Neira
● The extended products are used as template for the PCR using
three universal PCR primers: two forward primers, P1 and P2,
labeled with Cy3 and Cy5 and a reverse primer, P3.
● The PCR products are again bound and the single-stranded
dye-labeled DNAs are eluted and prepared for hybridization to
their complementary bead type via their unique address
sequences on the micron silica beads or cylindrical glass micro-
beads, using a beadChip (BeadArray technology) or microtiter
plate (Veracode technology), respectively.
References
8. Fallin MD et al (2005) Bipolar I disorder and genotyping assay. PLoS One 6(6):e20251,
schizophrenia: a 440-single-nucleotide poly- Epub 2011 Jun 6
morphism screen of 64 candidate genes among 10. Designing custom GoldenGate genotyping
Ashkenazi Jewish case-parent trios. Am J Hum assay. Technical note (from www.illumina.com)
Genet 77(6):918–936, Epub 2005 Oct 28
11. GoldenGate® assay workflow. Technical note
9. Campino S et al (2011) Population genetic (from www.illumina.com)
analysis of Plasmodium falciparum parasites 12. Analyzing GoldenGate genotyping data.
using a customized Illumina GoldenGate Technical note (from www.illumina.com)
Chapter 10
Abstract
With novel genome-wide technologies it is nowadays possible to perform detailed molecular analyses of
normal and malignant tissues. Acute myeloid leukemia (AML) is a heterogeneous group of diseases with
variable response to therapy. Gene expression profiling and genome-wide genotyping have recently been
successfully applied to unravel the heterogeneity of AML. This chapter gives instructions and recommen-
dations for genome-wide gene expression analyses, genotyping, and copy number analyses, as performed
for AML using Affymetrix GeneChips.
Key words Affymetrix GeneChips, Affymetrix DNA mapping arrays, Gene expression profiling,
Genome-wide genotyping, Copy number analyses, Acute myeloid leukemia
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_10, © Springer Science+Business Media, LLC 2013
155
156 Mathijs A. Sanders and Peter J.M. Valk
2 Materials
2.3 DNA Isolation 1. DNA lysis buffer: 12.5 ml 100 mM EDTA and 3.75 ml 1 M
NaCl in 50 ml water.
2. Proteinase K (20 mg/ml).
3. SDS (20 %).
4. Saturated NaCl (87.7 g NaCl in 250 ml water).
5. 96 % ethanol.
6. TE buffer (10 mM Tris–HCl, 1 mM EDTA).
2.4 Labeling and Hybridization and staining of cRNA were performed exactly
Hybridization according to the manufacturer of the Affymetrix GeneChips
Procedures [Affymetrix (Santa Clara, CA, USA)] [10].
2.4.2 Hybridization and 1. Acetylated bovine serum albumin (BSA) solution (50 mg/ml).
Staining Gene Expression 2. Herring Sperm DNA (10 mg/ml).
Profiling
3. GeneChip® Eukaryotic Hybridization Control Kit (contains
Hybridization Control cRNA and Control Oligo B2), 150 reactions, Affymetrix.
4. 5 M NaCl (RNase-free/DNase-free).
5. MES-Free Acid Monohydrate SigmaUltra.
6. MES Sodium Salt.
7. EDTA Disodium Salt, 0.5 M solution.
2.5 Labeling, Labeling of genomic DNA, hybridization, and staining for genome-
Hybridization, and wide genotyping were performed exactly according to the manu-
Staining Genome-Wide facturer of the Affymetrix GeneChips [Affymetrix (Santa Clara,
Genotyping CA, USA)] [11].
3.2 RNA Isolation 1. Lyse mononuclear cells (see Note 6) in 6 ml RNA lysis buffer
(See Note 5) by adding the lysis buffer, resuspending the cells and vortexing
for 1 min.
2. Pipet in open centrifuge tubes for SW41Ti rotor, 3 ml cesium
chloride.
3. Pipet carefully the lysate on top of the cesium chloride.
4. Weigh the tubes and make them equal in weight with lysis buf-
fer (maximal variation: 0.1 g).
5. Centrifuge for 18 h at 32,000 rpm (17,500 × g) at room
temperature.
6. After centrifuging the RNA pellet is on the bottom, DNA is
halfway and proteins are in the upper phase.
7. Pipet off the upper fluid and turn the tube upside down. DNA
that is left over in the tube cannot contaminate the RNA
pellet.
8. Cut the bottom of the tube containing the RNA pellet with a
sterile lancet.
9. Collect the RNA pellet into a sterile eppendorf tube by wash-
ing the bottom twice with RNase-free water. Keep on ice.
10. Precipitate the RNA by adding 40 μl 3 M NaAc pH 5.0 and
1 ml 96 % ethanol. Mix well by turning upside down.
11. Incubate for 30 min at −70 °C.
12. Centrifuge for 15 min at 16200 × g at 4 °C.
13. Wash pellet with 500 μl 70 % ethanol. Centrifuge for 10 min at
13,000 rpm at 4 °C.
14. Dissolve the pellet in 25 μl RNase-free water, by pipetting up
and down.
15. Measure the RNA concentration using a 1:10 dilution and
determine quality (see Note 7).
16. Store RNA at −70 °C.
3.3 DNA Isolation 1. Pellet mononuclear cells (see Note 9) by centrifugation 5 min
(See Note 8) at 4 °C at 1,200 rpm (300 × g).
2. Remove supernatant and resuspend 10–100× 106 cells in 3 ml
DNA lysis buffer.
3. Add 25 μl proteinase K (20 mg/ml).
4. Add 150 μl SDS (20 %).
5. Incubate overnight at 37 °C.
6. Add 1 ml saturated NaCl and vortex.
7. Centrifuge for 15 min at 2,500 rpm (1,100 × g).
8. Repeat steps 6 and 7 until the supernatant is clear.
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 161
Fig. 1 Overview of Affymetrix genome-wide expression profiling with U133-plus2.0 GeneChip microarray.
Affymetrix gene expression arrays use a standardized biotin labeling protocol, which utilizes an Oligo(dT)-T7
promoter primed, in vitro transcription based linear amplification strategy. The procedure consists of reverse
transcription with an oligo(dT) primer bearing a T7 promoter. Subsequently, the cDNA is subjected to second
strand synthesis and cleanup to become a template for in vitro transcription (IVT) with T7 RNA Polymerase in
the presence of biotinylated nucleotides. Following this, strict Affymetrix protocols are utilized by the standard
fluidics and scanning stations
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 163
3.4.2 Hybridization and Hybridization and staining of cRNA were performed exactly
Staining Gene Expression according to the manufacturer of the Affymetrix GeneChips
Profiling [Affymetrix (Santa Clara, CA, USA)] [10].
3.5 Labeling, Labeling of genomic DNA, hybridization, and staining for genome-
Hybridization, and wide genotyping were performed exactly according to the manu-
Staining Genome-Wide facturer of the Affymetrix GeneChips [Affymetrix (Santa Clara,
Genotyping CA, USA)] (Fig. 2) [11].
Fig. 2 Overview of Affymetrix genome-wide genotyping with human SNP array 6.0 array [11]. Genomic DNA is
digested with NspI and StyI, followed by adapter ligation, linear amplification, and labeling, before hybridization
on the GeneChip
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 165
3.7 Analyses of Gene In the following paragraphs important recommendations are given
Expression Profiling how to successfully accomplish the major study objectives when
Data analyzing gene expression data, i.e., class discovery, class compari-
son, and class prediction. A full overview of all types of gene expres-
sion data analyses is given in [13].
3.7.1 Normalization and Pre-processing microarrays is a vital step in acquiring the measure-
Summarization ments. To truly understand the output of the whole data-generating
process and the reason for pre-processing it is necessary to first
explain the fundaments of microarray analysis. On a microarray
each probe pair consists of a perfect match and a mismatch probe.
The signal intensity emitted from these probes are read by the
microarray scanner and condensed into a signal intensity file, also
called .CEL files. This .CEL file contains the signal intensity mea-
surement for each probe situated on the microarray and is there-
fore pivotal for research. These probes belong to a probe set, which
is directly related to a known transcript, as has been stated above.
Hence, the individual probes associated to one probe set can be
summarized to one intensity value reflecting the expression level of
the associated gene. Before summarization is it of utmost impor-
tance that the microarrays are normalized. Normalization is a type
of “calibration” that serves to remove nonbiological or systematic
variation between samples, such as differences in the background
and noise levels, hybridization conditions, handling and instru-
mentation consistencies. Most normalization procedures perform
the following pre-processing steps:
Step 1: Background correction of the probes.
Step 2: Normalization within the chip to correct technical variation
or to facilitate between-array comparison. Frequently used meth-
ods use statistical techniques such as “quantiles” or “invariant set
of genes.”
Step 3: Perfect Match correction methods, e.g., subtracting the
Mismatch probe from the Perfect Match probe.
Step 4: Summarization, e.g., “average difference” and “median
polish” which converts the 11–22 probe pair intensities into one
probe set value.
166 Mathijs A. Sanders and Peter J.M. Valk
3.7.2 Class Discovery Usually the first step after pre-processing is the use of techniques
to perform unsupervised analysis. One of these techniques is called
clustering, which is a tool that aims at dividing the data in such a
way that items (e.g., samples or genes) fall into the same group and
that dissimilar items fall into different groups. Clustering is an
unsupervised technique when prior information, such as pheno-
type, molecular subtype or any clinical parameter, is not taken into
account. It is an easy way to infer if samples with similar subtypes
of disease are grouping together, hinting the researcher that there
is information in the data that could discriminate these subtypes.
There are many different techniques to perform clustering. Most
frequently the technique called hierarchical clustering is performed.
This method divides the data set into clusters, which are further
subdivided into smaller clusters, resulting in a dendrogram. To
cluster the data the method needs a similarity/dissimilarity matrix.
Mostly this distance metric between samples is based on a subset of
the genes measured using the microarrays. Particular metrics that
are frequently used are:
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 167
NN (percentage)
B (percentage)
abnormality
A
285 AML patients
Karyotype
FLT3TKD
FLT3ITD
CEBPA(53) 67 4
61 5
AML patients
FLT3 ITD(100) 88 6
67 7
8
inv(16)(100) 9
EV11(45) 10
78 11
t(15:17)(100) 12
t(8:21)(100) 13
14
CEBPA(63) 63 15
t(11q23)(45) 16
CD34
NBM
AML patients
Fig. 3 Unsupervised cluster analyses of 285 cases of primary AML. (a) Correlation view of 285 AML patients
(2856 probe sets) [19]. The Correlation visualization tool displays pair-wise correlations between the samples.
The cells in the visualization are colored by Pearson’s correlation coefficient values with deeper colors indicat-
ing higher positive (red) or negative (blue) correlations. The scale bar indicates 100 % correlation (red) towards
100 % anti-correlation (blue). One hundred percent anti-correlation would indicate that genes with high
expression in one sample would always have low expression in the other sample and vice versa. The red
diagonal displays the comparison of an AML patient with itself, i.e., 100 % correlation. In order to reveal cor-
relation patterns, a matrix ordering method is applied to rearrange the samples. The ordering algorithm starts
with the most correlated sample pair and, through an iterative process, sorts all the samples into correlated
blocks. Each sample is joined to a block in an ordered manner so that a correlation trend is formed within a
block with the most correlated samples at the center. The blocks are then positioned along the diagonal of the
plot in a similar ordered manner. (b) Adapted correlation view (2856 probe sets) of 285 AML patients (right
panel) and the expression levels of the top 40 genes defining the 16 individual clusters of patients (left panel).
FAB classification and karyotype based on cytogenetics are depicted in the columns along the original diagonal
of the correlation view (FAB M0-red, M1-green, M2-purple, M3-orange, M4-yellow, M5-blue, M6-grey; karyo-
type: normal-green, inv(16)-yellow, t(8;21)-purple, t(15;17)-orange, t(11q23)/MLL abnormalities-blue, 7(q)
abnormalities-red, +8-pink, complex-black, other-grey). FLT3 ITD, FLT3 TKD, N-RAS, K-RAS and CEBPA muta-
tions and EVI1 overexpression are depicted in the same set of columns (red bar: positive and green bar:
negative). (Reprinted with permission from Valk et al., Copyright 2004, Massachusetts Medical Society.)
3.7.3 Class Comparison Class comparison involves the discovery of differentially expressed
genes among different classes of samples. Analysis methods are
“supervised” when they include prior classification information.
This may be different cell or tissue types or experimental/treat-
ment conditions. For example, when looking at tissues of normal
breast and cancerous breast, the genes that are consistently differ-
entially expressed between them, may be involved in the initiation
or progression of cancer.
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 169
3.7.4 Class Prediction Class prediction is a supervised technique which allows the discov-
ery of genes that alone, or in combination can predict which class
a sample belongs to. Please note that the genes which most opti-
mally classify the samples are not necessarily the most differentially
expressed. These techniques are particularly useful in diagnostics as
specific profiles can be inferred, with the ability to predict subtypes
or which patient will be effectively treated.
Many classification algorithms have been developed during the
last decades. Most methods select, by means of cross-validation, an
appropriate subset of genes which most optimally classifies the
samples. To infer if the prediction algorithm works well the
researcher has to split his data set into a training set and test set.
The model is trained on the training set by means of cross-valida-
tion to select the most appropriate subset of genes and finally infers
the model accuracy on the test set. The prediction accuracy should
never be inferred from the training data from which the model is
constructed, as it will lead to overfitting and an over-estimation of
the prediction accuracy.
There are different methods to perform the classification, but
they can generally be divided into two groups when it comes to
gene selection.
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 171
3.8.1 Genotyping SNP microarrays grant the ability to infer the genotype of SNPs.
SNP arrays have been used en masse to determine genotypes for
large numbers of SNPs per individual (~900,000 for Affymetrix
SNP 6.0). After pre-processing and labeling the DNA the scanner
determines the signal intensities of the probes that are tilled over
the SNP, as illustrated in Fig. 2. Based on the signal intensities the
researcher can then determine if the SNP allele has the genotype;
major/major (AA), major/minor (AB), minor/minor (BB), and
noCall (undetermined genotype).
These genotype calls can be used to perform Genome-Wide
Association (GWAS) analysis to infer if a particular genotype of a
SNP or a combination of SNP’s is associated with the outcome,
e.g., disease. This in particular is very difficult due to issues of sta-
tistical power. Using microarrays we are generating more than
900,000 observations. To infer if there is an association between a
SNP and the outcome we need to perform a statistical test. Since
we are performing this test on all these SNPs we will need to per-
form multiple testing correction, as shown above. This will result
in strong p-value corrections, because the large number of mea-
sured SNPs, resulting in no statistical significant association
between the SNP and the outcome. This is one of the reasons why
these studies need many samples.
Genotyping of SNPs makes it also possible to infer Loss of
Heterozygosity (LOH). In cancer research this is of particular
interest since these regions may contain tumor suppressor genes
with equal or similar mutations on both alleles. In the case of
paired samples, e.g., normal–cancerous tissue or diagnostic–remis-
sion sample, this can easily be done by determining regions of
homozygous genotypes (AA or BB) in the case samples which are
not observed in the control sample. If no control sample is avail-
able the regions of LOH can be determined by hidden Markov
models (HMM).
Outline inference genotype:
Step 1: The Affymetrix software package, GCOS, always contains
the algorithms to determine the genotype calls. Also, the stand-
alone software package Affymetrix-Power-Tools (APT) [28] has
the ability to determine the genotype calls per sample.
Step 2: Using these algorithms, it is best to determine the genotype
calls in a large batch of microarrays, as these algorithms are strongly
dependent on using the distribution of signal intensities over all
samples to optimally determine the genotype calls.
Genome-Wide Gene Expression Profiling, Genotyping, and Copy Number Analyses… 173
3.8.2 Copy Number One vital, but very difficult, step in SNP microarray analysis is
Analysis determining the copy number variations (CNVs). In particular
types of cancer genomic regions that are recurrently deleted or
amplified could pinpoint towards tumor suppressor genes or onco-
genes. Determining CNVs is by far the most difficult aspect of
analyzing SNP arrays.
There are multiple algorithms to determine CNVs from SNP
arrays, such as CNAG [29] and dChip [16]. In this section we will
focus on determining the CNVs using dChip. It is imperative,
especially for determining CNVs, to have a correct experimental
design. To optimally determine the CNVs per sample there must
be an appropriate reference batch (e.g., multiple samples) from
which the signal distribution per SNP can be determined under the
assumption that the reference has a normal copy number, i.e., dip-
loid. In a particular case it could happen that one or more samples
in the reference batch are not diploid for a particular individual
SNP. Even in this case the algorithm can accurately estimate the
copy number of a sample, and will work as long as this does not
occur in a too large proportion of reference samples.
Additionally, these reference samples should not be selected
from an online database. Sources of variation, such as different
methods of DNA isolation, and batch effects will result in very bad
estimations of CNVs. The most appropriate way to generate your
reference batch is to run some SNP arrays for normal karyotype,
remission, or healthy samples. Most effective would be to also have
a 1:1 male–female ratio in the reference batch to accurately esti-
mate CNVs for the X and Y chromosomes.
The outline for CNVs estimation in dChip [16]:
Step 1: Construct an appropriate experimental design. Determine
your reference set on the basis of DNA quality, expected normal
copy number (remission or healthy samples), and similar DNA iso-
lation and pre-processing methods.
Step 2: Gather all necessary files to load your SNP microarrays into
dChip. Next to your .CEL files generated by the scanner, you will
need a CDF file (library file describing the array) from the
Affymetrix Web site (select your array type), and a patient info file
describing which sample belongs to the reference batch and
gender.
Step 3: When all samples are loaded one should check if none of the
samples contain irregularities. It is possible to visualize the signal
intensities of the array in dChip. This grants the ability to look for
artifacts, such as blobs or scratches, resulting in wrongly measured
signal intensities. In the case a reference samples shows these arti-
facts, then it should be removed from the reference batch.
Step 4: Normalize to a reference sample. Just like for gene expres-
sion, microarrays the signal intensity distribution must be made
174 Mathijs A. Sanders and Peter J.M. Valk
4 Notes
References
1. http://www.affymetrix.com genome-wide expression patterns. Proc Natl
2. http://www.home.agilent.com/agilent/home Acad Sci USA 95(25):863–868
3. http://www.illumina.com 19. Valk PJM, Verhaak RGW, Beijen MA,
4. Millenaar FF et al (2006) How to decide? Erpelinck CAJ, Barjesteh van Waalwijk van
Different methods of calculating gene expres- Doorn-Khosrovani S, Boer JM, Beverloo HB,
sion from short oligonucleotide array data will Moorhouse MJ, van der Spek PJ, Löwenberg
give different results. BMC Bioinformatics B, Delwel R (2004) Prognostically useful
7:137 gene-expression profiles in acute myeloid leu-
kemia. N Engl J Med 350:1617–1628
5. Lipshutz RJ et al (1999) High density syn-
thetic oligonucleotide arrays. Nat Genet 21(1 20. Suzuki R, Shimodaira H (2006) Pvclust: an R
Suppl):20–24 package for assessing the uncertainty in hierar-
chical clustering. Bioinformatics 22(12):
6. Lipshutz RJ (2000) Applications of high- 40–42
density oligonucleotide arrays. Novartis Found
Symp 229:84–90, discussion 90–3 21. Smyth GK (2004) Linear models and empiri-
cal Bayes methods for assessing differential
7. Burnett A, Wetzler M, Lowenberg B (2011) expression in microarray experiments. Stat
Therapeutic advances in acute myeloid leuke- Appl Genet Mol Biol 3(1):Article 3
mia. J Clin Oncol 29(5):487–494
22. Holm S (1979) A simple sequentially rejective
8. Marcucci G, Haferlach T, Dohner H (2011) multiple test procedure. Scand J Stat
Molecular genetics of adult acute myeloid leu- 6(2):65–70
kemia: prognostic and therapeutic implica-
tions. J Clin Oncol 29(5):475–486 23. Benjamini Y, Hochberg Y (1995) Controlling
the false discovery rate: a practical and power-
9. Wouters BJ, Löwenberg B, Delwel R (2009) A ful approach to multiple testing. J R Stat Soc
decade of genome-wide gene expression pro- Ser 57(1):289–300
filing in acute myeloid leukemia: flashback and
prospects. Blood 113(2):291–298 24. Tusher VG, Tibshirani R, Chu G (2001)
Significant analysis of microarrays applied to
10. http://media.af fymetrix.com/suppor t/ the ionizing radiation response. Proc Natl
downloads/manuals/expression_analysis_ Acad Sci USA 98(9):5116–5121
technical_manual.pdf
25. Goeman JJ, van der Geer SA, de Kort F, van
11. http://media.af fymetrix.com/suppor t/ Houwelingen HC (2004) A global test for
downloads/manuals/genomewidesnp6_man- groups of genes: testing association with a clin-
ual.pdf ical outcome. Bioinformatics 20(1):93–99
12. Harmer SL, Kay SA (2000) Microarrays: 26. Tibshirani R (1996) Regression shrinkage and
determining the balance of cellular transcrip- selection via the lasso. J R Stat Soc Ser
tion. Plant Cell 12(5):613–616 58(1):267–288
13. WB van Leeuwen, C Vink (2009) Molecular 27. http://www.r-project.org/
Diagnostics—Techniques & Applications. IVA
Groep B.V. Rotterdam, the Netherlands. 28. http://www.affymetrix.com/partners_pro-
ISBN 978-90-6464-340-8 grams/programs/developer/tools/pow-
ertools.affx
14. Clarke JD, Zhu T (2006) Microarray analysis
of the transcriptome as a stepping stone 29. Nannya Y, Sanada M, Nakazaki K, Hosoya N,
towards understanding biological systems; Wang L, Hangaishi A, Kurokawa M, Chiba S,
practical considerations and perspectives. Plant Bailey DK, Kennedy GC, Ogawa S (2005) A
J 45(4):630–650 robust algorithm for copy number detection
using high-density oligonucleotide single
15. Wu Z, Irizarry RA, Gentleman R, Martinez- nucleotide polymorphism genotyping arrays.
Murillo F, Spencer F (2004) Model-based Cancer Res 65:6071–6079
background adjustment for oligonucleotide
expression arrays. J Am Stat Assoc 99:909 30. Sanders MA, Verhaak RGW, Geertsma-
Kleinekoort WM, Abbas S, Horsman S, van der
16. http://www.dchip.org Spek PJ, Löwenberg B, Valk PJM (2008)
17. http://www.omniviz.com SNPExpress: integrated visualization of genome-
18. Eisen MB, Spellman PT, Brown PO, Botstein wide genotypes, copy numbers and gene expres-
D (1998) Cluster analysis and display of sion levels. BMC Genomics 25(9):41
Chapter 11
Abstract
Pharmacoepigenetics is an emerging field, which can be studied by several approaches. Addressing DNA
methylation status of drug-metabolizing enzymes and transporters (DMET) is challenging and might
provide answers in relation to interindividual differences in pharmacokinetics and pharmacodynamics.
Studying genetic variation in DMET genes in relation to drug response has been the main focus of phar-
macogenetics laboratories; it is, however, expected that epigenetic modifications will play a role in drug
responses as well. Some of the variations in drug-responses cannot be explained by genetic variation in
DMET genes. For those particular genes it might be interesting to examine the DNA methylation status
in relation to pharmacokinetics. In this chapter we discuss the methods available and provide a protocol to
quantify DNA methylation status of CpG sites in candidate genes, which can readily be applied to most
pharmacogenetics laboratories. In addition, we provide details about optimization and validation of the
method in terms of technical specificity and technical sensitivity and precision of the method.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_11, © Springer Science+Business Media, LLC 2013
179
180 Sandra G. Heil
1.1.1 Bisulfite Treatment Treatment of genomic DNA with sodium bisulfite converts
unmethylated cytosines into uracil remaining methylated cytosines
as cytosines. The bisulfite-converted genomic DNA is then sub-
jected to PCR, in which uracil residues will be amplified as thymine-
residues and methylated cytosines as cytosines, enabling simple
discrimination by detection techniques like Sanger sequencing [9].
Bisulfite treatment can be performed by the initial protocol
described by Frommer et al. [9]. However, also bisulfite modifica-
tion kits are available that enable high-throughput bisulfite-
modification and provide good quality results (e.g., Zymo
Research). Importantly, a control reaction should be performed to
assess whether all unmethylated cytosines are modified into thy-
mines after PCR. Inefficient bisulfite-modification might result in
false-positive calls due to incomplete conversion of unmethylated
cytosines into uracils.
Fig. 1 Detection of DNA methylation by generally applied PCR-based quantitative methods. Genomic DNA is
treated with bisulfite which modifies unmethylated cytosines into uracil leaving methylated cytosines in a CpG
content unchanged. After PCR the uracils will be amplified as thymines. Several quantitative detection meth-
ods can be applied to discriminate between cytosines and thymines (e.g., cloning followed by Sanger sequenc-
ing, sequencing-by-synthesis based approaches such as Pyrosequencing®, mass spectrometry by epiTYPER™,
and real-time quantitative PCR based approaches such as MethyLight)
2 Materials
2.1 Control DNA Unmethylated and methylated human DNA from Zymo Research
(Cat No. D5014, ZymoResearch, BaseClear Leiden, The
Netherlands) can be used as control DNA during bisulfite treat-
ment. This bisulfite-treated control DNA is subsequently used as
control DNA in the PCR reaction.
2.2 Bisulfite ●● Zymo EZ DNA methylation direct kit (Cat. No. D5021,
Modification Zymo Research).
●● Eppendorf microcentrifuge tubes 1.5 and 2.0 mL.
●● Absolute ethanol (Cat.No. 1.00983.2500, Merck).
3 Methods
3.1 DNA Isolation High-quality genomic DNA is preferred for quantification of DNA
methylation. Isolation can be performed by several protocols as
long as the A260/280 ratio and the A260/230 ratio are around
1.8. DNA quality can be checked by an UV spectrophotometer,
for example with the NanoDrop (NanoDrop, Thermo Scientific).
3.2 Bisulfite Control DNA (unmethylated and methylated) and sample DNA is
Modification treated with bisulfite using the Zymo EZ DNA methylation direct
kit (Cat. No. D5021, Zymo Research). Reagents are prepared
according to the manufacturer’s instructions.
Epigenetic Techniques in Pharmacogenetics 183
Recovery =
[ Amount of DNA after bisulfite treatement (ng)] × 100 %
[ Amount of DNA used in bisulfite treatement (ng)]
3.3 Real-Time qPCR can be used to quantify the amount of cytosines and thymines
Quantitative PCR in the GOI. This method was originally described by Laird and
coworkers as MethyLight [10, 13]. qPCR conditions should be
optimized for the GOI and for the reference gene. We provide a
general protocol to optimize the qPCR assay including PCR effi-
ciency, validation and calculation of percentage methylated refer-
ence (PMR) [13] but do not provide sequence specific information
like primer and probe sequences as this is dependent upon the GOI.
1. Design primers based at the bisulfite-converted sequence of the
GOI and of the reference gene. We generally apply MethPrimer
software [14]. Choose primers for bisulfite-sequencing to
obtain primers that do not contain CpG sites (see Note 3).
2. Design a probe specific for methylated cytosines within the
GOI. We generally apply Taqman probes labelled with FAM
and a Black Hole Quencher (BHQ-1).
3.
Design a probe specific for a bisulfite-treated reference
sequence to control for amount of input DNA. See Note 4.
We generally apply Taqman probes for beta-actin labelled with
FAM-BHQ1.
4.
Optimize the PCR reaction using standard protocols and
chemicals using 1–5 μL of bisulfite-treated control DNA. Run
each reaction in triplicate.
5.
After optimization and validation (see Subheading 3.3.1–
3.3.3) run each sample in triplicate for each gene (monoplex).
Run standard curves in triplicate for GOI and reference gene
on each plate (see Note 5).
3.3.1 PCR Efficiency 1. Make a five-times dilution series of the methylated control
DNA (e.g., we prefer dilution series of undiluted and 5, 25,
125, 625, and 3,125 times dilution).
2. Run the optimized PCR protocol for both the GOI and the
reference gene at the ABI Prism 7000 Sequence detection
system.
3. Plot the threshold cycle (CT value) against the log of the dilu-
tion factor and calculate the coefficient of determination (R2)
and slope using linear regression (e.g., Excel or Analyse-it).
PCR efficiency can be calculated by the following formula
[15] (see Note 6):
4 Notes
1. Use a new tube for each step and use 1.5 mL or 2.0 mL
Eppendorf tubes instead of the original collection tubes.
2. Bisulfite-treated DNA resembles the characteristics of RNA
(e.g., contains uracil and is single-stranded). For that reason
we apply the RNA-40 factor in UV-spectrophotometry to cal-
culate the concentration (i.e., 1 OD260 Unit = 40 ng/μL
bisulfite-treated DNA).
3. Universal primers in combination with a probe containing one
or multiple CpG sites results in quantification of one to five
CpG sites at once depending on the sequence of the GOI.
A disadvantage of this approach is that quantification of meth-
ylation status of a single CpG is difficult to obtain due to the
presence of multiple CpG sites within the probe sequence.
Other methods such as (pyro)sequencing are available for
quantification of single CpG sites.
4. A frequently used reference gene used for quantification of
methylation is beta-actin. However, as a reference gene is
applied to control for the amount of input DNA, each gene
can theoretically be chosen as long as multiple copies of the
gene have not been described. The primers and probe of the
reference gene do not contain CpG sites and thus are specific
Epigenetic Techniques in Pharmacogenetics 187
Acknowledgment
References
1. Russo VEA, Martienssen RA, Riggs AD (1996) 7. Kristensen LS, Hansen LL (2009) PCR-based
Epigenetic mechanisms of gene regulation. Cold methods for detecting single-locus DNA meth-
Spring Harbor Press, Cold Spring Harbor, NY ylation biomarkers in cancer diagnostics,
2. Chen ZX, Riggs AD (2011) DNA methylation prognostics, and response to treatment. Clin
and demethylation in mammals. J Biol Chem Chem 55:1471–1483
286:18347–18353 8. Feinberg AP, Vogelstein B (1983) Hypomethy
3. Gomez A, Ingelman-Sundberg M (2009) lation distinguishes genes of some human can-
Pharmacoepigenetics: its role in interindividual cers from their normal counterparts. Nature
differences in drug response. Clin Pharmacol 301:89–92
Ther 85:426–430 9. Frommer M et al (1992) A genomic sequenc-
4. Ingelman-Sundberg M, Sim SC, Gomez A, ing protocol that yields a positive display of
Rodriguez-Antona C (2007) Influence of cyto- 5-methylcytosine residues in individual
chrome P450 polymorphisms on drug thera- DNA strands. Proc Natl Acad Sci USA 89:
pies: pharmacogenetic, pharmacoepigenetic and 1827–1831
clinical aspects. Pharmacol Ther 116:496–526 10. Eads CA et al (2000) MethyLight: a high-
5. Shen L, Waterland RA (2007) Methods of throughput assay to measure DNA methyla-
DNA methylation analysis. Curr Opin Clin tion. Nucleic Acids Res 28:E32
Nutr Metab Care 10:576–581 11. Colella S, Shen L, Baggerly KA, Issa JP, Krahe
6. Laird PW (2010) Principles and challenges of R (2003) Sensitive and quantitative universal
genomewide DNA methylation analysis. Nat Pyrosequencing methylation analysis of CpG
Rev Genet 11:191–203 sites. Biotechniques 35:146–150
188 Sandra G. Heil
Abstract
Since the human genome has been fully sequenced, and presence of single nucleotide polymorphisms
(SNPs) appeared abundant, many studies are associating SNPs with clinical response or even with disease.
For some diseases or drug treatments these associations are clear, so that genetic screening for such SNPs
or mutations is a standard procedure. For that reason, many different techniques have been developed for
fast and easy screening for such specific SNPs/mutations. For reliable screening, the use of controls with
known genotypes is indispensable. Plasmids are an ideal tool for making controls which can serve as an
inexhaustible source, making new validation superfluous.
In this chapter we describe how plasmid controls can be made using DNA with a heterozygous
genotype, and also from DNA of which only one allele is available.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_12, © Springer Science+Business Media, LLC 2013
189
190 Tahar van der Straaten and Henk-Jan Guchelaar
Xmnl 2009
T7 1 start
Scal 1890 Nael 2707 Apal 14
Aatll 20
f1 ori Sphl 26
BstZl 31
Ncol 37
BstZl 43
Ampr Notl 43
pGEM -T Easy lacZ Sacll 49
EcoRl 52
(3015bp)
Spel 64
EcoRl 70
Notl 77
BstZl 77
Pstl 88
ori Sall 90
Ndel 97
Sacl 109
1473VA05_6A
BstXl 118
Nsil 127
141
SP6
Fig. 1 pGEM-T easy vector from Promega. Within the Lac-operon the multiple
cloning site is opened and a thymidine is added at the 3′-ends
Plasmid Derived External Quality Controls for Genetic Testing 191
2 Materials
2.1 Generation In order to establish plasmid controls that can be used for several
of Plasmid Controls genotyping techniques, we suggest primers that are located about
500 nucleotides up and downstream of the SNP. As an example we
took DPYD gene rs3918290 (Fig. 2) to insert into Promega’s
pGEM-Teasy vector. Described below are two approaches, (1)
using a DNA sample that has been previously genotyped as hetero-
zygous. In this example the DNA sample was genotyped heterozy-
gous for rs3918290 by means of Taqman analysis (Lifetech,
Nieuwerkerk aan de IJssel, The Netherlands) and confirmed by
pyrosequencing (Qiagen, Venlo, The Netherlands), (2) Insert the
mutation at the SNP site in case of low minor allele frequency and
only one genotype is available.
Of note, for testing genotyping methods or validation of meth-
ods it is advised to use a standard control DNA panel of healthy
volunteers. This can be commercially available reference material
(i.e., from Coriell [13]); Gentris [14]; or GeT-RM cell lines col-
lected by the CDC [15], or, as we did, we took 94 blood samples
from blood-donors who gave informed consent for research use.
Fig. 2 DPYD exon 14 deletion. SNP rs3918290 (C/T) is shown in bold and capital. In bold and underlined for-
ward and reverse PCR are primers are shown
Plasmid Derived External Quality Controls for Genetic Testing 193
3 Methods
3.1 Generate 1. Perform regular PCR as follows: in one reaction tube add
Genotype 5 pmol of PCR primers forward and reverse, 10 μl Hotstar
Control Using mastermix, 10 ng of chromosomal DNA and add sterile water
Heterozygous DNA to a total volume of 20 μl.
2. Run a standard PCR program as follows: 15 min at 95 °C, 30
cycles of 95 °C—55 °C—72 °C for 30 s—30 s—60 s, respec-
tively, followed by a final extension at 72 °C for 10 min (impor-
tant for the addition of Adenosine at 3′-ends).
3. Analyze PCR product by gel electrophoresis.
4. Insert the PCR product into pGEM-T easy vector as follows:
in one tube at 3 μl PCR product, 1 μl pGEM-T vector, 5 μl
ligase buffer, and 1 μl ligase. Incubate for at least 2 h at room
temperature.
5. Defreeze competent E. coli cells and add to the ligation mix-
ture. Follow the procedure as described for these cells which
depends on the way how they are prepared, for example heat
shock or electroshock (see Note 2 and 4).
6. pGEM-T vector allows blue/white screening when comple-
mentary competent cells are used and substrate is added to the
growth plate. pGEM-T that has no insert will give blue colo-
nies, whereas an insertion will disturb the LacZ gene and yields
white colonies. Using heterozygous DNA, theoretically, 50 %
of (white) colonies will contain allele 1 and the other 50 %
allele 2 (Fig. 3). Grow four colonies and next day isolate plas-
mid DNA by standard methods [12] and check for genotype.
Usually, about 50 pg of plasmid in a PCR reaction is sufficient
for standard genotyping methods.
3.2 Generate Since one allele is available, the other allele has to be created. This
Genotype Control can be done by PCR where one primer (forward in this example)
Using DNA of Which is overlapping the SNP/mutation and contains the mutant nucleo-
One Allele Is Available tide at that position (Fig. 4). The resulted PCR product will con-
tain the mutation and can be used as a primer with the original
forward primer (Fig. 5).
1. Perform regular PCR as follows: in one reaction tube add
5 pmol of PCR primers mutant forward and reverse, 10 μl
Hotstar mastermix, 10 ng of chromosomal DNA (add sterile
water to a total volume of 20 μl).
2. Run a standard PCR program as follows: 15 min at 95 °C, 30
cycles of 95 °C—55 °C—72 °C for 30 s—30 s—60 s, respec-
tively, followed by a final extension at 72 °C for 10 min.
3. Analyze PCR product by gel electrophoresis (see Note 3).
194 Tahar van der Straaten and Henk-Jan Guchelaar
C T A C A
A A A C
A G A G A
C A
A
T A C A T A A G
G A A A G A A
T A
A A T A C A T A
T A A A G A A T A
A
A A T A C A C A A A
A A A G A G T
T C T C A
A A A A A A
A A A G A A A G
C A T A C A
C AA G C A A A A G
A G A G T A
T A A A
C A A A T A
A G A A
C A T A
TA TA A
G T T
Fig. 3 Mixture of PCR products containing either a C or a T at the SNP site. Per plasmid only one PCR strand
can be inserted, thus either a C or a T
T
5’ C 3’
Primer design 3’ G 5’
3’ 5’
Annealing and 5’ 3’
elongation
T
5’ 3’
3’ G 5’
New strands 5’ T 3’
3’ A 5’
after number of 5’ T 3’
cycles 3’ A 5’
5’ T 3’
3’ A 5’
5’ T 3’
3’ A 5’
Fig. 4 Introduction of mutation in DNA. A primer is chosen with the nucleotide of interest at the SNP site. This
mutant primer will bind on the complementary strain and elongate. This new strand, containing a thymidine at
the SNP site is a template for the reverse primer in the next PCR cycle. The next cycle will use both new strands
to get double stranded PCR with the mutation inserted
3’ A 5’
5’ 3’
3’ 5’
Fig. 5 PCR product with the inserted mutation will serve as a reverse primer in combination with the original used
forward primer, yielding a PCR product as was described for Fig. 4 but with a T instead of a C at the SNP site
196 Tahar van der Straaten and Henk-Jan Guchelaar
4 Notes
References
1. Sanger F, Coulson AR (1975) A rapid method 3. van der Straaten T, Swen J et al (2008) Use of
for determining sequences in DNA by primed plasmid-derived external quality control samples
synthesis with DNA polymerase. J Mol Biol in pharmacogenetic testing. Pharmacogenomics
94:441–448 9:1261–1266
2. van der Heiden I, van der Werf M et al (2004) 4. van der Straaten T, van Schaik RH (2010)
Sequencing: not always the “gold standard”. Genetic techniques for pharmacogenetic analy-
Clin Chem 50:248–249 ses. Curr Pharm Des 16:231–237
Plasmid Derived External Quality Controls for Genetic Testing 197
Abstract
A growing number of noncoding variants are found to influence the susceptibility to common diseases and
interindividual variation in drug response. However, the mechanisms by which noncoding variation affects
cellular and clinical phenotypes remain to be elucidated. Allele-specific assays allow testing directly the dif-
ferential properties of the alleles at a regulatory variant, which are detected as an allelic imbalance. Two
widely used allelic imbalance assays target cDNA and DNA from chromatin immunoprecipitation (ChIP)
experiments, and therefore revealing allele-specific gene expression and transcription factor binding,
respectively. The throughput of allelic imbalance assays ranges from single variant to the genome scale,
which are made possible by the recent advances in genotyping and sequencing technologies (e.g., genome-
wide quantitative cDNA genotyping, ChIP-seq).
Key words Polymorphism, Chromatin immunoprecipitation, RNA, cDNA, Quantitative PCR, Gene
expression
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_13, © Springer Science+Business Media, LLC 2013
201
202 Francesca Luca and Anna Di Rienzo
2 Materials
2.1 Cell Culture The following protocol uses lymphoblastoid cell lines (LCLs).
However, allelic imbalance assays can be performed also in other
cell lines as well as primary cells. Suggestions on how to modify the
protocol when using different cell types are provided throughout
the text.
1. LCLs from individuals carrying the heterozygous genotype at
the SNP(s) of interest (see Note 1).
2. RPMI 16490 (Gibco), supplemented with 15 % FBS and
0.1 % Gentamycin.
ChIP and RNA/cDNA Allelic Imbalance 203
2.2 Preparation of In the current protocol the Upstate (Millipore) ChIP Assay Kit
the Sample for Allelic reagents are used; however, details to prepare the reagents are also
Imbalance (AI) provided.
2.2.2 Preparation of the 1. LCLs in mid-log exponential phase (Approx total RNA yield:
Sample for RNA/cDNA AI 3 μg/106 cells).
2. Qiagen RNeasy Plus mini kit.
3. High-Capacity cDNA Reverse Transcription Kits from Applied
Biosystems.
2.3 AI Assay 1. Taqman® Universal PCR Master Mix with no AmpErase® UNG.
2. Taqman® 40× SNP Genotyping Assay.
3 Methods
3.1 Cell Culture This method describes the protocol for LCLs grown in suspension;
however, other cell types can be used (see Note 2). LCLs are seeded
at 0.5 × 106 and, once in mid-log exponential phase, may be stimu-
lated according to experimental design (for example with dexa-
methasone) (see Note 3) prior to harvesting.
3.2 Preparation of Alternative ChIP protocols are successfully used in other laborato-
the Sample for AI ries and can replace the one described here.
3.2.1 Preparation Formaldehyde Cross-linking
of the Sample for ChIP AI
1. In the tissue culture hood, add formaldehyde to culture flasks
(final concentration of 1 %). Swirl and incubate for 20 min at
37 °C in the tissue culture incubator (see Note 4).
2. Add 1.25 M (10×) Glycine to the flasks, to a final concentra-
tion of 0.125 M (1×). Glycine quenches the cross-linking
reaction. Swirl and incubate at room temperature for 5 min.
3. Meanwhile, add protease inhibitors to PBS (need about 20 ml
PBS per 75 cm2 flask) and cool the centrifuge to 4 °C.
4. Collect the cells by centrifugation at 290 × g for 7 min at 4 °C.
5. Wash cells twice with 10 ml of ice-cold PBS containing prote-
ase inhibitors.
6. Cell pellet can be stored at −80 °C.
ChIP and RNA/cDNA Allelic Imbalance 205
22. Pellet agarose (5,000 × g for 1 min or less at 4 °C) and place
supernatant in new tubes.
23. Remove 20 μl for the input control sample and store at −80 °C.
24. Add antibody (1–5 μg depending on antibody of choice) to
the supernatant and incubate overnight at 4 °C with rotation.
25. Add 45 μl Salmon Sperm DNA/Protein A Agarose Slurry for
1 h at 4 °C with rotation.
26. Pellet agarose (5,000 × g for 1 min at 4 °C). Aspirate out the
supernatant and wash for 3–5 min on a rotating platform with
1 ml of each of the buffers listed in the order given below:
(a) Low-Salt Immune Complex Wash Buffer, one wash
(b) High-Salt Immune Complex Wash Buffer, one wash
(c) LiCl Immune Complex Wash Buffer, one wash
(d) 1× TE, two washes
27. Meanwhile, prepare the Elution Buffer.
28. Elute by adding 250 μl elution buffer to the pelleted agarose/
antibody/protein complex. Vortex briefly and incubate at room
temperature for 15 min with rotation. Spin down the agarose,
and carefully transfer the supernatant fraction to another tube
and repeat elution. Combine eluates (total volume ~ 500 μl).
29. Thaw the input control samples.
30. Add 20 μl 5 M NaCl (2 μl for input) to the 500 μl eluates and
reverse cross-links by heating at 65 °C for 4 h or overnight (see
Note 8). One may store the sample at −20 °C and continue
the next day.
31. Add 1 μl RNase to each sample and incubate at room tempera-
ture for 30 min.
32. Add 10 μl 0.5 M EDTA (1 μl for input), 20 μl 1 M Tris–HCl
(2 μl for input), pH 6.5, and 2 μl of 10 mg/ml Proteinase K
(1 μl for input) to the combined eluates and incubate for 1 h
at 45 °C.
33. Recover DNA with the Qiagen PCR Purification Kit, follow-
ing the manufacturer’s instructions.
34. Before performing AI Assays on the ChIPed DNA, the quality
of the ChIP experiment should be assessed by performing
quantitative real time PCRs targeting a positive and a negative
control region.
3.2.2 Preparation of the 1. Extract total RNA with the Qiagen RNeasy Plus mini kit fol-
Sample for RNA/cDNA AI lowing the manufacturer’s protocol.
ChIP and RNA/cDNA Allelic Imbalance 207
2. Synthesize cDNA from total RNA (100 ng) using the High-
Capacity cDNA Reverse Transcription Kit (Applied Biosystems,
Foster City, CA) according to the manufacturer’s protocol.
Dilute cDNA samples 1:30 to perform AI assays.
3.3 AI Assay While ChIP AI assays target directly the candidate binding variant
of interest, cDNA AI assays are designed to target a coding SNP in
the gene that is differentially regulated by the two alleles at the
candidate regulatory variant.
1. The following criteria should be used to select a coding SNP
to be assayed in a cDNA AI assay:
(a) High linkage disequilibrium with the regulatory variant
(if phased genotype data including the regulatory SNP are
not available (see Note 9))
(b) High heterozygosity
(c) >40 bp away from exon boundary to allow designing
assays that will amplify both gDNA and cDNA thus
allowing for the use of a gDNA standard curve
2. In most cases, predesigned TaqMan genotyping assays are
available from Applied Biosystems. Alternatively, custom made
assays can be designed using the Custom TaqMan Assay
Design Tool.
3. Quantitative real-time PCR assays can be performed in either
96- or 384-well plates using any of the ABI systems (e.g., the
ABI PRISM 7900HT Sequence Detection System or the ABI
StepOnePlus™ Real-Time PCR System). Reactions are typi-
cally run in triplicates for each sample.
PCR mix for a sample run on a 96-well plate
Total volume: 20 μl
cDNA/ChIPed DNA: 4 μl
Taqman® Universal PCR Master Mix with no AmpErase®
UNG: 10 μl
Taqman® 40× SNP Genotyping Assay: 0.5 μl
Use Applied Biosystems standard recommended PCR cycling
conditions.
4. To account for differences between the two fluorochromes, a
standard curve should be built for each of the two alleles using
serial dilutions (see Note 10) of genomic DNA from an
individual heterozygous at the assayed SNP (Fig. 1). PCR
products are quantified for each allele separately in each
reaction and ratios between the two different alleles can be
calculated (Fig. 2). The results can then be averaged across
PCR replicates.
208 Francesca Luca and Anna Di Rienzo
Standard Curve
37
A
36
35 G
Input DNA
34
33
32
31
CT
30
29
28 IP DNA
27
26
25
24
Fig. 1 Example of the results from a ChIP AI assay at a regulatory variant for the
SGK1 gene [14]. The samples are plotted over the standard curves built for the
two alleles separately. An imbalance in the ChIPed DNA can be observed
4 Notes
Fig. 2 Example of the results of a cDNA AI assay targeting a coding SNP in link-
age disequilibrium with an interaction eQTL at the LSG1 gene [15]. The assay
was performed on samples cultured in two different conditions (with and without
dexamethasone). In this assay, the natural log-ratio between the two different
alleles was calculated and quantile normalized in each treatment condition sep-
arately. Two PCR replicates were performed and the results were averaged.
A significant difference between heterozygotes and homozygotes at the candi-
date regulatory variant in the presence of dexamethasone was observed
(p = 8.38 × 10−5). In each box, the horizontal line represents the median and the
whiskers represent the first and third quartile
Acknowledgments
References
Abstract
Genome-wide association (GWA) studies have identified thousands of genetic variants that contribute to
disease and pharmacologic traits. More recently, high-throughput sequencing studies promise to provide
a more complete catalog of genetic variants with roles in human phenotypic variation. Yet, characterizing
the influence of functional variants on genes, RNAs, proteins, and ultimately disease or pharmacologic
traits is a critical challenge for a vast majority of the implicated susceptibility loci. Here we describe SCAN,
a bioinformatics resource we have developed to elucidate the functional consequences of genetic variants
identified by genome-wide scans. In particular, this public resource implements a systems biology approach
to pharmacogenomic discovery.
Key words eQTLs, Pharmacogenomics, Expression profiling, Transcriptome, SNP function, Genetic
variation
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_14, © Springer Science+Business Media, LLC 2013
213
214 Eric R. Gamazon et al.
2 Features
2.1 Genetic The cell lines from the International HapMap Project [8] have
Variation: SNPs been the most frequently used for in vitro studies of gene expres-
and CNVs sion and pharmacologic phenotypes [6, 9, 10]. The cell lines are
Epstein-Barr virus transformed lymphocytes derived from appar-
ently healthy individuals of different ancestry. The HapMap LCLs
have been extensively utilized as a model for pharmacogenomic
studies, most notably on cellular toxicities of anticancer agents.
HapMap samples in 30 CEU (Utah residents with ancestry from
Northern and Western Europe) trios and in 30 YRI (Yoruba in
Ibadan, Nigeria) trios comprised the initial genotype dataset in
SCAN. Studies that have been conducted on these samples have
demonstrated that gene expression and drug response phenotypes
such as cytotoxicity are heritable and include an appreciable genetic
component. Extensive genotypic data are available on these samples
A Systems Biology Approach to Pharmacogenomics 215
2.2 Integration Given the proliferation of databases and resources for genomic
of Functional studies, it was felt that there was a need for consolidating publicly
Annotations of Genetic available functional annotations for loci emerging from genome-
Variation wide studies. We were particularly interested in developing novel
tools relevant to large-scale, whole-genome studies aimed at char-
acterizing susceptibility loci for complex traits. Functional classifi-
cation of variants is one of the primary challenges of such studies for
the purposes of prioritization for follow-up studies and of providing
mechanistic hypotheses for observed associations between SNPs
and disease or drug response. A database to assist in the identifica-
tion of the functional consequences or potential biological impact
of genetic variants from genome-wide scanning was a primary aim
in the development of SCAN.
A priori information on how genetic variation may impact
biological processes or molecular function may aid in the interpre-
tation of results from GWA studies. Indeed, GWA analyses
restricted to variants with increased probabilities of association may
be utilized to reduce the number of tests performed and increase
the power to detect susceptibility loci. Of course, many genetic
variants have no known functional effect on disease or therapeutic
outcome in humans. Furthermore, determining the influence of
genetic variation on genes, RNAs, or proteins, and ultimately disease
or drug response is a challenge of enormous complexity. SNPs
located in coding regions may be silent or synonymous, resulting in
no change in the gene product. A missense variant is clearly a useful
marker for disease. For example, a missense polymorphism causes a
change in amino acid from a glutamine in a normal hemoglobin
gene to a valine in a sickle cell hemoglobin gene; as a result, a
216 Eric R. Gamazon et al.
homozygote individual for the sickle cell allele develops sickle cell
anemia. On the other hand, a sickle cell homozygote individual or
a heterozygote individual at the hemoglobin gene locus may
develop better resistance to malaria. A nonsense variant results in a
premature termination codon. This type of polymorphism is
responsible for at least some forms of such diseases as Duchenne
Muscular Dystrophy (characterized by a damaged dystrophin gene)
and Cystic Fibrosis (caused by mutations in the CFTR gene).
On the other hand, nonsense-mediated decay (NMD) provides a
cellular machinery for detecting nonsense mutations and prevent-
ing the expression of an aberrant protein. A frameshift interferes
with the triplet nature of gene expression and causes a change in
the reading frame of the codons (thus, a change in the resulting
translation). A frameshift mutation in NOD2 has been shown to be
associated with Crohn’s disease [17, 18]. NOD2 is an outstanding
candidate for inflammatory bowel disease (IBD), as tumor necrosis
factor signaling and nuclear factor (NF) κB activation in mononu-
clear cells are critical for IBD pathophysiology. Yet the frameshift in
NOD2 was found to be associated solely with Crohn’s disease, and
not with ulcerative colitis. SCAN annotates a coding variant with
information on how it exerts its functional effect on its host gene by
altering the gene product.
An SNP may have other effects on biological molecules.
A transcript may contain variation in a region that is not translated.
An SNP in a 5′ untranslated (5′ UTR) region may alter a binding
site for a protein, thus affecting mRNA stability; alternatively, it may
promote or inhibit the initiation of translation. A recent study has
shown that 5′ UTR SNPs in nuclear transcripts encoding both mito-
chondrial and secreted proteins may influence gene regulation at the
level of mRNA export [19]. Similarly, an SNP in a 3′ untranslated
(3′ UTR) region may affect the binding site of a microRNA
(miRNA), a posttranscriptional regulator that binds to the target
transcript to induce translational repression or gene silencing.
Dysregulation of miRNAs has been associated with higher tumor
proliferation in human epithelial ovarian cancer [20] and with the
etiologies of cardiovascular diseases [21] and of a psychiatric pheno-
type [22]. SCAN annotates noncoding SNPs with information on
location in the genome relative to the most proximal transcript.
3 Implementation
SCAN provides a set of query tools (see Fig. 2) using the following
interfaces, which are all available in batch query mode:
1. An SNP Query that returns physical and functional annotation,
host and flanking genes, and genes whose expression levels are
predicted by the variant, at a user-specified P-value threshold.
2. A Gene Query that retrieves all variants within and up to a
user-specified distance (in kilobases) of the gene, maps the gene
218 Eric R. Gamazon et al.
Fig. 1 Platform coverage. SCAN enables the interpretation of results from GWA studies by assessing, for a
given set of SNPs, the coverage of high-throughput genotyping platforms relative to a reference panel
Fig. 2 Functional annotation. SCAN provides a set of interfaces for annotating SNPs, genes, genomic regions
and CNVs with high-throughput molecular phenotypes (e.g., gene expression)
4 Applications
4.1 eQTLs A GWA study of childhood onset asthma [29] implicated ORMDL3
for Known Asthma as a susceptibility gene. Multiple SNPs located on chromosome
Susceptibility Gene 17q21 have been found to be strongly and reproducibly associated
with risk of disease [30]. ORMDL3 is a gene that encodes a trans-
membrane protein anchored in the endoplasmic reticulum.
Particularly, genetic variants regulating ORMDL3 expression in cis
were hypothesized to be determinants of disease susceptibility
[29]. We sought to identify regulators of this gene in LCLs to
validate the initial findings.
220 Eric R. Gamazon et al.
Protocol:
(a) In SCAN’s Gene Query tool, enter the gene of interest
(ORMDL3). SCAN queries can also be done in batch mode by
entering or uploading a list, but we restrict the present analysis
to this particular gene.
(b) Select “include gene start, end, and chromosome” option.
(c) Select “include SNPs that predict expression with p-value less
than” and enter 0.0001 as SNP eQTL p-value threshold.
(d) Select “include CNVs that predict expression with p-value less
than” and enter 0.01 as CNV eQTL p-value threshold.
(e) Select “Restrict to eQTLs on the same chromosome”.
Results:
The experiment we just conducted replicates many of the eQTLs
reported for ORMDL3 in the original study (see Fig. 3), including
rs9303277 on IKZF3 (p = 4 × 10−10), rs2290400 on GSDMB
Fig. 3 Novel susceptibility loci identified from SCAN’s annotation pipeline. SCAN replicates many of the eQTLs
reported for ORMDL3 in the original GWA study (Moffatt, M.F., et al., 2007) of childhood onset asthma. In addi-
tion to the reported loci, we identified 2 CNVs on the same chromosome (namely, CNVR7003.1 and CNVR7095.3)
that predict ORMDL3 expression. These are outstanding candidate loci for follow-up studies on asthma
susceptibility
A Systems Biology Approach to Pharmacogenomics 221
4.2 Functional The identification of patients who are likely to experience adverse
Annotation of events from a particular chemotherapeutic treatment is an impor-
Chemotherapeutic tant step in the individualization of cancer chemotherapeutics.
Susceptibility A recent study [31] has applied an in vitro genome-wide cell-based
Associated SNPs model system to identify pharmacogenetic variants that can serve
as germ-line genetic biomarkers of carboplatin susceptibility in
head and neck cancer (HNC) patients. Two SNPs, rs2551038 and
rs6870861, identified in a cell-based model were found to be asso-
ciated with overall response to carboplatin-based therapy in HNC
patients. We sought mechanistic hypotheses for the published SNP
associations with chemoresponse.
Protocol:
(a) In SCAN’s SNP Query tool, enter the SNPs of interest
(rs2551038 and rs6870861). The SNPs should be white space-
or comma-delimited.
(b) Select “include SNP info” option.
(c) Select “include host gene and SNP function” option.
(d) Select “include left- and right-flanking genes” option.
(e) Select “include genes that SNP predicts expression for with
p-value less than” option and enter 0.0001 as SNP eQTL
p-value threshold.
Results:
This protocol identified rs2551038 as an intronic SNP within
HINT1, a gene that has been shown to act as a tumor suppressor and
to exert a pro-apoptotic function [32]. On the other hand, rs6870861
is located outside of a gene, but is flanked on the right by HINT1.
The two SNPs are regulating the expression of at least ten genes,
mostly in trans, including TLR4 (p = 5 × 10−7), the most significantly
222 Eric R. Gamazon et al.
5 Conclusion
Acknowledgments
References
16. Gamazon ER, Huang RS et al (2011) Copy 27. Stranger BE et al (2007) Population genomics
number polymorphisms and anticancer phar- of human gene expression. Nat Genet 39:
macogenomics. Genome Biol 12:R46 1217–1224
17. Ogura Y et al (2001) A frameshift mutation in 28. Storey JD et al (2007) Gene-expression varia-
NOD2 associated with susceptibility to tion within and among human populations.
Crohn’s disease. Nature 411:603–606 Am J Hum Genet 80:502–509
18. Hampe J et al (2001) Association between 29. Moffatt MF et al (2007) Genetic variants regu-
insertion mutation in NOD2 gene and Crohn’s lating ORMDL3 expression contribute to the
disease in German and British populations. risk of childhood asthma. Nature 448:470–473
Lancet 357:1925–1928 30. Bouzigon E et al (2008) Effect of 17q21 vari-
19. Cenik C (2011) Genome analysis reveals inter- ants and smoking exposure in early-onset
play between 5′UTR introns and nuclear asthma. N Engl J Med 359:1985–1994
mRNA export for secretory and mitochondrial 31. Ziliak D et al (2011) Germline polymorphisms
genes. PLoS Genet 7:e1001366 discovered via a cell-based, genome-wide
20. Zhang L et al (2008) Genomic and epigenetic approach predict platinum response in head
alterations deregulate microRNA expression in and neck cancers. Transl Res 157:265–272
human epithelial ovarian cancer. Proc Natl 32. Weiske J, Huber O (2006) The histidine triad
Acad Sci USA 105:7004–7009 protein Hint1 triggers apoptosis independent
21. Yang B et al (2007) The muscle-specific of its enzymatic activity. J Biol Chem
microRNA miR-1 regulates cardiac arrhyth- 281:27356–27366
mogenic potential by targeting GJA1 and 33. Kelly MG et al (2006) TLR-4 signaling pro-
KCNJ2. Nat Med 13:486–491 motes tumor growth and paclitaxel chemore-
22. Hansen T et al (2007) Brain expressed microR- sistance in ovarian cancer. Cancer Res 66:
NAs implicated in schizophrenia etiology. 3859–3868
PLoS One 2:e873 34. Willer CJ et al (2009) Six new loci associated
23. Pickrell JK et al (2010) Understanding mech- with body mass index highlight a neuronal
anisms underlying human gene expression influence on body weight regulation. Nat
variation with RNA sequencing. Nature 464: Genet 41:25–34
768–772 35. Yang T, Sauve AA (2006) NAD metabolism
24. Spielman RS et al (2007) Common genetic and sirtuins: metabolic regulation of protein
variants account for differences in gene expres- deacetylation in stress and toxicity. AAPS J
sion among ethnic groups. Nat Genet 39: 8:E632–E643
226–231 36. Pietilainen KH et al (2008) Global transcript
25. Zhang W et al (2009) Identification of common profiles of fat in monozygotic twins discordant
genetic variants that account for transcript for BMI: pathways behind acquired obesity.
isoform variation between human populations. PLoS Med 5:e51
Hum Genet 125:81–93 37. Hayes MG et al (2007) Identification of type 2
26. Duan S et al (2008) Genetic architecture of diabetes genes in Mexican Americans through
transcript-level variation in humans. Am J genome-wide association studies. Diabetes
Hum Genet 82:1101–1113 56:3033–3044
Chapter 15
Abstract
Clinical studies have strongly suggested that genetic polymorphisms and/or mutations of certain
ATP-binding cassette (ABC) transporter genes might be regarded as significant factors affecting patients’
responses to medication and/or the risk of diseases. In the case of ABCG2, certain single nucleotide poly-
morphisms (SNPs) in the encoding gene alter the substrate specificity and/or enhance endoplasmic
reticulum-associated degradation (ERAD) of the de novo synthesized ABCG2 protein via the ubiquitin-
mediated proteasomal proteolysis pathway. Hitherto accumulated clinical data imply that several nonsyn-
onymous SNPs affect the ABCG2-mediated clearance of drugs or cellular metabolites, although some
controversies still exist. Therefore, we recently developed high-speed functional screening and ERAD of
ABC transporters so as to evaluate the effect of genetic polymorphisms on their function and protein
expression levels in vitro. In this chapter we present in vitro experimental methods to elucidate the impact
of nonsynonymous SNPs on protein degradation of ABCG2 as well as on its transport function.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_15, © Springer Science+Business Media, LLC 2013
225
226 Toshihisa Ishikawa et al.
Fig. 1 Flowchart for experimental procedures for studing protein quality control
and transport function to validate clinically important SNPs. The protein quality
control and ERAD of SNP variants of the ABC transporters can be studied by using
Flp-In-293 cells, whereas the transport function assay is carried out by using plasma
membrane vesicles prepared from insect Sf9 cells and high-speed screening/QSAR
analysis technologies. QSAR quantitative structure–activity relationship, Q-RT-PCR
quantitative reverse-transcription polymerase chain reaction
2 Materials
2.1.3 pOG44 Vector pOG44 is a 5.8-kb Flp recombinase expression vector (Invitrogen,
Carlsbad, CA, USA). The FLP gene was originally isolated from
the S. cerevisiae 2-μ plasmid [20, 21] and encodes a site-specific
recombinase that is a member of the integrase family of recombi-
nases [22]. The Flp recombinase mediates a site-specific recombi-
nation reaction between interacting DNA molecules via the pairing
of interacting FRT sites [11, 23]. The native FLP gene encodes a
protein of 423 amino acids with a calculated molecular mass of
49 kDa. The FLP gene expressed from pOG44 encodes a
temperature-sensitive Flp recombinase, which carries a point muta-
tion (flp-F70L) that results in a change in amino acid 70 from Phe
to Leu [24]. The flp-F70L protein expressed from pOG44 exhibits
increased thermostability at 37 °C in mammalian cells when com-
pared with the native Flp recombinase [24].
2.1.4 Reagents The following reagents are available from commercial sources.
1. Dulbecco’s modified Eagle’s medium (D-MEM).
2. 10 % (v/v) heat-inactivated fetal calf serum (FCS).
3. L-Glutamine (2 mM).
4. Penicillin (100 U/ml).
5. Streptomycin (100 μg/ml).
6. Zeocin (100 μg/ml).
7. Hygromycin B (100 μg/ml).
8. Amphotericin B, 250 ng/ml.
9. Trypan Blue dye.
Methods to Examine the Impact of Nonsynonymous SNPs on Protein Degradation… 229
10. Lipofectamine-2000.
11. MG132 (inhibitor for proteasomal degradation of proteins).
12. Bafilomycin A1 (inhibitor for lysosomal degradation of proteins).
13. 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide
(MTT reagent).
2.2 Materials for We usually infect Sf9 cells (1 × 106 cells/ml) with human ABCG2-
Functional Evaluation of recombinant baculovirus and culture them at 27 °C with gentle
Nonsynonymous SNPs shaking [29, 30].
3 Methods
3.1 Methods SNP data on the polymorphisms of human ABCG2 gene were
for Evaluation obtained from the NCBI dbSNP database and publications [31].
of the Impact of Figure 2 depicts nonsynonymous polymorphisms and acquired
Nonsynonymous SNPs mutations in the human ABCG2 gene.
on Protein Degradation
3.1.1 SNP Data on
Nonsynonymous
Polymorphisms of Human
ABCG2 Gene
N590Y
Plasma
Membrane
G51C inside
COOH
B F489L
A
V12M R482G
H2N C R482T
Q126stop E334stop
Q141K S248P
T153M Q166E
Table 1
PCR primers and conditions for site-directed mutagenesis to create variants of ABCG2
Toshihisa Ishikawa et al.
Fig. 3 Flp-mediated integration of the ABCG2 cDNA into FRT-tagged genomic DNA. Flp-In-293 cells were
co-transfected with the pcDNA5/FRT vector carrying the ABCG2 cDNA and the Flp recombinase expression
plasmid pOG44. Flp recombinase mediates insertion of the expression construct with the ABCG2 cDNA into
the genome at the integrated FRT site through site-specific DNA recombination
3.1.5 Measurement The mRNA levels of ABCG2 and GAPDH are measured by quan-
of mRNA Levels by titative PCR, and the ratios of ABCG2 variants vs. GAPDH are
Quantitative RT-PCR plotted.
1. Determine the mRNA levels of ABCG2 and GAPDH by using
the 7500 Fast Real Time-PCR System (Applied Biosystems,
Foster City, CA, USA), TaqMan® Fast Universal Master Mix
(Applied Biosystems), and TaqMan® probes (ABCG2;
Methods to Examine the Impact of Nonsynonymous SNPs on Protein Degradation… 235
3.1.7 Immunofluo- 1. ABCG2 expressing Flp-In-293 cells are seeded onto collagen
rescence Microscopy type I-coated cover glasses and incubated under the above-
mentioned culture conditions for 24 h.
236 Toshihisa Ishikawa et al.
3.2.2 Expression Figure 4 demonstrates the strategy for the expression of ABCG2
of ABCG2 Variants variants in Sf9 cells.
in Sf9 Cells
1. Competent DH10Bac E. coli cells are transformed by the variant
ABCG2 plasmids. The variant ABCG2 cDNA is then trans-
posed into a bacmid, which is a baculovirus shuttle vector
carrying the baculovirus genome, in DH10Bac cells with the
aid of a helper plasmid.
Methods to Examine the Impact of Nonsynonymous SNPs on Protein Degradation… 237
ABCG2
pFastBac1-
ABCG2
plasmid
Competent DH10BacTM E. Coli cells
Recombinant Baculovirus
Recombinant Bacmid DNA
Prepare
plasma membrane vesicles Viral Amplification
3.2.3 Preparation of the Plasma membrane vesicles are prepared from ABCG2-expressing
Plasma Membrane Vesicles Sf9 cells as described previously [29, 30]. The use of low ionic
from Sf9 Cells strength buffers during the membrane preparation steps promotes
the formation of open membrane sheets and inside-out membrane
vesicles. It is important to maintain high integrity of the plasma
membrane vesicles used in the transport assay. In other words, the
membrane vesicles must be completely sealed. Figure 5
Centrifugation
2,000 x g, 10 min
Fig. 5 Preparation of membrane vesicles from ABCG2-expressing Sf9 cells. The frozen cell pellet was thawed
quickly, diluted 40-fold with the hypotonic buffer, and then homogenized with a Potter-Elvehjem homogenizer.
After centrifugation at 2,000 × g, the supernatant was further centrifuged at 100,000 × g for 30 min. The crude
membrane fraction was layered over 40 % (w/v) sucrose solution and centrifuged at 100,000 × g for 30 min.
The turbid layer at the interface was collected and then centrifuged at 100,000 × g for 30 min. The membrane
fraction was collected and resuspended in a small volume (150–250 μl) of 0.25 M sucrose containing 10 mM
Tris/HEPES (pH 7.4)
Methods to Examine the Impact of Nonsynonymous SNPs on Protein Degradation… 239
3.2.4 Immunological The amount of ABCG2 expressed in the cell membrane vesicles is
Detection of ABCG2 in determined by immunoblotting with BXP-21 (SIGNET, Dedham,
Plasma Membrane Vesicles MA, USA), a specific antibody to human ABCG2, as described
above. To quantitatively analyze the transport activity of ABCG2
variants, it is critically important to normalize the expression level
of each variant protein. There is a linear relationship between the
signal intensity of immunoblotting and the logarithmic value of
the amount of protein applied to the electrophoresis [29, 30].
Based on this linear relationship, the expressed levels of ABCG2
and its variants in different plasma membrane preparations can be
quantitatively estimated and normalized [29, 30].
30 ml/well
Incubate (PCR machine): 500 ml/well Sephadex G-25
4°C 10 sec (Bed volume: 100 ml)
37°C 10 min
1,600×g, 4°C, 5 min
0.25 M Sucrose,
Stop solution: 80 ml/well 10 mM Tris/HEPES (pH 7.4)
10 mM EDTA, 0.25 M Sucrose, ×3
1,600×g, 4°C, 5 min
10 mM Tris/HEPES (pH 7.4)
50 ml/well
Separation plate
96-well microplate
1,600×g, 4°C, 5 min
Fig. 6 Detection of ATP-dependent hematoporphyrin transport into plasma membrane vesicles by using a
96-well separation plate. Plasma membrane vesicles expressing human ABCG2 were prepared from Sf9 insect
cells. ATP-dependent transport of hematoporphyrin into the vesicles mediated by the action of ABCG2 was
measured by measuring the fluorescence of hematoporphyrin incorporated into the membrane vesicles [37]
3.2.6 High-Speed To detect the drug transport activity of ABCG2 WT and SNP
Screening to Measure the variants, we used methotrexate (MTX) as a model substrate.
Transport Activity of ABCG2 Figure 7 illustrates the procedure of the functional assay. This high-
and Its Variants speed screening method can be used to investigate drug-ABCG2
interactions based on quantitative structure–activity relationship
(QSAR) analysis [38].
1. The frozen stocked membrane is first thawed quickly, and then
membrane vesicles are formed by passing the membrane
suspension through a 27-gauge needle.
2. To measure the ABCG2-mediated MTX transport, the stan-
dard incubation medium should contain plasma membrane
vesicles (10 or 50 μg of protein), 200 μM [3′,5′,7′-3H]MTX
(Amersham, Buckinghamshire, UK), 0.25 M sucrose, 10 mM
Tris/HEPES, pH 7.4, 10 mM MgCl2, 1 mM ATP, 10 mM
creatine phosphate, and 100 μg/ml of creatine kinase in a final
volume of 100 μl. The incubation is carried out at 37 °C.
Incubation medium:
plasma membrane vesicles (50 mg of protein)
Preparation on ice 100 ml 0.25M sucrose, 10 mM Tris/HEPES (pH 7.4),
1 mM ATP, 10 mM MgCl2,
10 mM creatine phosphate, creatine kinase (100 mg/ml),
100 mM [3H]Methotrexate (MTX)
Incubation at 37°C for 20 min
1 ml
Stop solution: 0.25 M sucrose
(ice-cold) 2 mM EDTA
Quick mixing 10 mM Tris/HCl (pH 7.4)
270 ml / well
MultiScreenTM plate
200 ml / well 0.25 M sucrose
10 mM Tris/HCl (pH 7.4)
Aspiration
ATP Rinse 4 times
[3H]MTX
ADP, Pi
Fig. 7 High-speed screening method to study the transport activity of human ABCG2 and its SNP variants.
ATP-dependent transport of [3H]methotrexate (MTX) into plasma membrane vesicles mediated by the action of
ABCG2 was measured by counting the radioactivity remaining on the filter of MultiScreen™ plates. Inhibition
of MTX transport was detected by adding a test compound into the reaction mixture [38]
242 Toshihisa Ishikawa et al.
3. After a specified time (20 min for the standard condition), the
reaction medium is mixed with 1 ml of the ice-cold stop solution
(0.25 M sucrose, 10 mM Tris/HEPES, pH 7.4, and 2 mM
EDTA) to terminate the transport reaction. Subsequently, ali-
quots (270 μl per well) of the resulting mixture are transferred
to MultiScreen™ plates (Nihon Millipore KK, Tokyo, Japan).
4. Under aspiration, each well of the plate is then rinsed with the
0.25 M sucrose solution containing 10 mM Tris/HEPES, pH
7.4, four times (4 × 200 μl for each well) in an EDR384S system
(BioTec, Tokyo, Japan).
5. [3H]MTX thus incorporated into the vesicles is measured by
counting the radioactivity remaining on the filter of
MultiScreen™ plates, where each filter is placed in 2 ml of liq-
uid scintillation fluid (Ultima Gold, Packard BioScience).
4 Notes
4.1 Notes for The Flp-In-293 cell line is a useful cell system for studying the
Valuation of the Impact molecular mechanism of protein misfolding and the subsequently
of Nonsynonymous occurring ERAD process. Flp-In-293 cells are not polarized cells.
SNPs on Protein Therefore, for studying the apical or basolateral localization of
Degradation membrane proteins, MDCK (Madin-Darby canine kidney) and
LLC-PK1 (porcine kidney) cells may be applicable
4.1.1 Flp-In Cell Lines
1. The Flp-In method is based on the exchange of an expression
cassette within a previously tagged FRT site. M-FISH revealed
that ABCG2 cDNA was incorporated into the telomeric region
of chromosome 12p in Flp-In-293 cells [26, 27].
2. As shown in Fig. 8a, mRNA levels of ABCG2 WT and SNP
variants (V12M, Q141K, F208S, S248P, F431L, S441N, and
F489L) were evenly represented in Flp-In-293 cells. On the
other hand, ABCG2 WT and those SNP variants as well as
GAPDH proteins were detected by immunoblotting, and their
expression levels should be quantified. For this purpose, we
treated all of the samples with PNGase F and mercaptoethanol
to remove glycomoieties and to break the cysteinyl disulfide
bond forming a homodimer. Since there was a linear relation-
ship between the signal intensity of immunoblotting and the
logarithmic value of the amount of ABCG2 protein applied to
the electrophoresis, the expression level of ABCG2 or GAPDH
in cell lysate samples could be quantitatively estimated based on
the linear relationship [28, 29, 32]. The relative values of
protein levels were then normalized to the ratio of ABCG2
WT/GAPDH. Although mRNA levels were almost the same in
the WT and SNP variants (F208S and S441N), protein levels of
those variants were markedly decreased (Fig. 8b). The protein
level of Q141K variant was about half that of the WT level.
Methods to Examine the Impact of Nonsynonymous SNPs on Protein Degradation… 243
Q141K
Q141K
V12M
V12M
Mock
Mock
WT
WT
ABCG2 ABCG2
GAPDH GAPDH
2 2
1 1
*
0 0 *
S441N
S248P
S248P
F208S
F208S
F431L
F489L
F431L
F489L
Mock
Mock
WT
WT
ABCG2 ABCG2
GAPDH GAPDH
Relative protein level
Relative mRNA level
2 2
1 1
*
0 0 * *
Fig. 8 mRNA and protein expression levels (a) as well as immunofluorescence images of Flp-In-293 cells
expressing ABCG2 WT or SNP variants (b). (a) Relative levels of mRNA were detected by RT-PCR with specific
primers for ABCG2 and GAPDH. Data are calculated as ratios by referring to the GAPDH mRNA levels in
Flp-In-293 cells and normalized to the ratio of ABCG2/GAPDH. Data are expressed as mean values ± SD (n = 4).
Relative levels of ABCG2 protein were detected by immunoblotting. ABCG2 protein was detected by immunob-
lot analysis with BXP-21 monoclonal antibody. Data are calculated as ratios by referring to the GAPDH protein
levels in Flp-In-293 cells and normalized to the ratio of ABCG2/GAPDH. Data are expressed as means ± SD in
triplicate experiments. Statistical significance (*P < 0.05) was evaluated by Student’s t-test. (b) The ABCG2
protein was immunologically linked with Alexa Fluor 488 (green fluorescence), and nuclei were stained with
propidium iodide (red fluorescence). Horizontal bars correspond to 20 μm
244 Toshihisa Ishikawa et al.
Fig. 8 (continued)
Fig. 9 Effect of bafilomycin A1 (BMA) and MG132 (MG) on the protein levels of ABCG2 WT, F208S, and S441N
(a) as well as a schematic illustration of plausible pathways involved in the degradation of ABCG2 protein (b).
(a) Flp-In-293 cells expressing WT, F208S, or S441N were incubated in the absence or presence of BMA
(10 nM) or MG (2.0 μM) for 24 h. ABCG2 WT, F208S, and S441N variant proteins were analyzed by immunoblotting
with the ABCG2-specific monoclonal antibody (BXP-21) after PNGase F treatment. The ABCG2 protein level in the
cell lysate of each cell population was analyzed by immunoblotting with the ABCG2-specific monoclonal antibody
(BXP-21) or the GAPDH-specific antibody after PNGase F treatment. The signal intensity ratio (ABCG2/GAPDH)
was normalized to the control level (labeled as “None”). Data are expressed as means ± SD in triplicate experi-
ments. (b) The correctly processed ABCG2 WT is finally destined to reach the plasma membrane and is then
degraded by the endosome–lysosome pathway after remaining in the plasma membrane domain for a certain
period. In contrast, the misfolded ABCG2 protein undergoes ubiquitination-mediated proteasomal degradation.
Bafilomycin A1 (BMA) and MG132 inhibit lysosomal and proteasomal degradation, respectively
4.2 Notes To examine the quality of plasma membrane vesicles prepared from
for Functional Sf9 cells, we used scanning electron microscopy (SEM) technolo-
Evaluation of gies and identified the optimal conditions required to prepare the
Nonsynonymous SNPs membrane vesicles. SEM revealed that well-sealed membrane
vesicles have an average size (diameter) of about 200 nm [38].
4.2.1 Quality of Plasma
The timing of harvesting Sf9 cells after baculovirus infection is very
Membrane Vesicles
critical. The membrane morphology of infected Sf9 cells changed
Prepared from Sf9 cells
greatly; in particular, numerous pores were observed after day 5.
Membrane vesicles prepared from those cells (>day 5) are useless
for our purpose.
1. It is important to prepare membrane vesicles in the presence of
serine/cysteine protease inhibitors. Leupeptin (10 μg/ml)
inhibits the degradation of ABCG2 protein in membrane
vesicles prepared from baculovirus-infected Sf9 cells during
repetitive freeze–thaw cycles.
246 Toshihisa Ishikawa et al.
4.2.2 Gel-Filtration Assay Whereas the rapid filtration method is widely used for the transport
Method for the Transport of assay, it is not applicable for assaying porphyrin transport. Since
Hydrophobic Compounds hematoporphyrin is bound to the filter membrane surface, it causes
high background levels in the transport measurements [30, 37].
Therefore, we applied gel-filtration to the porphyrin transport
assay, as shown in Fig. 6. Based on our experiences, we recommend
the gel-filtration method when hydrophobic compounds are used
as substrates of the transporter of interest.
4.2.3 Quantitative To quantitatively analyze the transport activity per each SNP
Analysis of the Transport variant, it is important to quantitatively analyze the immunoblot-
Activity of SNP Variants ting intensities vs. ABCG2 protein levels. There is a linear relation-
ship between the signal intensity of immunoblotting and the
logarithmic value of the amount of protein applied to the electro-
phoresis [30]. Based on this linear relationship, the expression
levels of ABCG2 and its variants in different plasma membrane
preparations can be quantitatively estimated and normalized.
1. Figure 10 demonstrates the ATP-dependent transport of
hematoporphyrin (upper panel) and methotrexate (lower
panel) mediated by ABCG2 and its variants. Plasma membrane
vesicles (50 μg of protein) expressing ABCG2 and its variants
are incubated with 20 μM hematoporphyrin or 200 μM [3H]
methotrexate in the presence of 1 mM ATP. Each transport
activity is calculated by considering the normalized levels of
ABCG2 protein expression [30].
2. It is important to note that the variants Q126stop, F208S,
S248P, E334stop, and S441N lack substantial transport activity
for both hematoporphyrin and methotrexate. Interestingly, the
F489L variant, which does not transport methotrexate, exhibits
impaired hematoporphyrin transport (Vmax = 0.058 nmol/min/
mg protein, Km = 8.6 μM for F489L vs. Vmax = 0.654 nmol/min/
mg protein, Km = 17.8 μM for WT).
3. The F431L variant as well as the acquired mutants R482G and
R482T transport hematoporphyrin (upper panel), although
they do not transport methotrexate (lower panel). These
results provide evidence that certain nonsynonymous SNPs
Methods to Examine the Impact of Nonsynonymous SNPs on Protein Degradation… 247
0.5
Porphyrin
(nmol/min/mg protein)
0.4
Porphyrin transport
0.3
0.2
0.1
0.0
1.5
Methotrexate
Methotrexate transport
(nmol/min/mg protein)
1.0
0.5
0.0
WT
Mock
I206L
F571I
V12M
G51C
F431L
F489L
F208S
S248P
R482T
S441N
N590Y
Q141K
T153M
Q166E
D620N
R482G
E334stop
Q126stop
Fig. 10 ATP-dependent transport of hematoporphyrin (upper panel) and methotrexate (lower panel) mediated
by ABCG2 and its variants. Plasma membrane vesicles (50 μg of protein) were incubated with 20 μM hemato-
porphyrin or 200 μM [3H]MTX in the presence or absence of 1 mM ATP in the standard incubation medium at
37 °C for 10 min (hematoporphyrin) or 20 min (MTX). The ATP-dependent transport of hematoporphyrin or MTX
is normalized for the amount of ABCG2 protein as described previously [6]. Data are expressed as means ± SD
in triplicate experiments
Fig. 11 Characterization of ABCG2 WT and SNP variants. The properties of ABCG2 WT and SNP variants were
characterized as + (positive), − (negative), or ± (marginal) according to the following indexes: protein expression,
transport of methotrexate (MTX) or porphyrin, resistance to SN-38, mitoxantrone (MX), doxorubicin, or dauno-
rubicin, and prazosin-stimulated ATPase activity. Data are from ref. 28
for elevated serum uric acid levels and the risk of gout [46, 47].
Uric acid is the end product of purine metabolism in humans.
Two-thirds of the uric acid in the human body is normally excreted
through the kidney, whereas one-third gains entrance to the gut
where it undergoes uricolysis (decomposition of uric acid). Because
of impaired expression of the SNP variant (Q141K) of ABCG2,
elevated serum uric acid levels cause gout and are a risk factor for
cardiovascular disease and diabetes. This provides evidence that
ABCG2 expressed on the apical side of the proximal tubular cells
in human kidney plays a pivotal role in the renal excretion of serum
uric acid.
Acknowledgments
References
functional analysis approaches. Pharmacoge- 20. Broach JR, Hicks JB (1980) Replication and
nomics 5:67–99 recombination functions associated with the
6. Nakagawa H, Toyoda Y, Wakabayashi-Nakao yeast plasmid, 2 mu circle. Cell 21:501–508
K et al (2011) Ubiquitin-mediated protea- 21. Broach JR, Guarascio VR, Jayaram M (1982)
somal degradation of ABC transporters: a new Recombination within the yeast plasmid 2 mu
aspect of genetic polymorphisms and clinical circle is site-specific. Cell 29:227–234
impacts. J Pharm Sci 100:3602–3619 22. Argos P, Landy A, Abremski K et al (1986)
7. Ellgaard L, Molinari M, Helenius A (1999) The integrase family of site-specific recombi-
Setting the standards: quality control in the nases: regional similarities and global diversity.
secretory pathway. Science 286:1882–1888 EMBO J 5:433–440
8. Mori K (2000) Tripartite management of 23. Craig NL (1988) The mechanism of conserva-
unfolded proteins in the endoplasmic reticulum. tive site-specific recombination. Annu Rev
Cell 101:451–454 Genet 22:77–105
9. Hampton RY (2002) ER-associated degrada- 24. Buchholz F, Ringrose L, Angrand PO et al
tion in protein quality control and cellular (1996) Different thermostabilities of FLP and
regulation. Curr Opin Cell Biol 14:476–482 Cre recombinases: implications for applied
10. Kleizen B, Braakman I (2004) Protein folding site-specific recombination. Nucleic Acids Res
and quality control in the endoplasmic reticu- 24:4256–4262
lum. Curr Opin Cell Biol 16:343–349 25. Mitomo H, Kato R, Ito A et al (2003) A func-
11. Sauer B (1994) Site-specific recombination: tional study on polymorphism of the
developments and applications. Curr Opin ATP-binding cassette transporter ABCG2:
Biotechnol 5:521–527 critical role of arginine-482 in methotrexate
transport. Biochem J 373:767–774
12. Gronostajski RM, Sadowski PD (1985)
Determination of DNA sequences essential for 26. Tamura A, Wakabayashi K, Onishi Y et al
FLP-mediated recombination by a novel (2006) Genetic polymorphisms of human ABC
method. J Biol Chem 260:12320–12327 transporter ABCG2: development of the stan-
dard method for functional validation of SNPs
13. Jayaram M (1985) Two-micrometer circle site- by using the Flp recombinase system. J Exp
specific recombination: the minimal substrate Ther Oncol 6:1–11
and the possible role of flanking sequences.
27. Wakabayashi-Nakao K, Tamura A, Koshiba S
Proc Natl Acad Sci USA 82:5875–5879
et al (2010) Production of cells with targeted
14. Senecoff JF, Bruckner RC, Cox MM (1985) integration of gene variants of human ABC
The FLP recombinase of the yeast 2-micron transporter for stable and regulated expression
plasmid: characterization of its recombination using the Flp recombinase system. Methods
site. Proc Natl Acad Sci USA 82:7270–7274 Mol Biol 648:139–159
15. Andrews BJ, Proteau GA, Beatty LG et al 28. Tamura A, Wakabayashi K, Onishi Y et al
(1985) The FLP recombinase of the 2 micron (2007) Re-evaluation and functional classifica-
circle DNA of yeast: interaction with its target tion of non-synonymous single nucleotide
sequences. Cell 40:795–803 polymorphisms of the human ATP-binding
16. Boshart M, Weber F, Jahn G et al (1985) cassette transporter ABCG2. Cancer Sci
A very strong enhancer is located upstream of an 98:231–239
immediate early gene of human cytomegalovirus. 29. Ishikawa T, Sakurai A, Kanamori Y et al
Cell 41:521–530 (2005) High-speed screening of human
17. Nelson JA, Reynolds-Kohler C, Smith BA ATP-binding cassette transporter function
(1987) Negative and positive regulation by a and genetic polymorphisms: new strategies
short segment in the 5'-flanking region of the in pharmacogenomics. Methods Enzymol
human cytomegalovirus major immediate- 400:485–510
early gene. Mol Cell Biol 7:4125–4129 30. Tamura A, Watanabe M, Saito H et al (2006)
18. Andersson S, Davis DL, Dahlback H et al Functional validation of the genetic polymor-
(1989) Cloning, structure, and expression of the phisms of human ATP-binding cassette (ABC)
mitochondrial cytochrome P-450 sterol transporter ABCG2: identification of alleles
26-hydroxylase, a bile acid biosynthetic enzyme. that are defective in porphyrin transport. Mol
J Biol Chem 264:8222–8229 Pharmacol 70:287–296
19. Gritz L, Davies J (1983) Plasmid-encoded 31. Ishikawa T, Tamura A, Saito H et al (2005)
hygromycin B resistance: the sequence of Pharmacogenomics of the human ABC trans-
hygromycin B phosphotransferase gene and its porter ABCG2: from functional evaluation to
expression in Escherichia coli and Saccharomyces drug molecular design. Naturwissenschaften
cerevisiae. Gene 25:179–188 92:451–463
250 Toshihisa Ishikawa et al.
32. Nakagawa H, Tamura A, Wakabayashi K et al 40. Elbein AD, Pan YT, Pastuszak I et al (2003)
(2008) Ubiquitin-mediated proteasomal New insights on trehalose: a multifunctional
degradation of non-synonymous SNP variants molecule. Glycobiology 13:17R–27R
of human ABC transporter ABCG2. Biochem 41. Furuki T, Oku K, Sakurai M (2009)
J 411:623–631 Thermodynamic, hydration and structural
33. Wang P, Hammer DA, Granados RR (1997) characterization of alpha, alpha-trehalose.
Binding and fusion of Autographa californica Front Biosci 14:3523–3535
nucleopolyhedrovirus to cultured insect cells. 42. Guo N, Puhlev I, Brown DR et al (2008)
J Gen Virol 78:3081–3089 Trehalose expression confers desiccation toler-
34. Blissard GW, Wenz JR (1992) Baculovirus ance on human cells. Nat Biotechnol 18:
gp64 envelope glycoprotein is sufficient to 168–171
mediate pH-dependent membrane fusion. 43. Deghan A, Köttgen A, Yang Q et al (2008)
J Virol 66:6829–6835 Association of three genetic loci with uric acid
35. Lanir LM, Volkman LE (1998) Actin binding concentration and risk of gout: a genome-wide
and nucleation by Autographa californica M association study. Lancet 372:1953–1961
nucleopolyhedrovirus. Virology 243:167–177 44. Kolz M, Johnson T, Sanna S et al (2009)
36. Whittaker GR, Helenius A (1998) Nuclear Meta-analysis of 28,141 individuals identifies
import and export of viruses and virus common variants within five new loci that
genomes. Virology 246:1–23 influence uric acid concentrations. PLoS Genet
37. An R, Hagiya Y, Tamura A et al (2009) Cellular 5:e1000504
phototoxicity evoked through the inhibition 45. Stark K, Reinhard W, Grassi M et al (2009)
of human ABC transporter ABCG2 by cyclin- Common polymorphisms influencing serum
dependent kinase inhibitors in vitro. Pharm uric acid levels contribute to susceptibility to
Res 26:449–458 gout, but not to coronary artery disease. PLoS
38. Saito H, Hirano H, Nakagawa H et al (2006) One 4:e7729
A new strategy of high-speed screening and 46. Woodward O, Köttgen A, Coresh J et al
quantitative structure-activity relationship (2009) Identification of a urate transporter,
analysis to evaluate human ATP-binding cas- ABCG2, with a common functional polymor-
sette transporter ABCG2-drug interactions. phism causing gout. Proc Natl Acad Sci USA
J Pharmacol Exp Ther 317:1114–1124 106:10338–10342
39. Saito H, Hirano H, Shin W et al (2009) Technical 47. Matsuo H, Takada T, Ichida K et al (2009)
pitfalls and improvements in high-speed screen- Common defects of ABCG2, a high-capacity
ing and QSAR analysis to predict durg–drug urate exporter, cause gout. A function-based
interactions of ABC transporter ABCB11 (bile genetic analysis in a Japanese population. Sci
salt export pump). AAPS J 11:581–589 Transl Med 1:5ra11
Chapter 16
Abstract
Metabolism catalyzed by the cytochrome P450 enzymes (CYPs) represents the most important pathway
for drug metabolism and elimination in humans. Identification of the CYPs responsible for metabolism of
existing and novel drugs is critical for the prediction of adverse reactions caused by drug–drug interactions
or individual genetic polymorphism. An integrated approach is described for CYP-mediated metabolic
reaction phenotyping using both recombinant enzymes and human liver microsomes in combination of
selective inhibitors or inhibitory antibodies. The in vitro method described includes screening of recombi-
nant CYPs for metabolic activity, chemical inhibition or antibody neutralization, and correlation analysis
with isoform-selective marker activities. The primary focus is on identification of the most common
enzymes including CYP1A2, 2C9, 2C19, 2D6, and 3A4, although the same strategy could potentially be
used for identification of other isoforms.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_16, © Springer Science+Business Media, LLC 2013
251
252 Zhengyin Yan and Gary W. Caldwell
2 Materials
2.1 Buffers, All reagents were obtained from Sigma-Aldrich (St. Louis, MO)
Cofactors, and Stop except for those specified.
Solution
1. 0.5 M Potassium phosphate buffer, pH 7.4 is prepared as the
following:
(a) 0.5 M Potassium phosphate, KH2PO4, monobasic. Dissolve
34 g KH2PO4 in 450 mL deionized water, and then bring
the final volume to 500 mM with deionized water.
(b) 0.5 M Potassium phosphate, K2HPO4, dibasic. Dissolve
57 g K2HPO4·3H2O in 450 mL deionized water, and then
bring the final volume to 500 mM with deionized water.
(c) Mix 60 mL 0.5 M KH2PO4 with 280 mL 0.5 M K2HPO4,
and check with a pH meter for a pH value of 7.4. If
necessary, adjust pH with either KH2PO4 or K2HPO4.
2. 5 mM Sodium citrate, tribasic. Dissolve 14.7 mg sodium
citrate in 100 mL deionized water, and store at 4 °C.
3. Co-factors: Dissolve 400 mg nicotinamide adenine dinucleo-
tide phosphate (NADP+), 400 mg glucose-6-phosphate, and
266 mg MgCl2·6H2O in 18 mL deionized water, and then
adjust the final volume to 20 mL with deionized water. Aliquot
and store at −20 °C.
4. Glucose-6-phosphate dehydrogenase (G6PDH): 40 U/mL,
prepared in 5 mM sodium citrate. Aliquot and store at −20 °C.
5. Stop solution: acetonitrile containing 0.5 μM propranolol or
an equivalent as an internal standard for LC-MS/MS analysis.
2.2 CYP Inhibitors, 1. CYP Selective inhibitors: All inhibitors and their effective con-
Substrates, and centrations are listed in Table 1.
Antibodies 2. CYP Marker substrates (optional).
(a) Phenacetin (CYP1A2).
(b) Coumarin (CYP2A6).
(c) (S)-Mephenytoin (CYP2C19 and CYP2B6).
(d) Paclitaxel (CYP2C8).
(e) Diclofenac (CYP2C9).
(f) Bufuralol (CYP2D6).
(g) Chlorzoxazone (CYP2E1).
(h) Testosterone (CYP3A4).
3. Inhibitory antibodies. Polyclonal or monoclonal antibodies
raised against CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1,
and 3A4 were purchased from XenoTech, LLC (Lenexa, KS) or
other supplier (BD Biosciences, Woburn, MA).
254 Zhengyin Yan and Gary W. Caldwell
Table 1
Chemical inhibitors and effective concentrations for in vitro
CYP-phenotyping [10]
Inhibitor
CYP Inhibitor concentration (μM)
1A2 α-Napthoflavone 1
2A6 Methoxsalen 1
2B6 ThioTEPA 50
2C8 Montelukast 0.1
2C9 Sulphaphenazole 10
2C19 N-3-Benzylphenobarbital 1
2D6 Quinidine 1
2E1 Diethyldithiocarbamate 50
3A4/5 Ketoconazole 1
2.3 CYP Enzymes 1. Pooled human liver microsomes: HLM prepared from 20 to
and Human Liver 50 donors was obtained from BD Biosciences (Woburn, MA)
Microsomes and stored at −80 °C.
2. cDNA-expressed cytochrome P450: Supersomes™ enzymes
such as CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and
3A4 was all purchased from BD Biosciences (Woburn, MA)
and stored at −80 °C (see Note 1).
3. Individual human liver microsomal panel: Individual liver
microsomes prepared from 10 to 15 different donors. Each
HLM preparation was fully characterized using marker
substrates for CYP- specific activity including CYP1A2, 2A6,
2B6, 2C8, 2C9, 2C19, 2D6, 2E1, and 3A4/5 (BD Biosciences).
at the initial condition. During the run, the divert valve was acti-
vated to direct the HPLC eluant to the waste line for the first
1.5 min of elution and then switched to the mass spectrometer for
analysis. LC-MS data were processed by Analyst 1.4.2 (ABI Sciex)
to obtain peak areas of each analyte that were normalized relative
to the internal standard.
3 Methods
Table 2
Expression levels of different CYP enzymes in liver [10]
Mean level
(pmol/mg protein) Relative level (%)
4 Notes
References
1. Lin JH, Lu AY (1997) Role of pharmacokinet- microsomes: a re-evaluation of P450 isoform
ics and metabolism in drug discovery and selectivity. Eur J Drug Metab Pharmacokinet
development. Pharmacol Rev 49:403–449 36:1–16
2. Yan Z, Caldwell GW (2001) Metabolism pro- 8. Vermeir M, Hemeryck A, Cuyckens F,
filing, and cytochrome P450 inhibition & Francesch A, Bockx M, Van Houdt J, Steemans
induction in drug discovery. Curr Top Med K, Mannens G, Aviles P, De Coster R (2009)
Chem 5:403–425 In vitro studies on the metabolism of trabect-
3. Rannug A, Alexandrie A-K, Persson I, edin (YONDELIS) in monkey and man,
Ingelman-Sundberg M (1995) Genetic poly- including human CYP reaction phenotyping.
morphism of cytochromes P450 1A1, 2D6 Biochem Pharmacol 77:1642–1654
and 2E1: regulation and toxicological signifi- 9. Yan Z, Caldwell GW (2004) Evaluation of
cance. J Occup Environ Med 37:25–36 cytochrome P450 inhibition in human liver
4. Murray M (2006) Role of CYP pharmacoge- microsomes. Optimization in Drug Discovery,
netics and drug–drug interactions in the efficacy Humana, Totowa, NJ, pp 231–244
and safety of atypical and other antipsychotic 10. Zhang H, Davis CD, Sinz MW, Rodrigues AD
agents. J Pharm Pharmacol 58:871–885 (2007) Cytochrome P450 reaction-
5. Hamdy SI, Hiratsuka M, Narahara K, El-Enany phenotyping: an industrial perspective. Expert
M, Moursi N, Ahmed MS-E, Mizugaki M Opin Drug Metab Toxicol 3:667–687
(2002) Allele and genotype frequencies of 11. Harper TW, Brassil PJ (2008) Reaction phe-
polymorphic cytochromes P450 (CYP2C9, notyping: current industry efforts to identify
CYP2C19, CYP2E1) and dihydropyrimidine enzymes responsible for metabolizing drug
dehydrogenase (DPYD) in the Egyptian popu- candidates. AAPS J 10:200–207
lation. Br J Clin Pharmacol 53:596–603 12. Kumar V, Rock DA, Warren CJ, Tracy TS,
6. Ingelman-Sundberg M, Sim SC, Gomez A, Wahlstrom JL (2006) Enzyme source effects
Rodriguez-Antona C (2007) Influence of on CYP2C9 kinetics and inhibition. Drug
cytochrome P450 polymorphisms on drug Metab Dispos 34:1903–1908
therapies: pharmacogenetic, pharmacoepi- 13. Yan Z, Caldwell GW, Wu W, McKown L,
genetic and clinical aspects. Pharmacol Ther Rafferty B, Jones W, Masucci JA (2002) In
116:496–526 vitro identification of metabolic pathways and
7. Khojasteh SC, Prabhu S, Kenny JR, Halladay cytochrome P450 isoforms involved in the
JS, Lu AYH (2011) Chemical inhibitors of metabolism of etoperidone. Xenobiotica
cytochrome P450 isoforms in human liver 32(11):949–962
Chapter 17
Abstract
The identification of causative genes underlying biomedically relevant phenotypes, particularly complex
multigenic traits, is of vital interest to modern medicine. Using genome-wide association analysis, many
studies have successfully identified thousands of loci (called quantitative trait loci or QTL), some of these
associating with drug response phenotypes. However, the determination and validation of putative genes
has been much more challenging. The actions of drugs, both efficacious and deleterious, are complex
phenotypes that are controlled or influenced in part by genetic mechanisms.
Investigation for genetic correlates of complex traits and pharmacogenetic traits is often difficult to
perform in human studies due to cost, availability of relevant sample population, and limited ability to
control for environmental effects. These challenges can be circumvented with the use of mouse models for
pharmacogenetic studies. In addition, the mouse can be treated at sub- and supratherapeutic doses and
subjected to invasive procedures, which can facilitate measures of drug response phenotypes, making iden-
tification of pharmacogenetically relevant genes more feasible. The availability of multiple mouse genetic
and phenotypic resources is an additional benefit to using the mouse for pharmacogenetic studies.
Here, we describe the contribution of animal models, specifically the mouse, towards the field of
pharmacogenetics. In this chapter, we describe different mouse models, including the knockout mouse,
recombinant mouse inbred strains, in vitro mouse cell-based assays, as well as novel experimental approaches
like the Collaborative Cross recombinant mouse inbred panel, which can be applied to preclinical pharma-
cogenetics research. These approaches can be used to assess drug response phenotypes that are difficult to
model in humans, thereby facilitating drug discovery, development, and application.
Key words Quantitative trait loci (QTL), Quantitative trait genes (QTG), Knockout (KO) mouse,
Recombinant inbred strain, Genome-wide association mapping, High content screening (HCS),
Collaborative cross (CC), Cell barcoding
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_17, © Springer Science+Business Media, LLC 2013
263
264 Amber Frick et al.
1 Introduction
Drug response and toxicity are complex traits, highly variable across
individuals, partly attributable to heredity and genetic diversity [1].
Experiments using model organisms can complement human
genomic studies with unique advantages, including the ability to
circumvent some issues that arise in clinical trials due to administra-
tion of potentially toxic or narrow therapeutic index drugs, allow for
risky or invasive procedures, control environmental factors that influ-
ence drug response such as diet, and reduce experimental cost [2].
Many mouse inbred strains have been well characterized both geno-
typically and phenotypically. These genetically diverse and stable
mouse populations are powerful tools for genome-wide association
pharmacogenetic studies. Classical laboratory mouse strains exhibit
variation in multiple phenotypes and have long been used for
genetic analysis of human disease. Furthermore, the mouse genome
can be easily manipulated, which makes them robust models to
identify and validate specific causative genes underlying toxic and
variable drug responses in humans. Detailed information regarding
mouse genomics can be found in Silver’s Mouse Genetics: Concepts
and Applications (1995) [3] and Hedrich’s The Laboratory Mouse
(Handbook of Experimental Animals) (2004) [4].
More recently, inbred strains derived from wild mice have been
created, adding genetic and phenotypic diversity to the pool of
available laboratory mice. Ideally, we can measure multiple drug
response phenotypes in vitro or in vivo across a panel of mouse
inbred strains to identify genes underlying variable responses to
drugs. Findings from mouse studies and high-throughput mouse
cell-based screens can help identify which genetic variants deter-
mine positive, negative, or non-response to pharmacologic agents.
This information can be used to develop and design subsequent
clinical trials. The use of pharmacogenetic information in clinical
trials can help ensure that only patients who are likely to respond
or patients who are less likely to display toxicity will be tested with
the novel agent. This approach will minimize drug exposure to
patients who are less likely to benefit from the new drug.
Additionally, conducting clinical trials in a targeted patient popula-
tion can make research efforts more cost-effective. Fewer patients
may be required to observe an effect, which will reduce cost and
shorten the time required to complete the study.
Targeted drug trial designs and personalized drug therapy
are possible through the use of pharmacogenetic information.
Therefore, it is important to use and develop innovative preclinical
tools that will facilitate pharmacogenetic research. This chapter
provides an initial guide as to how mouse models can be used for
identification of pharmacogenetically relevant genes, thereby facili-
tating efforts to advance drug development and medication ther-
apy management.
In Vitro and In Vivo Mouse Models 265
2 Methods
2.2 Knockout Mice Generation of genetically altered mice has been extremely useful for
analyzing gene function and mapping complex traits. Presently,
knockout (KO) mice are readily available, and the methodology for
generating new KO lines is well established. A review by Liggett
(2004) [4] provides practical guidance regarding the use of geneti-
cally modified mouse models for pharmacogenomic research. KO
mice are generally used in pharmacogenetic studies to examine the
effects of specific genes in mediating pharmacotherapeutic out-
comes. To generate KO mouse models, a modified segment of the
mouse gene is transfected into embryonic stem (ES) cells. Some
cells will then incorporate the transfected DNA in the target chro-
mosomal region through homologous recombination. Subsequently,
cells with altered DNA are isolated and implanted into mice in a
state of pseudopregnancy where the corpus luteum persists without
an embryo following estrus and breeding with an infertile male.
Characterization of the gene’s pharmacologic function is performed
by comparing drug response phenotypes between control and KO
mice [4]. For example, Hernandez et al. characterized the role of
flavin-containing monooxygenase family genes (FMOs) in mediat-
ing imipramine metabolism and central nervous system effects by
using mice lacking different Fmo genes [5]. Another group showed
that genetic ablation of glutathione S-transferase Pi (GstP1/P2(−/−))
led to resistance to acetaminophen-induced liver damage [6].
266 Amber Frick et al.
2.3 Quantitative Trait KO mice or transformed ES cells are restricted to assessing single-
Loci Identification gene effects. It is not often viable to ablate multiple genes to evalu-
ate multigenic effects on drug response. KO mouse models are
amenable to pharmacogenetic studies of genes with known or sus-
pected function. Often times, however, we do not have prior
knowledge regarding which genes may influence drug response,
thus necessitating the use of genome-wide association analysis for
identification of genetic loci significantly linked with drug response
in quantitative trait locus (QTL) regions. The most common
approach for QTL identification is to study a specific mouse refer-
ence population that was generated from strategic breeding experi-
ments. The aim is to establish a reference mouse population that
exhibits phenotypic and genotypic variation, providing the basis for
QTL mapping analysis. For a review regarding the use of different
mouse breeding approaches, the readers are referred to Darvasi
(1998) [9]. Typically, two parental mouse inbred strains that exhibit
significant difference in drug response phenotypes are mated (out-
cross), which leads to generation of a recombinant mouse popula-
tion (F1 or filial generation 1). Well-established linkage analysis
methodologies are available for genetic mapping studies of out-
cross-backcross (N2 or nuclear generation 2, resulting from an F1
mated to its parent) and outcross-intercross (F2, resulting from F1
brother–sister mating) mouse populations [10], and many of these
QTL mapping studies have successfully identified genomic regions
closely linked with drug response. For example, Haston et al.
mapped regions that influence differences in bleomycin-induced
pulmonary fibrosis by using two inbred strains with differential sus-
ceptibility to this drug [11]. QTL underlying variations in pheno-
types related to ethanol [12] and cocaine consumption [13] have
also been identified using F2 mouse populations.
In Vitro and In Vivo Mouse Models 267
2.4 High-Throughput The use of in vitro cell-based assays for pharmacogenomics studies
in Vitro Cell-Based provides unprecedented opportunities for researchers to assess
Assays for molecular response to drugs. In comparison to in vivo models, cell-
Characterization of based assays have higher assay versatility and scalability. In vitro
Inter-individual Drug cell-based assays can be conducted in a high-throughput fashion,
Responses Using allowing for multiple endpoints to be measured simultaneously.
Mouse Embryonic Importantly, large cell-based in vitro screens can be performed for
Fibroblasts from comparison of intra-individual cellular responses to drugs and tox-
Recombinant Inbred ins, thereby making identification of drug response QTG feasible.
Mouse Strains Cell-based assays can be developed for in vitro characterization
of pharmacological and cytotoxic responses. There is a broad selec-
tion of drug response phenotypes, including mutagenicity, carcino-
genicity, cytotoxicity, and teratogenecity that can be measured in
cell-based assays. To obtain biologically relevant results, it is impor-
tant to choose the appropriate endpoints to measure. Table 1 lists
a number of different drug response phenotypes that can be easily
measured through the use of commercially available cell-based kits.
270 Amber Frick et al.
Table 1
Endpoints commonly measured in cell assays and high content screening
Cellular process or
response Biomarkers and endpoints measured
Apoptosis Cell loss
Cell viability
Nuclear morphology
DNA content
Cell permeability
Mitochondrial mass
Mitochondrial membrane potential
Changes in the actin cytoskeleton
Caspase-3 activation
Caspase-9 activation
Cytochrome c localization
Cell cycle control p53 detection
p21 detection
Autophagy LC3B protein quantification
Cell proliferation BrdU incorporation
Ki-67 antigen quantification
3H-thymidine incorporation (DNA synthesis)
14C-methionine incorporation (protein synthesis)
Cell morphology F-actin and microtubule rearrangements
Oxidative stress Superoxide formation
MnSOD production
Phospho-H2AX detection
Cytochrome C reduction
Loss of critical molecules ATP depletion
Glutathione depletion
Cell membrane integrity LDH release assay
Membrane-impermeable DNA stain
different ethnic groups. These cell lines can be obtained from the
Coriell Institute repository (http://ccr.coriell.org). Several studies
have used these cell lines to perform genome-wide association
studies, investigating genetic variants linked with differential
responses to cisplatin, carboplatin, and etoposide [36–38].
Immortalized cell lines are an important tool for pharmacoge-
nomic research. However, immortalized cell lines do not exhibit
normal in vivo cellular functions and have dysfunctional apoptotic
and cell cycle control mechanisms. Primary cultures derived from
human and animal tissues are an important alternative to immor-
talized cells because these cell lines more closely mimic normal cell
functions and are thus more physiologically relevant experimental
models for drug screening, in vitro mechanistic characterization,
or gene discovery [39]. For detailed protocols on cell isolation and
culture from a variety of mouse tissues, please refer to Ward and
Tosh’s Mouse Cell Culture: Methods and Protocols [40]. Mouse
embryonic fibroblasts (MEFs) are an example of an effective pri-
mary cell line currently utilized for these purposes. MEFs are
advantageous because they exhibit features of primary cultures and
are easily manipulated for experimental purposes. Here, we pro-
vide a methodology that can assist in the design of a pharmacoge-
nomic high-throughput screen using MEFs.
2.5.1 Cell Culture One of the most important and challenging aspects in using cell
assays for high-throughput pharmacogenomic QTL mapping is to
use cells that are in the same growth phase at the time they are
plated for HCS. To ensure consistency in conditions across all cell
lines, all cells must have been through the same number of pas-
sages when expanded and have the same confluency before the
HCS procedure, thus minimizing environmental and experimental
variation between multiple cell lines. In addition, primary cells are
less accessible or robust and have a limited life span compared to
immortalized or tumor cell lines [41]. Therefore, the time required
for cell growth and culture is an important experimental
consideration.
In Vitro and In Vivo Mouse Models 273
2.5.2 Dosing the Cells After a recovery period, which varies between cell types, treat the
cells with the drug of interest by either adding the drug to the
existing media or replacing the media with a media combined with
drug. A serial dilution of the drug is most commonly used to dose
each of the cell lines.
2.5.3 Cell Assay 1. HCS and additional methodologies like flow cytometry can be
and Phenotyping easily multiplexed. There are multiple drug response pheno-
types that can be measured (Table 1). Caution should be exer-
cised to minimize experimental variation.
2. Collect cellular response data using an automated microscope,
flow cytometer, fluorescence plate reader, or other types of
detection instruments.
2.5.4 Data Analysis 1. Data collected should be processed according to the type of
assay used. In the case of HCS, the images obtained are pro-
cessed by segmentation algorithms that quantify different
aspects of cellular morphology.
2. Normalize experimental values to reference wells, which are
treated with vehicle. This data normalization approach mini-
mizes experimental variation and other variance unrelated to
drug response.
3. If cells were treated with multiple drug concentrations, a
dose–response curve can be obtained (Fig. 1). A single pheno-
typic value, usually IC50, is then calculated from the curve.
4. Calculated phenotypic values can be used for QTL mapping
using the methods previously described (Fig. 2).
274 Amber Frick et al.
Fig. 1 Dose response curves and IC50 values for eight rotenone treated MEF cell
lines obtained using high-content image screening. MEFs from 32 inbred mouse
strains were plated on 384-well clear bottom plates and treated with nine differ-
ent concentrations of rotenone, ranging from 0.015 to 100 µM. Triplicate wells
were used for each concentration of the compound. The cells were fixed and
stained after 72 hours of treatment, and plates were subsequently imaged using
an automated fluorescence high-content imaging microscope (BD Pathway 435).
The number of cells was estimated by software analysis of Hoechst-stained
nuclei within collected images. (a) Dose-response curves were calculated using
the responses at each dose normalized by the vehicle-only wells (DMSO treat-
ment), results for eight of the cell lines are presented on this graph. QTL mapping
was performed using the IC50 values obtained for all of the strains. Data points
have been slightly skewed on the x-axis to prevent overlapping of SEM bars and
enhance clarity. (b) IC50 results from 30 strains are presented including SEM
bars. The responses from two strains failed the curve fitting. Observed differ-
ences are statistically significant
In Vitro and In Vivo Mouse Models 275
Fig. 2 Genome-wide association analysis for rotenone IC50 values. IC50 values obtained from rotenone dose–
response curves were used for SNPster analysis. (a) Association plot between haplotypes inferred from a
sliding 3-SNP window across the entire mouse genome and rotenone IC50 values. The arrow indicates the
genomic region with the strongest association signal. (b) Amplification of the indicated region in the X chromo-
some, showing known and predicted genes and transcripts that lie in this interval and could potentially explain
the phenotypic variation
2.5.5 Candidate QTG The identification of candidate genes is usually based on biological
Identification and relevance of the QTL genes to the phenotype being studied.
Validation Pathway analysis tools (GeneGO, http://www.genego.com and
Ingenuity, http://www.ingenuity.com) can be useful to discover
such relationships. These tools contain rich databases of known
interactions that allow the researcher to quickly find connections
between the genes of interest and pathways affected by the drug.
In cell-based studies, the first step to validate the selected can-
didate genes is usually the knockdown and overexpression of the
genes, followed by drug treatment of the cell line used. This can be
achieved by the transfection of siRNA oligonucleotides or DNA
vectors. Different methods of transfection can be used. For a
review on this subject, please refer to Kim and Eberwine[42]. The
response of the transfected cells to drug treatment is then com-
pared with cells that have a normal expression of the gene; changes
are suggestive of gene involvement in drug response.
276 Amber Frick et al.
3 Future Directions
4 Conclusion
Acknowledgments
References
1. Zhang W, Dolan ME (2009) Use of cell lines 6. Henderson CJ et al (2000) Increased resis-
in the investigation of pharmacogenetic loci. tance to acetaminophen hepatotoxicity in mice
Curr Pharm Des 15(32):3782–3795 lacking glutathione S-transferase Pi. Proc Natl
2. Bogaards JJ et al (2000) Determining the best Acad Sci USA 97(23):12741–12745
animal model for human cytochrome P450 7. Eisener-Dorman AF et al (2009) Cautionary
activities: a comparison of mouse, rat, rabbit, insights on knockout mouse studies: the gene
dog, micropig, monkey and man. Xenobiotica or not the gene? Brain Behav Immun
30(12):1131–1152 23(3):318–324
3. Cheung C, Gonzalez FJ (2008) Humanized 8. Skarnes WC et al (2011) A conditional knockout
mouse lines and their application for predic- resource for the genome-wide study of mouse
tion of human drug metabolism and gene function. Nature 474(7351):337–342
toxicological risk assessment. J Pharmacol Exp 9. Darvasi A (1998) Experimental strategies for
Ther 327(2):288–299 the genetic dissection of complex traits in ani-
4. Liggett SB (2004) Genetically modified mouse mal models. Nat Genet 18(1):19–24
models for pharmacogenomic research. Nat 10. Zou F (2009) QTL mapping in intercross and
Rev Genet 5(9):657–663 backcross populations. Methods Mol Biol
5. Hernandez D et al (2009) Deletion of 573:157–173
the mouse Fmo1 gene results in enhanced 11. Haston CK et al (2002) Bleomycin hydrolase
pharmacological behavioural responses to and a genetic locus within the MHC affect risk
imipramine. Pharmacogenet Genomics 19(4): for pulmonary fibrosis in mice. Hum Mol
289–299 Genet 11(16):1855–1863
278 Amber Frick et al.
12. Drews E et al (2010) Quantitative trait loci con- 29. Mayr LM, Bojanic D (2009) Novel trends in
tributing to physiological and behavioural etha- high-throughput screening. Curr Opin
nol responses after acute and chronic treatment. Pharmacol 9(5):580–588
Int J Neuropsychopharmacol 13(2):155–169 30. Zock JM (2009) Applications of high content
13. Jones BC et al (1999) Quantitative-trait loci screening in life science research. Comb Chem
analysis of cocaine-related behaviours and neu- High Throughput Screen 12(9):870–876
rochemistry. Pharmacogenetics 9(5):607–617 31. Zanella F et al (2010) High content screening:
14. Boyle AE, Gill K (2001) Sensitivity of AXB/ seeing is believing. Trends Biotechnol 28(5):
BXA recombinant inbred lines of mice to the 237–245
locomotor activating effects of cocaine: a 32. Taylor DL, Giuliano KA (2005) Multiplexed
quantitative trait loci analysis. Pharmacogenetics high content screening assays create a systems
11(3):255–264 cell biology approach to drug discovery. Drug
15. McClurg P et al (2006) Comparative analysis Discov Today Techn 2(2):149–154
of haplotype association mapping algorithms. 33. Giuliano KA et al (2006) Systems cell biology
BMC Bioinformatics 7:61 based on high-content screening. Methods
16. Guo Y et al (2006) In silico pharmacogenetics: Enzymol 414:601–619
warfarin metabolism. Nat Biotechnol 24(5): 34. Abraham VC et al (2004) High content
531–536 screening applied to large-scale cell biology.
17. Harrill AH et al (2009) Mouse population- Trends Biotechnol 22(1):15–22
guided resequencing reveals that variants in 35. Shapiro HM (2003) Practical flow cytometry.
CD44 contribute to acetaminophen-induced Wiley, Hoboken
liver injury in humans. Genome Res 19(9): 36. Huang RS et al (2007) Identification of
1507–1515 genetic variants contributing to cisplatin-
18. Zhang X et al (2011) In silico and in vitro induced cytotoxicity by use of a genomewide
pharmacogenetics: aldehyde oxidase rapidly approach. Am J Hum Genet 81(3):427–437
metabolizes a p38 kinase inhibitor. 37. Shukla SJ et al (2009) Whole-genome
Pharmacogenomics J 11(1):15–24 approach implicates CD44 in cellular resis-
19. Kang HM et al (2008) Efficient control of tance to carboplatin. Hum Genomics 3(2):
population structure in model organism asso- 128–142
ciation mapping. Genetics 178(3):1709–1723 38. Bleibel WK et al (2009) Identification of
20. Pletcher MT et al (2004) Use of a dense single genomic regions contributing to etoposide-
nucleotide polymorphism map for in silico induced cytotoxicity. Hum Genet 125(2):
mapping in the mouse. PLoS Biol 2(12):e393 173–180
21. McClurg P et al (2007) Genomewide associa- 39. Marshak DR, Greenwalt DE (2007)
tion analysis in diverse inbred mice: power and Differentiating primary human cells in rapid-
population structure. Genetics 176(1):675–683 throughput discovery applications. Methods
22. Grubb SC et al (2009) Mouse phenome data- Mol Biol 356:121–128
base. Nucleic Acids Res 37(Database issue): 40. Ward A, Tosh D (2010) Mouse cell culture:
D720–D730 methods and protocols, 1st edn. Humana
23. Blake J.A., et al. (2009) The Mouse Genome Press, New York
Database genotypes::phenotypes. Nucleic 41. Freshney R (2005) Culture of animal cells—a
Acids Res 37(Database issue):D712-9. manual of basic technique, 5th edn. Wiley,
24. Olson H et al (2000) Concordance of the tox- Hoboken, NJ
icity of pharmaceuticals in humans and in ani- 42. Kim TK, Eberwine JH (2010) Mammalian cell
mals. Regul Toxicol Pharmacol 32(1):56–67 transfection: the present and the future. Anal
25. Jaeschke H et al (2002) Mechanisms of hepa- Bioanal Chem 397(8):3173–3178
totoxicity. Toxicol Sci 65(2):166–176 43. Churchill GA et al (2004) The Collaborative
26. Rausch O (2006) High content cellular screen- Cross, a community resource for the genetic
ing. Curr Opin Chem Biol 10(4):316–320 analysis of complex traits. Nat Genet 36(11):
27. Bullen A (2008) Microscopic imaging tech- 1133–1137
niques for drug discovery. Nat Rev Drug 44. Aylor DL et al (2011) Genetic analysis of com-
Discov 7(1):54–67 plex traits in the emerging collaborative cross.
28. Abraham VC et al (2008) Application of a Genome Res 21:1213–1222
high-content multiparameter cytotoxicity assay 45. Krutzik PO, Nolan GP (2006) Fluorescent cell
to prioritize compounds based on toxicity barcoding in flow cytometry allows high-
potential in humans. J Biomol Screen 13(6): throughput drug screening and signaling
527–537 profiling. Nat Methods 3(5):361–368
Chapter 18
Abstract
The hydrodynamic tail vein injection is a technique that is used to deliver nucleic acids into live mice.
Delivery through this method results in the in vivo transfection of foreign DNA primarily in the liver.
Here, we describe the use of this technique to test for regulatory activity of liver promoters and enhancers,
using a dual luciferase reporter system as the readable/measureable output and how this application can
be used for pharmacogenomic studies.
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_18, © Springer Science+Business Media, LLC 2013
279
280 Mee J. Kim and Nadav Ahituv
2 Materials
2.1 For the Injection Nucleic acid (10 μg/mouse of assayed plasmid; Renilla control,
such as pGL4.74[hRluc/TK] (Promega): 2 μg/mouse).
Heat source (heat lamp or heat box with 120 W bulb, such as:
Aladin Enterprises, Inc., Cat. # RHB.1812).
Heat pad(s) (such as reusable pads from SnapHeat.com; Cat. #
SH812 & SH88).
Source of anesthesia (e.g., isoflurane), gas chamber, and mouth piece.
Scale.
Mice: 21–25 g (see Note 1).
3 mL syringes (Becton Dickinson; Cat. #: 309585).
27½ gauge needles (Becton Dickinson; Cat. #: 305109).
Medical gauze pads (Kendall; Cat. #: 2187).
Delivery Solution (Mirus TransIT®-EE In Vivo; Cat. #: Mir5340;
[18]).
5 mL sterile centrifuge/plastic tubes to hold injection solution
(Argos Technologies; Cat. #: T2076S).
2.3 Nucleic Acid 1. For the analysis of promoters and their variants, it is typical to
Preparation clone at least 250 bp upstream of the transcriptional start site
(TSS) and approximately 100 bp downstream of the TSS.
2.3.1 Promoters
2. Promoter sequences and their variants are cloned into the
pGL4.11b [luc2P] (Promega) vector that contains the luciferase
reporter gene (Fig. 1).
3. An empty pGL4.11b [luc2P] vector (without an insert) is used
as a negative control for promoter assays. In addition, the
reference sequence of the assayed promoter is typically used
as a baseline to compare the promoter activity of nucleotide
variants [19].
2.3.2 Enhancers 1. For enhancers, there is a variety of methods and resources from
which one can select candidate sequences to test for enhancer
activity. These include but are not restricted to: comparative
genomics, ChIP-seq datasets for enhancer marks, DNase
hypersensitive sites and transcription factor binding site analy-
sis. For pharmacogenomic purposes, we have focused on using
some of these approaches to analyze regulatory sequences
around genes that are involved in ADME, such as liver mem-
brane transporters [19, 20].
2. The sequences that are tested for enhancer activity and their
variants are cloned into the pGL4.23 [luc/minP] (Promega)
PROMOTER Luciferase
OR
ENHANCER minP Luciferase
Fig. 1 A schematic illustration of the hydrodynamic tail vein injection assay. Promoters or putative enhancer
sequences are cloned into a luciferase reporter plasmid and co-injected with a Renilla luciferase reporter
plasmid (not shown) into the tail vein of the mouse. Luciferase activity is assayed 24 h post injection and
measured by a luminometer. minP: minimal promoter
282 Mee J. Kim and Nadav Ahituv
3 Methods
3.1 Day 1: The total volume required for injection (formula adopted from
Hydrodynamic Tail Mirus; see Note 4 [18]):
Vein Injection
mouse weight ( g )
Total volume (mL ) = 10% + 0.1mL delivery solution
3.1.1 Sample 10 g / mL
Preparation
However, to determine the actual volume of delivery solution,
the formula is rearranged and the addition of nucleic acids is taken
into account:
⎡volume of 10 mg test plasmid(mL) ⎤
Delivery solution (mL) = total volume(mL) − ⎢ ⎥
⎣+volume of 2mg Renilla plasmid(mL)⎦
Example 3A
A mouse to be injected weighs 24 g. The DNA concentration of
the test construct (TC) plasmid is 500 ng/μL and the Renilla
plasmid (RP) concentration is 200 ng/μL. To inject 10 μg of the
test construct and 2 μg of the Renilla, the total injection mix
volume would be:
24 g
Total volume (mL ) = 10% + 0.1mL delivery solution
10 g / mL
= 2.5mL
Hydrodynamic Tail Vein Assay for Pharmacogenomics 283
Example 3B
Construct ID Test 1
Construct volume (μL) 20
Renilla volume (μL) 10
Weight of mouse (g) Mirus (mL)
21.0 2.17
21.5 2.22
22.0 2.27
22.5 2.32
23.0 2.37
23.5 2.42
24.0 2.47
24.5 2.52
25.0 2.57
Construct DNA concentration (ng/μL) 500
Volume for 10 μg (μL) 20
3.2 Preparation 1. Dilate the tail vein by warming the mouse with a heat source,
of the Animal for such as a heat box (Fig. 2), prior to administering the gaseous
Injection anesthetic (see Note 5). Following 3–5 min in the heat box,
transfer the mouse into the anesthesia chamber (Fig. 3b).
The chamber used to administer the anesthetic can also be
fitted with a heat pad for optimal dilation.
2. After the mouse is anesthetized in the chamber, as it is transferred
to the injection station, weigh the animal on a scale to determine
Fig. 2 (a) Heat box (Aladin Enterprises, Inc., Cat. # RHB.1812); (b) mice being warmed up for tail dilation
Fig. 3 (a) Gaseous anesthesia machine; (b) anesthesia box with heat pad; (c) injection station with anesthesia
mouth piece and heat pad
Hydrodynamic Tail Vein Assay for Pharmacogenomics 285
Fig. 4 Anesthetized mouse on injection station, ready for tail vein injection
Fig. 5 Mouse tail vein injection. (a) Entry of the needle is approximately at a 45° angle; (b) As the needle is
inserted into the vein, the needle becomes more parallel with the tail and the solution is injected rapidly
between 4 and 8 s
thinner. As the needle inserts into the vein, move the needle
nearly parallel to the tail and insert the entire length of the
needle into the vein (see Fig. 5b); inject the injection solution
into the tail (see Note 6).
5. Inject the entire contents of the syringe within 4–8 s at a
constant rate.
6. Stop the bleeding by applying the medical gauze to the
injection site.
7. Take the mouse off the anesthetic (optional; we usually label
each mouse with a different number using a permanent marker
so as to know what construct was injected) and allow it to
recover in a new cage (see Note 7).
3.3 Day 2: Based on Herweijer et al. [3], whose study determined that the
Harvesting the Livers optimal liver expression levels of injected DNA is 24 h post injection,
and Luciferase livers are harvested at this time point.
Measurements
3.3.1 Preparation 1. Before sacrificing the mice, dilute lysis buffer in water to working
of Reagents concentration and aliquot 3 mL into labeled liver collection
tubes (see Note 8).
2. Prepare the Luciferase Assay Reagent II and Stop N Glo®
Reagent solutions, according to manufacturer’s protocol
(see Note 9).
3. Sacrifice mice according to approved animal protocols, dissect
the liver and place it in the numbered liver collection tubes
containing 3 mL of cold lysis buffer on ice (make sure all of the
liver tissue is entirely immersed in the lysis buffer).
4. Homogenize the livers for 1 min at high speed until there are
no observed liver chunks, keeping samples on ice before and
after homogenization. Use 70 % ethanol to clean the homog-
enizer between samples.
5. Transfer 1 mL of the liver homogenate to a labeled 1.5 mL
microcentrifuge tube and centrifuge at 4 °C for 30 min at
14,000 rpm.
6. During this centrifugation, aliquot 380 μL lysis buffer to a
newly labeled 1.5 mL microcentrifuge tube per sample and
store at 4 °C.
7. Upon completion of centrifugation of the liver homogenate,
transfer 20 μL of the supernatant (liquid in the top phase of the
homogenate) to the 1.5 mL microcentrifuge tube containing
380 μL lysis buffer that has been chilled to 4 °C and vortex
briefly (see Note 10).
8. Administer the appropriate volume of Luciferase Assay and
Stop-N-Glo® reagents into the diluted liver supernatant from
Hydrodynamic Tail Vein Assay for Pharmacogenomics 287
3.4 Analysis The background blank readings of lysis buffer are routinely sub-
of Luciferase Activity tracted from both the Luciferase and Renilla activity readings.
Readings In addition, the Luciferase values are divided by the Renilla activity
values (within each sample) to get a normalized relative Luciferase
activity value and the replicates are averaged together.
An alternate method of quantifying luciferase activity is a real
time approach, using in vivo imaging technology such as the IVIS
optical imaging system by Caliper Life Sciences [22]. This system
allows for the quantification of bioluminescence and/or lumines-
cence in vivo and avoids sacrificing mice to measure reporter gene
activity.
4 Notes
prior to their sedation. Do not keep mice in the heat box for
more than 20 min.
6. If the needle is inserted properly, the blood in the vein should
clear and injection of the solution should be without resistance.
If there is resistance upon pushing the plunger, the needle is not
placed properly into the vein. This is also evident, if, during
the injection the tail becomes swollen locally and appears to be
“perspiring” the solution that is injected out of the tail’s pores.
Likewise, if the injection is improperly administered, the anal
area of the mouse may also become swollen. Resistance may also
be experienced mid-injection, perhaps by the movement of the
needle out of the vein. If that is the case, continue to inject
the solution but pull the needle slightly out. This may alleviate
the resistance and allow for a successful injection.
7. The mouse should recover within 5 min of the injection. The
heart rate may slow or increase rapidly within the first minute
post injection, however, this should normalize. If the mouse
appears to be seizing after the injection, this may be an indicator
that either an air bubble or an impurity entered the circulation
and the mouse may not survive. Careful monitoring of the
mice post injection is necessary.
8. The collection tube used may be dependent on the homoge-
nizer; a 14 mL Falcon tube (Becton Dickinson, Cat. #: 352001)
is adequate for a rotor stator homogenizer. Additional lysis buf-
fer will be needed to dilute the supernatant in step 7 and to use
as a blank/plate control during the luminescence read.
9. Keep both solutions away from direct light and heat. They can
be stored at 4 °C until they are ready to aliquot.
10. Dilution of the supernatant may not be necessary, depending
on the luminometer’s range.
References
1. Zhang G, Budker V, Wolff JA (1999) High 6. Lewis DL et al (2002) Efficient delivery of
levels of foreign gene expression in hepato- siRNA for inhibition of gene expression in
cytes after tail vein injections of naked plasmid postnatal mice. Nat Genet 32:107–108
DNA. Hum Gene Ther 10:1735–1737 7. McCaffrey AP et al (2002) RNA interference
2. Eggenhofer E et al (2009) High volume naked in adult mice. Nature 418:38–39
DNA tail-vein injection restores liver function 8. Sen A et al (2003) Inhibition of hepatitis C
in Fah-knock out mice. Hepatology 25: virus protein expression by RNA interference.
1002–1008 Virus Res 96:27–35
3. Herweijer H, Zhang G et al (2001) Time course 9. Song E et al (2003) RNA interference target-
of gene expression after plasmid DNA gene ing Fas protects mice from fulminant hepatitis.
transfer to the liver. J Gene Med 3(3):280–291 Nat Med 9:347–351
4. Herweijer H, Wolff JA (2003) Progress and 10. Zender L et al (2003) Caspase 8 small inter-
prospects: naked DNA gene transfer and therapy. fering RNA prevents acute liver failure in
Gene Ther 10:453–458 mice. Proc Natl Acad Sci USA 100:
5. Herweijer H, Wolff JA (2007) Gene therapy 7797–7802
progress and prospects: hydrodynamic gene 11. Xu J et al (2005) Reduction of PTP1B by RNAi
delivery. Gene Ther 14:99–107 upregulates the activity of insulin controlled
Hydrodynamic Tail Vein Assay for Pharmacogenomics 289
fatty acid synthase promoter. Biochem Biophys 18. MIRUS, TransIT®-EE Hydrodynamic
Res Commun 329:538–543 Delivery Solution Protocol. http://www.
12. Keng VW et al (2011) Modeling hepatitis B mirusbio.com/assets/cms_files/protocols/
virus X-induced hepatocellular carcinoma in ML043.pdf. Accessed July 2011
mice with the sleeping beauty transposon 19. Choi JH et al (2009) Identification and charac-
system. Hepatology 53:781–790 terization of novel polymorphisms in the basal
13. Zhou T et al (2010) Intracellular gene transfer promoter of the human transporter, MATE1.
in rats by tail vein injection of plasmid DNA. Pharmacogenet Genomics 19:770–780
AAPS J 12:692–698 20. Kim MJ et al (2011) Functional characteriza-
14. Chalasani N et al (2010) Genome-wide asso- tion of liver enhancers regulating drug-
ciation study identifies variants associated with associated transporters. Clin Pharmacol Ther
histologic features of nonalcoholic fatty liver 89:571–578
disease. Gastroenterology 139:1567–1576 21. Simonet WS et al (1993) A far-downstream
15. Zhang H et al (2010) Genome-wide associa- hepatocyte-specific control region directs
tion study identifies 1p36.22 as a new suscep- expression of the linked human apolipoprotein
tibility locus for hepatocellular carcinoma in E and C-I genes in transgenic mice. J Biol
chronic hepatitis B virus carriers. Nat Genet Chem 268:8221–8229
42:755–758 22. Caliper Life Sciences, IVIS® Lumina II. http://
16. Chen WM et al (2008) Variations in the www.caliperls.com/products/preclinical-
G6PC2/ABCB11 genomic region are associ- imaging/ivis-luminaii.htm. Accessed July 2011
ated with fasting glucose levels. J Clin Invest 23. Promega Corporation, Dual-Luciferase®
118:2620–2628 Reporter Assay Technical Manual. http://
17. Schadt EE et al (2008) Mapping the genetic www.promega.com/resources/protocols/
architecture of gene expression in human liver. technical-manuals/0/dual-luciferase-reporter-
PLoS Biol 6:1020–1032 assay-system-protocol/. Accessed July 2011
Part IV
Abstract
Human genomics research has produced vast amounts of data that can be applied to or used to inform
pharmacogenomic studies. The Internet is an extremely useful resource for pharmacogenomics as many
Web sites provide access to data from genomic and clinical studies or host tools which can be used to
interpret findings or generate hypotheses. Human genetic variation can now easily be explored or visual-
ized through genome browsers and Web-based repositories which store the details of millions of human
germ-line and somatic genetic variants. Gene expression data from many different tissue and cell types are
available through Web-based repositories, and human genetic variants that associate with mRNA expres-
sion can be identified using Web data portals. Pharmacogenetic associations can be explored through
publically available data repositories and the functionality of genetic variants predicted through Web-based
bioinformatic tools. Furthermore, resources relating to currently used genetic tests are available online.
Large clinical and population studies, many linked to medical records, can be queried for the availability of
biospecimens or data. In the future, as the amount of genomic and associated clinical data increases, there
is little doubt that Web-based resources will continue to evolve and overcome barriers hindering their
efficient use, leading to systems-based approaches to pharmacogenomics.
Key words Genome browser, Genetic variation, Genotypes, Gene expression, eQTLs, Genetic
association studies, GWAS, Biorepositories, Bioinformatic tools
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_19, © Springer Science+Business Media, LLC 2013
293
294 Dylan M. Glubb et al.
Fig. 1 Publications related to genomic resources. PubMed searches for associated terms such as “genome
browser” (a) and “expression database” (b) show their rapid growth since the beginning of the twenty-first
century
Table 1
Repositories of germ-line DNA sequence and genetic variation
Table 1
(continued)
Table 2
Repositories of somatic (cancer) variation
Table 3
Repositories of gene expression data
lines is also available in the browser. The NCI60 cell lines have
been used to screen >100,000 chemicals and Cell Miner database
contains drug concentrations which inhibit cell growth by 50 %
(GI50), expression (gene, protein and miRNA), DNA methylation
and fingerprinting data generated from these cell lines. Queries can
be made by drug, gene, protein, miRNA, tissue and the corre-
sponding data is available for download. The Systems Biology and
Medicine Database (SBM DB) is another repository which stores
expression data from cell lines. SBM DB contains mRNA expres-
sion profiles from 112 different (normal and tumor) tissues and
cell lines measured using Affymetrix U133 microarrays. Searches
can be made by gene and relative expression visualized across the
different tissues and cells.
Table 4
Repositories of eQTL data
Table 5
Repositories of GWAS and clinical genetic associations
Table 6
Tools to predict SNP function
Table 7
Web sites related to pharmacogenetic testing
Table 8
Web sites for biorepositories and large cohorts with linked medical information
References
9. Myers RM et al (2011) A user’s guide to the 12. Angrist M (2009) Eyes wide open: the
encyclopedia of DNA elements (ENCODE). personal genome project, citizen science and
PLoS Biol 9:e1001046 veracity in informed consent. Per Med 6:
10. Nakamura Y (2007) The BioBank Japan 691–699
project. Clin Adv Hematol Oncol 5:696–697 13. Kho AN et al (2011) Electronic medical
11. Ollier W, Sprosen T, Peakman T (2005) UK records for genetic research: results of the
Biobank: from concept to reality. Pharmaco eMERGE consortium. Sci Transl Med 3:
genomics 6:639–646 79re1
Chapter 20
Abstract
The Pharmacogenomics Knowledge Base, PharmGKB, is an interactive tool for researchers investigating
how genetic variation affects drug response. The PharmGKB Web site, http://www.pharmgkb.org, dis-
plays genotype, molecular, and clinical knowledge integrated into pathway representations and Very
Important Pharmacogene (VIP) summaries with links to additional external resources. Users can search
and browse the knowledgebase by genes, variants, drugs, diseases, and pathways. Registration is free to the
entire research community, but subject to agreement to use for research purposes only and not to redis-
tribute. Registered users can access and download data to aid in the design of future pharmacogenetics and
pharmacogenomics studies.
1 Background
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_20, © Springer Science+Business Media, LLC 2013
311
312 Caroline F. Thorn et al.
Fig. 1 The PharmGKB homepage, http://www.pharmgkb.org, contains directed search boxes where users can
also browse from lists of genes, variants, drugs, or diseases
2 Overview
3 Initial Interactions with the PharmGKB Web Site: Gene, Drug, and Disease Pages
Fig. 2 The TMPT gene page showing genomic variants and related drugs with links to the annotations and
haplotypes and tabs for clinical pharmacogenomics, pharmacogenomics research, overview, VIP, haplotypes,
pathways, related drugs and diseases, and downloads and links out
PharmGKB: The Pharmacogenomics Knowledge Base 315
4 Curated Knowledge
4.1 Literature A basic literature annotation captures the genes, drugs, and diseases
Annotations involved in a single article from Pubmed and the category (or catego-
ries) of evidence that describe the type of relationships measured.
Our current process for literature annotation uses NLP to suggest
possible genes, drugs, and diseases to the curator [14, 15] but after
reading the article the curator decides which are appropriate.
4.2 Genomic Variant In addition to tagging articles for basic relationships curators can
Annotations and Very also describe in detail the relationships for individual variants and
Important their effects on drug response. The variant is mapped to the dbSNP
Pharmacogenes identifier and controlled vocabularies are used to define the alleles
or genotypes observed in the paper and their response to drug,
in the particular population studied. Information about the popu-
lation size, location or race and ethnicity, allele frequencies and
statistical measures can be captured and stored in the database.
Although time consuming, the benefit of annotating each indi-
vidual publication in such a detailed manner is that it will allow for
all kinds of computational analyses. PharmGKB currently has over
5,000 genomic variant annotations [April 2013].
316 Caroline F. Thorn et al.
4.3 Clinical Once there is sufficient evidence available from variant annotations
Annotations for a given variant and drug combination a clinical annotation is
written. This is a summary of the clinical relevance for each of the
individual genotypes that may be observed for a given gene variant
and drug combination. The PharmGKB’s clinical annotations reflect
expert consensus based on clinical evidence and peer-reviewed lit-
erature available at the time they are written and are intended only
to assist clinicians in decision-making and to identify questions for
further research. A strength of evidence score is given for clinical
annotations based on the type of study, number of study subjects,
and statistical significance reported.
Liver cell: Model human liver cell showing blood, bile and intestinal compartments, indicating tissue
specific involvement of genes in the irinotecan pathway.
Neutropenia
CES1 SN-38
Irinotecan CES2 BCHE
Cell Membrane
SLCO1B1 ABCC1
APC
M4
NPC
CYP3A4 CYP3A4 Liver cell
© Pharm
mG
mGKB
CYP3A4
CES2
UGT1A1
Irinotecan
SN-38
CES1 UGT1A9
CES2
SN-38G
Bile
ABCC2 Via
ABCB1
ABCC2 ABCG2
ABCB1 ABCC2
e
stin
Inte
UGT1A1
SN-38G
UGT1A10
Irinotecan
CES1 SN-38
CES2
Diarrhea
Fig. 3 The Irinotecan Pathway, view of a model human liver cell showing blood, bile, and intestinal compartments,
indicating tissue-specific involvement of genes in the irinotecan pathway. Drugs are depicted by purple boxes,
transporter genes by turquoise ovals, genes coding for metabolic enzymes by blue ovals. http://www.pharmgkb.
org/do/serve?objId=PA2001&objCls=Pathway
318 Caroline F. Thorn et al.
5 Future Directions
Since the year 2000, the PharmGKB has become the “go to” site
for pharmacogenetics and pharmacogenomics knowledge [36,
37]. In response to assessment of the field and feedback from
users, the priorities for the next 5 years include:
● Supporting data-sharing consortia in which multiple investigators
pool their data in collaboration with PharmGKB to answer
specific questions that require large datasets, not typically avail-
able to single research groups.
● Developing algorithms for text mining in order to identify
appropriate pharmacogenomics literature, and begin the process
of extracting the key genes, variations, drugs, and phenotypes
that form the basis for our curator annotations.
● Creating algorithms for the analysis of rare variations that
emerge from whole exome and whole genome sequencing
efforts. Most of the efforts to date in pharmacogenomics have
focused on the analysis of common variants, but the era of
genome sequencing has made it clear that a primary challenge
will be interpreting rare or novel variations found in individual
genomes.
● Helping lead the clinical implementation and impact of phar-
macogenomics knowledge in clinical settings. The contents of
PharmGKB can provide a base of peer-reviewed information
from which clinical guidelines can be constructed.
● Studying the molecular and cellular mechanisms of drug
response in order to provide the knowledgebase required to
understand the systemic effects of drugs, their side effects, and
their unexpected interactions.
Finally, we will evaluate how these and other activities impact
the requirements for the PharmGKB Web site, and consider its evo-
lution from a purely research repository of knowledge to a more
integrated research and clinical resource for personalized medicine.
Acknowledgments
References
1. NIH. Goals for the PGRN. http://www. 14. Garten Y, Altman RB (2009) Pharmspresso: a
nigms.nih.gov/Research/FeaturedPrograms/ text mining tool for extraction of pharmacoge-
PGRN/ nomic concepts and relationships from full
2. Altman RB, Klein TE (2002) Challenges for text. BMC Bioinformatics 10(Suppl 2):S6
biomedical informatics and pharmacogenom- 15. Coulet A et al (2010) Using text to build
ics. Annu Rev Pharmacol Toxicol 42:113–133 semantic networks for pharmacogenomics.
3. Povey S et al (2001) The HUGO Gene J Biomed Inform 43:1009–1019
Nomenclature Committee (HGNC). Hum 16. Chen J et al (2010) Interethnic comparisons of
Genet 109:678–680 important pharmacology genes using SNP
4. Wishart DS et al (2006) DrugBank: a compre- databases: potential application to drug regu-
hensive resource for in silico drug discovery latory assessments. Pharmacogenomics 11:
and exploration. Nucleic Acids Res 1077–1094
34:D668–D672 17. Sissung TM et al (2010) Clinical pharmacol-
5. Eichelbaum M et al (2009) New feature: path- ogy and pharmacogenetics in a genomics era:
ways and important genes from PharmGKB. the DMET platform. Pharmacogenomics 11:
Pharmacogenet Genomics 19:403 89–103
6. Sangkuhl K et al (2008) PharmGKB: under- 18. Gamazon ER et al (2009) A pharmacogene
standing the effects of individual genetic vari- database enhanced by the 1000 Genomes
ants. Drug Metab Rev 40:539–551 Project. Pharmacogenet Genomics 19:
7. Relling MV, Klein TE (2011) CPIC: clinical 829–832
pharmacogenetics implementation consortium 19. Feng J et al (2010) Compilation of a compre-
of the pharmacogenomics research network. hensive gene panel for systematic assessment of
Clin Pharmacol Ther 89:464–467 genes that govern an individual’s drug
8. Swen JJ et al (2011) Pharmacogenetics: from responses. Pharmacogenomics 11:1403–1425
bench to byte–an update of guidelines. Clin 20. Demir E et al (2010) The BioPAX community
Pharmacol Ther 89:662–673 standard for pathway data sharing. Nat
9. Bolton E, Wang Y, Thiessen PA, Bryant SH Biotechnol 28:935–942
(2008) PubChem: integrated platform of small 21. Desta Z et al (2009) Antiestrogen pathway
molecules and biological activities. In: Annual (aromatase inhibitor). Pharmacogenet
Reports in Computational Chemistry. Genomics 19:554–555
American Chemical Society, Washington, DC 22. Thorn CF, Klein TE, Altman RB (2009)
10. (US), N. L. o. M. MeSH Browser http:// Codeine and morphine pathway. Pharmacogenet
www.nlm.nih.gov/mesh/MBrowser.html Genomics 19:556–558
11. Organisation, I. H. T. S. D. SNOMED CT 23. Yang J et al (2009) Etoposide pathway.
http://www.ihtsdo.org/snomed-ct/ Pharmacogenet Genomics 19:552–553
12. Rubin DL et al (2005) A statistical approach 24. Marsh S et al (2009) Platinum pathway.
to scanning the biomedical literature for phar- Pharmacogenet Genomics 19:563–564
macogenetics knowledge. J Am Med Inform 25. Sangkuhl K, Klein TE, Altman RB (2009)
Assoc 12:121–129 Selective serotonin reuptake inhibitors path-
13. Altman RB et al (2003) Indexing pharmaco- way. Pharmacogenet Genomics 19:907–909
genetic knowledge on the World Wide Web. 26. Zaza G et al (2010) Thiopurine pathway.
Pharmacogenetics 13:3–5 Pharmacogenet Genomics 20:573–574
320 Caroline F. Thorn et al.
27. Gong L, Altman RB, Klein TE (2011) 33. Sangkuhl K, Klein TE, Altman RB (2011)
Bisphosphonates pathway. Pharmacogenet PharmGKB summary: citalopram pharmacoki-
Genomics 21:50–53 netics pathway. Pharmacogenet Genomics
28. Maitland ML et al (2010) Vascular endothelial 21(11):769–772
growth factor pathway. Pharmacogenet 34. Thorn CF et al (2011) Doxorubicin pathways:
Genomics 20:346–349 pharmacodynamics and adverse effects.
29. Sangkuhl K, Klein TE, Altman RB (2010) Pharmacogenet Genomics 21(7):440–446
Clopidogrel pathway. Pharmacogenet Genomics 35. Thorn CF et al (2011) PharmGKB summary:
20:463–465 fluoropyrimidine pathways. Pharmacogenet
30. Sangkuhl K et al (2011) Platelet aggregation Genomics 21:237–242
pathway. Pharmacogenet Genomics 21(8): 36. Sim SC, Altman RB, Ingelman-Sundberg M
516–521 (2011) Databases in the area of pharmacoge-
31. Oshiro C et al (2009) Taxane Pathway. netics. Hum Mutat 32:526–531
Pharmacogenet Genomics 19:979–983 37. Thorn CF, Klein TE, Altman RB (2010)
32. Mikkelsen TS et al (2011) PharmGKB summary: Pharmacogenomics and bioinformatics:
methotrexate pathway. Pharmacogenet PharmGKB. Pharmacogenomics 11:
Genomics 21(10):679–686 501–505
Chapter 21
Abstract
Pharmacogenomics studies how the variations of the individuals’ genetic makeup are correlated with a
person’s response to certain drugs in relation to the therapeutic efficiency, clinical outcome, or even sur-
vival, and how they affect drug metabolism, transport, or clearance. Yet, since the incidence of these poly-
morphisms, being either single-point variations or small insertions/deletions, varies among different
populations, a systematic collection and documentation of these variations is warranted, in order to facili-
tate implementation of pharmacogenomics in different populations. Here we review the existing electronic
databases related to pharmacogenomics and pay particular attention in the description of the pharmacoge-
nomics module Frequency of Inherited Disorders database (FINDbase), which documents curated allelic
frequency data pertaining to 144 pharmacogenomics markers across 14 genes, representing approximately
87,000 individuals from 150 populations and ethnic groups worldwide. Long-term sustainability of these
resources aims to contribute to the design, development, and implementation of pharmacogenomics test-
ing towards the application of personalized approaches in medical treatment.
Key words Database, Pharmacogenomics, Markers, Allelic frequencies, Populations, Ethnic groups,
Genes
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_21, © Springer Science+Business Media, LLC 2013
321
322 Marianthi Georgitsi and George P. Patrinos
5 Database Overview
5.1 Data Collection The main body of data is derived from the published literature
(http://www.ncbi.nlm.nih.gov/pubmed), mainly from original
reports or, occasionally, from review articles if the original publica-
tions were not accessible, according to the following criteria:
● The population and ethnicity should be clearly stated.
● The cohorts should be ethnically homogeneous.
● The subjects should be unrelated.
● Each population should be represented by a sufficient sample
size [i.e., ≥50 subjects (100 chromosomes)], with exceptions
for smaller cohort sizes made in the case of isolated popula-
tions or tribes studied less commonly.
● Each population should be represented only once for each
gene in the final data, by the largest available cohort, in order
to avoid redundant cases.
Data curation pertained initially to a careful selection of only
the pharmacogenomically relevant variants, followed by a reevalu-
ation of the calculated allelic frequencies per study, as some incon-
sistencies were identified pertaining to either the reported allele
frequencies in different parts of the same article (i.e., text and
tables), or the number of samples based on which rare allele fre-
quencies were eventually calculated, or the nomenclature used for
326 Marianthi Georgitsi and George P. Patrinos
5.2 System Design FINDbase-PGx is a publicly available database that is accessible via
and Access the URL http://www.findbase.org, and is being hosted at the
Golden Helix server (http://www.goldenhelix.org). There are no
registration requirements for data querying. The system architec-
ture and database schema were detailed in the original publication
Genetic Databases in Pharmacogenomics… 327
Table 1
Well-established pharmacogenes currently included in FINDbase-PGx, presented according
to their role in drug metabolism, transport, or action (adapted from ref. 21)
Fig. 1 Outline of the entire FINDbase-PGx data collection, consisting of variation boxes (see also Fig. 2)
Fig. 2 Pharmacogenomic markers are presented in FINDbase-PGx as “Variation cards.” (a) Example of a card
representing the NAT2 variation rs1801280 (alternatively known as NAT2*5), in the Portuguese population. (b)
The corresponding information box with data regarding this particular marker appears upon zooming in the
corresponding “Variation card”
5.3 Querying Engine FINDbase-PGx gives the user the possibility to view, organize, cat-
egorize, and reorganize data dynamically, owing to the various fil-
ters provided in the left side menu (Fig. 3), as detailed earlier.
Currently, it is not yet possible to download data, but the users are
provided with a user-friendly environment for on-site data analysis,
by sorting the acquired data, with the help of the options provided
in the upper right drop-down menu (Fig. 4). The queries may be
simple, such as to observe variants from a certain population only
(Fig. 4), specific allelic variants (Fig. 5), rare variants with minor
allele frequency <10 % (Fig. 6), variants associated with a certain
drug, variants included in a particular publication, and others.
However, queries may also be compound, including various com-
binations of the above.
One querying example is presented in Fig. 4, pertaining spe-
cifically to Chinese. In this example, the Chinese population is rep-
resented by a total of 226 alleles, as depicted by an equal number
of display items across 13 genes (presented in alphabetical order):
eight alleles for CYP1A2, 125 alleles for CYP2D6, 20 alleles for
CYP2E1, three alleles for CYP3A4, five alleles for CYP3A5, six
alleles for DPYD, four alleles for NAT2, one allele for PON1, two
330 Marianthi Georgitsi and George P. Patrinos
Fig. 4 Database query based on a specific population. A query for retrieving all pharmacogenetically relevant
variations for the Chinese population alone (left red arrow), sorted by gene name (right red arrow), returns 226
alleles in equal number of display items
Genetic Databases in Pharmacogenomics… 331
Fig. 5 Database querying based on pharmacogenomic marker allele frequency: Presentation of data pertaining
to marker rs2306283 (c.388A>G, alternatively known as *1b) of SLCO1B1, filtered by rare allele frequency
data (upper right sorting drop-down menu, reading “Sort:RareAlleleFrequency”)
Fig. 6 Database querying based on rare allele frequency: Presentation of all markers with rare allele frequency
0–10.44 %, sorted by gene name
alleles for PON2, four alleles for SLCO1B1, 10 alleles for TPMT,
34 alleles for TYMS, and four alleles for UGT1A1. These data can
be further scrutinized by zooming into specific ethnic groups
within the Chinese population (Han, Yao, Uygur), or based on the
geographical region (Chinese people from China, Hong Kong,
Singapore, or Malaysia).
332 Marianthi Georgitsi and George P. Patrinos
8 Conclusions
Acknowledgments
We wish to thank Sjozef van Baal for providing the building blocks
for FINDbase and FINDbase-PGx and for his continuous efforts.
A part of our own work has been funded by the European
Commission [ITHANET (FP6-026539), EuroGenTest (FP6-
512148), and GEN2PHEN (FP7-200754) projects], and the
Golden Helix Institute of Biomedical Research. Dr. Marianthi
Georgitsi is the recipient of a State Scholarship Foundation (IKY)
postdoctoral grant.
References
1. McKusick VA, Mendelian Inheritance in Man 6. Klein TE et al (2001) Integrating genotype
(1966) A catalog of human genes and genetic and phenotype information: an overview of
disorders. Johns Hopkins University Press, the PharmGKB project. Pharmacogenetics
Baltimore, MD Research Network and Knowledge Base.
2. Hamosh A et al (2005) Online mendelian Pharmacogenomics J 1:167–170
inheritance in man (OMIM), a knowledgebase 7. Davis A, Long R (2001) Pharmacogenetics
of human genes and genetic disorders. Nucleic research network and knowledge base: 1st
Acids Res 33:D514–D517 annual scientific meeting. Pharmacogenomics
3. Hardison RC et al (2002) HbVar: a relational 2:285–289
database of human hemoglobin variants and 8. George RA et al (2008) General mutation
thalassemia mutations at the globin gene databases: analysis and review. J Med Genet
server. Hum Mutat 19:225–233 45:65–70
4. Patrinos GP et al (2004) Improvements in the 9. Stenson PD et al (2003) Human Gene
HbVar database of human hemoglobin vari- Mutation Database (HGMD): 2003 update.
ants and thalassemia mutations for population Hum Mutat 21:577–581
and sequence variation studies. Nucleic Acids 10. Claustres M, Horaitis O, Vanevski M, Cotton
Res 32:D537–D541 RG (2002) Time for a unified system of muta-
5. Cotton RG, McKusick V, Scriver CR (1998) tion description and reporting: a review of
The HUGO mutation database initiative. locus-specific mutation databases. Genome
Science 279:10–11 Res 12:680–688
336 Marianthi Georgitsi and George P. Patrinos
11. Cotton RG, Phillips K, Horaitis O (2007) A using SNP databases: potential application to
survey of locus-specific database curation. drug regulatory assessments. Pharmacoge-
Human Genome Variation Society. J Med nomics 11:1077–1094
Genet 44:e72 23. Georgitsi M et al (2011) Transcriptional regula-
12. Patrinos GP (2006) National and ethnic muta- tion and pharmacogenomics. Pharmacogenomics
tion databases: recording populations’ genog- 12:655–673
raphy. Hum Mutat 27:879–887 24. Bell M (2010) SOA modeling patterns for
13. Wheeler DL et al (2008) Database resources of service-oriented discovery and analysis. Wiley
the National Center for Biotechnology & Sons, Inc., Hoboken, NJ
Information. Nucleic Acids Res 36:D13–D21 25. Giardine B et al (2011) Systematic documen-
14. Thorisson GA et al (2009) HGVbaseG2P: a tation and analysis of human genetic variation
central genetic association database. Nucleic in hemoglobinopathies using the microattri-
Acids Res 37:D797–D802 bution approach. Nat Genet 43:295–301
15. Sipila K, Aula P (2002) Database for the muta- 26. Lagoumintzis G, Poulas K, Patrinos GP
tions of the Finnish disease heritage. Hum (2010) Genetic databases and their potential
Mutat 19:16–22 in pharmacogenomics. Curr Pharm Des 16:
16. Patrinos GP et al (2005) The Hellenic national 2224–2231
mutation database: a prototype database for 27. Squassina A et al (2010) Realities and expecta-
mutations leading to inherited disorders in the tions of pharmacogenomics and personalized
Hellenic population. Hum Mutat 25: medicine: impact of translating genetic knowl-
327–333 edge into clinical practice. Pharmacogenomics
17. Kleanthous M et al (2006) The cypriot and 11:1149–1167
Iranian National Mutation databases. Hum 28. Stein L (2002) Creating a bioinformatics
Mutat 27:598–599 nation. Nature 417:119–120
18. Patrinos GP, Kollia P, Papadakis MN (2005) 29. Ring HZ, Kwok PY, Cotton RG (2006)
Molecular diagnosis of inherited disorders: les- Human Variome Project: an international col-
sons from hemoglobinopathies. Hum Mutat laboration to catalogue human genetic varia-
26:399–412 tion. Pharmacogenomics 7:969–972
19. van Baal S et al (2007) FINDbase: a relational 30. Horaitis O et al (2007) A database of locus-
database recording frequencies of genetic specific databases. Nat Genet 39:425
defects leading to inherited disorders world- 31. Kaput J et al (2009) Planning the human vari-
wide. Nucleic Acids Res 35:D690–D695 ome project. The spain report. Hum Mutat
20. Georgitsi M et al (2011) FINDbase: a world- 30:496–510
wide database for genetic variation allele fre- 32. Patrinos GP, Wajcman H (2004) Recording
quencies updated. Nucleic Acids Res 39: human globin gene variation. Hemoglobin
D926–D932 28:5–7
21. Georgitsi M et al (2011) Population-specific 33. Axton M (2008) Human variome microattri-
documentation of pharmacogenomic markers bution reviews. Nat Genet 40:1
and their allelic frequencies in FINDbase. 34. Patrinos GP, Petricoin EF (2009) A new scien-
Pharmacogenomics 12:49–58 tific journal linked to a genetic database:
22. Chen J, Teo YY et al (2010) Interethnic com- towards a novel publication modality. Hum
parisons of important pharmacology genes Genomics Proteomics 1:e597478
Chapter 22
Abstract
In this chapter, we use calculation of estimated warfarin maintenance dosage as an example to illustrate
how to develop a multiple linear regression model to quantify the relationship between several indepen-
dent variables (e.g., patients’ genotype information) and a dependent variable (e.g., measureable clinical
outcome).
Key words Multiple regression, Warfarin dosing algorism, CYP2C9, VKORC1, INR, Pharmacogenetics,
Personalized medicine
1 Introduction
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_22, © Springer Science+Business Media, LLC 2013
337
338 Lu Yang and Mark W. Linder
2 Study Design
2.1 Note ● Ideally the mechanism of effect for a predictive genetic factor
on the outcome is the primary consideration for inclusion
within the model. However, validated statistical association in
the absence of mechanistic explanation may provide equivalent
input to the model but may limit opportunities for appropriate
accommodation in practice. For example, the knowledge that
CYP2C9 genetic variants reduce S-warfarin clearance not only
contributes to estimation of maintenance dose, but also is
instructive in terms of timing INR measurements with time to
reach steady state [11].
● The study population size depends on the allele frequency of
interest and might be relatively large if the genetic variants are
rare.
● Experimental approaches for data collection need to be selected
appropriately in terms of efficiency, reliability, and economic
situation.
340 Lu Yang and Mark W. Linder
3 Model Fitting
4 Model Validation
5 Summary
References
Abstract
Implementing pharmacogenetics in daily clinical practice has the potential to improve patient care. The
translation of results of pharmacogenetic studies into practical pharmacotherapeutic recommendations is
essential. These recommendations are preferably available at the time of drug prescribing and drug dis-
pensing. This chapter describes a process of developing evidence based drug dosing and pharmacothera-
peutic guidelines per genotype by the Dutch Pharmacogenetics Working Group. It is aimed to provide
recommendations in case drugs are prescribed to a patient whose genotype is known. Furthermore, several
examples are given. Many drugs are metabolized by the Cytochrome P450 CYP2D6 enzyme. Carriage of
genetic variants of the CYP2D6 gene can result in a predicted phenotype of poor, intermediate or ultrar-
apid metabolizer. Dose adjustments, pharmacotherapeutic and monitoring recommendations are described
for several CYP2D6 substrates, when initiated in patients with the above mentioned phenotypes.
1 Introduction
In the past years, both the number and quality of published studies
on the association between genetic variants and the pharmacokinet-
ics or pharmacodynamics of drugs has increased enormously. Studies
include larger populations and clinically relevant endpoints are more
frequently evaluated. The FDA and EMA have encouraged and
requested to add pharmacogenetic information to drug labels.
Pharmacogenetic testing has become cheaper en more readily avail-
able. Many researchers, health care providers and those involved in
health care policy have stated that implementing pharmacogenetics
in daily clinical practice is a major step forward to personalized med-
icine. By implementing pharmacogenetics, one aims to improve
The Pharmacogenetics Working Group is part of the Pharmacogenetics Project of the Royal Dutch Association for the
Advancement of Pharmacy
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7_23, © Springer Science+Business Media, LLC 2013
345
346 Vera H.M. Deneer and Ron H.N. van Schaik
Predicting
drug Better,
concentration Better patient Better public
PGx testing individualized,
or outcome health
prescribing
response
patient care and therefore improve public health (Fig. 1). However,
the implementation of pharmacogenetics in routine patient care is
still limited. One of the main reasons may be that studies have not
resulted in practical dose prescribing recommendations per geno-
type. In 2005, The Royal Dutch association for the advancement
of Pharmacy initiated the Pharmacogenetics Project for which
they established a multidisciplinary working group (Dutch
Pharmacogenetics Working Group; DPWG) in which (hospital)
pharmacists, medical doctors, clinical pharmacologists, clinical (bio)
chemists, and epidemiologists participate. The aim of the project
was (1) to evaluate gene–drug interactions by systematic review, (2)
to give drug dosing and pharmacotherapeutic recommendations
per genotype, (3) to make recommendations available at the time of
prescribing drugs by medical doctors and drug dispensing by phar-
macies. This approach did not include recommendations on whether
or not genotyping is thought to be necessary in case of certain drugs
or certain clinical conditions of patients, but aimed at providing
drug dosing and other pharmacotherapeutic recommendations in
case the genotype of the patient is known. It was anticipated that in
the near future genotyping is more often applied as part of routine
patient care in case of problems regarding pharmacotherapy such as
the development of side effects or insufficient efficacy of drugs.
Once a patient’s genotype is known, it should be taken into account
at the time other drugs are initiated.
Monitoring of pharmacotherapy of individual patients is rou-
tinely performed by using electronic drug prescribing systems or
drug dispensing systems with an incorporated database. The pre-
scribing and dispensing systems contain patient characteristics such
as prescribed drugs, a patient’s genotype. The incorporated data-
base includes information regarding drug interactions and gene–
drug interactions (Fig. 2). Implementation in daily practice means
Evidence Based Drug Dosing and Pharmacotherapeutic Recommendations… 347
Fig. 2 Implementation of pharmacogenetics database into drug prescribing and dispensing software
Table 1
Scoring system for level of evidence of gene–drug interaction [4]
Level of evidence
4 Published controlled studies of good qualitya relating to phenotyped and/or genotyped patients
or healthy volunteers, and having relevant pharmacokinetic or clinical endpoints
3 Published controlled studies of moderate qualityb relating to phenotyped and/or genotyped
patients or healthy volunteers, and having relevant pharmacokinetic or clinical endpoints
2 Published case reports, well documented, and having relevant pharmacokinetic or clinical
endpoints. Well documented case series
1 Published incomplete case reports. Product information
0 Data on file
– No evidence
Population size was not assessed when assigning the level of evidence but dose adjustments were calculated as the popu-
lation size-weighted mean
a
“Good quality” criteria include:
− The use of concomitant medication with a possible effect on the phenotype is reported in the manuscript
− Confounders are reported (e.g., smoking status)
− The reported data are based on steady-state kinetics
− Results are corrected for dose variability
b
Wherever one or more of these “good quality” criteria was missing, the quality of the study was considered to be
“moderate”
Table 2
Scoring system for clinical relevance of gene–drug interaction [4]
Table 2
(continued)
Fig. 3 Calculation of dose adjustment for a phenotype based on one article using AUC as pharmacokinetic
parameter [4]. D dose, PM poor metabolizer, AUC area under the plasma concentration versus time curve, EM
extensive metabolizer. The calculated dose adjustment is the percentage of the dose usually prescribed or
recommended
Fig. 4 Calculation of the overall dose adjustment for a phenotype as the population size-weighted mean of
calculated dose adjustments per article [4]. D dose, PM poor metabolizer, N(a), number of subjects of study “a”;
DPM(a), calculated dose adjustment for PMs based on the results of study “a,” “b,” “c,”…“x” represent other
articles. The calculated overall dose adjustment is the percentage of the dose usually prescribed or recom-
mended. The percentage by which the dose should be reduced is calculated as 100 % minus DPM(%)
Table 3
Classification of gene–drug interactions on whether an interaction
exists or an action is required in case the combination occurs
in an individual patient
Table 4
Gene–drug interactions of several CYP2D6 substrates [4]
References
1. Sachse C et al (1997) Cytochrome P450 2D6 haloperidol treatment. Clin Pharmacol Ther
variants in a Caucasian population: allele fre- 72:438
quencies and phenotypic consequences. Am J 4. Swen J et al (2008) Pharmacogenetics: from
Human Genet 60:284 bench to byte. Clin Pharmacol Ther 83:781
2. Kirchheiner J et al (2003) Effects of polymor- 5. Kirchheiner J et al (2004) Pharmacogenetics of
phisms in CYP2D6, CYP2C9, and CYP2C19 antidepressants and antipsychotics: the contri-
on trimipramine pharmacokinetics. J Clin bution of allelic variations to the phenotype of
Psychopharmacol 23:459 drug response. Mol Psychiatry 9:442
3. Brockmöller J et al (2002) The impact of 6. Swen J et al (2011) Pharmacogenetics: from
the CYP2D6 polymorphism on haloperidol bench to byte—an update of guidelines. Clin
pharmacokinetics and on the outcome of Pharmacol Ther 89:662
INDEX
Federico Innocenti and Ron H.N. van Schaik (eds.), Pharmacogenomics: Methods and Protocols,
Methods in Molecular Biology, vol. 1015, DOI 10.1007/978-1-62703-435-7, © Springer Science+Business Media, LLC 2013
355
PHARMACOGENOMICS: METHODS AND PROTOCOLS
356 Index
Copy number variations (CNVs)..............71, 72, 82, 84, 156, DMET. See Drug metabolizing enzyme and transporter
172–175, 214, 215, 219–222, 296, 298, 300, 302 (DMET)
CP. See Competitive probe (CP) DNA
CpG sites.................................................. 179–182, 184, 186 chromosomal .............................................. 190, 193, 231
Crohn’s disease ......................................................... 129, 216 genomic .........................28, 51, 57, 61–64, 67, 78, 79, 83,
CT value ....................................................................184–186 88–93, 95, 96, 132, 135, 159, 164, 176, 180–183,
Custom multiplex single nucleotide polymorphism 207, 227, 231, 234
mutation assay.........................................115–126 methylation..............................9, 179–182, 185, 302, 306
CYP enzymes sequencing ..................... 10, 143, 229, 267, 295, 297–299
CYP1A2 ..................................................... 251, 254, 256 DPWG.
CYP2C9 ............................................. 251, 252, 254, 256 See Dutch Pharmacogenetics Working Group (DPWG)
CYP2C9 variant status .........................................252 Drugbank ................................................................. 313, 315
CYP2D6......................................251, 252, 254, 256, 347 Drug development ................. 5, 10, 13, 16–17, 264, 267, 325
CYP2D6 PMs ......................................................348 Drug-induced liver injury (DILI).................................11, 18
CYP2E1 ............................................................. 254, 256 Drug interactions...............................4, 16, 17, 251, 346–352
CYP3A4 ............................................................. 251, 256 Drug metabolism....................................5, 11, 55, 76, 87–96,
CYP3A5 ............................................................. 251, 256 251–261, 265, 316, 327
CYPs. See Cytochrome P450s (CYPs) Drug metabolism genotyping assays.............................87–96
CYP-selective inhibitors................................... 256, 257, 260 Drug metabolizing enzyme and transporter
Cytochrome P450s (CYPs) ................................ 15, 251–261 (DMET) ......................................... 142, 180, 182
genes ................................................................... 180, 182
D Drug-metabolizing enzymes (DMEs)................. 4, 6, 12, 55,
Database of Genomic Variants (DGV) .................... 295, 296 87, 142, 225, 265, 279, 325–327
Database of Genotypes and Phenotypes Drug response
(dbGaP) .................................. 295, 298, 303, 304 phenotypes................... 214, 264–266, 268, 269, 273, 277
Databases phenotypes, multiple........................................... 264, 277
incorporated................................................................346 Drug transporters ..............................225–227, 279, 326, 327
locus-specific databases (LSDBs) ............... 323, 332–333 Dutch Pharmacogenetics Working Group
mutation databases (DPWG) ................................ 314, 346–348, 350
human gene mutation database (HGMD) ...........323
E
national/ethnic mutation databases
(NEMDBs) .................................... 323–325, 328 Efficient Mixed Model Association (EMMA) ................268
relational .....................................................................324 EGFR. See Epidermal growth factor receptor (EGFR)
Data quality ...........................................82–84, 315, 323, 324 Encyclopedia of DNA elements
dbGaP. See Database of Genotypes and Phenotypes (ENCODE) ........................................... 305, 306
(dbGaP) Endoplasmic reticulum (ER) .........14, 62, 219, 226–227, 244
Deletions ...........................9, 27, 71, 72, 75, 77, 88, 101, 116, endoplasmic reticulum-associated
137, 140, 175, 192, 196, 222, 296, 300, 326 degradation (ERAD) .............. 226, 227, 242, 244
Denaturing high-performance liquid chromatography Enhancers ................................................30, 52, 53, 279–288
(DHPLC) Epidermal growth factor receptor (EGFR) ..................13, 14
denaturation step, initial .........................................36, 37 ER. See Endoplasmic reticulum (ER)
mutation detection..................................................25–53 Ethnic groups ............................... 8–10, 12, 13, 66, 272, 297,
single-base extension (SBE) genotyping ......................27 322–326, 328, 331–333
SURVEYOR Nuclease ......................... 27–28, 30, 43, 53 Ethnic populations ........................................... 217, 295, 297
DGV. See Database of Genomic Variants (DGV) Exomes ......................................134, 136–142, 297, 298, 318
Diseases sequencing ...................................134, 136–142, 297, 298
cardiovascular.......................216, 247–248, 269, 333–334 Expression quantitative trait loci
gout.....................................................................247–248 (eQTLs)...................214, 217–221, 223, 302–303
human................................................. 127, 264, 277, 334 data .....................................................................302–303
inflammatory bowel disease (IBD) ..................... 141, 216
Parkinson’s disease (PD) .............................................136 F
Disorders, complex ........................................... 128, 129, 136 False discovery rate (FDR) ....................................... 169, 170
DMEs. See Drug-metabolizing enzymes (DMEs) Flow cytometry................................................. 116, 271, 273
PHARMACOGENOMICS: METHODS AND PROTOCOLS
Index
357
Frequency of Inherited Disorders database high-throughput SNP genotyping ..................... 217, 218
(FINDbase) ............................ 296, 297, 321–335 platform .............................................. 214, 217, 218, 298
FINDbase-PGx .................................. 325–330, 332, 333 GEO. See Gene Expression Omnibus (GEO)
GoldenGate genotyping ...........................................147–152
G GenomeStudio software .............................................151
Lymphoblastoid cell lines (LCLs) ................... 202–204, 209, Pharmacogenetics ....................... 4–6, 8–10, 16, 17, 115–126,
214, 217, 219, 302, 303 179–187, 221, 263–277, 293, 294, 296, 297, 304,
306–307, 311, 314–317, 322, 324, 337–340,
M 345–348, 350, 351
MAF. See Minor allele frequencies (MAF) Pharmacogenetics and pharmacogenomics knowledge base
MALDI. See Matrix-assisted laser desorption/ionisation (PharmGKB) ..................142, 294, 306, 311–319,
(MALDI) 322, 327, 328, 332, 333
Matrix-assisted laser desorption/ionisation (MALDI) Pharmacogenetics project ......................... 345, 346, 348, 350
mass differences ............................................................74 Dutch Pharmacogenetics Working
MassEXTEND primers .........................................77, 79 Group (DPWG) .....................................346–348
mass signals ..................................................................79 Pharmacogenetics Research Network (PGRN)....... 311, 313,
Mean threshold cycle................................................ 185, 186 316, 322
Membrane vesicles ....................226, 227, 237–241, 245–247 Pharmacogenomics
Metabolism .......................5–7, 11, 16, 17, 55, 71, 76, 87–97, applications ..............................10–17, 219–222, 297, 332
142, 248, 251–261, 265, 267, 268, 271, 279, 316, clinical ............................................................................5
327, 332 implementation of ...........................5, 217–219, 318, 325
Methotrexate (MTX) ............................................... 241, 248 Plasma membrane vesicles ................ 226, 238–241, 245–247
Microarrays..................... 9, 10, 155, 156, 162, 164–166, 169, Plasmid controls .......................................................190–192
171–176, 300–302 genotype control ......................................... 189, 193–195
Minor allele frequencies (MAF) ....................... 130, 148, 149 Polymerase chain reaction (PCR)
Minor groove binder (MGB) .......................................88–90 amplification .......................... 36, 72, 88, 90–94, 118, 120
Misfolded proteins ..................................................... 62, 227 asymmetric .........................................................118–121
Mouse embryonic fibroblasts (MEFs) ......................269–275 buffer ........................................................ 52, 77, 79, 121
mRNA levels ............................................ 201, 234, 242, 243 efficiency............................................. 184, 185, 187, 191
Multiplex ligation-dependent probe primers...............31, 32, 36, 45, 49, 74, 75, 77–79, 83, 97,
amplification (MLPA) ....................................118 101–105, 111–113, 118, 120, 124, 150, 151,
Mutations 192–195, 231–233
causative.............................................. 128, 134, 136, 324 real-time quantitative..................181, 182, 184–186, 202,
thalassemia..................................................................332 206, 207
Polymorphisms
N copy number ..................................9, 65, 66, 71, 156, 222
microsatellite ................................................................65
National Human Genome Research Institute (NHGRI)
multinucleotide (MNPs) ..............................................88
catalog ............................................. 303, 304, 308
nonsynonymous ...............................66, 67, 226, 230, 247
Natural language processing (NLP) .................................315
regulatory.............................................. 88, 201, 202, 217
NCI60 cell lines................................................ 299, 301, 302
single nucleotide (SNPs) .......... 7, 9, 71, 75, 88, 115–126,
Next-generation sequencing (NGS) ......8, 127–144, 299, 300
128, 156, 182, 225–248, 267, 312, 322, 326
NLP. See Natural language processing (NLP)
Poor metabolizers (PMs) .....................5–8, 16, 346, 348, 351
Non-small cell lung cancer (NSCLC) ..........................13–14
Predictive model .......................................................337–343
Non-synonymous SNPs ......................61, 225–248, 304, 305
Promoters ..............................64–66, 162, 179, 228, 279–288
NSCLC. See Non-small cell lung cancer (NSCLC)
Pyrosequencing
O assays ....................... 98, 99, 101, 102, 105–107, 111–114
dispensation order....................................... 106, 108, 114
Online Mendelian inheritance in mis-priming ........................................................ 112, 113
man (OMIM) ................................. 303, 322, 328 predicted histograms................................... 106, 109–111
primer design ........................................ 98, 101, 102, 112
P
PCR. See Polymerase chain reaction (PCR) Q
Personal genome project (PGP) ............................... 307, 308 Quantitative structure-activity
Personalized medicine .................. 55, 56, 144, 225, 318, 322, relationship (QSAR)............................... 226, 241
325, 333, 335, 337, 345–346 Quantitative trait genes (QTG) ............... 267–269, 275–277
PGP. See Personal genome project (PGP) identification of .................................. 267–269, 275–277
Pharmacoepigenetics ................................................ 180, 181 Quantitative trait loci (QTL) ............266–269, 274, 275, 277
Pharmacogenes ......................................... 315–316, 326, 327 mapping analysis................................. 266–269, 272, 273
PHARMACOGENOMICS: METHODS AND PROTOCOLS
Index
359
S TaqMan
drug metabolism enzymes (DME) genotyping
Sanger sequencing .............................130, 143, 180, 181, 190 assays...........................................................87–96
SCAN............................................................... 213–223, 302 genotyper software............................................ 88, 95, 96
Scanning electron microscopy (SEM), 245, 274 minor groove binder (MGB) probes .......................89, 90
Sequencing no-template controls (NTCs) ........................... 90, 93, 96
massively parallel ................................................ 130, 280 probes ..............................................88, 91, 182, 184, 234
technologies ................... 10, 128, 130, 133, 143, 202, 280 Transcription factor binding ..................... 201–210, 281, 306
whole-genome ............. 134, 136–143, 293, 297, 299, 318 Transport activity..............................226, 230, 239, 241–242,
Sequenom ..............................................67, 74, 77, 79, 82–84 246–247
Serious adverse drug reactions (SADRs) ................ 10, 12, 18
Significance analysis of microarrays (SAM) ............. 169, 170 U
Single nucleotide polymorphisms (SNPs) ......7, 9, 71, 75, 88,
UCSC genome browser ....................269, 294–296, 300, 301,
115–126, 128, 156, 182, 225–248, 267,
305, 306
312, 322, 326
UGT1A1 ............................. 7, 12, 65, 66, 326, 327, 331, 333
arrays ...........................................142, 156, 164, 172–174
UK Biobank ............................................................. 307, 308
coding ...........6, 87, 97, 207–210, 215, 216, 297, 305, 326
function, bioinformatic analyses of .............................305 V
genotyping assay ....................... 39, 77, 83, 87–91, 94, 95,
151, 204, 207 Very Important Pharmacogene (VIP) ......................313–316
regulatory.....................................207, 208, 210, 305, 326 Vitamin K oxide reductase (VKORC1) .............. 7, 8, 15–16,
tri-allelic ......................................................... 66, 67, 106 64, 65, 306, 326, 333, 337, 338, 340, 342
variants ............................... 62, 68, 72, 77, 134, 207, 217, W
226, 227, 231, 236, 241–244, 246–248, 304, 305
SmartAmp Warfarin ....... 7, 8, 15–16, 18, 64, 65, 267, 306, 334, 337–343
method ...................................................................55–68 WAVE system .......................................28, 29, 43, 44, 47, 53
primer design ................................................................64 Whole-exome sequencing ........................ 134, 136–142, 298