Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Dec 1.
Published in final edited form as: Semin Perinatol. 2014 Oct 3;38(8):534–540. doi: 10.1053/j.semperi.2014.08.017

Is Personalized Medicine Achievable in Obstetrics?

Sara K Quinney 1, David A Flockhart 2, Avinash S Patil 1
PMCID: PMC4252827  NIHMSID: NIHMS632844  PMID: 25282474

Abstract

Personalized medicine seeks to identify the right dose of the right drug for the right patient at the right time. Typically, individualization of therapy is based on the pharmacogenomic make-up of the individual and environmental factors that alter drug disposition and response. In addition to these factors, during pregnancy a woman’s body undergoes many changes that can impact the therapeutic efficacy of medications. Yet, there is minimal research regarding personalized medicine in obstetrics. Adoption of pharmacogenetic testing into the obstetrical care is dependent on evidence of analytical validity, clinical validity, and clinical utility. Here, we briefly present information regarding the potential utility of personalized medicine for treating the obstetric patient for pain with narcotics, hypertension, and preterm labor and discuss the impediments of bringing personalized medicine to the obstetrical clinic.


Obstetrics is a discipline focused on the care of women during pregnancy, an inherently normal phase of life. Unlike other medical specialties, obstetric care providers oversee a natural process which has been successfully navigated by women for thousands of years, even before modern medicine. Over time, care for the patient with abnormal pregnancy or medical complications of pregnancy has been added to the spectrum of care provided by obstetricians. These conditions are often corrected through procedural interventions on the mother or fetus, or through medical management.

Perhaps as a consequence of the perception of normalcy and concerns for harming the fetus, only a limited number of therapies have been developed to treat the complications of pregnancy. Medications approved by the US Food and Drug Administration (FDA) for use in pregnancy fall into a small number of major categories: tocolytics, antiemetics, labor induction agents, and a few others. In comparison to many other fields of medicine, the rate of approval of new medications for obstetrical indications has been slow. Uncharacteristically, the past four years has seen the FDA approval of 2 medications indicated for use in pregnancy: hydroxyprogesterone caproate (Makena®, Ther-Rx Corporation) and doxylamine + pyridoxine (Diclegis®, Duchesnay USA, Inc.). However, both medications represent a repackaging of previously accepted therapies. Hydroxyprogesterone caproate has been used since the late-20th century as a progestational agent, though it was not until the turn of the century that usage became more widespread1. Diclegis® is a variant of Bendectin®, a medication previously approved for treatment of hyperemesis gravidarum in pregnancy. The story of Bendectin® has been previously outlined extensively and represents an important lesson in the dangers inherent in fear about teratogenicity from medication usage in pregnancy2. The components of these two medications have been used off-label for years, though safety concerns still lingered among clinicians. The FDA approval of Makena® and Diclegis® formalize the acceptance of use of the medications in pregnancy. The approval of these two agents, with a long history of off-label use in pregnancy, also accentuates the difficulty in developing drugs to treat conditions in pregnancy. Truly innovative development of drug therapy in obstetrics leading to drug approval by the FDA has not occurred in many years.

Beyond medications used for obstetric indications, pregnant women are also exposed to treatments for medical co-morbidities complicating pregnancy. The selection of a medication to treat a given pathology (e.g., lupus, hypertension, seizure disorders) rests on evidence of efficacy in the non-pregnant population, balanced against teratogenicity or correlation with poor obstetric outcomes. Historically, the FDA has maintained a pregnancy drug rating system to summarize the known information on risk of drug usage in pregnancy. This A-B-C-D-X letter rating has been misinterpreted by many clinicians and has led to the withholding of therapy due to the perceived risk of medications within specific classes3. As early as 1997, the FDA recommended changes in drug labeling to move towards a descriptive rating system to encourage clinicians to more carefully consider the evidence supporting the use of a drug in pregnancy4,5.

The variability in patient response to medications that occurs in general, and to an even greater extent in pregnancy, has been a factor in the increased perception of risk during this period. In addition to baseline genetic diversity, the physiologic changes of pregnancy can accentuate inter-individual differences in drug response. These physiologic changes can alter the ADME (absorption, distribution, metabolism, elimination) properties of a drug and its resultant efficacy. For example, 17α-hydroxyprogesterone caproate administered to prevent recurrent preterm delivery may have a decreased effect in women with particular progesterone receptor subtypes6. The variability in patient response to 17α-hydroxyprogesterone caproate may be further compounded by inter-individual differences in creatinine clearance, volume of distribution, or other contributing factors during pregnancy. Betamethasone is provided to promote fetal maturity in cases of anticipated preterm delivery; however, the effects are not uniform in all treated fetuses. Recently, genetic variations have been identified that may influence the success of this intervention7.

The unpredictability of adverse events related to patient variability in obstetrics has led to undesirable consequences. Providers favor older drugs with minimal risks over newer, untested medications. Litigation resulting from drug-related adverse events has further dampened enthusiasm to provide appropriate therapy to pregnant patients. Paradoxically, few drugs are routinely monitored during pregnancy despite the increased variability. Therapeutic drug monitoring is limited by a lack of available clinical assays for most drugs.

Despite these obstacles, developments in obstetric therapeutics now occur against the background of a growing body of pharmacogenomic data on drugs in general, which can be used to improve the therapeutic range of the available therapies for pregnant women and their children. Persuasive arguments have been advanced in support of using genetic information to guide the choice of drug, the dose involved and the need for monitoring8,9. The FDA has included pharmacogenomic information in over 130 drug labels with over 50 genes implicated in drug efficacy or safety10. A large portion of pharmacogenomics information in drug labels relates to drug metabolism enzymes and many are in the form of “black box” warnings. It is estimated that one fourth of all outpatients receive one or more drugs with pharmacogenomic information in the label for that drug11.

Personalized Medicine in Obstetrics

The pregnant woman differs in many aspects from the non-pregnant patient. Not only are there longitudinal physiologic changes that effect drug pharmacokinetics12,13, but the genetic make-up of the fetus differs from that of the mother. While in some cases fetal genetics may play a role in fetal response to drugs14, maternal genetics will likely play the larger role in determining disposition and response. In addition, although pharmacogenomics is increasingly recognized as a key determinant of response for drugs, few studies have been conducted in pregnant women. Thus, as with many areas of obstetrics, we must extrapolate knowledge of pharmacogenomics to this orphaned population.

The FDA labeling for several drugs commonly prescribed by obstetricians contains pharmacogenomics information (Table 1). Below, we provide examples of pharmacogenetic information of potential value to pregnant women and their health care providers, including data relevant to the use of narcotics to treat severe pain and the treatment of hypertension and preterm labor.

Table 1.

Pharmacogenomic information in the FDA label of drugs commonly administered to pregnant women.

Drug Name Gene Pharmacogenomic Information in FDA Label
Tramadol CYP2D6 Concentrations in PM’s were 20% higher than in EM’s
Codeine CYP2D6 Respiratory depression and death have occurred in UM children and in breast-fed infants whose mothers are UM’s
Hydralazine NAT1-2 Mean absolute bioavailability varies from 10–26% with higher percentages in PM’s; EM’s have lower exposure
Metoprolol CYP2D6 EM’s who concomitantly take CYP2D6 inhibitors and PM’s have increased concentrations, decreasing metoprolol’s cardioselectivity
Glyburide G6PD Hemolytic anemia linked to G6PD deficiency
Esomeprazole & Omeprazole CYP2C19 Induction of CYP3A4 by St. John’s wort led to 37.9% decrease in omeprazole AUC in PM’s and a 43.9% decrease of AUC in EM’s
Lansoprazole CYP2C19 Concomitant administration with tacrolimus may increase whole blood levels of tacrolimus, especially in transplant patients who are IM’s or PM’s. Coadministration to EM’s taking clopidogrel reduced the AUC of clopidogrel’s active metabolite by 14%.
Pantoprazole CYP2C19 PM’s have elimination half-life of 3.5 to 10 hours; in EM’s, 71% of the dose is excreted in urine and 18% through biliary excretion
Metoclopramide CYB5R1-4 G6PD Patients with NADH-cytochrome b5 reductase deficiency are at an increased risk of developing methemoglobinemia and/or sulfhemoglobinemia when metoclopramide is administered. In patients with G6PD deficiency who experience metoclopramide- induced methemoglobinemia, methylene blue treatment is not recommended.
Nitrofurantoin G6PD Hemolytic anemia linked to G6PD deficiency
Citalopram CYP2C19 Cmax and AUC increased by 68% and 107% in PM’s. Highest recommended dose in PM’s is 20 mg/d due to risk of QT prolongation
Fluoxetine CYP2D6 PM’s have higher concentrations of S-fluoxetine, and lower concentrations of S-norfluoxetine, at steady state. There is no effect of CYP2D6 metabolism status on pharmacodynamics of fluoxetine.
Paroxetine CYP2D6 In EM’s, concomitant administration of paroxetine increased the AUC and Cmax of atomoxetine.

CYP2D6: cytochrome P450 2D6; NAT1-2: N-acetyltransferase 1 and 2; G6PD: glucose-6-phosphate dehydrogenase; CYP2C19: cytochrome P450 2C19; CYB5R1-4: cytochrome b5 reductase 1–4; PM: poor metabolizer; EM: extensive metabolizer; UM: ultra-rapid metabolizer; AUC: area under the plasma concentration-time curve; Cmax: maximum plasma concentration

Narcotics

Peripartum pain is commonly treated by narcotic pain relievers, such as codeine and hydrocodone. These prodrugs require biotransformation through CYP2D6 metabolism to their active moieties, morphine and hydromorphone, respectively. CYP2D6 activity is induced during pregnancy15. In addition, more than 80 pharmacogenomic variants have been reported for CYP2D6 (http://www.cypalleles.ki.se.cypalleles.com), many of which alter the activity of the enzyme (Table 2). Approximately 7% of Caucasians have a variant that leads to a CYP2D6 poor metabolizer (PM) phenotype, which results in reduced enzyme activity. For instance, these individuals have reduced capacity to convert codeine to morphine16,17 and do not obtain adequate pain relief from codeine. Conversely, about 2–3% of Caucasians possess multiple copies of active CYP2D6 alleles, leading to an ultrarapid metabolism (UM) phenotype. In these individuals, codeine is rapidly converted to morphine, potentially leading to toxicity16,17. While rare, cases of death in UM individuals treated with clinical doses of codeine have been reported1820. In some of these individuals, the presence of a variant that reduces the activity of UDP-Glucuronosyltransferase-2B7 (UGT2B7), the enzyme responsible for inactivation of morphine, may have also contributed to the toxic concentrations of morphine. After the report of an infant death associated with the CYP2D6 UM genotype of a breastfeeding mother taking codeine for post-Cesarean pain relief21, the U.S. FDA issued a Public Health Advisory cautioning women on the use of narcotic analgesics during breastfeeding22. Additionally, the Clinical Pharmacogenomics Implementation Consortium (CPIC) has issued guidelines on the use of codeine with respect to CYP2D6 genotype17. While hydrocodone and oxycodone undergo similar metabolic activation via CYP2D6, there are not adequate data regarding the consequences of PM or UM phenotype for use of these agents.

Table 2.

Common CYP2D6 alleles, functional effect, and frequency in African Americans and Caucasiansa

Allele Activity Allele Frequency, Mean(Range)%b
African Americans Caucasians
*1 Normal 40 (30–83) 54 (28–83)
*2 Normal 14 (4–29) 27 (10–40)
*3 Non-functional 0.3 (0–0.6) 1 (0–3)
*4 Non-functional 6.2 (4–8) 18 (10–33)
*5 Non-functional 6 (2–9) 3 (0–7)
*6 Non-functional 0.2 (0–1) 1 (0–3)
*9 Reduced 0.5 (0–1) 2 (0–5)
*10 Reduced 4 (3–8) 3 (0.4–15)
*17 Reduced 18 (13–26) 0.3 (0–1.1)
*36 Non-functional 0.6 (0–1) 0
*41 Reduced 9 (2–15) 9 (4–14)
*1×N Increased 0.4 (0–1.2) 0.8 (0–4)
*2×N Increased 1.6 (0.1–2) 1.3 (0–6)
*4×N Non-functional 2 (0.3–4) 0.3 (0–1)
a

Adapted from Clinical Pharmacogenetics Implementation Consortium Guidelines for Cytochrome P450 2D6 Genotype and Codeine Therapy: 2014 Update.17

Antihypertensives

Commonly used to treat hypertension in pregnancy, metoprolol is metabolized primarily by CYP2D6. A meta-analysis of studies in non-pregnant individuals recently identified a 15-fold difference in apparent oral clearance of metoprolol between ultrarapid and poor metabolizers for CYP2D623. In addition to the pharmacogenetic variation of CYP2D6, the enzyme’s increased activity in pregnancy15 may necessitate increased doses of metoprolol compared to those used in non-pregnant women.

Similar to metoprolol, labetalol’s half-life is decreased during pregnancy24. A recent study found that gestational age and lean body weight were significantly associated with oral clearance25. Labetalol is cleared predominantly by glucuronidation through UGT1A1 and UGT2B726. The documented increase in UGT1A1 expression during pregnancy has been attributed to the induction of UGT1A1 by progesterone26. A study in healthy Chinese males was unable to confirm an association with UGT1A1 genotype, but did find higher plasma concentrations of labetolol in CYP2C19*2*2 expressers, accounting for 60% of the total variation in plasma exposure27, indicating that oxidative metabolism may be an important component of labetalol’s clearance. To our knowledge, no studies of pharmacogenomics of labetalol in pregnancy have been conducted. Hydralazine, a vasodilator available for the treatment of hypertension since 1952, is one of the few medications available to treat hypertensive emergencies in pregnancy, including severe preeclampsia. Hydralazine is primarily metabolized and cleared from the body by the N-acetyltransferase enzyme28. Based on a small study examining the metabolism of caffeine, N-acetyltransferase activity does not appear to significantly change during pregnancy29. That said, functional polymorphisms in the genes encoding the two human n-acetyltransferases, NAT1 and NAT2, are common28. Over 50% of Caucasians are slow acetylators, leading to increased plasma concentrations of hydralazine, and therefore, increased risk of toxicity.

Treatment of Preterm Labor

While the β2 adrenergic receptor agonists, e.g. ritodrine, terbutaline, and hexoprenaline, have fallen out of favor in the treatment of preterm labor due to increased adverse effects and limited efficacy, it is important to note the potential for interindividual variability in response to the agents. There is evidence that polymorphisms in the β2 adrenergic receptor (ADRB2) are protective against preterm delivery3032. In addition, Landau et al. found that homozygosity in Arg16 improved the tocolytic response to hexoprenaline33. Additional studies are needed to examine the effect of polymorphisms in ADRB2 on response to β2 adrenergic receptor agonists. However, clinicians should be aware of the potential contribution of pharmacogenetics to the interindividual variability in response to these drugs, which may necessitate the increase of dose to improve efficacy or decreasing the dose to prevent adverse drug events.

The calcium channel blocker nifedipine is metabolized by CYP3A enzymes in the liver and gastrointestinal tract. CYP3A activity increases during pregnancy15,34, resulting in higher clearance of nifedipine35. Since CYP3A activity is highly variable within and between individuals, it is not surprising that plasma concentrations of nifedipine are highly variable (30–70%) among pregnant women. Studies of nifedipine in pregnant women have found that CYP3A5 genotype and concomitant administration of CYP3A inhibitors, such as clarithromycin, erythromycin, and fluoxetine, are associated with altered nifedipine pharmacokinetics36,37. In addition to the potential effect of polymorphisms in drug metabolizing enzymes, genetic polymorphisms in components of the L-type calcium channel (CANC1C, CACN1D) and the large-conductance calcium and voltage-dependent potassium channel ß1 subunit gene (KCNMB1) are associated with responsiveness and risk of cardiovascular side effects in patients taking calcium channel blockers for hypertension3840. At present, it is unknown if these variants influence the tocolytic response to nifedipine.

The prostaglandin inhibitor indomethacin is widely used as a tocolytic, and is primarily metabolized through O-demethylation by CYP2C941. The activity of CYP2C9 is increased during pregnancy12,42,43. Additionally, CYP2C9 is highly polymorphic with 10–20% of Caucasians and up to 6% of blacks expressing poor metabolism phenotypes arising from CYP2C9*2 or CYP2C9*3 alleles44. It is estimated that 50% of indomethacin clearance is due to CYP2C9, and poor metabolism status is predicted to increase exposure by 1.8-fold in non-pregnant individuals45. However, to our knowledge, no clinical studies have examined the effect of CYP2C9 genotype in pregnancy or on indomethacin tocolysis.

While its effectiveness as a tocolytic is marginal46, magnesium sulfate is commonly used for fetal neuroprotection4749. Magnesium sulfate is cleared through renal filtration with homeostatisis maintained by reabsorption through a variety of passive and active transport mechanisms50. To our knowledge, the pharmacogenomics of magnesium sulfate have not been examined.

Betamethasone and, less commonly, dexamethasone are administered to women at risk of preterm birth to promote fetal lung maturity. Several studies have examined pharmacogenomics of corticosteroids, although few have focused on this indication. Pharmacogenomic studies in asthma have associated variants in several genes in the corticosteroid pathway with responsiveness to inhaled corticosteroids: corticotropin releasing hormone receptor 1 (CRHR1)51; T-box 21 (TBX21)52; neurokinin receptor 2 (NK2R)52; Stress-induced phosphoprotein 1 (STIP1)53; dual specificity phosphatase 1 (DUSP1)54; the Low affinity IgE receptor, FCER2 (rs28364072)55; and the glucocorticoid-induced transcript 1 gene (GLCC1)56. In a study of 62 preterm neonates born to mothers who had received betamethasone, the I105V variant in glutathione-S-transferase-P1 (GST-P1) was associated with reduced occurrence of respiratory distress syndrome57. GST-P1 is the primary GST isoform expressed in the placenta, and this variant leads to reduced enzyme activity, which could result in increased fetal concentrations of betamethasone. Haas et al. conducted a pharmacogenomic study examining SNPs in the betamethasone metabolic pathway in cohort of 109 pregnant women and 117 maternal-neonatal pairs treated with betamethasone for fetal lung maturation14. A multivariate analysis controlling for various demographic and clinical factors found a statistically significant association of maternal CYP3A5 genotype, maternal N43C1 (rs41423247), fetal ADCY9 (rs2230739), and fetal CYP3A7*1E with neonatal respiratory distress syndrome (RDS). Of these genes, fetal CYP3A7*1E genotype was found to have the highest odds ratio of 23.68 (95% CI: 1.33–420.6) for development of RDS14, potentially due to an increased clearance of betamethasone by CYP3A7. Additional analysis of this same cohort identified SNPs in maternal or fetal importin 13 (IPO13) that were associated with outcomes such as surfactant use and bronchopulmonary dysplasia7.

Clinical Utility of Pharmacogenomic Tests in Obstetrics

Despite the widespread availability of a large number of pharmacogenetic tests58, the widespread use of pharmacogenomic testing is inherently dependent on the generation of evidence that supports it. Three key elements of evidence are required: analytical validity, clinical validity and clinical utility.

Analytical validity refers to the reproducibility of a given test in the laboratory. In contrast, clinical validity refers to the ability of a test to act as a robust predictor of a clinical parameter, for example, a pharmacokinetic variable that summarizes drug exposure such as the plasma half-life or area under the plasma drug concentration-time curve (AUC). Clinical utility assesses the ability of a test to reliably predict a change in treatment necessitated by the result of the genetic test. The treatment change may include a change in drug, dose, or monitoring59. While analytical validity is widely available and accepted for most clinically available pharmacogenetic tests, clinical validity and utility have been less well studied or documented59.

These criteria should be met in pregnant women prior to applying pharmacogenomics testing to obstetric therapeutics. However, research to document clinical utility will require significant effort and breadth of vision, since clinical utility has not been established in cases specific to pregnant patients. That said, this is also the case for many drugs used both in obstetrics and pediatrics where drugs are often prescribed “off label” because of the lack of trials in the appropriate populations. However, as with other aspects of drug therapy in pregnancy, it may be acceptable to extrapolate pharmacogenetic information from the non-pregnant population. For example, it is reasonable to presume that the pharmacogenetic testing for HLA-B*5701 that is standard of care before the use abacavir in HIV patients to prevent potentially fatal and disfiguring abacavir-related skin hypersensitivity60 should also be provided to pregnant women with HIV.

Economic considerations are also important. While many pharmacogenetic tests are now available in the United States for less than a few hundred dollars, this is not universally the case. Some institutions and laboratories continue to charge thousands of dollars for tests that cost much less to carry out. The economic value of most pharmacogenetic tests, even in general practice, remains a notably understudied area6163. There is an increasing realization that pharmacogenetic testing may be a means that can be used by large health care systems to improve the quality of care and save costs by reducing the significant human and economic damage brought about by adverse drug reactions64, and ensuring that appropriate therapies are given to the patients most likely to benefit.

Conclusion

Obstetrical patients have long been an orphaned population with respect to medical advancement. However, a recent directive from the Institute of Medicine’s committee on Women and Health Research that promotes the inclusion of pregnant women in clinical studies65 and the establishment of the NICHD’s Obstetric and Fetal Pharmacology Research Unit Network symbolize a recognition that the nearly four million women who are pregnant annually in the United States66 are not immune to diseases necessitating drug therapy. While obstetrics may trail behind other medical specialties in the development of personalized medicine, in some cases knowledge obtained from other therapeutic areas can be extrapolated to the pregnant population. Additionally, the principles of analytic, clinical, and perhaps economic validity of pharmacogenomic testing developed in other populations can guide the implementation of personalized medicine to obstetrical patients. Development of models that bring together an individual’s pharmacogenetic make-up and the physiologic changes associated with pregnancy may eventually guide individualization of drug selection and dosage during pregnancy to optimize drug benefit in the obstetric patient.

Acknowledgments

This work was supported by the Obstetric-Fetal Pharmacology Research Units Network grant number 1U10HD063094-0, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health. SKQ is also supported by 1 K23 HD071134-01A1, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosure

The authors report no proprietary or commercial interest in any product mentioned or concept discussed in this article.

References

  • 1.Hall NR. What agent should be used to prevent recurrent preterm birth: 17-P or natural progesterone? Obstetrics and gynecology clinics of North America. 2011 Jun;38(2):235–246. ix–x. doi: 10.1016/j.ogc.2011.02.014. [DOI] [PubMed] [Google Scholar]
  • 2.Bishai R, Mazzotta P, Atanackovic G, et al. Critical appraisal of drug therapy for nausea and vomiting of pregnancy: II. Efficacy and safety of diclectin (doxylamine-B6) The Canadian journal of clinical pharmacology = Journal canadien de pharmacologie clinique. 2000 Autumn;7(3):138–143. [PubMed] [Google Scholar]
  • 3.Doering PL, Boothby LA, Cheok M. Review of pregnancy labeling of prescription drugs: is the current system adequate to inform of risks? Am J Obstet Gynecol. 2002 Aug;187(2):333–339. doi: 10.1067/mob.2002.125740. [DOI] [PubMed] [Google Scholar]
  • 4.U.S. Food and Drug Administration. Pregnancy and lactation labeling. 2011 http://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/Labeling/ucm093307.htm. Accessed 02/17/2014.
  • 5.Department of Health and Human Services. Content and format of labeling for human prescription drug and biological products; requirements for pregnancy and lactation labeling (proposed rules) Federal Register. 2008;73(104):30831–30868. [Google Scholar]
  • 6.Manuck TA, Lai Y, Meis PJ, et al. Progesterone receptor polymorphisms and clinical response to 17-alpha-hydroxyprogesterone caproate. Am J Obstet Gynecol. 2011 Aug;205(2):135 e131–139. doi: 10.1016/j.ajog.2011.03.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Haas DM, Dantzer J, Lehmann AS, et al. The impact of glucocorticoid polymorphisms on markers of neonatal respiratory disease after antenatal betamethasone administration. Am J Obstet Gynecol. 2013 Mar;208(3):215 e211–216. doi: 10.1016/j.ajog.2012.12.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Collins F. Opportunities and challenges for the NIH–an interview with Francis Collins. Interview by Robert Steinbrook. The New England journal of medicine. 2009 Oct 1;361(14):1321–1323. doi: 10.1056/NEJMp0905046. [DOI] [PubMed] [Google Scholar]
  • 9.Green ED, Guyer MS. National Human Genome Research I. Charting a course for genomic medicine from base pairs to bedside. Nature. 2011 Feb 10;470(7333):204–213. doi: 10.1038/nature09764. [DOI] [PubMed] [Google Scholar]
  • 10.U.S. Food and Drug Administration. Pharmacogenomic Biomarkers in Drug Labeling. 2014 http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/ucm083378.htm. Accessed 01/31/2014.
  • 11.Frueh FW, Amur S, Mummaneni P, et al. Pharmacogenomic biomarker information in drug labels approved by the United States food and drug administration: prevalence of related drug use. Pharmacotherapy. 2008 Aug;28(8):992–998. doi: 10.1592/phco.28.8.992. [DOI] [PubMed] [Google Scholar]
  • 12.Anderson GD. Pregnancy-induced changes in pharmacokinetics: a mechanistic-based approach. Clin Pharmacokinet. 2005;44(10):989–1008. doi: 10.2165/00003088-200544100-00001. [DOI] [PubMed] [Google Scholar]
  • 13.Abduljalil K, Furness P, Johnson TN, Rostami-Hodjegan A, Soltani H. Anatomical, physiological and metabolic changes with gestational age during normal pregnancy: a database for parameters required in physiologically based pharmacokinetic modelling. Clin Pharmacokinet. 2012 Jun 1;51(6):365–396. doi: 10.2165/11597440-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 14.Haas DM, Lehmann AS, Skaar T, et al. The impact of drug metabolizing enzyme polymorphisms on outcomes after antenatal corticosteroid use. Am J Obstet Gynecol. 2012 May;206(5):447 e417–424. doi: 10.1016/j.ajog.2012.02.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hodge LS, Tracy TS. Alterations in drug disposition during pregnancy: implications for drug therapy. Expert opinion on drug metabolism & toxicology. 2007 Aug;3(4):557–571. doi: 10.1517/17425225.3.4.557. [DOI] [PubMed] [Google Scholar]
  • 16.Kirchheiner J, Seeringer A. Clinical implications of pharmacogenetics of cytochrome P450 drug metabolizing enzymes. Biochimica et biophysica acta. 2007 Mar;1770(3):489–494. doi: 10.1016/j.bbagen.2006.09.019. [DOI] [PubMed] [Google Scholar]
  • 17.Crews KR, Gaedigk A, Dunnenberger HM, et al. Clinical Pharmacogenetics Implementation Consortium Guidelines for Cytochrome P450 2D6 Genotype and Codeine Therapy: 2014 Update. Clin Pharmacol Ther. 2014 Jan 23; doi: 10.1038/clpt.2013.254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Madadi P, Shirazi F, Walter FG, Koren G. Establishing causality of CNS depression in breastfed infants following maternal codeine use. Paediatric drugs. 2008;10(6):399–404. doi: 10.2165/0148581-200810060-00007. [DOI] [PubMed] [Google Scholar]
  • 19.Madadi P, Sistonen J, Silverman G, et al. Life-threatening adverse events following therapeutic opioid administration in adults: is pharmacogenetic analysis useful? Pain research & management : the journal of the Canadian Pain Society = journal de la societe canadienne pour le traitement de la douleur. 2013 May-Jun;18(3):133–136. doi: 10.1155/2013/518012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kelly LE, Rieder M, van den Anker J, et al. More codeine fatalities after tonsillectomy in North American children. Pediatrics. 2012 May;129(5):e1343–1347. doi: 10.1542/peds.2011-2538. [DOI] [PubMed] [Google Scholar]
  • 21.Koren G, Cairns J, Chitayat D, Gaedigk A, Leeder SJ. Pharmacogenetics of morphine poisoning in a breastfed neonate of a codeine-prescribed mother. Lancet. 2006 Aug 19;368(9536):704. doi: 10.1016/S0140-6736(06)69255-6. [DOI] [PubMed] [Google Scholar]
  • 22.US FDA Public Health Advisory: Use of Codeine By Some Breastfeeding Mothers May Lead To Life-Threatening Side Effects In Nursing Babies. 2007 http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/PublicHealthAdvisories/ucm054717.htm. Accessed 1/14/2014.
  • 23.Blake CM, Kharasch ED, Schwab M, Nagele P. A meta-analysis of CYP2D6 metabolizer phenotype and metoprolol pharmacokinetics. Clin Pharmacol Ther. 2013 Sep;94(3):394–399. doi: 10.1038/clpt.2013.96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Rogers RC, Sibai BM, Whybrew WD. Labetalol pharmacokinetics in pregnancy-induced hypertension. Am J Obstet Gynecol. 1990 Feb;162(2):362–366. doi: 10.1016/0002-9378(90)90386-l. [DOI] [PubMed] [Google Scholar]
  • 25.Fischer JH, Sarto GE, Hardman J, et al. Influence of gestational age and body weight on the pharmacokinetics of labetalol in pregnancy. Clin Pharmacokinet. 2014 Apr;53(4):373–383. doi: 10.1007/s40262-013-0123-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Jeong H, Choi S, Song JW, Chen H, Fischer JH. Regulation of UDP-glucuronosyltransferase (UGT) 1A1 by progesterone and its impact on labetalol elimination. Xenobiotica; the fate of foreign compounds in biological systems. 2008 Jan;38(1):62–75. doi: 10.1080/00498250701744633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Chan SW, Hu M, Ko SS, et al. CYP2C19 genotype has a major influence on labetalol pharmacokinetics in healthy male Chinese subjects. Eur J Clin Pharmacol. 2013 Apr;69(4):799–806. doi: 10.1007/s00228-012-1428-x. [DOI] [PubMed] [Google Scholar]
  • 28.Spinasse LB, Santos AR, Suffys PN, Muxfeldt ES, Salles GF. Different phenotypes of the NAT2 gene influences hydralazine antihypertensive response in patients with resistant hypertension. Pharmacogenomics. 2014 Feb;15(2):169–178. doi: 10.2217/pgs.13.202. [DOI] [PubMed] [Google Scholar]
  • 29.Bologa M, Tang B, Klein J, Tesoro A, Koren G. Pregnancy-induced changes in drug metabolism in epileptic women. J Pharmacol Exp Ther. 1991 May;257(2):735–740. [PubMed] [Google Scholar]
  • 30.Landau R, Xie HG, Dishy V, et al. beta2-Adrenergic receptor genotype and preterm delivery. Am J Obstet Gynecol. 2002 Nov;187(5):1294–1298. doi: 10.1067/mob.2002.128524. [DOI] [PubMed] [Google Scholar]
  • 31.Doh K, Sziller I, Vardhana S, Kovacs E, Papp Z, Witkin SS. Beta2-adrenergic receptor gene polymorphisms and pregnancy outcome. Journal of perinatal medicine. 2004;32(5):413–417. doi: 10.1515/JPM.2004.138. [DOI] [PubMed] [Google Scholar]
  • 32.Ozkur M, Dogulu F, Ozkur A, Gokmen B, Inaloz SS, Aynacioglu AS. Association of the Gln27Glu polymorphism of the beta-2-adrenergic receptor with preterm labor. International journal of gynaecology and obstetrics: the official organ of the International Federation of Gynaecology and Obstetrics. 2002 Jun;77(3):209–215. doi: 10.1016/s0020-7292(02)00035-8. [DOI] [PubMed] [Google Scholar]
  • 33.Landau R, Morales MA, Antonarakis SE, Blouin JL, Smiley RM. Arg16 homozygosity of the beta2-adrenergic receptor improves the outcome after beta2-agonist tocolysis for preterm labor. Clin Pharmacol Ther. 2005 Dec;78(6):656–663. doi: 10.1016/j.clpt.2005.08.021. [DOI] [PubMed] [Google Scholar]
  • 34.Hebert MF, Easterling TR, Kirby B, et al. Effects of pregnancy on CYP3A and P-glycoprotein activities as measured by disposition of midazolam and digoxin: a University of Washington specialized center of research study. Clin Pharmacol Ther. 2008 Aug;84(2):248–253. doi: 10.1038/clpt.2008.1. [DOI] [PubMed] [Google Scholar]
  • 35.Quinney SK, Mohamed AN, Hebert MF, et al. A Semi-Mechanistic Metabolism Model of CYP3A Substrates in Pregnancy: Predicting Changes in Midazolam and Nifedipine Pharmacokinetics. CPT: Pharmacomet Syst Pharmacol. 2012;1:e2. doi: 10.1038/psp.2012.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Haas DM, Quinney SK, McCormick CL, Jones DR, Renbarger JL. A pilot study of the impact of genotype on nifedipine pharmacokinetics when used as a tocolytic. J Matern Fetal Neonatal Med. 2012 Apr;25(4):419–423. doi: 10.3109/14767058.2011.583700. [DOI] [PubMed] [Google Scholar]
  • 37.Haas DM, Quinney SK, Clay JM, et al. Nifedipine Pharmacokinetics Are Influenced by CYP3A5 Genotype When Used as a Preterm Labor Tocolytic. Am J Perinatol. 2013 Apr;30(4):275–282. doi: 10.1055/s-0032-1323590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bremer T, Man A, Kask K, Diamond C. CACNA1C polymorphisms are associated with the efficacy of calcium channel blockers in the treatment of hypertension. Pharmacogenomics. 2006 Apr;7(3):271–279. doi: 10.2217/14622416.7.3.271. [DOI] [PubMed] [Google Scholar]
  • 39.Beitelshees AL, Navare H, Wang D, et al. CACNA1C gene polymorphisms, cardiovascular disease outcomes, and treatment response. Circ Cardiovasc Genet. 2009 Aug;2(4):362–370. doi: 10.1161/CIRCGENETICS.109.857839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kamide K, Yang J, Matayoshi T, et al. Genetic polymorphisms of L-type calcium channel alpha1C and alpha1D subunit genes are associated with sensitivity to the antihypertensive effects of L-type dihydropyridine calcium-channel blockers. Circ J. 2009 Apr;73(4):732–740. doi: 10.1253/circj.cj-08-0761. [DOI] [PubMed] [Google Scholar]
  • 41.Nakajima M, Inoue T, Shimada N, Tokudome S, Yamamoto T, Kuroiwa Y. Cytochrome P450 2C9 catalyzes indomethacin O-demethylation in human liver microsomes. Drug Metab Dispos. 1998 Mar;26(3):261–266. [PubMed] [Google Scholar]
  • 42.Tomson T, Lindbom U, Ekqvist B, Sundqvist A. Disposition of carbamazepine and phenytoin in pregnancy. Epilepsia. 1994 Jan-Feb;35(1):131–135. doi: 10.1111/j.1528-1157.1994.tb02922.x. [DOI] [PubMed] [Google Scholar]
  • 43.Tomson T, Lindbom U, Ekqvist B, Sundqvist A. Epilepsy and pregnancy: a prospective study of seizure control in relation to free and total plasma concentrations of carbamazepine and phenytoin. Epilepsia. 1994 Jan-Feb;35(1):122–130. doi: 10.1111/j.1528-1157.1994.tb02921.x. [DOI] [PubMed] [Google Scholar]
  • 44.PharmGKB. The Pharmacogenomics Knowledgebase. 2014 www.pharmgkb.org. Accessed 1/14/2014.
  • 45.Rodrigues AD. Impact of CYP2C9 genotype on pharmacokinetics: are all cyclooxygenase inhibitors the same? Drug Metab Dispos. 2005 Nov;33(11):1567–1575. doi: 10.1124/dmd.105.006452. [DOI] [PubMed] [Google Scholar]
  • 46.Haas DM, Caldwell DM, Kirkpatrick P, McIntosh JJ, Welton NJ. Tocolytic therapy for preterm delivery: systematic review and network meta-analysis. BMJ. 2012;345:e6226. doi: 10.1136/bmj.e6226. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Conde-Agudelo A, Romero R. Antenatal magnesium sulfate for the prevention of cerebral palsy in preterm infants less than 34 weeks’ gestation: a systematic review and metaanalysis. Am J Obstet Gynecol. 2009 Jun;200(6):595–609. doi: 10.1016/j.ajog.2009.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Rouse DJ, Hirtz DG, Thom E, et al. A randomized, controlled trial of magnesium sulfate for the prevention of cerebral palsy. The New England journal of medicine. 2008 Aug 28;359(9):895–905. doi: 10.1056/NEJMoa0801187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Crowther CA, Hiller JE, Doyle LW, Haslam RR. Australasian Collaborative Trial of Magnesium Sulphate Collaborative G. Effect of magnesium sulfate given for neuroprotection before preterm birth: a randomized controlled trial. JAMA : the journal of the American Medical Association. 2003 Nov 26;290(20):2669–2676. doi: 10.1001/jama.290.20.2669. [DOI] [PubMed] [Google Scholar]
  • 50.de Baaij JHF, Hoenderop JGJ, Bindels RJM. Regulation of magnesium balance: lessons learned from human genetic disease. Clinical Kidney Journal. 2012 Feb 1;5(Suppl 1):i15–i24. doi: 10.1093/ndtplus/sfr164. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tantisira KG, Lake S, Silverman ES, et al. Corticosteroid pharmacogenetics: association of sequence variants in CRHR1 with improved lung function in asthmatics treated with inhaled corticosteroids. Human molecular genetics. 2004 Jul 1;13(13):1353–1359. doi: 10.1093/hmg/ddh149. [DOI] [PubMed] [Google Scholar]
  • 52.Ye YM, Lee HY, Kim SH, et al. Pharmacogenetic study of the effects of NK2R G231E G>A and TBX21 H33Q C>G polymorphisms on asthma control with inhaled corticosteroid treatment. Journal of clinical pharmacy and therapeutics. 2009 Dec;34(6):693–701. doi: 10.1111/j.1365-2710.2009.01054.x. [DOI] [PubMed] [Google Scholar]
  • 53.Hawkins GA, Lazarus R, Smith RS, et al. The glucocorticoid receptor heterocomplex gene STIP1 is associated with improved lung function in asthmatic subjects treated with inhaled corticosteroids. The Journal of allergy and clinical immunology. 2009 Jun;123(6):1376–1383 e1377. doi: 10.1016/j.jaci.2009.01.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jin Y, Hu D, Peterson EL, et al. Dual-specificity phosphatase 1 as a pharmacogenetic modifier of inhaled steroid response among asthmatic patients. The Journal of allergy and clinical immunology. 2010 Sep;126(3):618–625. e611–612. doi: 10.1016/j.jaci.2010.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Tantisira KG, Silverman ES, Mariani TJ, et al. FCER2: a pharmacogenetic basis for severe exacerbations in children with asthma. The Journal of allergy and clinical immunology. 2007 Dec;120(6):1285–1291. doi: 10.1016/j.jaci.2007.09.005. [DOI] [PubMed] [Google Scholar]
  • 56.Tantisira KG, Lasky-Su J, Harada M, et al. Genomewide association between GLCCI1 and response to glucocorticoid therapy in asthma. The New England journal of medicine. 2011 Sep 29;365(13):1173–1183. doi: 10.1056/NEJMoa0911353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Oretti C, Marino S, Mosca F, et al. Glutathione-S-transferase-P1 I105V polymorphism and response to antenatal betamethasone in the prevention of respiratory distress syndrome. Eur J Clin Pharmacol. 2009 May;65(5):483–491. doi: 10.1007/s00228-009-0617-8. [DOI] [PubMed] [Google Scholar]
  • 58.Flockhart DA, Skaar T, Berlin DS, Klein TE, Nguyen AT. Clinically available pharmacogenomics tests. Clin Pharmacol Ther. 2009 Jul;86(1):109–113. doi: 10.1038/clpt.2009.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Weiss ST, McLeod HL, Flockhart DA, et al. Creating and evaluating genetic tests predictive of drug response. Nature reviews Drug discovery. 2008 Jul;7(7):568–574. doi: 10.1038/nrd2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dello Russo C, Lisi L, Fabbiani M, et al. Detection of HLA-B*57:01 by real-time PCR: implementation into routine clinical practice and additional validation data. Pharmacogenomics. 2014 Feb;15(3):319–327. doi: 10.2217/pgs.13.242. [DOI] [PubMed] [Google Scholar]
  • 61.Olgiati P, Bajo E, Bigelli M, De Ronchi D, Serretti A. Should pharmacogenetics be incorporated in major depression treatment? Economic evaluation in high-and middle-income European countries. Progress in neuro-psychopharmacology & biological psychiatry. 2012 Jan 10;36(1):147–154. doi: 10.1016/j.pnpbp.2011.08.013. [DOI] [PubMed] [Google Scholar]
  • 62.Pink J, Pirmohamed M, Lane S, Hughes DA. Cost-Effectiveness of Pharmacogenetics-Guided Warfarin Therapy vs. Alternative Anticoagulation in Atrial Fibrillation. Clin Pharmacol Ther. 2014 Feb;95(2):199–207. doi: 10.1038/clpt.2013.190. [DOI] [PubMed] [Google Scholar]
  • 63.Thompson AJ, Newman WG, Elliott RA, Roberts SA, Tricker K, Payne K. The Cost-Effectiveness of a Pharmacogenetic Test: A Trial-Based Evaluation of TPMT Genotyping for Azathioprine. Value in health : the journal of the International Society for Pharmacoeconomics and Outcomes Research. 2014 Jan;17(1):22–33. doi: 10.1016/j.jval.2013.10.007. [DOI] [PubMed] [Google Scholar]
  • 64.Davies EC, Green CF, Mottram DR, Pirmohamed M. Adverse drug reactions in hospitals: a narrative review. Current drug safety. 2007 Jan;2(1):79–87. doi: 10.2174/157488607779315507. [DOI] [PubMed] [Google Scholar]
  • 65.Mastroianni AC, Faden R, Federman D. Women and health research: a report from the Institute of Medicine. Kennedy Institute of Ethics journal. 1994 Mar;4(1):55–62. doi: 10.1353/ken.0.0121. [DOI] [PubMed] [Google Scholar]
  • 66.Martin J, Hamilton B, Ventura S, Osterman M, Mathews T. Births: Final data for 2011. National Vital Statistics Reports. 2013;62(1) [PubMed] [Google Scholar]

RESOURCES