46

The lack of response demonstrated by P.F. to the standard dose of paroxetine fits

the phenotype of an ultrarapid metabolizer. The recommendation per the CPIC

guideline would be to choose another drug not primarily metabolized via the

CYP2D6 pathway.

47 This case provides a prime example of how time can be wasted

and patient morbidity increased due to lack of response when pharmacogenomic

markers are not considered prior to initiating therapy.

CASE 4-8

QUESTION 1: You have started work at a pediatric hospital and notice that codeine is not orderable in the

electronic medical record. When you ask a coworker, they inform you that the black box warning placed on all

codeine products by the FDA in 2012 has resulted in the majority of pediatric institutions removing the drug

from the formulary. What genotype is correlated with the risk of life-threatening apnea?

Codeine is metabolized by CYP2D6 to the active metabolite morphine, which

provides analgesic properties. For patients with the ultrarapid metabolizer status

described earlier, the amount of morphine generated can exceed the body’s ability to

convert to less toxic metabolites, resulting in life-threatening apnea.

48 The ultrarapid

metabolizer phenotype is found in up to 4% of the population.

49

In contrast, there are

several loss-of-function variants with far-ranging incidence across ethnicities. For

example, the *4 allele is found in 6% of African-Americans, 5% of Asians, and up to

18% of European-Americans/Europeans.

50 These variants confer little to no enzyme

function, resulting in lack of morphine formation. Patients with these genotypes will

not have the desired analgesic response to codeine.

50

DEVELOPMENTAL IMPACT AND

PHARMACOGENOMICS

CASE 4-9

QUESTION 1: As part of a voluntary research study in the NICU, pharmacogenomic results are returned to

the provider for a baby girl born at 30 weeks, who is now at corrected 36.5-week gestational age. The provider

notes that the genotype is CYP2C19 *17/*17 gain of function, indicating she is an ultrarapid metabolizer. The

provider states he would like to start her on voriconazole for a presumed fungal infection and asks what the

dosing should be in context of her genotype. What other information would be necessary to recommend a

voriconazole dose for this baby?

Voriconazole is metabolized by CYP3A4, CYP2C9, and CYP2C19, with the most

significant contribution coming from CYP2C19.

9 The Royal Dutch Pharmacists

Association has guidelines in place recommending monitoring of voriconazole serum

concentrations for CYP2C19 poor and intermediate metabolizers, as these patients

have been found to have higher concentrations of active drug and increased risk of

toxicity. The consideration for a person with the CYP2C19 *17/*17 is the potential

for increased breakdown of active drug and treatment failure.

51

Given this patient’s age, there are multiple complex factors that need to be

considered for dosing, both genetic and nongenetic. As voriconazole is dosed by

body weight, the actual body weight of the child is necessary for an appropriate dose

calculation.

52 Drug–drug interactions may also need to be considered in the event that

the patient is receiving medications that induce or inhibit enzymes in the drug’s

pathway. Other variables also contributing to complicated infant dosing and

clearance unpredictability include developing gastrointestinal and renal function and

fluctuations in body fat and water composition affecting volume of drug

distribution.

53 Additionally, although the patient’s genotype suggests that she will

have increased breakdown of the active drug, it is known that regardless of genotype,

humans are not born with fully functional CYP enzymes.

53

CYP enzyme development occurs at variable rates over time making it difficult to

determine the contribution of pharmacogenomic variants on optimal dosing (see Fig.

4-3). An excellent summary review of enzyme maturation by Kearns and colleagues

was published in the NEJM in 2003.

53 CYP1A2, for example, is virtually

undetectable at birth, with initial development of low-level enzyme concentrations

observed over the first month

p. 59

p. 60

of life.

54,55Although general trends are observed, it is also understood that among

individuals, development rates vary throughout childhood, particularly in the first

year of life.

54 Given this consideration, pharmacogenomics in the infant population

may be less of reliable factor than it is in adults, and its overall contribution remains

unpredictable as few studies are dedicated to the effects of pharmacogenomic

variants in younger individuals. Therefore, it is particularly important in children and

infants to monitor for drug concentrations, response, and toxicities, and to adjust dose

accordingly.

55 Voriconazole is of particular concern with this group, with evidence

suggesting that current pediatric dosing recommendations may lead to subtherapeutic

concentrations and a general increased risk of treatment failure.

55

Figure 4-3 CYP2C19 enzyme activity by age.

FAMILY IMPLICATIONS OF

PHARMACOGENETICS

CASE 4-10

QUESTION 1: A 12-year-old-girl has an operation and develops malignant hyperthermia (MH) upon initiation

of anesthesia. Subsequently, she is found to have a c. 7300G>A mutation in the RYR1 gene, a common variant

associated with predisposition for MH. Following her recovery and discussion of future medical and drug

management, what else would be important to discuss?

Genes are inherited; therefore, any discussion of genetic variants must be

undertaken with an awareness of the greater implications of any identified changes. In

this case especially, because the consequences of the gene change are so profound

and the mutation is a dominant one, it is likely that she inherited the change. An

exploration of the family history may suggest that one parent is more likely to be the

carrier than another, and counseling with a view toward testing may well be guided

by such information. Furthermore, one must consider the potential for siblings,

grandparents, and the extended family to be affected depending on the severity of the

variant status and the clinical picture.

In the situation where the individual is being counseled for pharmacogenomic

testing presymptomatically, it is important to raise the possibility that the results may

have implications for the greater family before testing is undertaken.

58

MAKING THE CASE FOR PHARMACOGENETICS

Pharmacogenomics has allowed pharmaceutical companies to design and develop

drugs specifically targeted at certain mutations. In order to determine the right patient

for the drug, companion tests (pharmacogenetic assays that target the variants of

interest) are usually required. The pharmacist must understand which medications

require companion testing and be able to interpret the results. Although up to 50% of

drug companies are pursuing drug targets that would require companion genetic tests,

there are several hindrances that must be addressed, including potential delays in

companion test approval and potentially decreasing the patient base by eliminating

those who would otherwise be prescribed the drug in the absence of knowledge of

the genetic marker presence.

2 Additionally, several studies have shown that patients

are more adherent with their medications after having personalized genotyping, even

if the results show “normal” enzyme function.

59

CASE 4-11

QUESTION 1: K.D. is a 22-year-old female with cystic fibrosis. Her pulmonologist is interested in the new

drug ivacaftor, but is unsure what testing is required to start therapy. What testing is necessary and which

variants qualify for ivacaftor therapy?

Ivacaftor (Kalydeco) is a drug for cystic fibrosis that targets ten specific variants

in the Cystic Fibrosis Transmembrane Regulator (CFTR) gene. If a patient has a

variant status other than G551D, G1244E, G1349D, G178R, G551S, R117H,

S1251N, S1255P, S549N, or S549R, the drug will be ineffective.

60

The CFTR gene codes for the CFTR protein on a variety of tissue surfaces,

including the lungs. When functioning properly, the CFTR protein is a key component

in maintaining intracellular salt balance.

61

In cystic fibrosis, because of a variety of

possible genetic mutations the CFTR protein fails to function properly leading to

fluid imbalances, the buildup of secretions, and several related complications.

Ivacaftor acts as a CFTR potentiator increasing chloride ion transport, restoring

electrolyte balance, reducing the buildup of secretions, and improving health

outcomes such as pulmonary function and weight gain for affected patients.

62 The

approval of this drug in 2012 for patients with the G551D mutation offered promise

for continued future development of therapies targeting causes of disease that are

genetically associated. This promise remains encouraging as the use of ivacaftor has

since been approved in the treatment of several additional gene mutations, and new

similar targeted therapies such as lumacaftor (Orkambi), a CFTR corrector, continue

to enter the market.

63

Another potential use of pharmacogenomics involves salvaging drugs with high

toxicity profiles. Historically, drugs have been taken off the market after an

unacceptable number of patients either suffered significant morbidity or mortality

secondary to the use of the drug. In some cases, pharmacogenomic studies may be

able to determine which patients could continue to benefit from the drug and which

for patients’ use must be avoided.

Despite all of the advances in testing and application, pharmacogenomic

preemptive testing (testing prior to the development of an adverse effect or lack of

response) is not currently in wide use. There are several reasons for this. The lack of

knowledge by healthcare professionals, including pharmacists, is a large barrier. One

recent study demonstrated that only 29% of the physicians surveyed had received any

formal education and only 10.3% felt knowledgeable enough to prescribe or discuss

the results of pharmacogenomic testing.

64 Another large impediment to

implementation is the lack of consistent reimbursement by insurance providers.

Consistent coverage by insurance companies will require regulatory effort and

additional proof that pharmacogenomic testing improves outcome and decreases

cost.

2

Therefore, at this time, the majority of preemptive testing is being done in

academic medical centers, in cancer centers, or by for-profit pharmacogenomic

testing companies that can either institutionally support the development of the

programs or can directly bill for the services not covered by insurance.

CASE 4-11, QUESTION 2: What references could be used to assist in interpretation of pharmacogenomic

markers and subsequent drug dosing recommendations?

Currently, the FDA lists pharmacogenomic markers in 190 drug labels and the

European Medicines Agency lists pharmacogenomic information in 78 drug

labels.

65,66

In order to be included in the FDA labeling, the pharmacogenomic marker

must have actionable data, such as an increase in adverse effects or reduction in

efficacy

p. 60

p. 61

in patients with certain variants. Guidelines for drug dosing and selection have

been developed by the Clinical Pharmacogenomics Implementation Consortium

(CPIC) and the Pharmacogenetics Working Groups of the Royal Dutch Pharmacists

Association (DPWG). Each guideline is rated on the level of evidence available to

support the recommendation. Currently, there are 36 CPIC Level I evidence

published guidelines available on www.pharmgkb.org, a website supported by the

NIH and hosted by Stanford University.

67 The site houses a wealth of information

including pathway diagrams, annotated bibliographies, and lookup tables. It is

important to note, the guidelines do not address who should be tested, but instead

focus on what to do with the data if it is available.

Incorporating pharmacogenomic data into the electronic medical record systems

used by many hospitals, clinics, pharmacies, and primary care offices is challenging

at best. Result reports from the laboratories capable of running these tests are rarely

in a machine-readable format and are generally scanned as pdf documents to the

medical record.

68 This becomes a major challenge for the system to be able to

provide relevant pharmacogenomic data at the time it is needed, usually during

prescribing of a drug. Results of this type that persist throughout a person’s lifetime

and are relevant all along the continuum require significant bioinformatics expertise

to house, retrieve, interpret, and present to the end user at the right time.

69

Additionally, understanding that the testing will evolve over time, either resulting in

the need to sequence a new sample or ideally, reanalyzing the previously deep

sequenced sample with new algorithms to apply the most recent variant knowledge.

A payment model for reinterpretation without retesting is extremely uncommon in the

field of laboratory medicine today.

Another extremely important consideration is data security and privacy. Data

security refers to the ability to protect information from breaches and inadvertent

dissemination. Data privacy refers to the ability to respect patient preference for data

sharing, both with the patient themselves and with the larger health care community.

How is the decision made to offer and incorporate pharmacogenomic testing into

practice? In some cases, it becomes forced upon the healthcare provider, such as a

pharmacist or primary care physician, who is handed a printed report by a patient

who has purchased direct to consumer pharmacogenetic testing services and insists

that the information be considered when prescribing and dispensing decisions are

made. Clearly, that is a less than ideal situation. Healthcare provider education has

begun to incorporate pharmacogenomic science into the core curriculums, but it is

often in the form of a single1-to 2-hour lecture. This will be inadequate preparation

to deal with the era of personalized medicine and the eventual day when everyone

will have access to their own genomic data.

When assessing a pharmacogenomic test for potential incorporation into practice,

several factors impact the return on investment and clinical outcome. These include

but are not limited to the number of patients needed to be tested in order to find one

patient with an actionable variant (also known as the incidence of variant status) and

the ethnic variation, being cognizant that we are becoming a very blended population

without full knowledge of our entire ancestry. Additionally, institutions must

investigate how much each test costs to run, if it will be reimbursed, and what are the

cost savings offset by averting serious adverse reactions or the consequences of

nonresponse.

2,70,71

Genetic testing can introduce ethical questions that can be challenging, particularly

when dealing with sequencing large portions of the DNA. Although most people want

to know whether or not they should take a medication, many do not wish to know

their risk of developing Alzheimer disease or breast cancer. This becomes a concern

with some of the broader sequencing tests such as whole exome or whole genome.

The American College of Medical Genetics and Genomics (ACMG) released a

position statement in 2013, recommending mandatory reporting in 24 categories of

diseases caused by genetic variants whenever a sequencing test is performed,

regardless of patient preference around wanting to know that information and

irrespective of age of the patient.

72 Arguments against this practice have included the

patient’s right to decide, informing the parents/caregivers of an adult-onset disease

for a child, and insurance discrimination risks.

73,74 Although it is illegal for health

insurance coverage to be denied due to genetic findings following the passage of the

Genetic Information Nondiscrimination Act (GINA) in 2008, this is currently not true

for life or long-term care insurance coverage.

75

CONCLUSION

Pharmacogenomics is simultaneously an exciting and challenging component of

personalized medicine and the practice of pharmacy. One of the most important

points to remember is that pharmacogenomic information is an additional clinical

marker, but is rarely the only answer. A patient’s organ function, disease state, diet,

smoking status, other environmental factors, and drug–drug interactions play a very

large role in the disposition of drugs. Age-based maturation of enzyme function must

also be accounted for when determining the impact of the genotype on drug

metabolism.

Pharmacists are uniquely qualified to interpret and apply pharmacogenetic findings

to medication recommendations. However, this will require adequate preparation,

application of validated algorithms, access to continually updated literature, and a

partnership with genetic experts, including geneticists and genetic counselors.

76

KEY REFERENCES AND WEBSITES

A full list of references for this chapter can be found at

http://thepoint.lww.com/AT11e. Below are the key references and websites for this

chapter, with the corresponding reference number in this chapter found in parentheses

after the reference.

Key References

Altman RB et al, eds. Principles of Pharmacogenetics and Pharmacogenomics. 1st ed. New York, NY: Cambridge

University Press; 2012.

Gaedigk A. Complexities of CYP2D6 gene analysis and interpretation. Int Rev Psychiatry. 2013;25(5):534–553.

(46)

Kearns GL et al. Developmental pharmacology—drug disposition, action, and therapy in infants and children. N

EnglJ Med. 2003;349:1157–1167. (53)

Samer CF et al. Applications of CYP450 testing in the clinicalsetting. MolDiagn Ther.2013;17:165–184. (6)

Key Websites

FDA Gene List.

http://www.fda.gov/Drugs/ScienceResearch/ResearchAreas/Pharmacogenetics/default.htm.

NIH Precision Medicine Initiative. http://www.nih.gov/precisionmedicine/.

PharmGKB.org. http://www.pharmgkb.org.

Warfarin Dosing. http://www.warfarindosing.org/Source/Home.aspx.

COMPLETE REFERENCES CHAPTER 4

PHARMACOGENOMICS AND PERSONALIZED MEDICINE

Hansen NT et al. Generating genome-scale candidate gene lists for pharmacogenomics. Clin Pharmacol Ther.

2009;86(2):183–189.

Shabaruddin FH et al. Economic evaluations of personalized medicine: existing challenges and current

developments. Pharmacogenomics Pers Med. 2015;8:115–126.

Scott S. Personalizing medicine with clinical pharmacogenetics. Genet Med. 2011;13(12):987–995.

Motulsky AG. Drug reaction enzymes and biochemical genetics. J Am Med Assoc. 1957;165:835–837.

Wessler S, Avioli LV. Pharmacogenetics. Glucose-6-phosphate dehydrogenase deficiency. J Am Med Assoc .

1968;205(10):679–683.

Samer CF et al. Applications of CYP450 testing in the clinicalsetting. Mol Diagn Ther. 2013;17:165–184.

Sim SC, Ingelmann-Sundberg M. The human cytochrome P450 (CYP) allele nomenclature website: a peerreviewed database of CYP variants and their associated effects. Hum Genomics. 2010;4(4):278–281.

Kohn LT et al, eds. To Err Is human: Building a Safer Health System. Washington, DC: National Academy of

Sciences; 2000.

Sikka R et al. Bench to bedside: pharmacogenomics, adverse drug interactions, and the cytochrome P450 system.

Acad Emerg Med. 2005;12(12):1227–1235.

Beitelshees AL, Veenstra DL. Evolving research and stakeholder perspectives on pharmacogenomics. J Am

Med Assoc. 2011;306(11):1252–1253.

Lexicomp. Isoniazid. Copyright 1978–2015. Hudson, OH: Lexicomp. Accessed September 8, 2015.

Daly AK. Drug-induced liver injury: past, present and future. Pharmacogenomics. 2010;11(5):607–611.

Pasipanodya JG et al. Meta-analysis of clinical studies supports the pharmacokinetic variability hypothesis for

acquired drug resistance and failure of antituberculosis therapy. Clin Infect Dis. 2012;55(2):169–177.

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more