Murray et al. Clinical practice guidelines for sustained neuromuscular blockade in the adult critically ill patient.

Crit Care Med. 2016;44(11):2079–2103.

Society of Critical Care Medicine and American Society of Health-Pharmacists. Clinical practice guidelines for

sustained neuromuscular blockade in the adult critically ill patient. Am J Health Sys Pharm. 2002;59:179–195.

Papazian L et al. ACURASYS study investigators. Neuromuscular blockers in early acute respiratory distress

syndrome. N EnglJ Med. 2010;363(12):1107–1116.

Sunjic KM et al. Pharmacokinetic and other considerations for drug therapy during targeted temperature

management. Crit Care Med. 2015;43:2228–2238.

Kisor DF et al. Clinical pharmacokinetics of cisatracurium besilate. Clin Pharmacokinet. 1999;36(1):27–40.

Lagneau F et al. A comparison of two depths of prolonged neuromuscular blockade induced by cisatracurium in

mechanically ventilated critically ill patients. Intensive Care Med. 2002;28:1735–1741.

Sessler CN. Train-of-four to monitor neuromuscular blockade? Chest. 2004;126(4):1018–1022.

Latcha S. Electrolyte disorders in critically ill patients. In: Oropello JM et al, eds. Critical Care New York, NY:

M c G r a w - H i l l ; http://accessmedicine.mhmedical.com.ezproxymcp.flo.org/content.aspx?

bookid=1944&sectionid=143517373. Accessed February 26, 2017.

Agents Acting at the Neuromuscular Junction and Autonomic Ganglia. In: Hilal-Dandan R et al, eds. Goodman

and Gilman’s Manual of Pharmacology and Therapeutics, 2e New York, NY: McGraw-Hill;

http://accesspharmacy.mhmedical.com.ezproxymcp.flo.org/content.aspx?

bookid=1810&sectionid=124490279. Accessed February 25, 2017.

Kindler CH et al. Additive inhitbition of nicotinic acetylcholine receptors by corticosteroids and the neuromuscular

blocking drug vecuronium. Anestheseiology. 2000;92:821–32.

Price D et al. A fresh look at paralytics in the critically ill: real promise and concern. Ann Intensive Care.

2012;2:43.

Waitz A et al. Biologic aspects of the interaction between gentamicin and carbenicillin. J Antibiot. 1974;25:219–

225.

Shwed JA et al. Lack of effect of clavulanic acid on aminoglycoside inactivation by ticarcillin. J Infect Dis

Pharmacother. 1995;1(2):35–43.

Walterspiel JN et al. Comparative Inactivation of Isepamicin, Amikacin, and Gentamicin by Nine β-Lactam and

Two β-Lactamase Inhibitors, Cilastatin and Heparin. Antimicrob Agents Chemother. 1991;35(9):1875–1878.

.

101.

102.

.

.

.

106.

.

.

.

.

.

.

.

.

.

.

.

Pickering LK et al. Effect of concentration and time upon inactivation of Tobramycin, Gentamicin, Netilmicin

and Amikacin by Azlocillin, Carbenicillin, Mecillinam, Mezlocillin and Piperacillin. J Pharmacol Exp Ther.

1981;217:345–349.

Gerald K et al, ed. 2017. AHFS Drug Information® - 59th Ed. Bethesda, MD. American Society of HealthSystem Pharmacists. ISBN-10: 1-58528-558-7, ISBN-13: 978-1-58528-558-7. ISSN: 8756-6028. STAT!Ref

Online Electronic Medical Library. http://online.statref.com.ezproxymcp.flo.org/Document.aspx?

docAddress=v7fPA2o3awdso_vsl1VHnA%3d%3d. 3/1/2017 7:28:14 PM CST (UTC -06:00).

Taber HW et al. Bacterial uptake of aminoglycoside antibiotics. Microbiol Rev. 1987;51(4):439–457.

Leggett JE. Aminoglycosides. In: Bennett JE et al, eds. Mandell, Douglas, and Bennett’s Principles and Practice

of Infectious Diseases. 8th ed. Philadelphia, PA: Churchill Livingstone Elsevier; 2015.

Davis BD. Bactericidal Synergism Between β-Lactams and Aminoglycosides: Mechanism and Possible

Therapeutic Implications. Rev Infect Dis. 1982;4(2):237–245.

Haouala A et al. Drug interactions with tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood.

2011;117(8):e75–e87.

Rochat B et al. Imatinib metabolite profiling in parallel to imatinib quantification in plasma of treated patients using

liquid chromatography-mass spectrometry. J Mass Spectrom. 2008;43(6):736–752.

Peng B et al. Clinical pharmacokinetics of imatinib. Clin Pharmacokinet. 2005;44(9):879–894.

Kompendium ch [homepage]. Switzerland: Compendium Suisse des medicaments 2010 [updated 2010].

http://www.kompendium.ch/. Accessed March 10, 2017.

Brendel C et al. Imatinib mesylate and nilotinib (AMN 107) exhibit high-affinity interaction with ABCG2 on

primitive hematopoietic stem cells. Leukemia. 2007;21(6):1267–1275.

Ozvergy-Laczka C et al. High-affinity interaction of tyrosine kinase inhibitors with the ABCG2 multidrug

transporter. Mol Pharmacol. 2004;65(6):1485–1495.

White DL et al. OCT-1-mediated influx is a key determinant of the intracellular uptake of imatinib but not

nilotinib (AMN 1078): educed OCT-1 activity is the cause of low in vitro sensitivity to imatinib. Blood.

2006;108(2):687–704.

Van Erp NP et al. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Tev. 2009;35(8):692–

706.

Petain A et al. Population pharmacokinetics and pharmacogenetics of imatinib in children and adults. Clin

Cancer Res. 2008;14(21):7102–7109.

Widmer N et al. Population pharmacokinetics of imatinib and the role of alpha-acide glycoprotein. Br J Clin

Pharmacol. 2006;62(1):97–112.

Widmer N et al. Relationship of imatinib-free plasma levels and target genotype with efficacy and tolerability. Br

J Cancer. 2008;98(10):1633–1640.

Azuma M et al. Role of alpha 1-acid glycoprotein in therapeutic antifibrotic effects of imatinib with macrolides in

mice. Am J Respir Crit Care Med. 2007;176(12):1243–1250.

Dutreix C et al. Pharmacokinetic interaction between ketoconazole and imatinib mesylate (Gleevec) in healthy

subjects. Cancer Chemother Pharmacol. 2004;54(4):290–294.

Frye RF et al. Effect of St. John’s wort on imatanib mesylate pharmacokinetics and protein binding of imatinib

mesylate. Pharmacotherapy. 2004;24(11):1508–1514.

Smith P et al. The influence of St. John’s wort on the pharmacokinetics and protein binding of imatinib mesylate.

Pharmacotherapy. 2004;24(11):1508–1514.

Recoche I et al. Drug-drug interactions with imatinib An observationalstudy. Medicine. 2016;95(40):1–5.

Cholongitas E et al. Dermatitis after suspected imatinib-levothyroxine interaction in a patient with gastrointestinal

stomal tumor. Cancer Chemother Pharmacol. 2008;61:1083–1084.

De Grrot JWB et al. Imatinib induces hypothyroidism in patient receiving levothyroxine. Clin Pharmacol Ther.

2005;78:433–438.

Horn JR et al. Proposal for a new tool to evaluate drug interaction cases. Ann Pharmacother. 2007;41(4):674–

680.

Tilson H et al. Recommendations for selecting drug-drug interactions for clinical decision support. Am J HealthSystem Pharm. 2016;73(8):576–585.

Hansten PD. Drug interaction management. Pharm World Sci. 2003;25(3):94–97.

p. 50


Pharmacogenomics is a single and important element of the broader

concept of personalized medicine, which incorporates a variety of

factors, both genetic and nongenetic, to guide targeted and individualized

therapeutic decisions.

Case 4-1(Question 1),

Figure 4-1,

Tables 4-1, 4-2, 4-3

Pharmacogenomic effects can be both pharmacokinetic and

pharmacodynamic in nature. Pharmacokinetic effects are observed

when variants affect the absorption, distribution, metabolism, or

excretion of a drug. Pharmacodynamic polymorphisms may result in

variable amounts of drug target enzymes or receptors as well as

possible changes in the drug target shape. These variations may render

therapy ineffective or necessitate dose changes in certain affected

populations.

Case 4-2 (Questions 1, 2)

DNA polymorphisms in drug-metabolizing enzymes can result in gain of

function, loss of function, or no effect on function of the enzyme

produced. In some cases, significant toxicity can result.

Case 4-3 (Question 1),

Figure 4-2,

Table 4-4,

Case 4-4 (Question 1), Table

4-5

Drug transport proteins and binding targets can also be affected by

pharmacogenomic variants.

Case 4-5 (Questions 1,2),

Table 4-6

HLA genes are unique in that the presence of a variant does not impact

the metabolism of a drug, but instead may indicate the likelihood of the

development of a serious or life-threatening reaction.

Case 4-6 (Questions 1, 2),

Table 4-7

Test interpretation is crucial to the practical application of

pharmacogenomic data. In particular, CYP2D6 is a well-described

cytochrome P450 enzyme responsible for approximately 25% of all drug

metabolism. However, CYP2D6 is a complicated gene locus subject to

multiple variants, pseudogene interference, and copy number variation.

Case 4-7 (Questions 1, 2),

Case 4-8 (Question 1), Table

4-8

Age-based development adds a level of complexity when assessing

pharmacogenomic markers in pediatric patients. Many of the

pharmacogenomic dosing guidelines have not been validated in infants

and children.

Case 4-9 (Question 1),

Figure 4-3

Implementation of pharmacogenomics into practice currently faces many

challenges including: the appropriate determination of when and whom

to test; storage, analysis, and security of genetic data; and

considerations for implementing pharmacogenomic data and

recommendations into practice.

Case 4-10 (Question 1),

Case 4-11 (Questions 1, 2)

“Pharmacogenomics” is the study and application of gene expression on drug

pharmacokinetics and pharmacodynamics. This term is often used interchangeably

with “pharmacogenetics.” In the strictest of definitions, “pharmacogenetics” is used

in the context of a drug response to a single gene, whereas “pharmacogenomics”

refers to the broader study of the full genomic impact on drug behavior.

1 Because

genetic variants are very specific to the individual, the use of pharmacogenomics in

clinical practice has become a core component of the personalized medicine and

precision medicine movements.

2,3

Ironically, the study of pharmacogenomics is not

new. There are cases in the literature from the 1950s and 1960s describing the

influence of a person’s genetics on drug toxicity.

4,5 With the advent of newer,

cheaper, faster deoxyribonucleic acid (DNA) sequencing techniques, the study of

pharmacogenomics has exploded. In recent years, we have been able to begin

transitioning that knowledge from the research realm to the clinic, although not

without challenges.

p. 51

p. 52

A brief review of the basics of human genetics in the context of application to

pharmacogenomics will be discussed in this chapter. Students are encouraged to

utilize the selected references provided for this chapter for further review if needed.

Although somatic “tumor” genetics, or the study of the mutations found in cancer

cells, is a very timely and important consideration in antineoplastic drug selection

and the treatment of oncologic processes, this chapter will primarily focus on the

germline mutations in human DNA affecting drug absorption, distribution,

metabolism, and excretion.

Human DNA is composed of 3 billion nucleotide base pairs, arranged on 46

chromosomes. There is significant variation in the genetic code from one person to

another, creating what makes each of us unique—such as brown eyes, red hair,

height, and heritable diseases. Included in the unique variations are changes in our

ability to process medications. These differences are largely a result of variants in

the nucleotide sequence on the genes responsible. The four nucleotides that make up

the genes are adenine (A), guanine (G), cytosine (C), and thymine (T).

In most cases, but not all, two copies of each gene are present, one inherited from

the mother and one from the father. Each copy is referred to as an “allele.” If both the

maternal and paternal alleles are the same, then the individual is considered

homozygous for that allele or gene. If the parental copies differ, then the patient is

considered to be heterozygous. Genes code for the production of proteins, such as

enzymes, the structural components of cells, hormones, antibodies, and transport

molecules. In pharmacogenomics, the gene and the enzyme often share the same name.

For example, the gene that codes for cytochrome P450 CYP3A4 is known as

CYP3A4 and the gene that codes for thiopurinemethyltransferase (TPMT) is known

as TPMT.

When there is a change in the sequence or code, it is referred to as a variant. A

change in a single base pair is commonly referred to as a single-nucleotide

polymorphism or SNP (pronounced “snip”) (Fig.4-1). Although the formal definition

requires the variant to occur in at least 1% of the population, common usage does not

differentiate on the basis of population frequency. A SNP that changes the function of

the gene product is referred to as a mutation, and although not all SNPs are mutations,

the terms are often used interchangeably. The location of the SNP is designated by a

reference sequence number abbreviated “rsID.” Many of the pharmacogenetic

variants discussed in this chapter are SNPs and were originally found through

genome-wide association studies (GWAS). Most GWAS are usually constructed to

find common SNPs in a group of patients with the condition of interest and then

subsequently define a level of association. These SNPs are responsible for 90% of

pharmacogenetic variability and can result in gain of function (greater production of

enzyme, or higher enzyme activity, than normal) or loss of function (decreased or no

production of enzyme compared to normal).

3 Other changes can include deletions or

insertions of larger segments of DNA and are commonly referred to as “indels.” In

many cases, indels terminate production of the protein.

In some cases, a change in a single allele is sufficient to affect drug metabolism.

This occurs in dominant disorders. These occur because new mutations are passed

from one individual to the next. Whether the ancestors exhibited symptoms or not

likely depends on their own history and, of course, may not be known. In other cases,

both copies of the gene must be defective in order for there to be a functional

consequence. This occurs in recessive disorders, in which the parents are typically

unaffected heterozygous carriers. One in four of their children will be at risk for

inheriting the change and potentially exhibiting symptoms. As discussed earlier, this

child may be homozygous if both parents have the same SNP, or it may be as a

compound heterozygote, if each parent has a different dysfunctional SNP or mutation

in the specific gene. Typically, in recessive disorders, there is otherwise no family

history of any affected individual.

The resultant call or read of the variants gives us a genotype. The phenotype refers

to the expression of that genotype. In some cases, this is visible, such as skin color or

another physical characteristic. In other cases it is invisible, such as blood type or a

less than normal amount of a Human Cytochrome P450 (CYP450) metabolizing

enzyme. In the context of pharmacogenomics and the metabolizing enzymes, patients

are often classified as having a phenotype of ultrarapid metabolizer, extensive

metabolizers, intermediate metabolizers, or poor metabolizers based on their

genotype.

6 There are several other phenotypes that will be discussed in the chapter,

particularly those that deal with drug targets or pharmacodynamic responses.

The vast majority of drug metabolism pathways are not simply, a single enzyme

resulting in one inactive metabolite that is immediately excreted by the body. Instead,

most drugs utilize multiple metabolizing enzyme pathways, rely on several drug

transporters, and have many metabolites of varying activity. Each of these steps may

be subject to variable gene expression, coupled with indirect genetic and

environmental effects on variables such as disease, weight, nutrition, age, and others.

Add this complexity and it becomes evident how complicated pharmacogenomic

interpretations can be.

Pharmacogenomics is unique in the field of genetics in that the representation of a

person’s genotype is noted by a “*,” known as a star allele in the literature. For

example, in many situations, the text representation of normal or “wild-type” variant

status is *1/*1.

7 As other alleles are discovered, they are numbered sequentially *2,

*3, etc. Inevitably, as discoveries are sometimes reported simultaneously around the

world, there is occasionally an overlap of star alleles and the variants represented.

There are several worldwide databases for reporting new pharmacogenomic

variants, including the CYP Allele Nomenclature Database

http://www.cypalleles.ki.se/ and the Database of Genomic Variants

http://dgv.tcag.ca/dgv/app/home.

During drug development and postmarketing analysis, it becomes clear that some

patients will have severe adverse drug reactions (including nonresponse) after

receiving the population-derived standard dose. These adverse reactions occur in the

absence of medication error or lack of proper adherence to the drug. The Institute of

Medicine “To Err is Human” report published in 2000 cited over 2 million reported

adverse drug reactions in the United States per year, resulting in approximately

100,000 deaths at a cost of 20.6 billion annually.

8

It is estimated that 52% of U.S.

adults are taking at least one prescription drug and 12% are taking more than five

prescription drugs. If herbal products and over-the-counter drugs are included, then

the estimates increase to over 80% and 29%, respectively.

6

It is well known that

adverse drug reactions often go unnoticed and even more frequently unreported,

making the actual numbers likely far higher. The clinical application of

pharmacogenomics not only has allowed us to explain some of these historical

reactions, but also helps clinicians predict who may be at higher risk for developing

an adverse reaction if exposed to a drug or drug class.

9 Avoidance of adverse drug

reactions has a measureable impact on a patient’s quality of life, decreases overall

health care costs, and lessens the time burden on health care providers managing the

adverse reaction. However, acceptance of clinical pharmacogenomic testing is not

universal and there remains the challenge of conclusively validating the genetic

variant associated with the adverse reaction.

10

Figure 4-1 Example of a single-nucleotide polymorphism (SNP).

p. 52

p. 53

PHARMACOKINETIC IMPLICATIONS

CASE 4-1

QUESTION 1: A.S. is a 2-year-old diagnosed with active multidrug-resistant tuberculosis (TB). The Centers

for Disease Control and Prevention treatment protocol recommends isoniazid, rifampin, ethambutol, and

pyrazinamide for initial therapy. Her mother is concerned because several family members in their home

country of China had developed serious “liver problems” when they took isoniazid and she insists on the “gene

test” for A.S. What genetic testing is A.S.’s mother referring to and what are the risks of toxicity in relation to

isoniazid therapy?

Isoniazid is part of the four-drug regimen used to treat active TB and latent TB

infection because it is bactericidal against Mycobacterium tuberculosis organisms.

11

One of the most commonly reported side effects leading to premature discontinuation

of the drug is drug-induced liver injury (DILI). Significantly elevated hepatic

enzymes are seen in up to 20% of patients, with progression to hepatitis in a smaller

percentage of those treated.

12

The metabolism of isoniazid is complex, with a key enzyme within the pathway

being N-acetyltransferase-2 (NAT2).

11 NAT2 acetylates both the parent drug and a

hydrazine metabolite, converting the hydrazine metabolite to acetyl hydrazine

(AcHz).

12An alternative metabolic pathway for hydrazine results in a toxic reactive

metabolite. When NAT2 function is decreased, this pathway dominates, resulting in

increased DILI and toxicity.

The production of the NAT2 enzyme is generated from a gene of the same name,

with several known polymorphisms leading to alleles associated with either slow or

rapid acetylation rates. Individuals homozygous for alleles associated with loss of

function are considered “slow acetylators,” those homozygous for alleles associated

with gain of function “rapid acetylators,” and heterozygotes “intermediate

acetylators” (Table 4-1). DILI has been found to be highest in patients who are slow

acetylators.

12

In addition to isoniazid, NAT2 is also a known acetylator of other

drugs and/or metabolites, although the clinical effects of NAT2 genotype on these

drugs are less well-studied, including: sulfonamides (sulfamethoxazole, metabolites

of sulfasalazine), aromatic and aliphatic amines (procainamide, dapsone, metabolite

of clonazepam, mescaline).

12

The “gene test” mentioned by A.S.’s mother is likely a test for the NAT2 gene. A

variety of pharmacogenomic tests are available to examine SNPs in the NAT2 gene

and assign a patient’s genotype, either as part of a more extensive gene panel or as a

targeted assay. Preemptive NAT2 testing prior to isoniazid therapy is not currently

the standard of care; however, evidence suggests that in some populations with a high

prevalence of DILI in slow acetylators, this may be beneficial.

14–18 Asian cohorts

have been found to have the highest rates of DILI in the setting of a slow acetylator

status (see summary of studies in Table 4-2). Although there are no formal dosing

guidelines set for isoniazid therapy in the context of genotype or acetylator status, a

study completed by Azuma et al. with 155 Japanese TB patients proposed a

modified, genotype-based dosing regimen with successful clinically and statistically

significant outcomes (results summarized in Table 4-3).

19

There remain several pending questions as to how to most safely and appropriately

incorporate NAT2 genotype results into a widespread clinical application. For

example, pediatric patients such as A.S. offer a unique set of challenges, because

recommended isoniazid starting doses are within a higher range (10–15 mg/kg), and

dose modifications based on genotype have not been studied in this population.

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