Raymond CB et al. Treatment of leg cramps in patients with chronic kidney disease receiving hemodialysis.

Cannt J. 2011;21(3):19.

El-Hennawy AS et al. A selected controlled trial of supplementary vitamin E for treatment of muscle cramps in

hemodialysis patients. Am J Ther. 2010;17(5):455.

Khajehdehi P et al. A randomized, double-blind, placebo-controlled trial of supplementary vitamins E, C and their

combination for treatment of haemodialysis cramps. Nephrol Dial Transplant. 2001;16:1448.

Arieff AI. Dialysis disequilibrium syndrome: current concepts on pathogenesis and prevention. Kidney Int.

1994;45:629.

Tseke P et al. Thrombolysis with alteplase: a non-invasive treatment for occluded arteriovenous fistulas and

grafts. Artif Organs. 2011;35(1):58.

Falk A et al. Reteplase in the treatment of thrombosed hemodialysis grafts. J Vasc Interv Radiol. 2001;12:1257.

Falk A et al. Tenecteplase in the treatment of thrombosed hemodialysis grafts. Cardiovasc Intervent Radiol.

2005;28(4):472.

Hilleman D et al. Efficacy, safety, and cost of thrombolytic agents for the management of dysfunctional

hemodialysis catheters: a systematic review. Pharmacotherapy. 2011;31(10):1031.

Hemmelgarn BR et al. Prevention of dialysis catheter malfunction with recombinant tissue plasminogen activator.

N EnglJ Med. 2011;364(4):303.

Dixon BS et al. Effect of dipyridamole plus aspirin on hemodialysis graft patency. N Engl J Med.

2009;360(21):2191.

Nee R et al. Cost-effectiveness of antiplatelet therapy to prolong primary patency of hemodialysis graft. Clin

Nephrol. 2014;81(1):38.

Crowther MA et al. Low-intensity warfarin is ineffective for the prevention of PTFE graft failure in patients on

hemodialysis: a randomized controlled trial. J Am Soc Nephrol. 2002;13:2331.

Schmitz PG et al. Prophylaxis of hemodialysis graft thrombosis with fish oil: double-blind, randomized, prospective

trial. J Am Soc Nephrol. 2002;13(1):184.

Lok CE et al. Effect of fish oil supplementation on graft patency and cardiovascular events among patients with

new synthetic arteriovenous hemodialysis grafts: a randomized controlled trial. JAMA. 2012;307(17):1809.

Mermel LA et al. Clinical practice guidelines for the diagnosis and management of intravascular catheter-related

infection: 2009 Update by the Infectious Diseases Society of America. Clin Infect Dis. 2009;49(1):1.

Wasserstein AG. L-carnitine supplementation in dialysis: treatment in quest of disease. Semin Dial. 2013;26(1):11.

Rabindranath KS et al. Automated vs continuous ambulatory peritoneal dialysis: a systematic review of

randomized controlled trials. Nephrol Dial Transplant. 2007;22(10):2991.

Cnossen TT et al. Comparison of outcomes on continuous ambulatory peritoneal dialysis versus automated

peritoneal dialysis: results from a USA database. Perit Dial Int. 2011;31(6):679.

Brophy DF et al. Automated peritoneal dialysis: new implications for pharmacists. Ann Pharmacother.

1997;31:756.

Blake PG, ed. Adequacy of peritoneal dialysis and chronic peritoneal dialysis prescription. In: Daugirdas JT et al,

eds. Handbook of Dialysis. Philadelphia, PA: Wolters Kluwer Health; 2015:464.

Port FK et al. Risk of peritonitis and technique failure by CAPD connection technique: a national study. Kidney

Int. 1992;42:967.

Brown EA et al. Survival of functionally anuric patients on automated peritoneal dialysis: The European APD

Outcome Study. J Am Soc Nephrol. 2003;14:2948.

Moberly JB et al. Pharmacokinetics of icodextrin in peritoneal dialysis patients. Kidney Int. 2002;62(Suppl

81):S23.

Finkelstein F et al. Superiority of icodextrin compared to 4.25% dextrose for peritoneal ultrafiltration. J Am Soc

Nephrol. 2005;16:546.

Cho KH et al. Effect of icodextrin dialysis solution on body weight and fat accumulation over time in CAPD

patients. Nephrol Dial Transplant. 2010;25(2):593.

Silver SA et al. Practical considerations when prescribing icodextrin: a narrative review. Am J Nephrol.

2014;39(6):515.

Firanek CA et al. Avoidable iatrogenic hypoglycemia in patients on peritoneal dialysis: the risks of nonspecific

glucose monitoring devices and drug-device interaction. J Patient Saf. 2014;10(4):218.

Li PK et al. Peritoneal dialysis-related infections recommendations: 2010 update. Perit Dial Int. 2010;30(4):393.

Badve SV et al. Use of aminoglycosides for peritoneal dialysis-associated peritonitis does not affect residual renal

function. Nephrol Dial Transplant. 2012;27(1):381.

Varghese JM et al. Pharmacokinetics of intraperitoneal gentamicin in peritoneal dialysis patients with peritonitis

(GIPD study). Clin J Am Soc Nephrol. 2012;7(8):1249.

Manley HJ et al. Treatment of peritonitis in APD: pharmacokinetic principles. Semin Dial. 2002;15:418.

Piraino B et al. ISPD position statement on reducing the risks of peritoneal dialysis-related infections. Perit Dial

Int. 2011;31(6):614.

Piraino B et al. ISPD guidelines/recommendations. Peritoneal dialysis-related infections recommendations: 2005

update. Perit Dial Int. 2005;25:107.

BernardiniJ et al. Randomized, double-blind trial of antibiotic exit site cream for prevention of exit site infection in

peritoneal dialysis patients. J Am Soc Nephrol. 2005;116:539.

Keane WF et al. Adult peritoneal dialysis-related peritonitis treatment recommendations: 2000 update. Perit Dial

Int. 2000;20:396.

p. 663

The pharmacokinetics and pharmacodynamics of many drugs are altered

in patients with impaired renal function (e.g., declining glomerular

filtration rate) or who are on renal replacement therapy.

Case 31-1 (Questions 1, 2),

Case 31-2 (Question 1),

Case 31-3 (Questions 1, 3),

Case 31-4 (Questions 1, 2),

Case 31-5 (Questions 1, 4),

Case 31-8 (Questions 1–3)

Clinicians should be aware of drugs that require dosage adjustment in the

setting of renal dysfunction to avoid adverse drug events and poor

patient outcomes.

Case 31-1 (Questions 1, 2),

Case 31-3 (Questions 1, 3),

Case 31-4 (Question 1, 2),

Case 31-5 (Questions 1, 4)

Dosages of drugs that are cleared by the kidneys should be adjusted

according to the patient’s renal function (e.g., creatinine clearance).

The initial dose can be determined using the manufacturer’s prescribing

information, published guidelines, or published literature.

Case 31-1 (Questions 1, 4),

Case 31-3 (Question 3),

Case 31-4 (Question 1),

Case 31-5 (Questions 1, 4, 5)

Many drugs have a narrow therapeutic window (range of drug

concentrations that will achieve the desired effect), for which there may

be lack of efficacy with subtherapeutic levels and adverse events

associated with elevated levels. Therapeutic drug concentration

monitoring should be performed to achieve the desired target

concentration.

Case 31-1 (Questions 2, 3),

Case 31-5 (Question 2),

Case 31-7 (Question 1),

Case 31-8 (Question 3)

Renal replacement therapy (e.g., hemodialysis, continuous venovenous

hemofiltration) can have a significant influence on the extracorporeal

removal of drug. Clinicians should be aware of the method of renal

replacement therapy and its impact on drug dosing.

Case 31-1 (Questions 5–8),

Case 31-2 (Questions 1, 2),

Case 31-3 (Questions 1, 4),

Case 31-5 (Questions 3, 4),

Case 31-6 (Question 1)

Biotransformation of drugs may be altered in patients with renal failure.

Active or toxic metabolites may accumulate in patients with renal

failure, leading to adverse effects. Excipients such as diluents can also

accumulate in the setting of renal failure, resulting in toxicity.

Case 31-3 (Question 2),

Case 31-8 (Questions 1, 2)

BASIC PRINCIPLES

The kidneys play an important role in the disposition of many drugs. It is important to

design specific pharmacotherapeutic regimens for patients with renal impairment.

Without careful dosing and therapeutic drug monitoring for select medications in

these patients, accumulation of drugs or toxic metabolites can occur, resulting in

serious adverse effects. Many patients are treated with multiple medications, which

may require even greater attention to dosage adjustment.

In addition to altered drug elimination, numerous other factors associated with

kidney disease predispose patients to potential drug toxicity by altering the

pharmacokinetic disposition and the pharmacodynamic effects of drugs. For example,

the physiologic changes associated with uremia can change drug absorption, protein

binding, distribution, or elimination. These physiologic effects can alter drug

concentrations in the plasma or blood, and at the targeted tissue site of activity,

thereby affecting drug efficacy and toxicity.

Less is known about the effect of renal disease on drug pharmacodynamics, i.e.,

the pharmacologic or toxicologic effects

p. 664

p. 665

produced relative to the drug concentration. Patients with renal disease can be

more sensitive to some drugs, and experience an increased frequency of adverse drug

reactions.

Effect of Renal Failure on Drug Disposition

BIOAVAILABILITY

Although several factors can potentially affect drug absorption in patients with

kidney disease, limited data are available describing altered bioavailability. For

example, drug absorption could be impaired in uremia by nausea, vomiting, diarrhea,

gastritis, and edema of the gastrointestinal (GI) tract, the latter condition being a

complication of nephrotic syndrome. Gastric and intestinal motility, as well as

gastric emptying time, can be altered by the neuropathy associated with uremia.

Uremia also can increase gastric ammonia, leading to an increased gastric pH, which

may affect the bioavailability of drugs that require an acidic environment for

absorption such as ferrous sulfate.

1 Similarly, calcium-containing antacids used by

patients with renal failure for GI symptoms and hyperphosphatemia neutralize

hydrochloric acid in the stomach and increase gastric pH. Patients with end-stage

renal disease (ESRD) often take oral phosphate binders, such as sevelamer and

lanthanum carbonate, which can impair the absorption of other medications.

2,3

The bioavailability of orally administered drugs also depends on the extent to

which the drug is eliminated by first-pass (presystemic) metabolism. The first-pass

hepatic metabolism of oral propranolol was found to be reduced in patients with

renal disease, leading to increased bioavailability.

4 Subsequent studies, however,

attributed the observed increased concentrations of propranolol in renal failure to a

significant increase in the blood to plasma ratio.

5

Intestinal P-glycoprotein activity

may be decreased as well.

6 Other drugs exhibiting increased bioavailability in renal

disease include cloxacillin, propoxyphene, dihydrocodeine, encainide, and

zidovudine (AZT). For example, the area under the concentration–time curve of

dihydrocodeine is increased by 70% in those patients with impaired renal function.

7

PROTEIN BINDING AND VOLUME OF DISTRIBUTION

The extent to which a drug exerts its pharmacologic effects is related to the amount of

free or unbound drug available for distribution to target tissues. Patients with renal

failure often have alterations in plasma protein binding, which can increase the

amount of unbound drug.

8 Clinically, this is most important for highly protein-bound

acidic drugs (>80%), whereas the binding of basic drugs is usually unchanged or

possibly decreased in renal disease. Decreased protein binding of affected drugs

results in increases in the free fraction of drug, an increase in the apparent volume of

distribution (Vd), and higher plasma clearance (Cl) for drugs with a low-extraction

ratio. However, the simultaneous increase in both the Vd and clearance results in

little or no change in the elimination half-life (t1/2

) of these drugs. Alternatively, the

Vd of high-extraction ratio drugs can increase without a concomitant change in

clearance. In this situation, the t1/2 would increase, based on the following

relationships, where Kd is the elimination rate constant of the drug:

In patients with renal failure, the accumulation of uremic toxins may also alter

protein binding. When the free fraction of drugs that are highly protein bound

changes, the interpretation of the total drug concentration must also be considered.

That is, with an increase in the free fraction, the total drug concentration necessary to

exert the desired pharmacologic effect is lower than that needed under normal

conditions.

Table 31-1

Plasma Protein Binding (%) of Acidic Drugs in Renal Failure

Drug Normal Renal Failure

Cefazolin 85 69

Cefoxitin 73 25

Clofibrate 97 91

Diazoxide 94 84

Furosemide 96 94

Pentobarbital 66 59

Phenytoin 88–93 74–84

Salicylate 87–97 74–84

Sulfamethoxazole 66 42

Valproic acid 92 77

Warfarin 99 98

Hypoalbuminemia is a common complication of renal failure. Because acidic

rather than basic drugs are bound to albumin, their protein binding tends to be altered

in patients with renal failure (Table 31-1).

9 Patients with uremia accumulate acidic

by-products that may inhibit binding or displace acidic drugs from albumin binding

sites. This is supported by the observed improvement in protein binding after

removal of uremic by-products by hemodialysis. Finally, the structural conformation

of albumin is altered in renal disease, which may reduce the number or affinity of

binding sites for drugs. Studies have demonstrated differences in the amino acid

composition of albumin between healthy people and patients with uremia.

10 The

anticonvulsant, phenytoin, is a classic example of a drug whose protein binding is

altered in renal disease.

11 This is discussed in more detail later in this chapter.

Renal disease can change the Vd of various drugs. The Vd or “apparent volume of

distribution” is the “volume” or size of a compartment necessary to account for the

total amount of drug in the body if it were present throughout the body at the same

concentration as that found in plasma. A decrease in the plasma protein binding of

highly protein-bound drugs, such as phenytoin, leads to an increase in the apparent

Vd.

Drugs that are not highly protein bound (e.g., gentamicin, isoniazid) have little

change in their Vd in renal disease. Digoxin is a unique exception in that its Vd is

decreased in renal disease. This is attributed to a decrease in myocardial tissue

uptake of digoxin, leading to a decrease in the myocardial or tissue to serum

concentration ratio.

12

ELIMINATION

The extent to which renal disease affects the elimination of a drug depends on the

amount of drug normally excreted unchanged in the urine and the degree of renal

impairment. As kidney disease progresses, the kidney’s ability to excrete uremic

toxins diminishes. Consequently, the ability to eliminate certain drugs that are renally

excreted also decreases. If the dose of these drugs is not modified for the patient’s

degree of renal dysfunction, these drugs will accumulate, potentially leading to an

increase in the pharmacologic effect and toxicity.

The kidney eliminates drugs primarily by filtration or active secretion.

Characteristics of a drug that determine its ability to be filtered include its affinity for

protein binding and its molecular weight (MW). Drugs with low protein binding or

those that are

p. 665

p. 666

displaced from proteins in the setting of renal disease are filtered more readily.

Molecules with a high MW (>20,000 Da) are not readily filtered because of their

large size. The reasons for how renal disease selectively alters the process of

glomerular filtration or tubular secretion of specific drugs are not well understood.

The elimination of drugs by the kidneys in patients with renal disease usually can be

estimated by measuring the ability of the kidney to eliminate substances such as

creatinine (i.e., creatinine clearance [CrCl]) (see Chapter 29, Acute Kidney Injury).

Organic anion transporters (OATs) are predominantly found in the basolateral

membrane of the renal tubules. OATs facilitate the uptake of small organic anions

into renal tubular cells. Decreased OAT activity as a result of acute kidney injury can

decrease the renal secretion of various drugs such as methotrexate, nonsteroidal

antiinflammatory drugs, and acetylsalicylic acid.

13

Renal disease can also have an important impact on the elimination of drugs that

are primarily metabolized by the liver.

14 Metabolic processes, such as hydroxylation

and glucuronidation, often produce inactive, more polar compounds that can be

eliminated by the kidney. The metabolites of some drugs (e.g., meperidine, morphine,

procainamide) are pharmacologically active or toxic. In patients with renal disease,

these metabolites may accumulate, leading to an increase in pharmacologic activity

and adverse effects.

15,16 For example, the central nervous system (CNS) toxicity

observed in renal disease has been attributed to accumulation of the morphine

metabolite, morphine-6-glucuronide. Therefore, careful dosing modifications or

avoidance of these drugs is warranted in patients with renal impairment. Metabolic

enzymes have been found within renal tissue, and may play a role in the metabolism

of some drugs.

17,18 For example, the nonrenal clearance of drugs (e.g., acyclovir)

decreases in patients with renal impairment, and is believed to be caused by a

decrease in “renal” metabolism.

19

Excipients used to formulate medications should also be considered. For example,

the pharmacokinetics of itraconazole and voriconazole are not significantly altered in

the setting of renal dysfunction. However, the parenteral formulations of itraconazole,

posaconazole, and voriconazole contain the solubilizing agent, β-cyclodextrin, which

is normally rapidly eliminated by glomerular filtration but can accumulate in patients

with renal impairment, causing GI disturbances.

20

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