(Go: >> BACK << -|- >> HOME <<)

SlideShare a Scribd company logo
Drugs pharmacology in kidney disease
Drugs Pharmacology
in Kidney Disease
By
M. H. Farjoo M.D., Ph.D., Bioanimator
Shahid Beheshti University of Medical Sciences
Drugs Pharmacology in Kidney Disease
 Weak Acids & Weak Bases
 The Order of Drug Metabolism
 Protein Binding
 Creatinine
 Drug Dosing
 Drug Absorption
 Renal Excretion, Reabsorption and Metabolism
 Drug Induced Renal Injuries
 Prescribing in Renal Disease
 Therapeutic Drug Monitoring
 Drug Selection
Weak Acids & Weak Bases
 The protonated form of a weak acid is neutral.
 The unprotonated form of a weak base is neutral.
 The neutral (uncharged) form is more lipid-soluble.
 Acids are more lipid-soluble at acid pH, and bases are
more lipid-soluble at alkaline pH.
 Weak acids and weak bases gain or lose protons
depending on the pH.
 So their movement between aqueous & lipid
mediums varies with the pH.
Weak Acids & Weak Bases
 If kidney filters the drug, by changing the urine pH
the drug can be "trapped" in the urine (in overdose).
 Weak acids are excreted faster in alkaline urine and
vise versa.
 The ionized particles cannot pass easily through renal
tubular membranes.
 Therefore, less drug is reabsorbed into the blood and
more is excreted by the kidneys.
Weak Acids & Weak Bases
 Sodium bicarbonate + phenobarbital → increased
excretion of phenobarbital.
 The sodium bicarbonate alkalinizes the urine, raising
the number of barbiturate ions in the renal filtrate.
 A large number of drugs are weak bases, and most of
them are amine-containing molecules.
 Primary, secondary, and tertiary amines undergo
protonation and vary their solubility with pH.
 Quaternary amines are always in the poorly lipid-
soluble charged form.
Drugs pharmacology in kidney disease
Drugs pharmacology in kidney disease
Weak Acids & Weak Bases
Body Fluid Range of pH
Fluid: Blood
Ratios for
Sulfadiazine
(acid, pKa 6.5)
Fluid: Blood
Ratios for
Pyrimethamine
(base, pKa 7.0)
Urine 5.0-8.0 0.12-4.65 0.79-72.24
Breast milk 6.4-7.6 0.2-1.77 0.89-3.56
Jejunum, ileum
contents
7.5-8.0 1.23-3.54 0.79-0.94
Stomach contents 1.92-2.59 0.11 18,386-85,993
Prostatic secretions 6.45-7.4 0.21 1.0-3.25
Vaginal secretions 3.4-4.2 0.11 452-2848
Acidification of Urine
 Increases elimination of amphetamine, methylene
dioxymethamfetamine (MDMA or ‘ecstasy’),
dexfenfluramine, quinine and phencyclidine.
 Oral NH4Cl, taken with food to avoid vomiting,
acidifies the urine.
 It should not be given to patients with impaired renal or
hepatic function.
 Other means include arginine hydrochloride, Ascorbic
acid and calcium chloride by mouth.
Alkalinisation of Urine
 Increases the elimination of salicylate, phenobarbital,
and some herbicides.
 Treats crystal nephropathy by increasing drug
solubility of methotrexate, sulphonamides and
triamterene.
 Reduces irritation of an inflamed urinary tract
 Discourages the growth of E. coli.
 The urine can be made alkaline by sodium bicarbonate
i.v., or by potassium citrate by mouth.
The Order of Drug Metabolism
 The rate at which drugs are metabolized, is called “the
order” of reaction.
 Two orders exist:
 First-order (exponential): a constant fraction of drug is
transported or metabolised in unit time.
 Zero-order (saturation kinetics): a constant amount of
drug is transported or metabolised in unit time.
 In doses used clinically, most drugs show first order
processes.
 Enzyme-mediated reactions often show zero-order (rate
limitation) dynamics.
Changes in plasma concentration following an intravenous bolus
injection of a drug in the elimination phase.
As elimination is a first-order process, the time for any concentration
point to fall by 50% (t½) is always the same.
Drugs pharmacology in kidney disease
The Order of Drug Metabolism
 Zero-order kinetic has important consequences for
drugs such as:
 Alcohol
 Phenytoin
 Aspirin
 In first order kinetic, because the rate of reaction is
constant, it is dose independent,
 By approaching zero order kinetic, increase in dose,
alters the kinetics, and it become dose dependent.
 Albumin is the main drug-binding plasma protein for
acidic drugs.
 Protein binding in uremic patients is decreased for all
acidic drugs.
 This is due to decreased albumin or reduced binding
capacity.
 Albumin decrease occurs in renal failure (albumin is
lost in the urine).
Protein Binding
 The reduced binding is because of:
 Reductions in albumin concentration.
 Structural changes in the albumin molecule.
 Uremic toxins compete with drugs for protein binding.
 Reductions in albumin binding, increases volume of
distribution (Vd)of drugs.
 Edema fluid (an ECF) in renal impairment also increases
Vd of water-soluble drugs.
 So more unbound drug distributes into sites of elimination.
Protein Binding
Protein binding Pic.
 The elimination clearance of drugs is increased.
 Drug half-life and therapeutic effects is decreased.
 The higher levels of unbound drug can result in
toxicity.
 However, protein binding changes do not affect
distribution volume or clearance of unbound drug.
Protein Binding
Protein binding Pic.
 The protein binding of basic drugs tends to be normal
or even increased.
 For basic drugs (clindamycin, propafenone), alpha1-
acid glycoprotein (AAG) is the main binding protein.
 The amount of AAG increases in renal renal failure.
 In these patients larger amounts of a basic drug is
bound and a smaller amount is free to exert an effect.
Protein Binding
 Alterations in tissue binding of drugs (digoxin) also
occurs.
 Renal failure decreases digoxin volume of distribution.
 Digoxin is displaced from tissues by:
 Metabolic products that cannot be excreted by impaired
kidneys
 A reduction in tissue levels of Na/K-ATPase, the
tissue-binding site for digoxin.
Protein Binding
Protein Binding:
Comparison of Free and Total Phenytoin Levels
Protein
Bound
Phenytoin
 Renal status should be monitored in any client with
risk factors for renal insufficiency.
 Signs and symptoms of renal failure include:
 Decreased urine output (<600 mL/24 hours)
 Increased BUN or increased creatinine (>2 mg/dL)
Creatinine
Creatinine
 Renal impairment accounts for one-third of the
prescribing errors.
 Dose assessment solely based on serum creatinine (Cr)
without estimation of Cr clearance is misleading.
 Creatinine is determined by both muscle mass and
GFR.
 So its measurement cannot be used as the sole indicator
of renal function.
Drugs pharmacology in kidney disease
 Serum creatinine is a relatively unreliable indicator of
renal function in elderly clients.
 Because of diminished muscle mass, they may have a
normal creatinine even if their GFR is markedly
reduced.
 Some drugs (cimetidine and trimethoprim) increase
creatinine and create a false impression of renal
failure.
 They interfere with secretion of creatinine into kidney
tubules.
Creatinine
 Estimations of creatinine clearance are more accurate
for:
 Clients with stable renal function (stable serum
creatinine).
 Average muscle mass (for their age, and BMI).
 Estimations are less accurate for:
 Emaciated and obese clients.
 For those with changing renal function (as in acute
illness).
Creatinine
Drug Dosing
 The steady state of drug is only determined by drug
dose and elimination clearance (CLE).
 Since the metabolism of most drugs is by first-order,
Drug metabolism is characterized by clearance.
 Non-renal clearance is usually by the liver.
 It also includes hemodialysis and other methods of
drug removal.
Drug Dosing
 For determining the required dose in renal failure, the
Dettli method is used.
 The reliability of the Dettli method of predicting drug
clearance depends on two critical assumptions:
1. The non-renal clearance of the drug remains constant
when renal function is impaired.
2. ClE (total) declines in a linear fashion with ClCr.
the relationship between total systemic drug clearance (CLTOT)
and creatinine clearance (CLCR).
CLR, renal clearance; CLNR, nonrenal clearance.
The Dettli method of drug dosing
A Case for Cimetidine
 A functionally anephric patient postoperatively
complained of GE reflux, so cimetidine was started.
 Because of renal failure, the dose was halved.
 Three days later, the patient was confused and
diagnosis of dialysis dementia was made.
 Later the suggestion was made that cimetidine be
discontinued.
 Two days later the patient was alert and was
discharged from the hospital.
A Case for Cimetidine
 The Cimetidine label states that Patients with creatinine
clearance <30 cc/min should receive half the
recommended dose.
 However, under “Pharmacokinetics” it indicates that
75% of the drug is excreted via kidneys.
 Only one fourth of the dose is eliminated by non-renal
mechanisms.
 So patients who receive half the usual dose, still will
receive twice the adequate dose!
Cimetidine elimination clearance
Drug Absorption
 The bioavailability of most drugs does not change in
impaired renal function.
 Absorption of oral drugs may be decreased indirectly
by:
 Delayed gastric emptying
 Vomiting and diarrhea
 Edema of the GI tract (with generalized edema).
 Changes in gastric pH
 Changes in gastric pH in CRF occurs by:
 Oral alkalinizing agents (bicarbonate, citrate).
 Use of antacids for phosphate-binding effects.
 This causes decreased absorption of oral acidic drugs
and increases absorption of alkaline drugs.
 Increased bioavailability of drugs with first-pass
metabolism is possible (impairment of metabolizing
enzymes).
Drug Absorption
Renal Excretion
 Glomerular filtration affects all drugs of small
molecular size and is restrictive.
 Restrictive means that glomerular filtration is
influenced by protein binding of drug.
 Renal tubular secretion is non-restrictive, and both
protein-bound and free drug are eliminated.
 Competition by drugs for renal tubular secretion is an
important cause of drug–drug interactions.
Renal Excretion
 Anionic drugs compete with other anionic drugs for
active transport pathways, as do cationic drugs.
 When two drugs secreted by the same pathway are
given together, the renal clearance of each of them
decreases.
 For example, renal clearance of methotrexate is halved
when salicylate is co-administered with it.
Renal Excretion
 ClCr reflects GFR and renal drug clearance correlates
with the reciprocal of serum creatinine or ClCr.
 ClCr is also a guide to the renal clearance of drugs with
extensive renal tubular secretion or reabsorption.
 This is the result of glomerulo-tubular balance (GTB)
so drug Cl declines fairly linearly with reductions in
ClCr.
 GTB is the ability of the proximal tubule to reabsorb a
constant fraction of glomerular filtrate delivered to it.
Renal Excretion
 Based on renal clearance values, two conclusions can
be made:
 If renal clearance exceeds drug filtration rate, there
is net renal tubular secretion of the drug.
 If renal clearance is less than drug filtration rate,
there is net renal tubular reabsorption of the drug.
 Excretion of many drugs is reduced in renal failure.
 The kidneys normally excrete both the parent drug
and metabolites produced by the liver.
 Renal excretion mechanisms includes:
 Glomerular filtration
 Tubular secretion
 Tubular reabsorption
 All of these mechanism are affected by renal
impairment.
Renal Excretion
 An adequate fluid intake is required to excrete drugs
by the kidneys.
 Any factor that depletes ECF increases the risk of
worsening renal impairment which include:
 Inadequate fluid intake
 Diuretic drugs
 Loss of body fluids (bleeding, vomiting, diarrhea)
Renal Excretion
 Lithium (Li) is entirely excreted by the kidneys and
has a very narrow therapeutic range.
 80% of Li is reabsorbed in the proximal tubules.
 The amount of reabsorption depends on the
concentration of sodium in the proximal tubules.
 Deficiency of sodium or co-administration of
diuretic: sodium loss↑, Li excretion↓ => Li toxicity↑
 Excessive sodium intake lowers Li to non therapeutic
ranges.
Renal Reabsorption
Renal Reabsorption
 Proximal tubular endocytosis, is important in removing
proteins that pass through glomerular pores.
 Aminoglycosides are filtered at the glomerulus but are
nephrotoxic because of active uptake by endocytosis.
 This uptake is saturable, so aminoglycosides are less
nephrotoxic in single daily doses than separate doses.
 Metabolism may, or may not change by renal failure
because:
 In uremia, in liver, reduction and hydrolysis is
slower, but oxidation by CYPs and conjugation are
normal.
 Impaired kidney may not eliminate metabolites of
the parent drug with pharmacologic activity.
 The kidney contains many of the same
metabolizing enzymes found in the liver (eg: renal
CYP enzymes).
Renal Metabolism
Drugs pharmacology in kidney disease
Non-renal Metabolism
 Hepatic drug clearance (ClH) may be altered in chronic
renal failure.
 This may be due to depression in hepatic organic anion
transport polypeptides, or increase in P-gp expression.
 Impaired renal function impacts hepatic clearance in
both Phase I, and Phase II metabolization .
 This is particularly pronounced in end-stage renal
disease (ESRD) and Phase I metabolization.
Non-renal Metabolism
 The effects of impaired renal function on hepatic
clearance is due to:
 Accumulation of parathyroid hormone (PTH)
 Cytokines
 Toxins
 Alteration in gene transcription.
Drugs pharmacology in kidney disease
Drugs pharmacology in kidney disease
Direct Drug-induced Renal Disease
 Heavy metals: mercury, gold, iron, lead.
 Antimicrobials: aminoglycosides, amphotericin,
cephalosporins.
 Iodinated contrast media: agents for the biliary tract
 Solvents: carbon tetrachloride, ethylene glycol.
 NSAID combinations.
 NSAIDs can cause renal impairment even though
they are eliminated mainly by hepatic metabolism.
 Acetaminophen is nephrotoxic in overdose because of
a metabolite that may cause kidney necrosis.
 Aspirin is nephrotoxic in high doses, and its protein
binding is reduced in renal failure.
Direct Drug-induced Renal Disease,
NSAIDs
Drugs pharmacology in kidney disease
 NSAIDs can decrease blood flow in the kidneys by
inhibiting synthesis of prostaglandins (PG) that dilate
renal blood vessels.
 When renal blood flow is normal, these PGs have
limited activity.
 When renal blood flow is decreased, their synthesis is
increased to protect the kidneys from ischemia.
 In those who depend on PGs to maintain renal blood
flow, NSAIDs result in decreased GFR, and retention
of salt and water.
Direct Drug-induced Renal Disease,
NSAIDs
Drugs pharmacology in kidney disease
 NSAIDs can also cause kidney damage by a
hypersensitivity reaction that leads to ARF.
 NSAIDs may adversely affect a fetus’s kidneys when:
 Given during late pregnancy to prevent premature
labor.
 Given after birth for PDA.
Direct Drug-induced Renal Disease,
NSAIDs
Indirect Drug-induced Renal Disease
 Cytotoxic drugs and uricosurics may cause urate to be
precipitated in the tubule.
 Calciferol may cause renal calcification by inducing
hypercalcaemia.
 Diuretic and laxative abuse can cause tubular damage
secondary to potassium and sodium depletion.
 Anticoagulants may cause haemorrhage into the
kidney.
Other Drug-induced Kidney Lesions
 Vasculitis: allopurinol, isoniazid, sulphonamides.
 Allergic interstitial nephritis: penicillins, thiazides,
allopurinol, phenytoin, sulphonamides.
 Drug-induced lupus erythematosus: hydralazine,
procainamide, sulfasalazine.
Drug Induced Renal Syndromes
 Acute renal failure: aminoglycosides, cisplatin.
 Chronic renal failure: NSAIDs.
 Nephrotic syndrome: penicillamine, gold,
 Functional impairment:
 Reduced ability to dilute and concentrate urine: lithium
 Potassium loss in urine: loop diuretics
 Acid–base imbalance: acetazolamide
Drug t½ (h) in normal and severely impaired renal
function
Prescribing in Renal Disease
 Adjustment of the loading dose is generally
unnecessary (Vd usually does not change).
 Digoxin is an exception because the Vd is contracted in
uremic patients due to altered tissue binding.
 For maintenance dose either reduce each dose or
lengthen the time between doses.
 Dosage of many drugs needs to be decreased in renal
failure including:
 Aminoglycoside antibiotics
 Most cephalosporin antibiotics
 Fluoroquinolones
 Digoxin
Prescribing in Renal Disease
Prescribing in Renal Disease
 Special caution is needed in:
 hypoproteinemia and the drug is extensively plasma
protein bound
 Advanced renal disease when accumulated
metabolic products may compete for protein binding
sites.
 The early stages of dosing until response to the drug
can be gauged.
 Elderly, kidney blood flow, GFR, and tubular
secretion of drugs is decreased (accumulation of
drug).
Therapeutic Drug Monitoring
 Patients differ greatly in the dose of drug required to
achieve the same response.
 The dose of warfarin varies up to five-fold between
individuals.
 The question is: how optimal drug effect can be achieved
quickly for the individual patient?
 For drugs with short t½, both peak (15 min after an IV
dose) and trough (just before the next dose) concentrations
should be known.
 For drugs with long t½, it is best to sample just before a
dose is due.
Therapeutic Drug Monitoring
 Sometimes TDM does not worth the cost:
 Plasma concentration may not be worth measuring
(antihypertensive [blood pressure], hypoglycemics [blood
glucose], diuretics [urine output], warfarin [INR]).
 Plasma concentration has no correlation with effect (Aspirin
as antiplatelet, anticancer drugs).
 Plasma concentration may correlate poorly with effect
(Diazepam with many active metabolites).
 Drug selection is guided by renal function and the
effects of drugs on renal function.
 Many commonly used drugs may adversely affect
renal function (NSAIDs or OTC drugs).
 Some drugs are excreted exclusively by the kidneys
(aminoglycosides, lithium).
 Some drugs are contraindicated in renal impairment
(tetracyclines except doxycycline).
Drug Selection
 Drugs can be used if guidelines are followed
(reducing dosage, using TDM and renal function
tests, avoiding dehydration).
 Drugs known to be nephrotoxic should be avoided.
 Sometime there are no substitutes and nephrotoxic
drugs must be given.
 Some commonly used nephrotoxic drugs include
aminoglycosided, amphotericin B, and cisplatin.
Drug Selection
Drugs pharmacology in kidney disease
Thank you
Any question?
Sites of action of diuretic drugs.
ENaC, epithelial sodium channel; NCCT, thiazide-sensitive Na–Cl co-transporter;
NKCC2, Na–K–2Cl co-transporter; ROMK, rectifying outer medullary potassium
channel.
Drugs pharmacology in kidney disease

More Related Content

What's hot

Hypolipidemic agents
Hypolipidemic agentsHypolipidemic agents
Hypolipidemic agents
Dr.Bhargav Purohit
 
Hypolipidaemic vinay
Hypolipidaemic vinayHypolipidaemic vinay
Hypolipidaemic vinay
vinay tuteja
 
Powerpoints Hypolipidemics
Powerpoints HypolipidemicsPowerpoints Hypolipidemics
Powerpoints Hypolipidemics
MD Specialclass
 
Hypolipidemic drugs MBBS and PG Dr. Resu Neha Reddy
Hypolipidemic drugs MBBS and PG Dr. Resu Neha ReddyHypolipidemic drugs MBBS and PG Dr. Resu Neha Reddy
Hypolipidemic drugs MBBS and PG Dr. Resu Neha Reddy
Dr Resu Neha Reddy
 
Ezetimibe And Lipids - Dr Vivek Baliga Reviews
Ezetimibe And Lipids - Dr Vivek Baliga ReviewsEzetimibe And Lipids - Dr Vivek Baliga Reviews
Ezetimibe And Lipids - Dr Vivek Baliga Reviews
Dr Vivek Baliga
 
Mahi ddmp
Mahi ddmpMahi ddmp
Mahi ddmp
mahender Kotte
 
16.hypolipidemics
16.hypolipidemics16.hypolipidemics
16.hypolipidemics
Dr.Manish Kumar
 
Estimation of various biochemical parameter in serum
Estimation of various biochemical parameter in serumEstimation of various biochemical parameter in serum
Estimation of various biochemical parameter in serum
Sayanti Sau
 
Hypolipidaemic drugs
Hypolipidaemic drugsHypolipidaemic drugs
Hypolipidaemic drugs
drnutan goswami
 
Antilipemics
AntilipemicsAntilipemics
Antilipemics
raj kumar
 
Class hypolipidemics
Class hypolipidemicsClass hypolipidemics
Class hypolipidemics
Raghu Prasada
 
lipid-lowering drugs
lipid-lowering drugslipid-lowering drugs
lipid-lowering drugs
Elham Khaled
 
Liver function tests 2020
Liver function tests 2020Liver function tests 2020
Liver function tests 2020
subramaniam sethupathy
 
Applications of pharmacokinetics parameters in disease states
Applications of pharmacokinetics parameters in disease statesApplications of pharmacokinetics parameters in disease states
Applications of pharmacokinetics parameters in disease states
Umair hanif
 
Hypolipidaemic drugs
Hypolipidaemic drugsHypolipidaemic drugs
Hypolipidaemic drugs
Shweta Supriya Pradhan
 
Enzymes in liver disorders presentation with cases
Enzymes in liver disorders presentation  with casesEnzymes in liver disorders presentation  with cases
Enzymes in liver disorders presentation with cases
Sri Devraj Urs medical college, Kolar, Karnataka , India
 
lipid lowering agents
lipid lowering agentslipid lowering agents
lipid lowering agents
srikalayenigalla
 
Hypolipidemic drugs
Hypolipidemic drugsHypolipidemic drugs
Hypolipidemic drugs
ajaykumarbp
 
Hyperlipidemia
Hyperlipidemia Hyperlipidemia
Hyperlipidemia
PriyanshiPatel18
 
bile acid sequestrants
bile acid sequestrantsbile acid sequestrants
bile acid sequestrants
Shirsak Mondal
 

What's hot (20)

Hypolipidemic agents
Hypolipidemic agentsHypolipidemic agents
Hypolipidemic agents
 
Hypolipidaemic vinay
Hypolipidaemic vinayHypolipidaemic vinay
Hypolipidaemic vinay
 
Powerpoints Hypolipidemics
Powerpoints HypolipidemicsPowerpoints Hypolipidemics
Powerpoints Hypolipidemics
 
Hypolipidemic drugs MBBS and PG Dr. Resu Neha Reddy
Hypolipidemic drugs MBBS and PG Dr. Resu Neha ReddyHypolipidemic drugs MBBS and PG Dr. Resu Neha Reddy
Hypolipidemic drugs MBBS and PG Dr. Resu Neha Reddy
 
Ezetimibe And Lipids - Dr Vivek Baliga Reviews
Ezetimibe And Lipids - Dr Vivek Baliga ReviewsEzetimibe And Lipids - Dr Vivek Baliga Reviews
Ezetimibe And Lipids - Dr Vivek Baliga Reviews
 
Mahi ddmp
Mahi ddmpMahi ddmp
Mahi ddmp
 
16.hypolipidemics
16.hypolipidemics16.hypolipidemics
16.hypolipidemics
 
Estimation of various biochemical parameter in serum
Estimation of various biochemical parameter in serumEstimation of various biochemical parameter in serum
Estimation of various biochemical parameter in serum
 
Hypolipidaemic drugs
Hypolipidaemic drugsHypolipidaemic drugs
Hypolipidaemic drugs
 
Antilipemics
AntilipemicsAntilipemics
Antilipemics
 
Class hypolipidemics
Class hypolipidemicsClass hypolipidemics
Class hypolipidemics
 
lipid-lowering drugs
lipid-lowering drugslipid-lowering drugs
lipid-lowering drugs
 
Liver function tests 2020
Liver function tests 2020Liver function tests 2020
Liver function tests 2020
 
Applications of pharmacokinetics parameters in disease states
Applications of pharmacokinetics parameters in disease statesApplications of pharmacokinetics parameters in disease states
Applications of pharmacokinetics parameters in disease states
 
Hypolipidaemic drugs
Hypolipidaemic drugsHypolipidaemic drugs
Hypolipidaemic drugs
 
Enzymes in liver disorders presentation with cases
Enzymes in liver disorders presentation  with casesEnzymes in liver disorders presentation  with cases
Enzymes in liver disorders presentation with cases
 
lipid lowering agents
lipid lowering agentslipid lowering agents
lipid lowering agents
 
Hypolipidemic drugs
Hypolipidemic drugsHypolipidemic drugs
Hypolipidemic drugs
 
Hyperlipidemia
Hyperlipidemia Hyperlipidemia
Hyperlipidemia
 
bile acid sequestrants
bile acid sequestrantsbile acid sequestrants
bile acid sequestrants
 

Similar to Drugs pharmacology in kidney disease

Pharmakokinetic Variations in Kidney diseases.
Pharmakokinetic Variations in Kidney diseases.Pharmakokinetic Variations in Kidney diseases.
Pharmakokinetic Variations in Kidney diseases.
Maleha Sial
 
Effects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokineticsEffects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokinetics
Dr. Ramesh Bhandari
 
Effects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokineticsEffects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokinetics
Dr. Ramesh Bhandari
 
Elimination
EliminationElimination
Elimination
Kailas Mali
 
Drugs pharmacology in liver disease
Drugs pharmacology in liver diseaseDrugs pharmacology in liver disease
Drugs pharmacology in liver disease
Drugs pharmacology in liver diseaseDrugs pharmacology in liver disease
dosage adjustment in renal and hepatic failure for medical student
dosage adjustment in renal and hepatic failure for medical studentdosage adjustment in renal and hepatic failure for medical student
dosage adjustment in renal and hepatic failure for medical student
DeepaJoshi41
 
Drug Dosing in Renal Failure.pdf
Drug Dosing in Renal Failure.pdfDrug Dosing in Renal Failure.pdf
Drug Dosing in Renal Failure.pdf
samthamby79
 
EFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptx
EFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptxEFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptx
EFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptx
Dr. Sabareesh Subramanian A
 
HepaticDosageAdjustment.pdf
HepaticDosageAdjustment.pdfHepaticDosageAdjustment.pdf
HepaticDosageAdjustment.pdf
Helena949653
 
Dosing of drugs in liver failure
Dosing of drugs in liver failureDosing of drugs in liver failure
Dosing of drugs in liver failure
YOUAN BI BENIET MARIUS
 
Drug Elimination
Drug EliminationDrug Elimination
Drug Elimination
UsmanKhalid135
 
Variations in Pk's in disease states.pptx
Variations in Pk's in disease states.pptxVariations in Pk's in disease states.pptx
Variations in Pk's in disease states.pptx
SARADPAWAR1
 
excretion
excretionexcretion
excretion
Pinkesh Patel
 
Drugs and kidney
Drugs and kidneyDrugs and kidney
Drugs and kidney
Mahmoud El-saharty
 
Variations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdfVariations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdf
SARADPAWAR1
 
Variations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdfVariations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdf
SARADPAWAR1
 
Handling of drugs in Renal Failure (kinetics and dynamics)
Handling of drugs in Renal Failure (kinetics and dynamics) Handling of drugs in Renal Failure (kinetics and dynamics)
Handling of drugs in Renal Failure (kinetics and dynamics)
Bilal AL-mosheqh
 
HYPERURICAEMIA + all related brand training material.pptx
HYPERURICAEMIA  + all related brand training material.pptxHYPERURICAEMIA  + all related brand training material.pptx
HYPERURICAEMIA + all related brand training material.pptx
Pabitra Thapa
 
Geriatrics
GeriatricsGeriatrics
Geriatrics
Abhinav Sawhney
 

Similar to Drugs pharmacology in kidney disease (20)

Pharmakokinetic Variations in Kidney diseases.
Pharmakokinetic Variations in Kidney diseases.Pharmakokinetic Variations in Kidney diseases.
Pharmakokinetic Variations in Kidney diseases.
 
Effects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokineticsEffects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokinetics
 
Effects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokineticsEffects of liver diseases on pharmacokinetics
Effects of liver diseases on pharmacokinetics
 
Elimination
EliminationElimination
Elimination
 
Drugs pharmacology in liver disease
Drugs pharmacology in liver diseaseDrugs pharmacology in liver disease
Drugs pharmacology in liver disease
 
Drugs pharmacology in liver disease
Drugs pharmacology in liver diseaseDrugs pharmacology in liver disease
Drugs pharmacology in liver disease
 
dosage adjustment in renal and hepatic failure for medical student
dosage adjustment in renal and hepatic failure for medical studentdosage adjustment in renal and hepatic failure for medical student
dosage adjustment in renal and hepatic failure for medical student
 
Drug Dosing in Renal Failure.pdf
Drug Dosing in Renal Failure.pdfDrug Dosing in Renal Failure.pdf
Drug Dosing in Renal Failure.pdf
 
EFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptx
EFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptxEFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptx
EFFECT OF HEPATIC DISEASE ON PHARMACOKINETICS.pptx
 
HepaticDosageAdjustment.pdf
HepaticDosageAdjustment.pdfHepaticDosageAdjustment.pdf
HepaticDosageAdjustment.pdf
 
Dosing of drugs in liver failure
Dosing of drugs in liver failureDosing of drugs in liver failure
Dosing of drugs in liver failure
 
Drug Elimination
Drug EliminationDrug Elimination
Drug Elimination
 
Variations in Pk's in disease states.pptx
Variations in Pk's in disease states.pptxVariations in Pk's in disease states.pptx
Variations in Pk's in disease states.pptx
 
excretion
excretionexcretion
excretion
 
Drugs and kidney
Drugs and kidneyDrugs and kidney
Drugs and kidney
 
Variations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdfVariations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdf
 
Variations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdfVariations in Pk's in disease states.pdf
Variations in Pk's in disease states.pdf
 
Handling of drugs in Renal Failure (kinetics and dynamics)
Handling of drugs in Renal Failure (kinetics and dynamics) Handling of drugs in Renal Failure (kinetics and dynamics)
Handling of drugs in Renal Failure (kinetics and dynamics)
 
HYPERURICAEMIA + all related brand training material.pptx
HYPERURICAEMIA  + all related brand training material.pptxHYPERURICAEMIA  + all related brand training material.pptx
HYPERURICAEMIA + all related brand training material.pptx
 
Geriatrics
GeriatricsGeriatrics
Geriatrics
 

More from Mohammad Hadi Farjoo MD, PhD, Shahid behehsti University of Medical Sciences

Applied statistics part 5
Applied statistics part 5Applied statistics part 5
Applied statistics part 4
Applied statistics part  4Applied statistics part  4
Applied statistics part 3
Applied statistics part 3Applied statistics part 3
Applied statistics part 2
Applied statistics  part 2Applied statistics  part 2
Applied statistics part 1
Applied statistics part 1Applied statistics part 1
Drugs used in disorders of coagulation
Drugs used in disorders of coagulationDrugs used in disorders of coagulation
Agents used in anemias hematopoietic growth factors
Agents used in anemias hematopoietic growth factorsAgents used in anemias hematopoietic growth factors
Agents used in anemias hematopoietic growth factors
Mohammad Hadi Farjoo MD, PhD, Shahid behehsti University of Medical Sciences
 
Drugs used in dyslipidemia
Drugs used in dyslipidemiaDrugs used in dyslipidemia
Immunopharmacology
Immunopharmacology Immunopharmacology
Management of the poisoned patient.
Management of the poisoned patient.Management of the poisoned patient.
Rational prescribing & prescription writing
Rational prescribing & prescription writingRational prescribing & prescription writing
Drug use in pregnancy and lactation part 2
Drug use in pregnancy and lactation part 2Drug use in pregnancy and lactation part 2
Drug use in pregnancy and lactation part 1
Drug use in pregnancy and lactation part 1Drug use in pregnancy and lactation part 1
Drug use in pregnancy and lactation part 3
Drug use in pregnancy and lactation part 3Drug use in pregnancy and lactation part 3
Drugs causing methemoglobinemia
Drugs causing methemoglobinemiaDrugs causing methemoglobinemia
Drugs pharmacology in lung disease
Drugs pharmacology in lung diseaseDrugs pharmacology in lung disease
Drugs pharmacology in heart disease
Drugs pharmacology in heart diseaseDrugs pharmacology in heart disease
Academic writing 2nd part 6 bahman 1398
Academic writing 2nd part 6 bahman 1398Academic writing 2nd part 6 bahman 1398
Academic writing part 1
Academic writing part 1Academic writing part 1
treatment of cardiac arrhythmias 2
treatment of cardiac arrhythmias 2treatment of cardiac arrhythmias 2

More from Mohammad Hadi Farjoo MD, PhD, Shahid behehsti University of Medical Sciences (20)

Applied statistics part 5
Applied statistics part 5Applied statistics part 5
Applied statistics part 5
 
Applied statistics part 4
Applied statistics part  4Applied statistics part  4
Applied statistics part 4
 
Applied statistics part 3
Applied statistics part 3Applied statistics part 3
Applied statistics part 3
 
Applied statistics part 2
Applied statistics  part 2Applied statistics  part 2
Applied statistics part 2
 
Applied statistics part 1
Applied statistics part 1Applied statistics part 1
Applied statistics part 1
 
Drugs used in disorders of coagulation
Drugs used in disorders of coagulationDrugs used in disorders of coagulation
Drugs used in disorders of coagulation
 
Agents used in anemias hematopoietic growth factors
Agents used in anemias hematopoietic growth factorsAgents used in anemias hematopoietic growth factors
Agents used in anemias hematopoietic growth factors
 
Drugs used in dyslipidemia
Drugs used in dyslipidemiaDrugs used in dyslipidemia
Drugs used in dyslipidemia
 
Immunopharmacology
Immunopharmacology Immunopharmacology
Immunopharmacology
 
Management of the poisoned patient.
Management of the poisoned patient.Management of the poisoned patient.
Management of the poisoned patient.
 
Rational prescribing & prescription writing
Rational prescribing & prescription writingRational prescribing & prescription writing
Rational prescribing & prescription writing
 
Drug use in pregnancy and lactation part 2
Drug use in pregnancy and lactation part 2Drug use in pregnancy and lactation part 2
Drug use in pregnancy and lactation part 2
 
Drug use in pregnancy and lactation part 1
Drug use in pregnancy and lactation part 1Drug use in pregnancy and lactation part 1
Drug use in pregnancy and lactation part 1
 
Drug use in pregnancy and lactation part 3
Drug use in pregnancy and lactation part 3Drug use in pregnancy and lactation part 3
Drug use in pregnancy and lactation part 3
 
Drugs causing methemoglobinemia
Drugs causing methemoglobinemiaDrugs causing methemoglobinemia
Drugs causing methemoglobinemia
 
Drugs pharmacology in lung disease
Drugs pharmacology in lung diseaseDrugs pharmacology in lung disease
Drugs pharmacology in lung disease
 
Drugs pharmacology in heart disease
Drugs pharmacology in heart diseaseDrugs pharmacology in heart disease
Drugs pharmacology in heart disease
 
Academic writing 2nd part 6 bahman 1398
Academic writing 2nd part 6 bahman 1398Academic writing 2nd part 6 bahman 1398
Academic writing 2nd part 6 bahman 1398
 
Academic writing part 1
Academic writing part 1Academic writing part 1
Academic writing part 1
 
treatment of cardiac arrhythmias 2
treatment of cardiac arrhythmias 2treatment of cardiac arrhythmias 2
treatment of cardiac arrhythmias 2
 

Recently uploaded

BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATIONBASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
JaiDivyaTella
 
Juvenile Myelomonocytic Leukemia (JMML)
Juvenile Myelomonocytic Leukemia   (JMML)Juvenile Myelomonocytic Leukemia   (JMML)
Juvenile Myelomonocytic Leukemia (JMML)
Reenaz Shaik
 
virus pharmaceutical microbiology .pdf..
virus pharmaceutical microbiology .pdf..virus pharmaceutical microbiology .pdf..
virus pharmaceutical microbiology .pdf..
Bhagyashree Gajbhare
 
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptxWhy Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
AmandaChou9
 
Hepcidin - Regulation and its role in Iron metabolism
Hepcidin - Regulation and its role in Iron metabolismHepcidin - Regulation and its role in Iron metabolism
Hepcidin - Regulation and its role in Iron metabolism
Reenaz Shaik
 
Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...
Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...
Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...
PVI, PeerView Institute for Medical Education
 
How to build a successful medical tourism practice
How to build a successful medical tourism practiceHow to build a successful medical tourism practice
How to build a successful medical tourism practice
Arlen Meyers, MD, MBA
 
SNAKE BITE presentation... Ppt..........
SNAKE BITE presentation... Ppt..........SNAKE BITE presentation... Ppt..........
SNAKE BITE presentation... Ppt..........
kinggameone94
 
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.GawadHemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.Gawad
NephroTube - Dr.Gawad
 
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Mohamad محمد Al-Gailani الكيلاني
 
Introduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptxIntroduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptx
Shamsuddin Mahmud
 
Hb electrophoresis- Types, Procedure and Analysis
Hb electrophoresis- Types, Procedure and AnalysisHb electrophoresis- Types, Procedure and Analysis
Hb electrophoresis- Types, Procedure and Analysis
Reenaz Shaik
 
Gene therapy - Types, Advantages and genetic manifestations in molecular med...
Gene therapy - Types, Advantages and  genetic manifestations in molecular med...Gene therapy - Types, Advantages and  genetic manifestations in molecular med...
Gene therapy - Types, Advantages and genetic manifestations in molecular med...
Reenaz Shaik
 
TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...
TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...
TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...
rightmanforbloodline
 
Best All Range PCD Pharma Franchise in India
Best All Range PCD Pharma Franchise in IndiaBest All Range PCD Pharma Franchise in India
Best All Range PCD Pharma Franchise in India
See Ever Healthcare
 
A comparative study on uroculturome antimicrobial susceptibility in apparentl...
A comparative study on uroculturome antimicrobial susceptibility in apparentl...A comparative study on uroculturome antimicrobial susceptibility in apparentl...
A comparative study on uroculturome antimicrobial susceptibility in apparentl...
Bhoj Raj Singh
 
Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)
MuhammedMNasser
 
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.GawadHemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
NephroTube - Dr.Gawad
 
Hepatocarcinoma today between guidelines and medical therapy. The role of sur...
Hepatocarcinoma today between guidelines and medical therapy. The role of sur...Hepatocarcinoma today between guidelines and medical therapy. The role of sur...
Hepatocarcinoma today between guidelines and medical therapy. The role of sur...
Gian Luca Grazi
 
chemistry of amino acids and proteins for I AHS.pdf
chemistry of amino acids and proteins for I AHS.pdfchemistry of amino acids and proteins for I AHS.pdf
chemistry of amino acids and proteins for I AHS.pdf
Santosh kumar Pasagadugula
 

Recently uploaded (20)

BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATIONBASIC PRINCIPLES OF CELL INJURY & ADAPTATION
BASIC PRINCIPLES OF CELL INJURY & ADAPTATION
 
Juvenile Myelomonocytic Leukemia (JMML)
Juvenile Myelomonocytic Leukemia   (JMML)Juvenile Myelomonocytic Leukemia   (JMML)
Juvenile Myelomonocytic Leukemia (JMML)
 
virus pharmaceutical microbiology .pdf..
virus pharmaceutical microbiology .pdf..virus pharmaceutical microbiology .pdf..
virus pharmaceutical microbiology .pdf..
 
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptxWhy Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
Why Does Seminal Vesiculitis Causes Jelly-like Sperm.pptx
 
Hepcidin - Regulation and its role in Iron metabolism
Hepcidin - Regulation and its role in Iron metabolismHepcidin - Regulation and its role in Iron metabolism
Hepcidin - Regulation and its role in Iron metabolism
 
Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...
Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...
Decoding Biomarker Testing and Targeted Therapy in NSCLC: The Complete Guide ...
 
How to build a successful medical tourism practice
How to build a successful medical tourism practiceHow to build a successful medical tourism practice
How to build a successful medical tourism practice
 
SNAKE BITE presentation... Ppt..........
SNAKE BITE presentation... Ppt..........SNAKE BITE presentation... Ppt..........
SNAKE BITE presentation... Ppt..........
 
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.GawadHemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 2 - Dr.Gawad
 
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
Giant Breast Lipoma Masquerading as Breast Enlargement ورم شحمي عملاق للثدي م...
 
Introduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptxIntroduction to Removable partial dneture.pptx
Introduction to Removable partial dneture.pptx
 
Hb electrophoresis- Types, Procedure and Analysis
Hb electrophoresis- Types, Procedure and AnalysisHb electrophoresis- Types, Procedure and Analysis
Hb electrophoresis- Types, Procedure and Analysis
 
Gene therapy - Types, Advantages and genetic manifestations in molecular med...
Gene therapy - Types, Advantages and  genetic manifestations in molecular med...Gene therapy - Types, Advantages and  genetic manifestations in molecular med...
Gene therapy - Types, Advantages and genetic manifestations in molecular med...
 
TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...
TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...
TEST BANK For Katzung's Basic and Clinical Pharmacology, 16th Edition By {Tod...
 
Best All Range PCD Pharma Franchise in India
Best All Range PCD Pharma Franchise in IndiaBest All Range PCD Pharma Franchise in India
Best All Range PCD Pharma Franchise in India
 
A comparative study on uroculturome antimicrobial susceptibility in apparentl...
A comparative study on uroculturome antimicrobial susceptibility in apparentl...A comparative study on uroculturome antimicrobial susceptibility in apparentl...
A comparative study on uroculturome antimicrobial susceptibility in apparentl...
 
Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)Handbook of Dental anatomy (practical part)
Handbook of Dental anatomy (practical part)
 
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.GawadHemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
Hemodialysis: Chapter 8, Complications During Hemodialysis, Part 3 - Dr.Gawad
 
Hepatocarcinoma today between guidelines and medical therapy. The role of sur...
Hepatocarcinoma today between guidelines and medical therapy. The role of sur...Hepatocarcinoma today between guidelines and medical therapy. The role of sur...
Hepatocarcinoma today between guidelines and medical therapy. The role of sur...
 
chemistry of amino acids and proteins for I AHS.pdf
chemistry of amino acids and proteins for I AHS.pdfchemistry of amino acids and proteins for I AHS.pdf
chemistry of amino acids and proteins for I AHS.pdf
 

Drugs pharmacology in kidney disease

  • 2. Drugs Pharmacology in Kidney Disease By M. H. Farjoo M.D., Ph.D., Bioanimator Shahid Beheshti University of Medical Sciences
  • 3. Drugs Pharmacology in Kidney Disease  Weak Acids & Weak Bases  The Order of Drug Metabolism  Protein Binding  Creatinine  Drug Dosing  Drug Absorption  Renal Excretion, Reabsorption and Metabolism  Drug Induced Renal Injuries  Prescribing in Renal Disease  Therapeutic Drug Monitoring  Drug Selection
  • 4. Weak Acids & Weak Bases  The protonated form of a weak acid is neutral.  The unprotonated form of a weak base is neutral.  The neutral (uncharged) form is more lipid-soluble.  Acids are more lipid-soluble at acid pH, and bases are more lipid-soluble at alkaline pH.  Weak acids and weak bases gain or lose protons depending on the pH.  So their movement between aqueous & lipid mediums varies with the pH.
  • 5. Weak Acids & Weak Bases  If kidney filters the drug, by changing the urine pH the drug can be "trapped" in the urine (in overdose).  Weak acids are excreted faster in alkaline urine and vise versa.  The ionized particles cannot pass easily through renal tubular membranes.  Therefore, less drug is reabsorbed into the blood and more is excreted by the kidneys.
  • 6. Weak Acids & Weak Bases  Sodium bicarbonate + phenobarbital → increased excretion of phenobarbital.  The sodium bicarbonate alkalinizes the urine, raising the number of barbiturate ions in the renal filtrate.  A large number of drugs are weak bases, and most of them are amine-containing molecules.  Primary, secondary, and tertiary amines undergo protonation and vary their solubility with pH.  Quaternary amines are always in the poorly lipid- soluble charged form.
  • 9. Weak Acids & Weak Bases Body Fluid Range of pH Fluid: Blood Ratios for Sulfadiazine (acid, pKa 6.5) Fluid: Blood Ratios for Pyrimethamine (base, pKa 7.0) Urine 5.0-8.0 0.12-4.65 0.79-72.24 Breast milk 6.4-7.6 0.2-1.77 0.89-3.56 Jejunum, ileum contents 7.5-8.0 1.23-3.54 0.79-0.94 Stomach contents 1.92-2.59 0.11 18,386-85,993 Prostatic secretions 6.45-7.4 0.21 1.0-3.25 Vaginal secretions 3.4-4.2 0.11 452-2848
  • 10. Acidification of Urine  Increases elimination of amphetamine, methylene dioxymethamfetamine (MDMA or ‘ecstasy’), dexfenfluramine, quinine and phencyclidine.  Oral NH4Cl, taken with food to avoid vomiting, acidifies the urine.  It should not be given to patients with impaired renal or hepatic function.  Other means include arginine hydrochloride, Ascorbic acid and calcium chloride by mouth.
  • 11. Alkalinisation of Urine  Increases the elimination of salicylate, phenobarbital, and some herbicides.  Treats crystal nephropathy by increasing drug solubility of methotrexate, sulphonamides and triamterene.  Reduces irritation of an inflamed urinary tract  Discourages the growth of E. coli.  The urine can be made alkaline by sodium bicarbonate i.v., or by potassium citrate by mouth.
  • 12. The Order of Drug Metabolism  The rate at which drugs are metabolized, is called “the order” of reaction.  Two orders exist:  First-order (exponential): a constant fraction of drug is transported or metabolised in unit time.  Zero-order (saturation kinetics): a constant amount of drug is transported or metabolised in unit time.  In doses used clinically, most drugs show first order processes.  Enzyme-mediated reactions often show zero-order (rate limitation) dynamics.
  • 13. Changes in plasma concentration following an intravenous bolus injection of a drug in the elimination phase. As elimination is a first-order process, the time for any concentration point to fall by 50% (t½) is always the same.
  • 15. The Order of Drug Metabolism  Zero-order kinetic has important consequences for drugs such as:  Alcohol  Phenytoin  Aspirin  In first order kinetic, because the rate of reaction is constant, it is dose independent,  By approaching zero order kinetic, increase in dose, alters the kinetics, and it become dose dependent.
  • 16.  Albumin is the main drug-binding plasma protein for acidic drugs.  Protein binding in uremic patients is decreased for all acidic drugs.  This is due to decreased albumin or reduced binding capacity.  Albumin decrease occurs in renal failure (albumin is lost in the urine). Protein Binding
  • 17.  The reduced binding is because of:  Reductions in albumin concentration.  Structural changes in the albumin molecule.  Uremic toxins compete with drugs for protein binding.  Reductions in albumin binding, increases volume of distribution (Vd)of drugs.  Edema fluid (an ECF) in renal impairment also increases Vd of water-soluble drugs.  So more unbound drug distributes into sites of elimination. Protein Binding
  • 19.  The elimination clearance of drugs is increased.  Drug half-life and therapeutic effects is decreased.  The higher levels of unbound drug can result in toxicity.  However, protein binding changes do not affect distribution volume or clearance of unbound drug. Protein Binding
  • 21.  The protein binding of basic drugs tends to be normal or even increased.  For basic drugs (clindamycin, propafenone), alpha1- acid glycoprotein (AAG) is the main binding protein.  The amount of AAG increases in renal renal failure.  In these patients larger amounts of a basic drug is bound and a smaller amount is free to exert an effect. Protein Binding
  • 22.  Alterations in tissue binding of drugs (digoxin) also occurs.  Renal failure decreases digoxin volume of distribution.  Digoxin is displaced from tissues by:  Metabolic products that cannot be excreted by impaired kidneys  A reduction in tissue levels of Na/K-ATPase, the tissue-binding site for digoxin. Protein Binding
  • 23. Protein Binding: Comparison of Free and Total Phenytoin Levels Protein Bound Phenytoin
  • 24.  Renal status should be monitored in any client with risk factors for renal insufficiency.  Signs and symptoms of renal failure include:  Decreased urine output (<600 mL/24 hours)  Increased BUN or increased creatinine (>2 mg/dL) Creatinine
  • 25. Creatinine  Renal impairment accounts for one-third of the prescribing errors.  Dose assessment solely based on serum creatinine (Cr) without estimation of Cr clearance is misleading.  Creatinine is determined by both muscle mass and GFR.  So its measurement cannot be used as the sole indicator of renal function.
  • 27.  Serum creatinine is a relatively unreliable indicator of renal function in elderly clients.  Because of diminished muscle mass, they may have a normal creatinine even if their GFR is markedly reduced.  Some drugs (cimetidine and trimethoprim) increase creatinine and create a false impression of renal failure.  They interfere with secretion of creatinine into kidney tubules. Creatinine
  • 28.  Estimations of creatinine clearance are more accurate for:  Clients with stable renal function (stable serum creatinine).  Average muscle mass (for their age, and BMI).  Estimations are less accurate for:  Emaciated and obese clients.  For those with changing renal function (as in acute illness). Creatinine
  • 29. Drug Dosing  The steady state of drug is only determined by drug dose and elimination clearance (CLE).  Since the metabolism of most drugs is by first-order, Drug metabolism is characterized by clearance.  Non-renal clearance is usually by the liver.  It also includes hemodialysis and other methods of drug removal.
  • 30. Drug Dosing  For determining the required dose in renal failure, the Dettli method is used.  The reliability of the Dettli method of predicting drug clearance depends on two critical assumptions: 1. The non-renal clearance of the drug remains constant when renal function is impaired. 2. ClE (total) declines in a linear fashion with ClCr.
  • 31. the relationship between total systemic drug clearance (CLTOT) and creatinine clearance (CLCR). CLR, renal clearance; CLNR, nonrenal clearance. The Dettli method of drug dosing
  • 32. A Case for Cimetidine  A functionally anephric patient postoperatively complained of GE reflux, so cimetidine was started.  Because of renal failure, the dose was halved.  Three days later, the patient was confused and diagnosis of dialysis dementia was made.  Later the suggestion was made that cimetidine be discontinued.  Two days later the patient was alert and was discharged from the hospital.
  • 33. A Case for Cimetidine  The Cimetidine label states that Patients with creatinine clearance <30 cc/min should receive half the recommended dose.  However, under “Pharmacokinetics” it indicates that 75% of the drug is excreted via kidneys.  Only one fourth of the dose is eliminated by non-renal mechanisms.  So patients who receive half the usual dose, still will receive twice the adequate dose!
  • 35. Drug Absorption  The bioavailability of most drugs does not change in impaired renal function.  Absorption of oral drugs may be decreased indirectly by:  Delayed gastric emptying  Vomiting and diarrhea  Edema of the GI tract (with generalized edema).  Changes in gastric pH
  • 36.  Changes in gastric pH in CRF occurs by:  Oral alkalinizing agents (bicarbonate, citrate).  Use of antacids for phosphate-binding effects.  This causes decreased absorption of oral acidic drugs and increases absorption of alkaline drugs.  Increased bioavailability of drugs with first-pass metabolism is possible (impairment of metabolizing enzymes). Drug Absorption
  • 37. Renal Excretion  Glomerular filtration affects all drugs of small molecular size and is restrictive.  Restrictive means that glomerular filtration is influenced by protein binding of drug.  Renal tubular secretion is non-restrictive, and both protein-bound and free drug are eliminated.  Competition by drugs for renal tubular secretion is an important cause of drug–drug interactions.
  • 38. Renal Excretion  Anionic drugs compete with other anionic drugs for active transport pathways, as do cationic drugs.  When two drugs secreted by the same pathway are given together, the renal clearance of each of them decreases.  For example, renal clearance of methotrexate is halved when salicylate is co-administered with it.
  • 39. Renal Excretion  ClCr reflects GFR and renal drug clearance correlates with the reciprocal of serum creatinine or ClCr.  ClCr is also a guide to the renal clearance of drugs with extensive renal tubular secretion or reabsorption.  This is the result of glomerulo-tubular balance (GTB) so drug Cl declines fairly linearly with reductions in ClCr.  GTB is the ability of the proximal tubule to reabsorb a constant fraction of glomerular filtrate delivered to it.
  • 40. Renal Excretion  Based on renal clearance values, two conclusions can be made:  If renal clearance exceeds drug filtration rate, there is net renal tubular secretion of the drug.  If renal clearance is less than drug filtration rate, there is net renal tubular reabsorption of the drug.
  • 41.  Excretion of many drugs is reduced in renal failure.  The kidneys normally excrete both the parent drug and metabolites produced by the liver.  Renal excretion mechanisms includes:  Glomerular filtration  Tubular secretion  Tubular reabsorption  All of these mechanism are affected by renal impairment. Renal Excretion
  • 42.  An adequate fluid intake is required to excrete drugs by the kidneys.  Any factor that depletes ECF increases the risk of worsening renal impairment which include:  Inadequate fluid intake  Diuretic drugs  Loss of body fluids (bleeding, vomiting, diarrhea) Renal Excretion
  • 43.  Lithium (Li) is entirely excreted by the kidneys and has a very narrow therapeutic range.  80% of Li is reabsorbed in the proximal tubules.  The amount of reabsorption depends on the concentration of sodium in the proximal tubules.  Deficiency of sodium or co-administration of diuretic: sodium loss↑, Li excretion↓ => Li toxicity↑  Excessive sodium intake lowers Li to non therapeutic ranges. Renal Reabsorption
  • 44. Renal Reabsorption  Proximal tubular endocytosis, is important in removing proteins that pass through glomerular pores.  Aminoglycosides are filtered at the glomerulus but are nephrotoxic because of active uptake by endocytosis.  This uptake is saturable, so aminoglycosides are less nephrotoxic in single daily doses than separate doses.
  • 45.  Metabolism may, or may not change by renal failure because:  In uremia, in liver, reduction and hydrolysis is slower, but oxidation by CYPs and conjugation are normal.  Impaired kidney may not eliminate metabolites of the parent drug with pharmacologic activity.  The kidney contains many of the same metabolizing enzymes found in the liver (eg: renal CYP enzymes). Renal Metabolism
  • 47. Non-renal Metabolism  Hepatic drug clearance (ClH) may be altered in chronic renal failure.  This may be due to depression in hepatic organic anion transport polypeptides, or increase in P-gp expression.  Impaired renal function impacts hepatic clearance in both Phase I, and Phase II metabolization .  This is particularly pronounced in end-stage renal disease (ESRD) and Phase I metabolization.
  • 48. Non-renal Metabolism  The effects of impaired renal function on hepatic clearance is due to:  Accumulation of parathyroid hormone (PTH)  Cytokines  Toxins  Alteration in gene transcription.
  • 51. Direct Drug-induced Renal Disease  Heavy metals: mercury, gold, iron, lead.  Antimicrobials: aminoglycosides, amphotericin, cephalosporins.  Iodinated contrast media: agents for the biliary tract  Solvents: carbon tetrachloride, ethylene glycol.  NSAID combinations.
  • 52.  NSAIDs can cause renal impairment even though they are eliminated mainly by hepatic metabolism.  Acetaminophen is nephrotoxic in overdose because of a metabolite that may cause kidney necrosis.  Aspirin is nephrotoxic in high doses, and its protein binding is reduced in renal failure. Direct Drug-induced Renal Disease, NSAIDs
  • 54.  NSAIDs can decrease blood flow in the kidneys by inhibiting synthesis of prostaglandins (PG) that dilate renal blood vessels.  When renal blood flow is normal, these PGs have limited activity.  When renal blood flow is decreased, their synthesis is increased to protect the kidneys from ischemia.  In those who depend on PGs to maintain renal blood flow, NSAIDs result in decreased GFR, and retention of salt and water. Direct Drug-induced Renal Disease, NSAIDs
  • 56.  NSAIDs can also cause kidney damage by a hypersensitivity reaction that leads to ARF.  NSAIDs may adversely affect a fetus’s kidneys when:  Given during late pregnancy to prevent premature labor.  Given after birth for PDA. Direct Drug-induced Renal Disease, NSAIDs
  • 57. Indirect Drug-induced Renal Disease  Cytotoxic drugs and uricosurics may cause urate to be precipitated in the tubule.  Calciferol may cause renal calcification by inducing hypercalcaemia.  Diuretic and laxative abuse can cause tubular damage secondary to potassium and sodium depletion.  Anticoagulants may cause haemorrhage into the kidney.
  • 58. Other Drug-induced Kidney Lesions  Vasculitis: allopurinol, isoniazid, sulphonamides.  Allergic interstitial nephritis: penicillins, thiazides, allopurinol, phenytoin, sulphonamides.  Drug-induced lupus erythematosus: hydralazine, procainamide, sulfasalazine.
  • 59. Drug Induced Renal Syndromes  Acute renal failure: aminoglycosides, cisplatin.  Chronic renal failure: NSAIDs.  Nephrotic syndrome: penicillamine, gold,  Functional impairment:  Reduced ability to dilute and concentrate urine: lithium  Potassium loss in urine: loop diuretics  Acid–base imbalance: acetazolamide
  • 60. Drug t½ (h) in normal and severely impaired renal function
  • 61. Prescribing in Renal Disease  Adjustment of the loading dose is generally unnecessary (Vd usually does not change).  Digoxin is an exception because the Vd is contracted in uremic patients due to altered tissue binding.  For maintenance dose either reduce each dose or lengthen the time between doses.
  • 62.  Dosage of many drugs needs to be decreased in renal failure including:  Aminoglycoside antibiotics  Most cephalosporin antibiotics  Fluoroquinolones  Digoxin Prescribing in Renal Disease
  • 63. Prescribing in Renal Disease  Special caution is needed in:  hypoproteinemia and the drug is extensively plasma protein bound  Advanced renal disease when accumulated metabolic products may compete for protein binding sites.  The early stages of dosing until response to the drug can be gauged.  Elderly, kidney blood flow, GFR, and tubular secretion of drugs is decreased (accumulation of drug).
  • 64. Therapeutic Drug Monitoring  Patients differ greatly in the dose of drug required to achieve the same response.  The dose of warfarin varies up to five-fold between individuals.  The question is: how optimal drug effect can be achieved quickly for the individual patient?  For drugs with short t½, both peak (15 min after an IV dose) and trough (just before the next dose) concentrations should be known.  For drugs with long t½, it is best to sample just before a dose is due.
  • 65. Therapeutic Drug Monitoring  Sometimes TDM does not worth the cost:  Plasma concentration may not be worth measuring (antihypertensive [blood pressure], hypoglycemics [blood glucose], diuretics [urine output], warfarin [INR]).  Plasma concentration has no correlation with effect (Aspirin as antiplatelet, anticancer drugs).  Plasma concentration may correlate poorly with effect (Diazepam with many active metabolites).
  • 66.  Drug selection is guided by renal function and the effects of drugs on renal function.  Many commonly used drugs may adversely affect renal function (NSAIDs or OTC drugs).  Some drugs are excreted exclusively by the kidneys (aminoglycosides, lithium).  Some drugs are contraindicated in renal impairment (tetracyclines except doxycycline). Drug Selection
  • 67.  Drugs can be used if guidelines are followed (reducing dosage, using TDM and renal function tests, avoiding dehydration).  Drugs known to be nephrotoxic should be avoided.  Sometime there are no substitutes and nephrotoxic drugs must be given.  Some commonly used nephrotoxic drugs include aminoglycosided, amphotericin B, and cisplatin. Drug Selection
  • 70. Sites of action of diuretic drugs. ENaC, epithelial sodium channel; NCCT, thiazide-sensitive Na–Cl co-transporter; NKCC2, Na–K–2Cl co-transporter; ROMK, rectifying outer medullary potassium channel.

Editor's Notes

  1. So, sodium bicarbonate is administered in ASA or phenobarbital overdose to increase the ionization of these drugs. This therapeutic intervention also reduces reabsorption by increasing urine flow.
  2. Trapping of a weak base (pyrimethamine) in the urine when the urine is more acidic than the blood. In the hypothetical case illustrated, the diffusible uncharged form of the drug has equilibrated across the membrane, but the total concentration (charged plus uncharged) in the urine is almost eight times higher than in the blood.
  3. Acidification of Urine Is used as a test for renal tubular acidosis Ref: Clinical pharmacology by Peter Bennett 2012
  4. Alkalinisation of urine: Increases the elimination of salicylate, phenobarbital, and chlorophenoxy herbicides, e.g. 2,4-D, MCPA Sodium overload may exacerbate cardiac failure, and sodium or potassium excess are dangerous when renal function is impaired. Ref: Clinical pharmacology by Peter Bennett 2012
  5. First-order (exponential) processes In the majority of instances, the rates at which absorption, distribution, metabolism and excretion of a drug occur are directly proportional to its concentration in the body. In other words, transfer of drug across a cell membrane or formation of a metabolite is high at high concentrations and falls in direct proportion to be low at low concentrations (an exponential relationship). Processes for which the rate of reaction is proportional to the concentration of participating molecules are first-order processes. In doses used clinically, most drugs are subject to first order processes of absorption, distribution, metabolism and elimination Zero-order processes (saturation kinetics) As the amount of drug in the body rises, metabolic reactions or processes that have limited capacity become saturated. In other words, the rate of the process reaches a maximum amount at which it stays constant, e.g. due to limited activity of an enzyme, and any further increase in rate is impossible despite an increase in the dose of drug. In these circumstances, the rate of reaction is no longer proportional to dose, and exhibits rate-limited or dosedependent 5 or zero-order or saturation kinetics. In practice, enzyme-mediated metabolic reactions are the most likely to show rate limitation because the amount of enzyme present is finite and can become saturated. Ref: Clinical Pharmacology 2012 by Peter Bennett
  6. Fig. 8.2 Changes in plasma concentration following an intravenous bolus injection of a drug in the elimination phase (the distribution phase, see text, is not shown). As elimination is a first-order process, the time for any concentration point to fall by 50% (t½) is always the same. Order of reaction and t½. When a drug is subject to first order kinetics, the t½ is a constant characteristic, i.e. a constant value can be quoted throughout the plasma concentration range (accepting that there will be variation in t½ between individuals), and this is convenient. But if the rate of a process is not directly proportional to plasma concentration, then the t½ cannot be constant. Consequently, no single value for t½ describes overall elimination when a drug exhibits zeroorder kinetics. In fact, t½ decreases as plasma concentration falls and the calculations on elimination and dosing that are so easy with first-order elimination (see below) become more complicated. Ref: Clinical Pharmacology 2012 by Peter Bennett
  7. When a drug is given at a constant rate (continuous or repeated administration), the time to reach steady state depends only on the t½ and, for all practical purposes, after 5 t½ periods the amount of drug in the body is constant and the plasma concentration is at a plateau Ref: Clinical Pharmacology 2012 by Peter Bennett
  8. Alcohol (ethanol) is a drug whose kinetics has considerable implications for society as well as for the individual, as follows: Alcohol is subject to first-order kinetics with a t½ of about 1 h at plasma concentrations below 10 mg/dL (attained after drinking about two-thirds of a unit (glass) of wine or beer). Above this concentration the main enzyme (alcohol dehydrogenase) that converts the alcohol into acetaldehyde approaches and then reaches saturation, at which point alcohol metabolism cannot proceed any faster than about 10 mL or 8 g/h for a 70-kg man. If the subject continues to drink, the blood alcohol concentration rises disproportionately, for the rate of metabolism remains the same, as alcohol shows zero-order kinetics. An illustration. Consider a man of average size who drinks about half (375 mL) a standard bottle of whisky (40% alcohol), i.e. 150 mL alcohol, over a short period, absorbs it and goes drunk to bed at midnight with a blood alcohol concentration of about 250 mg/dL. If alcohol metabolism were subject to first-order kinetics, with a t½ of 1 h throughout the whole range of social consumption, the subject would halve his blood alcohol concentration each hour (see Fig. 8.2). It is easy to calculate that, when he drives his car to work at 08.00 hours the next morning, he has a negligible blood alcohol concentration (less than 1 mg/dL) though, no doubt, a hangover might reduce his driving skill. But at these high concentrations, alcohol is in fact subject to zero-order kinetics and so, metabolising about 10 mL alcohol per hour, after 8 h the subject has eliminated only 80 mL, leaving 70 mL in his body and giving a blood concentration of about 120 mg/dL. At this level, his driving skill is seriously impaired. The subject has an accident on his way to work and is breathalysed despite his indignant protests that he last touched a drop before midnight. Banned from the road, on his train journey to work he will have leisure to reflect on the difference between first-order and zero-order kinetics (though this is unlikely!). In practice. The example above describes an imagined event but similar cases occur in everyday therapeutics. Phenytoin, at low dose, exhibits a first-order elimination process and there is a directly proportional increase in the steady-state plasma concentration with increase in dose. But gradually the enzymatic elimination process approaches and reaches saturation, the process becoming constant and zero order. While the dosing rate can be increased, the metabolism rate cannot, and the plasma Ref: Clinical Pharmacology 2012 by Peter Bennett
  9. ECF = extra cellular fluid Most acidic drugs bind to the bilirubin binding site on albumin. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson Albumin decrease occurs in: Nephrotic states (albumin is lost in the urine). Hypermetabolic states (stress, trauma, sepsis) in which protein breakdown exceeds protein synthesis. Liver disease (decreased synthesis of albumin).
  10. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  11. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  12. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  13. Comparison of free and total plasma phenytoin levels in a patient with normal renal function and a functionally anephric patient who are both treated with a 300-mg daily phenytoin dose and have identical CLint. Although free phenytoin levels are 0.8 mg/mL in both patients, phenytoin is only 84% bound (16% free) in the functionally anephric patient, compared to 92% bound (8% free) in the patient with normal renal function. For that reason total phenytoin levels in the functionally anephric patient are only 5 mg/mL, whereas they are 10 mg/mL in the patient with normal renal function. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson Phenytoin is an acidic, restrictively eliminated drug that can be used to illustrate some of the changes in drug distribution and elimination that occur in patients with impaired renal function. In patients with normal renal function, 92% of the phenytoin in plasma is protein bound. However, the percentage that is unbound or “free” rises from 8% in these individuals to 16% (or more) in hemodialysis-dependent patients. In a study comparing phenytoin pharmacokinetics in normal subjects and uremic patients, Odar-Cederl€of and Borga° [49] administered a single low dose of this drug so that first-order kinetics were approximated. The results shown in Table 5.3 can be inferred from their study. The uremic patients had an increase in distribution volume that was consistent with the observed decrease in phenytoin binding to plasma proteins. The three-fold increase in hepatic clearance that was observed in these patients also was primarily the result of decreased phenytoin protein binding. Although CLint for this CYP2C9, CYP2C19, and P–gp substrate also appeared to be increased in the uremic patients, the difference did not reach statistical significance at the P ¼ 0.05 level. A major problem arises in clinical practice when only total (protein-bound þ free) phenytoin concentrations are measured and used to guide therapy of patients with severely impaired renal function. The decreases in phenytoin binding that occur in these patients result in commensurate decreases in total plasma levels (Figure 5.3). Even though therapeutic and toxic pharmacologic effects are correlated with unbound rather than total phenytoin concentrations in plasma, the decrease in total concentrations can mislead physicians into increasing phenytoin doses inappropriately. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  14. PK = Pharmacokinetics ClE = elimination clearance (total clearance) ClCr = Creatinine clearance
  15. Moderate renal insufficiency (creatinine clearance 10 to 50 mL/min.). Severe renal insufficiency (creatinine clearance < 10 mL/min.).
  16. BMI =body mass index In oliguria (<500 mL/24 hrs), the creatinine clearance is less than 10 mL/min., regardless of the creatinine concentration.
  17. ClE = elimination clearance (total clearance) The characterization of drug metabolism by a clearance term usually is appropriate, since the metabolism of most drugs can be described by first-order kinetics within the range of therapeutic drug concentrations
  18. ClE = elimination clearance (total clearance) ClCr = Creatinine clearance Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  19. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  20. A 67-year-old man had been functionally anephric, requiring outpatient hemodialysis for several years. He was hospitalized for revision of his arteriovenous shunt and postoperatively complained of symptoms of gastroesophageal reflux. This complaint prompted institution of cimetidine therapy. In view of the patient’s impaired renal function, the usually prescribed dose was reduced by half. Three days later, the patient was noted to be confused. An initial diagnosis of dialysis dementia was made and the family was informed that dialysis would be discontinued. On teaching rounds, the suggestion was made that cimetidine be discontinued. Two days later the patient was alert and was discharged from the hospital to resume outpatient hemodialysis therapy. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  21. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  22. Nomogram for estimating cimetidine elimination clearance (CLE) for a 70-kg patient with impaired renal function. The right-hand ordinate indicates cimetidine CLE measured in young adults with normal renal function, and the left-hand ordinate indicates expected cimetidine CLE in a functionally anephric patient, based on the fact that 23% of an administered dose is eliminated by non-renal routes in healthy subjects. The heavy line connecting these points can be used to estimate cimetidine CLE from creatinine clearance (CLCR). For example, a 70-kg patient with CLCR of 50 mL/min (large dot) would be expected to have a cimetidineCLE of 517 mL/min, and to respond satisfactorily to doses that are 60% (517/850=0.6) of those recommended for patients with normal renal function. Reproduced with permission from Atkinson AJ Jr, Craig RM. Therapy of peptic ulcer disease. In: Molinoff PB, editor. Peptic ulcer disease. Mechanisms and management. Rutherford, NJ: Healthpress Publishing Group, Inc.; 1990. pp. 83–112 [9]. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  23. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  24. Drug filtration rate = GFR*fu*[drug] (fu = free fraction) The proteins which actively transport drugs include: P-glycoprotein (P-gp), six multiple drug resistance proteins, five cation and nine organic anion transporters, along with a number of genetic variants Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  25. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  26. Cl Cr = creatinine clearance Cl = clearance Ref: Principles Of Clinical Pharmacology By Arthur Atkinson Glomerulotubular balance (GTB) is defined as the ability of each successive segment of the proximal tubule to reabsorb a constant fraction of glomerular filtrate and solutes delivered to it. Ref: https://www.ncbi.nlm.nih.gov › pubmed
  27. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  28. Adequate renal function is a prerequisite for lithium therapy. In renal impairment, TDM for lithium must be done. Lithium and bromide are examples of drugs that are extensively reabsorbed by active transport mechanisms. Present evidence suggests that lithium is reabsorbed at the level of the proximal tubule by a Na /H exchanger (NHE-3) at the brush border and extruded into the blood by sodium–potassium ATPase and the sodium bicarbonate cotransporter located at the basolateral membrane. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  29. Net drug elimination is affected by drug reabsorption in the distal nephron, by passive diffusion. Proximal tubular endocytosis, mediated by the apical cell membrane receptors megalin and cipolin, plays an important role in removing proteins and peptides that pass through glomerular filtration pores [22]. This accounts for the absence of protein in normal urine and for the essential conservation of protein-carrier-bound vitamins and trace elements which are returned to the systemic circulation. However, the absorbed peptides and proteins are degraded by lysosomal proteases within the renal tubular cells. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  30. Most drugs are lipid soluble which aids their movement across cell membranes. The kidneys can excrete only water soluble substances. One function of metabolism is to convert fat soluble drugs into water-soluble metabolites. The kidney plays a major role in the clearance of insulin from the systemic circulation, removing approximately 50% of endogenous insulin and a greater proportion of insulin administered to diabetic patients. Insulin is filtered at the glomerulus, reabsorbed by the proximal tubule cell endocytotic mechanism described above, and then degraded by lysosomal proteolytic enzymes. Consequently, insulin requirements are markedly reduced in diabetic patients with impaired renal function. Imipenem and other peptides and peptidomimetics are also filtered at the glomerulus, absorbed by endocytosis, then metabolized by proximal renal tubule cell proteases [27]. Cilastatin, an inhibitor of proximal tubular dipeptidases, is co-administered with imipenem to enhance the clinical effectiveness of this antibiotic. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  31. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  32. Hepatic drug clearance (CLH) is mediated by transporter uptake and excretion mechanisms, as well as by metabolic processes within hepatocytes. In some cases hepatocyte uptake is the rate limiting step in drug elimination, and the expression and uptake function of hepatic organic anion transport polypeptides (OATPs) has been shown to be depressed in an animal model of chronic renal failure whereas P-gp expression appeared to be increased. Impaired renal function impacts the hepatic clearance of drugs that are metabolized by a number of enzymatic pathways, as indicated in Table 5.2. Both Phase I biotransformations (cytochrome P450 (CYP) and non-CYP enzymes) and Phase II biotransformations (e.g., acetylation by NAT-2, glucuronidation by UGT2B) may be impaired to varying degrees, which are particularly pronounced in patients with end-stage renal disease (ESRD). The effects of impaired renal function on hepatic drug elimination have been attributed to the accumulation of 3–carboxy-4-methyl-5-propyl-2-furan propanoic acid (CMPF), indoxyl sulfate, parathyroid hormone (PTH), cytokines, and perhaps other toxins that inhibit drug metabolism and transport. On the basis of experiments in rodent and in vitro models of chronic renal failure it has been shown that impairment occurs in some cases at the level of gene transcription, as indicated by decreased levels of the mRNA that encodes OATP2, a number of CYP enzyme, and NAT2 Ref: Principles Of Clinical Pharmacology By Arthur Atkinson An organic-anion-transporting polypeptide (OATP) is a membrane transport protein or 'transporter' that mediates the transport of mainly organic anions across the cell membrane. Therefore, OATPs are present in the lipid bilayer of the cell membrane, acting as the cell's gatekeepers. Ref: https://en.wikipedia.org/wiki/Organic-anion-transporting_polypeptide Important drug metabolizing phase I enzymes such as cytochrome P450 enzymes (CYP2C9, CYP2C19, CYP2D6 and CYP3A4) and phase II enzymes n-acetyltransferase 2 (NAT2), UDP-glucuronosyltransferase (UGT), thiopurine S-methyltransferase (TPMT) are located in the liver. These enzymes are encoded by specific genes and polymorphism of such genes may inhibit or increase enzyme activity. Besides genetic polymorphism, environmental factors and concomitant drug intake can modulate the activity of drug metabolizing enzymes. In this context, drug interactions can occur indirectly mediated through genes. This covert gene-drug interaction is an area of great clinical importance and needs to be investigated in detail. NAT2 is located in the liver and catalyzes the acetylation of isoniazid (INH), hydralazine, sulphadoxine, procainamide, dapsone and other clinically important drugs. It also catalyzes the acetylation of aromatic and heterocyclic carcinogens. It is implicated in the modification of risk factors in the development of malignancies involving the urinary bladder, colorectal region, breast, prostate, lungs and the head and neck region. It is also shown to be involved in the development of Alzheimer's disease, schizophrenia, diabetes, cataract and parkinsonism Ref: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4989826/ NAT2 is one of only 2 N-acetyltransferase genes in humans; the other, NAT1, shows little variation between individuals, whereas NAT2 is known to have over 23 variants. N-acetyltransferases are enzymes acting primarily in the liver to detoxify a large number of chemicals, including caffeine and several prescribed drugs. Ref: https://www.snpedia.com › index.php › NAT2
  33. CYP enzymes vary in their sensitivity to the adverse effects of impaired renal function, but that the extent of their impairment increases as renal function deteriorates. Relatively little information is available about the effects of impaired renal function on Phase II metabolic pathways. In an early study, Gibson et al. [37] found that NAT2-mediated procainamide acetylation in hemodialysis- dependent patients was reduced by 61% in phenotypic slow acetylators and by 69% in rapid acetylators. Subsequently, Kim et al. [38] reported that isoniazid acetylation by NAT2 was decreased by 63% in ESRD slow acetylators but by only 23% in rapid acetylators. Osborne et al. [39] have shown that Phase II metabolismofmorphine to formglucuronide conjugates is reduced by 48% in functionally anephric patients. More importantly, these patients accumulated much higher concentrations of the morphine-6-glucuronide metabolite that is a much more potent narcotic than morphine. Both of these factors account for the serious adverse events that have been reported in some patients with severely impaired renal function who have been treated with morphine Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  34. Effect of increasing degrees of renal impairment on hepatic clearance mediated by different CYP enzymes. Moderate impairment ¼ CLCR 30–59 mL/min, Severe impairment ¼ CLCR < 30 mL/min, ▬▬ ▬ ▬ ▬▬ CYP3A4, ▬▬ ▬▬ CYP2D6, ▬▬▬ CYP2C9, ▬ ▬ ▬ ▬ CYP1A2, ,,,, CYP2C19, ▬ ▬▬ ▬ ▬▬ CYP2C8. Figure based on data from Rowland Yeo K, Aarabi M, Jamei M, Rostami-Hodjegan A. Expert Rev Clin Pharmacol 2011;4:261–74 [36]. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  35. Ref: Clinical pharmacology by Peter Bennett 2012
  36. PDA = patent ductus arteriosus
  37. Probenecid is a uricosuric and renal tubular blocking agent.  Ref: Clinical pharmacology by Peter Bennett 2012
  38. Ref: Clinical pharmacology by Peter Bennett 2012
  39. Ref: Clinical pharmacology by Peter Bennett 2012
  40. Ref: Clinical pharmacology by Peter Bennett 2012
  41. Vd = volume of distribution Ref: Clinical pharmacology by Peter Bennett 2012
  42. The time to reach steady-state blood concentration is dependent only on drug t½, and a drug reaches 97% of its ultimate steady-state concentration in 5 t½. Thus, if t½ is prolonged by renal impairment, so also will be the time to reach steady state. Ref: Clinical pharmacology by Peter Bennett 2012
  43. drug effect relates to free (unbound) concentration at the tissue receptor site, which in turn reflects (but is not necessarily the same as) the concentration in the plasma. For many drugs, correlation between plasma concentration and effect is indeed better than that between dose and effect. Yet monitoring therapy by measuring drug in plasma is of practical use only in selected instances. The underlying reasons repay some thought. Ref: Clinical Pharmacology 2012 by Peter Bennett
  44. Ref: Clinical Pharmacology 2012 by Peter Bennett
  45. Potassium depletion The loop diuretics produce a smaller fall in serum potassium concentration than do the thiazides, for equivalent diuretic effect, but have a greater capacity for diuresis, i.e. higher efficacy especially in large dose, and so are associated with greater decline in potassium levels. If diuresis is brisk and continuous, clinically important potassium depletion is likely to occur. When a thiazide diuretic is used for hypertension, there is probably no case for routine prescription of a potassium supplement if no predisposing factors are Present Low dietary intake of potassium predisposes to hypokalaemia; the risk is particularly notable in the elderly, many of whom ingest less than 50 mmol per day (the dietary normal is 80 mmol).   Hypokalaemia may be aggravated by other drugs, e.g. b2-adrenoceptor agonists, theophylline, corticosteroids, amphotericin. Hyperkalaemia Ciclosporin, tacrolimus, indometacin and possibly other NSAIDs may cause hyperkalaemia with the potassium-sparing diuretics. Treatment of hyperkalaemia Depends on the severity and the following measures are appropriate: • Any potassium-sparing diuretic should be discontinued. A cation-exchange resin, e.g. polystyrene sulphonate resin (Resonium A, Calcium Resonium, see below) can be used orally (more effective than rectally), to remove body potassium by the gut. • Potassium may be moved rapidly from plasma into cells by giving: n sodium bicarbonate, 50 mL 8.4% solution through a central line, and repeated in a few minutes if characteristic ECG changes persist n glucose, 50 mL 50% solution, plus 10 units soluble insulin by i.v. infusion n nebulised b2-agonist, salbutamol 5–10 mg, is effective in stimulating the pumping of potassium into skeletal muscle. • In the presence of ECG changes, calcium gluconate, 10 mL of 10% solution, should be given i.v. and repeated if necessary in a few minutes; it has no effect on the serum potassium but opposes the myocardial effect of a raised serum potassium level. Calcium may potentiate digoxin and should be used cautiously, if at all, in a patient taking this drug. Sodium bicarbonate and calcium salt must not be mixed in a syringe or reservoir because calcium precipitates. • Dialysis may be needed in refractory cases and is highly effective. Hyponatraemia may result if sodium loss occurs in patients who drink a large quantity of water when taking a diuretic. Other mechanisms are probably involved, including enhancement of antidiuretic hormone release. Such patients have reduced total body sodium and extracellular fluid and are oedema free. Discontinuing the diuretic and restricting water intake are effective. The condition should be distinguished from hyponatraemia with oedema, which develops in some patients with congestive cardiac failure, cirrhosis or nephrotic syndrome. Here salt and water intake should be restricted because extracellular fluid volume is expanded. The combination of a potassium-sparing diuretic and ACE inhibitor can also cause severe hyponatraemia – more commonly than life-threatening hyperkalaemia. Urate retention with hyperuricaemia and, sometimes, clinical gout occurs with thiazides and loop diuretics. The effect is unimportant or negligible with the low-efficacy diuretics, e.g. amiloride and spironolactone. Two mechanisms appear to be responsible. First, diuretics cause volume depletion, reduction in glomerular filtration and increased absorption of almost all solutes in the proximal tubule, including urate. Second, diuretics and uric acid are organic acids and compete for the transport mechanism that pumps such substances from the blood into the tubular fluid. Diuretic-induced hyperuricaemia can be prevented by allopurinol or probenecid (which also antagonises diuretic efficacy by reducing their transport into the urine). Magnesium deficiency Loop and thiazide diuretics cause significant urinary loss of magnesium; potassium-sparing diuretics probably also cause magnesium retention. Magnesium deficiency brought about by diuretics is rarely severe enough to induce the classic picture of neuromuscular irritability and tetany but cardiac arrhythmias, mainly of ventricular origin, do occur and respond to repletion of magnesium (2 g or 8 mmol of Mg2þ is given as 4 mL 50% magnesium sulphate infused i.v. over 10–15 min followed by up to 72 mmol infused over the next 24 h). Calcium homeostasis Renal calcium loss is increased by the loop diuretics; in the short term this is not a serious disadvantage and indeed furosemide may be used in the management of hypercalcaemia after rehydration has been achieved. In the long term, hypocalcaemia may be harmful, especially in elderly patients, who tend in any case to be in negative calcium balance. Thiazides, by contrast, decrease renal excretion of calcium and this property may influence the choice of diuretic in a potentially calcium-deficient or osteoporotic individual, as thiazide use is associated with a reduced risk of hip fracture in the elderly. The hypocalciuric effect of the thiazides has also been used effectively in patients with idiopathic hypercalciuria, the commonest metabolic cause of renal stones. Interactions Loop diuretics (especially as intravenous boluses) potentiate ototoxicity of aminoglycosides and nephrotoxicity of some cephalosporins. NSAIDs tend to cause sodium retention, which counteracts the effect of diuretics; the mechanism may involve inhibition of renal prostaglandin formation. Diuretic treatment of a patient taking lithium can precipitate toxicity from this drug (the increased sodium loss is accompanied by reduced lithium excretion). Ref: Clinical pharmacology by Peter Bennett 2012
  46. Renal drug handling. Drugs may be filtered from the blood in the renal glomerulus, secreted into the proximal tubule, reabsorbed from the distal tubular fluid back into the systemic circulation, and collected in the urine. Membrane transporters (OAT, OCT, MDR1, and MRP2, among others) mediate secretion into the proximal tubule (see Figures 5–12 and 5–13 for details). Reabsorption of compounds from the distal tubular fluid (generally acidic) is pH sensitive: Ionizable drugs are subject to ion trapping, and altering urinary pH to favor ionization can enhance excretion of charged species
  47. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson
  48. Ref: Principles Of Clinical Pharmacology By Arthur Atkinson