Renal Clearance

Total Page:16

File Type:pdf, Size:1020Kb

Renal Clearance Faculty version with model answers Clearance Bruce M. Koeppen, M.D., Ph.D. University of Connecticut Health Center 1. Inulin is used in an experiment to measure the glomerular filtration rate (GFR). Inulin is continuously infused to achieve a steady-state concentration in the plasma of 1.0 mg/dL. Urine is collected over a 10 hour period. The total volume of urine is 1.5 L, and the urinary concentration of inulin is 440 mg/L. What is the GFR, as determined from the inulin clearance? Inulin Clearance = GFR = _____110____ ml/min Calculations: Urine flow rate = 1,500 ml/600 min = 2.5 ml/min Urine [inulin] = 440 mg/L = 0.44 mg/ml Plasma [inulin] = 1.0 mg/dL = 0.01 mg/ml Inulin clearance = GFR = (2.5 ml/min x 0.44 mg/ml)/0.01 mg/ml = 110 ml/min 2. In clinical situations the GFR of a patient is usually determined by measuring the clearance of creatinine. Like inulin, creatinine is freely filtered at the glomerulus. However, in contrast to inulin, a small amount is also secreted (approximately 10% of all creatinine excreted in the urine reflects this secretory component). Therefore creatinine is not an ideal marker for GFR. Despite this error, creatinine is used to measure GFR because it is produced endogenously by skeletal muscle, and does not require an infusion. Therefore it is easy to do a creatinine clearance, because the patient can do it at home by simply collecting their urine (usually over a 24 hr period), and having a single blood sample obtained to measure the plasma [creatinine]. What would be the estimated GFR of a patient with the following values for plasma and urine [creatinine] and urine volume. Plasma [creatinine] = 1.5 mg/dL Urine volume = 1.5 L/24 hours Urine [creatinine] = 1.44 gm/L Creatinine Clearance?~ GFR = _____100____ ml/min Calculation: Urine flow rate = 1,500 ml/1440 min = 1.04 ml/min Urine [creatinine] = 1.44 gm/L = 1.44 mg/ml Plasma [creatinine] = 1.5 mg/dL = 0.015 mg/ml Creatinine Clearance ≈ GFR = (1.04 ml/min x 1.44 mg/ml) / 0.015 mg/ml = 100 ml/min 3. One important consequence of renal disease is loss of nephrons. As nephrons are lost the GFR will decrease (remember that GFR is the sum of the filtration rates of all the nephrons in the kidneys). Therefore, as renal disease progresses the GFR declines, and the plasma [creatinine] increases. The relationship between GFR and plasma [creatinine] is illustrated below. ©Bruce M. Koeppen, M.D., Ph.D., University of Connecticut Health Center -1- 10 8 Plasm a 6 [ creat inine] (mg/ dL) 4 2 20 40 60 80 100 120 GFR (ml/ min) How do you explain the shape of this relationship, and what limitations does this present when trying to estimate GFR from a single plasma creatinine value? This curve reflects the fact that to maintain steady-state balance, excretion of creatinine by the kidneys must equal its production by skeletal muscle, and remain constant (this assumes production rate is constant). Because the excretion rate of creatinine essentially equals the creatinine filtered load (ignoring the secretory components of creatinine excretion), the following relationship applies: Ucr x V = GFR x Pcr Since Ucr x V is constant (ie., steady-state balance), the plasma concentration of creatinine (Pcr) must increase as GFR declines. Note that when GFR first begins to decline with renal disease, the change in plasma [creatinine] is quite small. In fact an individual can lose 1/3 to 1/2 of renal function and still have a plasma [creatinine] that is only slightly elevated above the normal reference value. Similarly, large changes in plasma [creatinine] occur with advanced renal disease even when only a small fraction of additional nephrons are lost. Thus predicting a precise GFR value from a single plasma [creatinine] determination is relatively inaccurate. However, monitoring the change in plasma [creatinine] over time is a useful way to monitor progression (or resolution) of renal disease. 4. A 25 year old man weighing 60 kg has a plasma [creatinine] of 1.4 mg/dL (nl = 0.8 - 1.4 mg/dL). A 24 hour urine collection is done to determine his creatinine clearance, and thereby estimate his GFR. The following data are obtained: Urine [creatinine] = 833 mg/L Urine volume = 1,080 ml/24 hour What is his creatinine clearance? Creatinine clearance = _____44.6____ ml/min Calculation: Urine flow rate = 1,080 ml/1440 min = 0.75 ml/min Urine [creatinine] = 833 mg/L = 0.833 mg/ml Plasma [creatinine] = 1.4 mg/dL = 0.014 mg/ml ©Bruce M. Koeppen, M.D., Ph.D., University of Connecticut Health Center -2- Creatinine Clearance ~ GFR = (0.75 ml/min x 0.833 mg/ml) / 0.014 mg/ml = 44.6 ml/min If the normal range of GFR for adult males is 90 - 140 ml/min, what would you conclude about the renal function of this man? The value of 44.6 ml/min for a creatinine clearance would seem to indicate a significant reduction in renal function (perhaps 50%). While his plasma [creatinine] is within the normal range (upper limit of normal), it is difficult to interpret because there is no baseline value. For example, it may have been 0.8 mg/dL previous to this current determination. This emphasizes the point that early on in renal disease small increases in plasma [creatinine] can occur with large initial declines in renal function. What error could lead to the calculation of a creatinine clearance that does not reflect the actual GFR? How could you determine if such an error occurred in this man, without having to repeat the 24 hour urine collection? (Hint: Muscle creatinine production is relatively constant and proportional to muscle mass. In adult males, the creatinine production rate is 20 -25 mg/kg body weight/day, and in adult females it is 15 - 20 mg/kg body weight/day). A falsely low creatinine clearance will be calculated if the urine collection was incomplete (ie., collected for less than a 24 hr period). Because creatinine production is relatively constant and proportional to muscle mass a simple calculation of the total amount of creatinine excreted in the urine will allow you to determine if the collection was complete. For example, this man weighs 60 kg, and his creatinine production should be in the range of 1,200 - 1,500 mg/24 hr. This should equal his creatinine excretion rate. However, his excretion rate is only about 900 mg/24 hr (1,080 ml x 0.833 mg/ml). Therefore, it appears that the low value for creatinine clearance may not a reflection of decreased renal function, but the result of an incomplete urine collection. Note: there is some reason to believe that perhaps this man does have some degree of decreased renal function. By taking the expected creatinine excretion rate of 1,200 - 1,500 mg/24 hr, a predicted creatinine clearance of 60 - 75 ml/min can be calculated. Unfortunately, because it is not know exactly how much urine was not collected, an accurate measure can only be obtained after the man does an appropriate 24 hour urine collection. 5. The renal handling of Na+ and K+ and urea are examined in two individuals. Both individuals eat the same diet, and are in steady-state balance. However, they have different glomerular filtration rates. Subject A has a GFR of 170 L/day, while the GFR of subject B is 50 L/day. The following data are obtained. Na+ Intake K+ Intake Urea Production GFR Urine Flow Subject mEq/day mEq/day gm/day L/day L/day A 100 50 8.5 170 1 B 100 50 8.5 50 1 Serum [Na+] Serum [K+] Serum BUN* GFR Subject mEq/L mEq/L mg/dL L/day A 145 3.5 10 170 ©Bruce M. Koeppen, M.D., Ph.D., University of Connecticut Health Center -3- B 145 3.5 34 50 *BUN = blood urea nitrogen Assume that the kidneys represent the only excretion route for Na+, K+ and urea. Calculate the following: Subject A Subject B Clearance Na+ (L/day) ____0.69___ ____0.69___ K+ (L/day) ____14.3___ ____14.3___ Urea (L/day) _____85____ _____25____ Filtered Load Na+ (mEq/day) ___24,650__ ____7,250___ K+ (mEq/day) _____595___ _____175___ Urea (mg/day) ___17,000__ __17,000___ Fractional Excretion Na+ (%) _____0.4____ _____1.4____ K+ (%) _____8.4____ ____28.6____ Urea (%) _____50____ _____50____ Tubular Reabsorption Na+ (mEq/day) __24,550 _ ___7,150___ K+ (mEq/day) ___545 __ _____125___ Urea (mg/day) ___8,500 __ ____8,500___ The following formulae are used: Clearance = excretion rate / plasma concentration Filtered Load = GFR x serum concentration Fractional Excretion = excretion rate / filtered load x 100 Tubular reabsorption = filtered load - excretion rate Calculations subject A: Clearance Na+: 100 mEq/d / 145 mEq/L = 0.69 L/day K+ : 50 mEq/d / 3.5 mEq/L = 14.3 L/day ©Bruce M. Koeppen, M.D., Ph.D., University of Connecticut Health Center -4- Urea: 8,500 mg/d / 100 mg/L = 85 L/day Filtered Load Na+: 170 L/d x 145 mEq/L = 24,650 mEq/day K+: 170 L/d x 3.5 mEq/L = 595 mEq/day Urea: 170 L/d x 100 mg/L = 17,000 mg/day Fractional Excretion Na+: 100 mEq/d / 24,650 mEq/d x 100 = 0.4% K+: 50 mEq/d / 595 mEq/d x 100 = 8.4% Urea: 8,500 mg/d / 17,000 mg/day x 100 = 50% Tubular Reabsorption Na+: 24,650 mEq/d - 100 mEq/d = 24,550 mEq/day K+: 595 mEq/d - 50 mEq/d = 545 mEq/day Urea: 17,000 mg/d - 8,500 mg/d = 8,500 mg/day Calculations subject B: Clearance Na+: 100 mEq/d / 145 mEq/L = 0.69 L/day K+ : 50 mEq/d / 3.5 mEq/L = 14.3 L/day Urea: 8,500 mg/d / 340 mg/L = 25 L/day Filtered Load Na+: 50 L/d x 145 mEq/L = 7,250 mEq/day K+: 50 L/d x 3.5 mEq/L = 175 mEq/day Urea: 500 L/d x 340 mg/L = 17,000 mg/day Fractional Excretion Na+: 100 mEq/d / 7,250 mEq/d x 100 = 1.4% K+: 50 mEq/d / 175 mEq/d x 100 = 28.6% Urea: 8,500 mg/d / 17,000 mg/day x 100 = 50% Tubular Reabsorption Na+: 7,250 mEq/d - 100 mEq/d = 7,150 mEq/day K+: 175 mEq/d - 50 mEq/d = 125 mEq/day Urea: 17,000 mg/d - 8,500 mg/d = 8,500 mg/day Based on the above data, explain each of the following: Note: questions A-D are all related, the point of each is to understand how Na+, K+ and urea are handled by the kidney, and how this can be discerned from the above calculations.
Recommended publications
  • Clinical Pharmacology 1: Phase 1 Studies and Early Drug Development
    Clinical Pharmacology 1: Phase 1 Studies and Early Drug Development Gerlie Gieser, Ph.D. Office of Clinical Pharmacology, Div. IV Objectives • Outline the Phase 1 studies conducted to characterize the Clinical Pharmacology of a drug; describe important design elements of and the information gained from these studies. • List the Clinical Pharmacology characteristics of an Ideal Drug • Describe how the Clinical Pharmacology information from Phase 1 can help design Phase 2/3 trials • Discuss the timing of Clinical Pharmacology studies during drug development, and provide examples of how the information generated could impact the overall clinical development plan and product labeling. Phase 1 of Drug Development CLINICAL DEVELOPMENT RESEARCH PRE POST AND CLINICAL APPROVAL 1 DISCOVERY DEVELOPMENT 2 3 PHASE e e e s s s a a a h h h P P P Clinical Pharmacology Studies Initial IND (first in human) NDA/BLA SUBMISSION Phase 1 – studies designed mainly to investigate the safety/tolerability (if possible, identify MTD), pharmacokinetics and pharmacodynamics of an investigational drug in humans Clinical Pharmacology • Study of the Pharmacokinetics (PK) and Pharmacodynamics (PD) of the drug in humans – PK: what the body does to the drug (Absorption, Distribution, Metabolism, Excretion) – PD: what the drug does to the body • PK and PD profiles of the drug are influenced by physicochemical properties of the drug, product/formulation, administration route, patient’s intrinsic and extrinsic factors (e.g., organ dysfunction, diseases, concomitant medications,
    [Show full text]
  • Microrna Pharmacoepigenetics: Posttranscriptional Regulation Mechanisms Behind Variable Drug Disposition and Strategy to Develop More Effective Therapy
    1521-009X/44/3/308–319$25.00 http://dx.doi.org/10.1124/dmd.115.067470 DRUG METABOLISM AND DISPOSITION Drug Metab Dispos 44:308–319, March 2016 Copyright ª 2016 by The American Society for Pharmacology and Experimental Therapeutics Minireview MicroRNA Pharmacoepigenetics: Posttranscriptional Regulation Mechanisms behind Variable Drug Disposition and Strategy to Develop More Effective Therapy Ai-Ming Yu, Ye Tian, Mei-Juan Tu, Pui Yan Ho, and Joseph L. Jilek Department of Biochemistry & Molecular Medicine, University of California Davis School of Medicine, Sacramento, California Received September 30, 2015; accepted November 12, 2015 Downloaded from ABSTRACT Knowledge of drug absorption, distribution, metabolism, and excre- we review the advances in miRNA pharmacoepigenetics including tion (ADME) or pharmacokinetics properties is essential for drug the mechanistic actions of miRNAs in the modulation of Phase I and development and safe use of medicine. Varied or altered ADME may II drug-metabolizing enzymes, efflux and uptake transporters, and lead to a loss of efficacy or adverse drug effects. Understanding the xenobiotic receptors or transcription factors after briefly introducing causes of variations in drug disposition and response has proven the characteristics of miRNA-mediated posttranscriptional gene dmd.aspetjournals.org critical for the practice of personalized or precision medicine. The regulation. Consequently, miRNAs may have significant influence rise of noncoding microRNA (miRNA) pharmacoepigenetics and on drug disposition and response. Therefore, research on miRNA pharmacoepigenomics has come with accumulating evidence sup- pharmacoepigenetics shall not only improve mechanistic under- porting the role of miRNAs in the modulation of ADME gene standing of variations in pharmacotherapy but also provide novel expression and then drug disposition and response.
    [Show full text]
  • Toxicological Profile for Radon
    RADON 205 10. GLOSSARY Some terms in this glossary are generic and may not be used in this profile. Absorbed Dose, Chemical—The amount of a substance that is either absorbed into the body or placed in contact with the skin. For oral or inhalation routes, this is normally the product of the intake quantity and the uptake fraction divided by the body weight and, if appropriate, the time, expressed as mg/kg for a single intake or mg/kg/day for multiple intakes. For dermal exposure, this is the amount of material applied to the skin, and is normally divided by the body mass and expressed as mg/kg. Absorbed Dose, Radiation—The mean energy imparted to the irradiated medium, per unit mass, by ionizing radiation. Units: rad (rad), gray (Gy). Absorbed Fraction—A term used in internal dosimetry. It is that fraction of the photon energy (emitted within a specified volume of material) which is absorbed by the volume. The absorbed fraction depends on the source distribution, the photon energy, and the size, shape and composition of the volume. Absorption—The process by which a chemical penetrates the exchange boundaries of an organism after contact, or the process by which radiation imparts some or all of its energy to any material through which it passes. Self-Absorption—Absorption of radiation (emitted by radioactive atoms) by the material in which the atoms are located; in particular, the absorption of radiation within a sample being assayed. Absorption Coefficient—Fractional absorption of the energy of an unscattered beam of x- or gamma- radiation per unit thickness (linear absorption coefficient), per unit mass (mass absorption coefficient), or per atom (atomic absorption coefficient) of absorber, due to transfer of energy to the absorber.
    [Show full text]
  • Renal Effects of Atrial Natriuretic Peptide Infusion in Young and Adult ~Ats'
    003 1-3998/88/2403-0333$02.00/0 PEDIATRIC RESEARCH Vol. 24, No. 3, 1988 Copyright O 1988 International Pediatric Research Foundation, Inc. Printed in U.S.A. Renal Effects of Atrial Natriuretic Peptide Infusion in Young and Adult ~ats' ROBERT L. CHEVALIER, R. ARIEL GOMEZ, ROBERT M. CAREY, MICHAEL J. PEACH, AND JOEL M. LINDEN WITH THE TECHNICAL ASSISTANCE OF CATHERINE E. JONES, NANCY V. RAGSDALE, AND BARBARA THORNHILL Departments of Pediatrics [R.L.C., A.R.G., C.E.J., B. T.], Internal Medicine [R.M.C., J.M. L., N. V.R.], Pharmacology [M.J.P.], and Physiology [J.M.L.], University of Virginia, School of Medicine, Charlottesville, Virginia 22908 ABSTRAm. The immature kidney appears to be less GFR, glomerular filtration rate responsive to atrial natriuretic peptide (ANP) than the MAP, mean arterial pressure mature kidney. It has been proposed that this difference UeC~pV,urinary cGMP excretion accounts for the limited ability of the young animal to UN,V, urinary sodium excretion excrete a sodium load. To delineate the effects of age on the renal response to exogenous ANP, Sprague-Dawley rats were anesthetized for study at 31-32 days of age, 35- 41 days of age, and adulthod. Synthetic rat A* was infused intravenously for 20 min at increasing doses rang- By comparison to the adult kidney, the immature kidney ing from 0.1 to 0.8 pg/kg/min, and mean arterial pressure, responds to volume expansion with a more limited diuresis and glomerular filtration rate, plasma ANP concentration, natriuresis (I). A number of factors have been implicated to urine flow rate, and urine sodium excretion were measured explain this phenomenon in the neonatal kidney, including a at each dose.
    [Show full text]
  • Urate-2- C Transport in the Rat Nephron
    Urate-2-14C transport in the rat nephron Ronald A. Kramp, … , William E. Lassiter, Carl W. Gottschalk J Clin Invest. 1971;50(1):35-48. https://doi.org/10.1172/JCI106482. Research Article Intrarenal transport of urate-2-14C was studied in anesthetized rats using the microinjection technic. During saline diuresis, small volumes of urate-2-14C (0.24-0.48 mM) and inulin-3H were injected into surface proximal and distal convoluted tubules, and ureteral urine was collected serially. Total (74-96%) and direct (57-84%) urate recovery increased significantly the more distal the puncture site. Delayed recovery (±20%) remained approximately the same regardless of localization of the microinjection. After proximal injections, total and direct recoveries of urate-2-14C were significantly higher in rats treated with probenecid, pyrazinoate, or PAH than during saline diuresis alone, while the excretion rates were comparable after distal injection. Delayed recovery was not altered by drug administration. The decreased proximal reabsorption of urate is presumably due to an effect of the drugs on the luminal membrane of the nephron. For perfusion at high urate concentrations, nonradioactive urate was added to the injectate (0.89-1.78 mM). Urate-2-14C recovery was almost complete and there was no delayed excretion, demonstrating saturation kinetics. These findings are compatible with a carrier-mediated mechanism for urate transport probably located at the luminal border of the proximal tubular epithelium. No definitive evidence for urate secretion was found in these studies. Find the latest version: https://jci.me/106482/pdf Urate-2-14C Transport in the Rat Nephron RONALD A.
    [Show full text]
  • Clinical Pharmacology Elimination of Drugs
    BRITISH MEDICAL JOURNAL VOLUME 282 7 MARCH 1981 809 Br Med J (Clin Res Ed): first published as 10.1136/bmj.282.6266.809 on 7 March 1981. Downloaded from Today's Treatment Clinical pharmacology Elimination of drugs H E BARBER, J C PETRIE This article outlines the principles ofdrug elimination, principally influenced by the lipid solubility of the drug. The more lipid by the liver and kidneys. Most elimination processes of drugs soluble the drug is, the more distribution into lipid compart- follow first-order elimination kinetics. In such processes a ments takes place and hence the larger is the Vd of the drug, defined and constant fraction of a drug-for example, half-is thus affecting its elimination. irreversibly eliminated in a constant time. The half-life time (t1) Drug clearance is also influenced by the rate of delivery of the in such first-order processes is the time taken for the concen- drug to the organs of elimination, principally the liver and tration of the drug to fall to half its previous value, regardless kidney, and by the capacity of the organs to eliminate the drug. of the initial concentration of the drug. Some drugs-for In clinical practice most drugs are swallowed and with some, example, ethyl alcohol-follow zero-order elimination kinetics. for example, propranolol, the bioavailability is low because only In these processes elimination of the drug occurs at a constant a fraction of the oral dose administered reaches the systemic rate, because the enzymes responsible for metabolism of drugs, circulation due to extensive presystemic or first-pass elimination and the active excretion processes particularly in the kidney, during absorption from the gut and passage through the liver.
    [Show full text]
  • The Natriuretic Peptide Clearance Receptor Locally Modulates the Physiological Effects of the Natriuretic Peptide System
    Proc. Natl. Acad. Sci. USA Vol. 96, pp. 7403–7408, June 1999 Genetics The natriuretic peptide clearance receptor locally modulates the physiological effects of the natriuretic peptide system (gene targetingygene ‘‘knock out’’yguanylyl cyclase activityyurine osmolalityybone metabolism) NAOMICHI MATSUKAWA*†,WOJCIECH J. GRZESIK‡,NOBUYUKI TAKAHASHI*, KAILASH N. PANDEY§,STEPHEN PANG¶, i MITSUO YAMAUCHI‡, AND OLIVER SMITHIES* *Department of Pathology and Laboratory Medicine and ‡Dental Research Center, University of North Carolina, Chapel Hill, NC 27599-7525; §Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112; and ¶Department Anatomy and Cell Biology, Queen’s University, Kingston, ON, Canada K7L 3N6 Contributed by Oliver Smithies, April 26, 1999 ABSTRACT Natriuretic peptides (NPs), mainly produced NPRA responds to ANP and, to a 10-fold lesser degree, to in heart [atrial (ANP) and B-type (BNP)], brain (CNP), and BNP; NPRB responds primarily to CNP. NPRA is strongly kidney (urodilatin), decrease blood pressure and increase salt expressed in the vasculature, kidneys, and adrenal glands, and excretion. These functions are mediated by natriuretic peptide its stimulation mediates vasorelaxant and natriuretic functions receptors A and B (NPRA and NPRB) having cytoplasmic and decreases aldosterone synthesis. NPRB is strongly ex- guanylyl cyclase domains that are stimulated when the recep- pressed in the brain, including the pituitary gland, and may tors bind ligand. A more abundantly expressed receptor have a role in neuroendocrine regulation. A third natriuretic (NPRC or C-type) has a short cytoplasmic domain without peptide receptor (NPRC) has only a short cytoplasmic domain guanylyl cyclase activity. NPRC is thought to act as a clearance with no GC activity; it is generally thought to act as a clearance receptor, although it may have additional functions.
    [Show full text]
  • RECEPTOR ANTAGONISTS Pegvisomant
    11 RECEPTOR ANTAGONISTS Pegvisomant: structure and function S Pradhananga, I Wilkinson and R J M Ross Division of Clinical Sciences, University of Sheffield, Northern General Hospital, Sheffield S5 7AU, UK (Requests for offprints should be addressed to R Ross, Clinical Sciences, Northern General Hospital, Sheffield S5 7AU, UK; Email: [email protected]) Abstract Pegvisomant is the pegylated form of mutant growth hormone (B2036). B2036 has increased affinity in one binding site and lowered affinity in its second binding site, it has been shown that this molecule still enables dimerisation of the growth hormone receptor at the cell surface but does not allow the necessary conformational changes for signalling. Pegylation decreases the antagonistic activity of B2036, however the rate of clearance of the pegylated B2036 is greatly reduced compared to the unpegylated form. Even though the antagonistic activity of pegvisomant is lower than B2036, the reduced rate of clearance makes it an effective clinical drug for the treatment of conditions such as acromegaly. Journal of Molecular Endocrinology (2002) 29, 11–14 Introduction gated to this core B2036 molecule. Pegylation has little effect on the affinity of pegvisomant for GHBP The growth hormone (GH) antagonist, pegviso- but reduces affinity for the cell surface receptor. mant, is the first specific growth hormone receptor Therefore, there is a high dose requirement for antagonist to be developed for the treatment of pegvisomant in clinical practice; however pegy- acromegaly (Trainer et al. 2000, van der Lely et al. lation has a benefit in that it increases the half life 2001). In this review, the relationship between of clearance.
    [Show full text]
  • Absorption & Half-Life
    Slide 1 Absorption and Half-Life Nick Holford Dept Pharmacology & Clinical Pharmacology University of Auckland, New Zealand Slide 2 Objectives ➢ Understand the physiological determinants of extent and rate of absorption ➢ Be able to describe bolus, first-order and zero-order input processes ➢ Learn the definition of half-life ➢ Be able to describe the time course of drug accumulation during constant rate input and elimination after input stops ➢ Appreciate the applications of absorption and half-life concepts to clinical practice ©NHG Holford, 2021, all rights reserved. Slide Drug absorption can be described 3 by two quite distinct factors: • The extent of absorption Absorption reflects the total amount of drug entering the body. It is not time dependent. • The rate of absorption determines how quickly the ➢ Extent of Absorption drug enters the body. The rate typically changes with time. ➢ Rate of Absorption ©NHG Holford, 2021, all rights reserved. Slide The extent of oral absorption can 4 be considered in 2 parts. The first part is the fraction of drug Extent (F) absorbed across the gut wall (f). This describes how much drug gets from the gut into the portal venous system. It is determined in ➢ Fraction Absorbed (f) part by physicochemical properties. Small, unionized » into portal vein from gut molecules e.g. theophylline, are almost completely absorbed » physicochemistry across the gut wall. Large, ionized – theophylline (100%) (small,unionized) molecules like gentamicin cross – gentamicin (< 5%) (large, ionized) membranes with difficulty and only a small fraction is absorbed » metabolism/transport across the gut wall. Many drugs – simvastatin (50%?) (CYP3A4) can cross the luminal cell membrane but are then – digoxin (65%) (PGP transporter) metabolized in the gut wall (typically by CYP3A4 e.g.
    [Show full text]
  • The Half-Lives: Physical, Biological, and Effective
    The Half-Lives: Physical, Biological, and Effective Introduction: There are three half-lives that are important when considering the use of radioactive drugs for both diagnostic and therapeutic purposes. While both the physical and biological half-lives are important since they relate directly to the disappearance of radioactivity from the body by two separate pathways (radioactive decay, biological clearance), there is no half-life as important in humans as the effective half-life. As we will see shortly, this half-life takes into account not only elimination from the body but also radioactive decay. If there is ever a question about residual activity in the body, the calculation uses the effective half-life; in radiation dosimetry calculations, the only half- life that is included in the equation is the effective half-life. Let’s take a look individually at the three half-lives. Half Lives: Physical Physical half-life is defined as the period of time required to reduce the radioactivity level of a source to exactly one half its original value due solely to radioactive decay. The physical half-life is designated tphys or more commonly t1/2 . By default, the term t1/2 refers to the physical half-life and tphys is used when either or both of the other two half- lives are included in the discussion. There are a few things to note about the tphys : • The tphys can be measured directly by counting a sample at 2 different points in time and then calculating what the half-life is. • For example, if activity decreases from 100% to 25% in 24 hours, then the half- life is 12 hours since a decrease from 100% to 50% to 25% implies that 2 half- lives have elapsed.
    [Show full text]
  • Atrial Natriuretic Peptide and Blood Volume During Red Cell Transfusion
    Archives ofDisease in Childhood 1991; 66: 395-397 395 Atrial natriuretic peptide and blood volume during Arch Dis Child: first published as 10.1136/adc.66.4_Spec_No.395 on 1 April 1991. Downloaded from red cell transfusion in preterm infants W Rascher, N Lingens, M Bald, 0 Linderkamp Abstract 1710 g (range 650-2660). All infants were in Because raised plasma concentrations of good clinical condition. Four were mechanically atrial natriuretic peptide indicate volume ventilated through an intratracheal tube. None expansion, we studied the effect of red cell had renal disease, shock, sepsis or were being transfusion on plasma atrial natriuretic pep- treated with diuretics. The volume of trans- tide concentration, packed cell volume, and fused red cells was calculated to raise the packed intravascular volume in eight preterm infants. cell volume to 0-45 (haemoglobin to 155 g/l). Red cell transfusion increased red cell mass, The transfusion rate averaged 0-05 (0 01) packed cell volume and erythrocyte count, ml/min/kg. but decreased plasma volume. Total blood Before and one hour after red cell transfusion volume, plasma atrial natriuretic peptide con- blood was drawn in EDTA coated tubes (1 5-2 centration, urine flow rate, and urinary ml) for determination of plasma atrial natriur- sodium excretion did not change. etic peptide and haemoglobin concentrations We conclude that a slow transfusion of packed cell volume, haemoglobin F concentra- less than 10 ml red cells/kg body weight does tion, and plasma osmolality. In addition 0 5 ml not cause volume expansion with subsequent blood was drawn in plastic tubes for measure- atrial natriuretic peptide release thereby ment of serum sodium, potassium and creati- affecting the cardiovascular system.
    [Show full text]
  • The Impact of Micrornas and Alternative Splicing in Pharmacogenomics
    The Pharmacogenomics Journal (2009) 9, 1–13 & 2009 Nature Publishing Group All rights reserved 1470-269X/09 $32.00 www.nature.com/tpj PERSPECTIVE cogenetic history see2). Since then, the The impact of microRNAs and number of cases described in the literature has increased exponentially alternative splicing in and they were mainly focused on single-nucleotide polymorphism pharmacogenomics (SNP), insertions and deletions (indels) of nucleotides, copy number variation 1 2 3 (CNV) and missense mutations (for F Passetti , CG Ferreira and FF Costa review see Roses3 and Giacomini et al.4). 1 Laboratory of Bioinformatics and Computational Biology, Division of Clinical and The most studied gene family so far Translational Research, Research Coordination (CPQ), Instituto Nacional de Caˆncer, Rio is the cytochrome P450 enzyme5–7 (for de Janeiro, Brazil; 2Division of Clinical and Translational Research, Research Coordination more details see Table 2). The genetic (CPQ), Instituto Nacional de Caˆncer, Rio de Janeiro, Brazil and 3Cancer Biology and variability observed in this gene family Epigenomics Program, Children’s Memorial Research Center, Northwestern University’s has been associated with differences in Feinberg School of Medicine, Chicago, IL, USA drug metabolism in several patholo- gies, such as cancer, cardiovascular diseases and rheumatoid arthritis.5–8 The Pharmacogenomics Journal (2009) 9, more than half of the human genes, There is one SNP in CYP2C9 that 1–13; doi:10.1038/tpj.2008.14 thereby changing the sequence of key causes an amino acid change that proteins related to drug resistance, modify the ability of the enzyme to activation and metabolism. Further- metabolize the anticoagulant warfar- Keywords: polymorphisms; miRNAs; alter- more, alternative splicing and miRNAs in.9 This genetic variation has impor- native splicing; drug metabolism; drug can work together to differentially tant implications in the dose a patient resistance; complex diseases and therapy control genes.
    [Show full text]