BMD225 revision cards for week 1 -4

?
What are the main factors regulating insulin secretion?
Blood Glucose concentration - main, GIT - gastrrointestinal tract hormones (GIP, GLP-1), Sulfonylureas - release insulin
1 of 149
What does insulin do?
Increases glucose uptake by Glut-4 in adipose tissue and muscle, inhibits glycogen breakdown so makes glycogen, synthesises glycogen, anabolic hormone, has alpha and beta subunits
2 of 149
Describe the insulin receptor?
A large transmembrane glycoprotein complex belonging to the tyrosine kinase linked type 3 receptor superfamily
3 of 149
What is diabetes mellitus and what can it cause?
Total/relative insulin insufficiency, characterised by hyperglycaemia. Causes: polyuria, ketoacidosis,increased thirst and polydipsia, appearance of glucose in urine
4 of 149
What are the two forms of DM?
Type 1 (IDDM/ Juvenile Onset Diabetes) and Type 2 (NIDDM)
5 of 149
What are some facts about Type 1 diabetes?
Type 1; often autoimmune, absolute insulin deficiency , usually see in young but can be seen in all ages, not usually about obesity, problem is with B cells
6 of 149
What are some facts about Type 2 diabetes?
Mainly due to obesity, accompanied by insulin resistance and by impaired insulin secretion, B cell function declines, treatment is initially dietary then oral methods
7 of 149
On a graph comparing normal people with type 1 and type 2 diabetics what would you expect to see?
For normal - biphasic point in the 1st phase (rapid start then slow onset). Type 2; 1st phase missing, 2nd phase slightly present. Type 1; Both phases missing. In 1st phase also produced by AA, sulfonylureas, glucagon and GI tract
8 of 149
What are the different types of insulin?
Rapid acting, short acting (Regular), intermediate acting (NPH), intermediate and short acting mixtures, long acting
9 of 149
What are some diabetic complications (long term) ?
Neuropathy; nerve damage, can predispose someone to atherosclerosis, associated with accumulation of osmotically active metabolites of glucose produced by the action of aldose reductase. Nephropathy, Peripheral vascular disease, retinopathy, CHD,
10 of 149
In the pancreas where there is beta cell dysfunction what are the drgus that work to decrease glucose level?
Sulfonylureas, Glinides, DPP-4 inhibitors
11 of 149
In muscle and Fat, where there is insulin resistance, what are the drugs that work to decrease glucose level?
TZDs (Glitazones), Biguanides
12 of 149
In the gut where there is glucose absorption, what are the drugs that work to decrease glucose level?
Alpha glucosidase inhibitors, Biguanides
13 of 149
In the liver, where there is hepatic glucose overproduction, what are the drugs that work to decrease glucose level?
Biguanides, TDZs (Glitazones), DPP-4 inhibitors
14 of 149
What type of insulin has the greatest range of activity?
Rapid actin because of the surface area causing it to take a long time to reach the peak
15 of 149
What are the complications of insulin therapy?
Hypoglycaemia, Immune mediated responses
16 of 149
What are the causes, symptoms and treatment for hypoglycaemia?
Insulin dose too large, individual misses a meal, physical exertion. Mental confusion, palpitations,sweating,trembling,coma. Glucagon administrtion
17 of 149
#what nis the use of HbA(1c-smallscript)
Lowering it reduces risk of complications. It is called glysated haemoglobin and is often used as a marker for severity of a disease. The levels of it show a positive correlation with severity of diabetes.
18 of 149
Tell me about Biguanides - metformin?
They don't require a functioning Beta cell for their therapeutic effect, They: slow down GI absorption of glucose, increase the uptake of glucose into skeletal muscle, reduce glucagon, decrease hepatic gluconeogenesis, affects insulin.
19 of 149
What are the disadvantages/advantages of biguanides?
Not well tolerated by all which can lead to sulphonylureas. Adv: Doesn't cause hypoglycaemia/stimualte appetite.
20 of 149
First generation examples of sulphonylureas?
Tolbutamide, Chlorpropamide.
21 of 149
second generation examples of sulphonylureas?
Glibenclamide, Glipizide.
22 of 149
What are the disadvantages of sulphonylureas?
Pronounced hypoglycaemia, stimulate appetitie, often cause weight gain.
23 of 149
What is the mechanism of action for sulphonylureas?
Block ATP dependent K+ channels (that can be found on the surface membranes of B cells), Bind to an ABC causing channel to close, closure results in beta cell depolarisation, calcium entry and insulin release.
24 of 149
What makes sulphonylureas different and similar to biguanides?
Similar: Treat Type 2 diabetes in its early stage. Difference: Requires functional Beta cells.
25 of 149
What is the role of Glinides?
Modulate beta cell insulin release - regulates K+ efflux, control post prandial glucose increases - resotres initial insulin concentration.
26 of 149
Benefit of glinides?
Lower hypoglycaemia because they associate adn dissociate from binding site more rapidly so provide a better overall safety profile when looking at occupancy for binding sites.
27 of 149
Role of thiazolidinediones (Glitazones)?
Insulin sensitizers that act on the peroxisome proliferator activated receptor gamma. Enhance the effectiveness of endogenous insulin thereby reducing hepatic glucose output and increasing glucose uptake into muscle. Act on nuclear receptors.
28 of 149
Thiazolidinediones by reducing blood glucose concentration what is the implication?
Reduced insulin, free fatty acid concentration, decline of triglycerides, LDL and HDL are unchanged/slightly increased. The proportion of small dense LDL particles is reduced
29 of 149
Problem with thiazolidinediones?
Weight gain of approx 1-4 kg
30 of 149
Tell me about alpha-glucosidase inhibitors - acarbose?
helpful for obese type 2 diabetes, slow carb absorption.
31 of 149
What trial is used in relation to acarbose?
STOP_NIDDM) trial; therapy in orediabetic individualss, reduces new cases of type 2 diabetes, helps restore b cell function
32 of 149
What are the problems with alpha-glucosidase inhibitors?
Has no effect on insulin secretion/it's tissue action and is less effective for achieving glycaemic control, flatulance, loose stools/diarrhoea, abdominal pain, bloating, oral absorbtion is very low.
33 of 149
Benefit of alpha glucosidase inhibitors?
The slow carb absorption means the postprandrial increase in blood glucose is reduced.
34 of 149
What are the two different types of incretin mimetics and enhancers?
GLP-1 agonists, Dipeptidyl peptidase 4 inhibitors -mimics effect of GLPs, don't cause weight loss/gain
35 of 149
Of those types what are the examples?
1. Exrenatide - peptide found in saliva of Gilatide monsters, Liraglutide. 2. Vildagliptin, Sitagliptin - Both lower blood glucose by potentiating GLP1 + GIP which stimulate insulin secretion.
36 of 149
Problems that incretin mimetics and enhancers can cause?
GI effects, Pancreatitis, Occasional Liver Disease, Worsening of heart failure
37 of 149
What is amylin?
a peptide hormone cosecreted with insulin from the pancreatic Beta cell and is therefore deficient in diabetic people. It inhibits glucagon secretion, delays gastric emptying and acts as a safety agent.
38 of 149
What are the 3 types of chemical communication?
Endocrine, Paracrine, Neurotransmitters
39 of 149
What systems make up the autonomic nervous systerm and briefly what are their roles?
Sympathetic - Fight/Flight, Parasympathetic - Rest/Digest. ANS conveys message from CNS to rest of body, regulates contraction/relaxation etc.
40 of 149
What did Loewi figure out?
Stimulation of the vagus caused the appearance in the perfusote of the frog heart of a substance capable of producing in a second heart an inhibitory effect. He named it "vagusstoff"
41 of 149
What drugs inhibit cholinergic transmission?
ACHE (neostigmine) , Non depolarising blocking agents (Tubocuraine), Presynaptic toxins (Botulinum), Vesamicol, Hemicholinium
42 of 149
What drugs activate cholinergic transmission?
Depolarising blocking agents (Suxamethonium)
43 of 149
NAme some clinical uses of botulinum toxin?
Neuromuscular; Facial wrinkles, pain conditions. Autonomic - Hyperhidrosis, GI disorders
44 of 149
What are the 3 types of ACHE?
Short duration, medium duration, irreversible.
45 of 149
What are the criteria for identifying a neurotransmitter
Chemical must be found within a neuron, must be produced within a neuron. When a neuron is stimulated it must release the chemical. When chimcal is released it must act on a post-synaptic receptor and cause an effect. After release: inactivation
46 of 149
What are the differences in the ganglionic fibres and location between parasympathetic and sympathetic?
Parasympathetic - Long preganglionic fibres, ganglion in target tissue, short postganglionic fibres. Sympathethic - reverse.
47 of 149
In the autonomic nervous system what can be said about the other 2 ganglionic gibres of the efferent nerves.
To the somatic (voluntary) there is only a single fibre. To the adrenal medulla there is a shorter single fibre with the ganglion in the paravertebral chain.
48 of 149
In the autonomic nervous system what neurotransmitters are released and where?
All nerve fibers leaving CNS release Ach. Parasympathetic - ACH in both post and pre. Sympathetic - NA/ACh (sweat glands)
49 of 149
In the autonomic nervous system on what receptors do the neurotransmitters act on.
All nerve fibers leaving CNS act on nicotinic receptors. Parasympathetic: Post - acts on MUSCARINIC. Sympathethic - acts on alpha/beta adrenoreceptors or MUSCARINIC.
50 of 149
What are the steps in neurotransmission?
Uptake of precursors, NT synthesis, NT storage, Breakdown of surplus NT, DEP, influx of Ca, Exocytosis release, Diffusion, Interaction with Post Membrane, Inactivation, Reuptake, Uptake by non neuronal, Presynaptic receptor interaction
51 of 149
What are the two types of presynaptic modulation?
Heterotropic - can casue conversion between PS and S, A NT is affecting the release of another NT from a distinct site. Homotropic - causes feedback from site in which it's released.
52 of 149
What is noradrenaline, what is its role?
ANS - postganglionic sympathetic nerve endings. Acts of alpha and beta adrenoreceptors. alpha 1 - activate phospholipase C, alpha 2 - inhibits adenylate cyclase, Beta 1,2,3 - stimulate adenylate cyclase.
53 of 149
What is the route for synthesis of NA?
Tyrosine, DOPA, Dopamine, Noradrenaline, Adrenaline.
54 of 149
How is NA released?
It's stored in vesicles, presynaptic mechanism regulates it's release, monoamine oxidase/ catechol-O-methly transferase involved in it's metabolism.
55 of 149
In the smooth muscle how do adrenoreceptor agonists regulate it?
a1 - contraction, b2 - relaxation
56 of 149
In the cardiac muscle how do adrenoreceptor agonists regulate it?
B1 - inotropic and chronotropic effects
57 of 149
In metabolism how do adrenoreceptor agonists(sympathomimetic drugs) regulate it?
B1 - carbohydrate, B3 - lipid
58 of 149
In the nerve terminals how do adrenoreceptor agonist regulate it?
a2 - main affect that mediates, effect is inhibitory, basis of action of clonidine
59 of 149
What are the uses of b-adrenoreceptor antagonists?
In cardiovascular disease. Non selective - propranolol. B1 selective - atenolol, labetolol
60 of 149
In the synthesis and release of Ach what are the compounds involved and their roles?
Choline - start, CHat - adds Acetyl Coa, VACHT - puts drugs that affect Ach storage and release in vesicles. Vesamicol - blocks VACHT. HC-3 - hemicholine transporter. ACHE
61 of 149
What are the 2 different types of activity of exogenous Ach?
Muscarinic - aprasympathetic stimulation. Nicotinic - stimulation of autonomic ganglia.
62 of 149
What are muscarinic receptors and what are the different types?
GPCR, much slower time course of action than with transmission mediated by nicotin receptors. M1(neural), M2(Cardiac), M3(glandular/smooth muscle
63 of 149
What are some muscarinic agonists and their effects and uses?
Pilocarpine, Carbachol, Muscarine. Effects: parasympathetic: bradycardia, vasodilation, sweating. PEripheral actions mainly mediated by M2 and M3. Clinical uses: glaucoma and bladder hypotonia.
64 of 149
What are muscarinic antagonists?
Parasympatholytics - tachycardia, smooth muscle relaxation. All competitive. Clinical applications: atropine - adjunct for anaesthesia, HI hypermotility.
65 of 149
What differences can be seen between non depolarising and depolarising blocking agents?
Effect of anticholinesterase drugs, fasciculations as a prelude to paralysis.
66 of 149
What are some examples of nicotinc agonists and antagonists?
Suxamethonium. Trimetaphan, Pancuronium, Atracurium, Vercutonium, Mivacurium.
67 of 149
What differences can be seen between non depolarising and depolarising blocking agents?
Depolarising: bind to Ach receptors and generate AP. Not metabolised by ACHE so binding is prolonged; extended DP. NonDP - competitive antagonists - bind but unable to induce ion channel openings, prevent Ach from binding
68 of 149
How do you experimentally deterimine affinity?
Looking at toal bs non specific total bound against concentration. Subtracting the two gives a specificity bound graph. Then a scratchard plot can be made and K and Bmax identified.
69 of 149
How do you experimentally determine efficacy and potency?
"Take organs for a bath" - bioassay, using saline and adding drugs and measuring magnitude of concentration by force transducer.
70 of 149
In a concentration response curve what is Emax and EC50 a measure of?
Emax - A measure of efficacy. EC50 - a measure of potency - effective concentration that produces 50% of that maximum response. The smaller the Ec50 the more potent - gets to maximal resposne ar a smaller conc of drug.
71 of 149
Define efficacy and potency?
Efficacy: ability of a drug to cause a functional response as a result of binding to its receptor. Potency: a measure of drug activity governed by both affinity and efficacy.
72 of 149
What are the difference and similarities between partial agonsits and full agonists?Partial have a lower efficacy than full agonists bu the same occupancy.
Partial have a lower efficacy than full agonists bu the same occupancy.
73 of 149
What are the differences between reversible competitive antagonism and irreversible competitibe antagonism?
Reversibel: equilibrium can be shifted to the right by increasing the concentration of agonist and maximal response is unchanged. Irreversible: non equilibrium conditionds due to slow/no dissociation of antagonist from the receptor.
74 of 149
Can full agonist occupancy be achieved in irreversible competitive antagonism?
No, and the observed maximal response if reduced.
75 of 149
How do we quantify competitive antagonism?
Using a Schild Plot. Organ bath experiment needs to be conducted enabling the determination of a set of dose ratios plotted against antagonist conc.
76 of 149
How do we quantify competitive antagonism?
No efficacy so agonist needed, the action of which the antagonist of interest can inhibit. The antagonist potency can be expressed in the form of a PA2 value
77 of 149
What is the pA2 value?
The negative logarithm of the molar concentration of antagonist which will reduce the response of a tissue to a double dose of agonist to that of a singl dose of agonist.
78 of 149
What are some other types of antagonism?
Chemical, Pharmacokinetic, Physiological.
79 of 149
What is occupation governed by?
Affinity.
80 of 149
What is activation governed by?
Efficacy
81 of 149
What are the targets for drug interaction?
Receptors, Ion channels, enzymes, Carrier molecules/transporters
82 of 149
What does the law of mass action give rise to and what does it relate to?
It give rise to Kd (dissociation constant), It relates to the principle that the rate of a chemical reaction is directly proportional to the concentrations of the reacting substances.
83 of 149
What are the equations involved in the law of mass action?
D+R-->
84 of 149
If there is a high affinity what does that mean for Kd?
Low
85 of 149
What are the factors affecting drug metabolism?
Age, Disease State, Gender Differences, Diet, Environment, Drug Interactions, Genetic Differences
86 of 149
What are the implications of inducing Cp450 enzymes?
Reduction in pharmacodynamic effect of the inducing drug, reduction in effect of coadministered drugs which are metabolised by the same route
87 of 149
What happens to the laf life of warfarin after repeated rifampicin dosing?
Reduced plasma 1/2 life.
88 of 149
How does age affect drug metabolism?
Heptic function is lower at birth and old age. Chloramphenicol can cause grey baby syndrome. Diazepam can cause mental confusion in elderly. There is an increase variability and increased 1/2 life with agin.
89 of 149
How can environmental factors affect drug emtabolism?
They affect cP450 function - inducers/inhibitors. Examples: tobacco, heavy metals, industrial pollutants.
90 of 149
How can disease state affect drug metabolism?
Liver disease causes lower hepatic function - microsomal oxidases affected, decreased capacity to metabolise drugs. Viral hepatitis causes slower diazepam metabolism - overdose and coma maybe
91 of 149
How can diet affect drug metabolism?
GFJ interactions - increases plasma felodipine concentrations and alters it's oral bioavailability - increasing cardiac toxicity risk. Increases simvastatin serum concentration - prolonging life
92 of 149
How else can diet affect drug metabolism?
Glucosinolates in brussel sprouts can induce p450 enzymes that are metabolising enzymes so drug effects and duration of action are diminished.
93 of 149
How can genetic differences affect drug metabolism?
Causes a large fold variation in response to a drug, often due to SNPs
94 of 149
How else can genetic differences affect drug metabolism?
CYP2D6 polymorphisms are responsible for the metabolism of a lot of important drugs. Within a european pop 4 phenotypes: PM, IM, EM (highest frequency in caucasians), UM.
95 of 149
What is warfarin and how do genetic differences affect it's drug metabolism?
Anti coagulant, wide variation in dose requirements depending on haplotype. Gene guided dosing takes into account the patient genotype for CYP2C9 and VKORC1.
96 of 149
What is isoniazid and how do genetic differences affect it's metabolism?
Anti-TB drug. Metabolised by acetylation. Two types: fast acetylators (may lead to higher blood levels of drug and increase in toxicity), slow acetylators (60%, respond well) - types inherited, ratio varies
97 of 149
How does isoniazid compare with salicylate?
S - normally distributed. I- bimodial distribution.
98 of 149
Do receptor polymorphisms as part of genetic differences play a part in drug response?
yes, eg %-HT2C - can cause increased body fat.
99 of 149
How does gender affect drug metabolism?
Sex hormones can change expression pattern. In phase 1 and 2 only CYP2C9 shows no sex differences in clearance values. In phase 2 only conjugation N-acetyltransferase mixed with caffeine/dapsone will show no sex differences in clearing.
100 of 149
Which hepatic gene mixed with which model substrate show increase in clearance in women.
CYP3A4 - midazolam.
101 of 149
What are the different ways drugs can be administered?
Oral/Rectal, PErcutaneous, Intravenous, Intramuscular, Intrathecal, Inhalation.
102 of 149
How can drugs be eliminated?
Urine, faeces, milk/sweat, expired air
103 of 149
What is drug disposition?
The processes a drug is subjected to by the body upon administration. Includes: Absorption,Distribution,MEtabolism, Excretion.
104 of 149
What is the concept of bioavailability?
It is defined as the fraction (F) of the administered dose that reaches the systemic circulation.
105 of 149
Which route will have F as 1?
Intravenous injection
106 of 149
Which route will have F
Particularly oral
107 of 149
What does the bioavailability of
Gastirc inability of drug, 1st pass metabolism (liver), Reduced absorption into the circulation.
108 of 149
What are the factors that affect drug absorption?
Gastric motility, area of absorbing surface, blood flow at absorbing surface, ingestion of food, plasma protein binding, nature of the drug.
109 of 149
What are the different ways a drug can be absorbed across membranes?
Diffusion through lipid, diffusion through aqueous channel, via a carrier, pinocytosis.
110 of 149
What occurs when a drug is diffused down an ion channel/ aqueous pore?
Movement is down a concentration gradient, the membrane contains small pores which allow water soluble molecules of < 100Da through. Eg Lithium. NOT A MAJOR ROUTE OF DRUG PERMEATION.
111 of 149
What occurs when a drug moves via carrier mediated transport?
Active transport/facilitated diffusion. Important sites: renal tubule, BBB, GI tract. These transporters can become saturated limiting rate of drug permeation + competition can occur between two ligands that bind to such carriers.
112 of 149
What occurs when a drug moves via pinocytosis?
Uptakes macromolecules; 1000Da molecular weight or more) e.g insulin crossing the BBB. Attempts to utilise it as target drug uptake mechanism - incorporating the drug into lipid liposomes.
113 of 149
What occurs when a drug diffuses through lipid membranes?
Common route for most drugs, only small lipid soluble, non polar drugs. Depends on surface area and blood flow. Degree of permeation by this route dictated by Fick's Law.
114 of 149
What relationship does Fick's law follow?
The amount of drug diffusing with time is directly proportional to the permeability coefficient x membrane area x (concentration of drug either side of the membrane) but is INVERSELY PROPORTIONAL to the membrane thickness.
115 of 149
What is the implication of a highly lipid soluble drug?
It has a large transmembrane concentration gradient and will diffuse more rapidly than a drug of lower lipid solubility even when the concentration gradient on either side of the membrane is the same.
116 of 149
What is the effect of ph on drug ionisation?
It allow prediction of absorption.
117 of 149
What are the henderson hasselbach equations?
for a weak acid: pka=ph+log10[HA]/[A-], for weak base HA becomes BH+ and A- becomes B.
118 of 149
What is the pKa?
A dissociation constant, negative logarithm of Ka, equal to the ph at which 50% if the drug molecules are ionised.
119 of 149
What are ionised species and what is their implication?
BH+/A- --> have very low lipid solubility and are virtually unable to permeate membranes without a specific transport mechanism
120 of 149
What are the implications of the uncharged species?
Lipid solubility depends on their chemical nature, for many they are sufficiently lipid soluble.
121 of 149
In terms of distribution what body compartment has the largest?
Intracellular water.
122 of 149
Order the body compartments from largest percentage to lowest.
Intracellular water, Fat, Interstitial water, Plasma water, Transcellular water
123 of 149
What is the volume of distribution?
The volume of fluid required to contain the total amount of drug in the body at the same concentration as that present in the plasma (Cp)
124 of 149
How cna Vd be calculated?
Vd= Dose/Cp
125 of 149
What does the degree of ionisation give rise to?
Ion trapping
126 of 149
For aspirin which body compartment has it's highest concentration and why?
Urine because urine is alkaline and ionisation of the acidic drug is greatest at alkaline ph.
127 of 149
For pethidine which body compartment has it's highest concentration and why?
Gastric Juice because as the drug is a weak base ionisation is greatest at an acidic pH.
128 of 149
How is Vd derived experimentally?
By plotting plasma concentration vs time and then log plasma concentration versus time.
129 of 149
What are the sites of drug metabolism and what happens to drugs?
Few are eliminated from the body unchanged, most undergo biotransformation. The sites include: liver (major), kidney, lungs, skin, intestine.
130 of 149
What is the consequence of first pass metabolism, in which organ can it be found and what is the circulation of drugs in that organ?
It reduces bioavailability, in the liver, oral medication--> intestinal tract--> along portal vein to liver--> exits liver from hepatic vein --> systemic circulation--> hepatic artery back into liver.
131 of 149
What is the process of metabolism?
To change a drug into more water soluble molecules which increase polarity and aid excretion.
132 of 149
What occurs to drugs in certain instances?
1. Drugs can be activated by metabolism -prodrugs. 2. Metabolites can produce more active metaboliteswhich contribute to the action of the drug
133 of 149
What does metabolism often consist of?
Phase 1 and Phase 2. Drug --> Derivative (Phase 1) Derivative --> Conjugate (Phase 2)
134 of 149
What processes are used in phase 1?
Ocidation, Hydroxylation, Dealylation, Deamination, Hydrolysis.
135 of 149
What process is used in phase 2?
Conjugation.
136 of 149
What occurs in Phase 1 reactions?
Often enzyme catalysed. Most important: microsomal mixed function oxidase - resides on smooth ER in liver, requires a reducing agent (NADPH) and molecular oxygen. Of these, Cytochrome P450 (a haem protein) is most important.
137 of 149
What are the non mfo phase 1 reactions?
Oxidaative reactions, cytoplasmic in nature, metabolise ethanol. Mitochondrial: catecholamine metabolism by monoamine oxidase.
138 of 149
What occurs in Phase 2 reactions?
Occur mainly in the liver, conjugation reactions which occur at polar groups(introdcued during phase 1 reactions), conjugated products are very water soluble, excreted in urine /bile
139 of 149
What is the most common conjugation reaction and what does it utilise?
Glucoronidation. Uses an endogenous reactant (UDP glucorinic acid), attaches to N,O,S atoms.
140 of 149
Does paracetamol get metabolised via phase 1/ phase 2 reaction?
Both. Phase 2: Glucoronide/ Sulphate conjugation. Phase 1: via CP450.
141 of 149
What occurs in paracetamol metabolism?
90% conjugation. remainder oxidation - using CYP2E1 isoform
142 of 149
How do drugs get excreted?
Mainly in the kidney via clearance.
143 of 149
What is clearance?
A measure of elimination (depends on both metabolism and excretion). It can also be considered a proprtionality constant between the rate of elimination and the concentration of the drug.
144 of 149
Is there an equation for clearance?
Rate of elimination/Plasma Concentration
145 of 149
How can renal clearance be defined as?
A measure of elimination, defined as 'The volume of plasma cleared of drug by the kidney per unit time' (ml/min)
146 of 149
What are the factors influencing clearance?
Renal processes: Glomerular Filtration, Passive reabsorption, Active Tubular Secretion.
147 of 149
How can ion trapping cause the urinary pH to influence clearance.
In the example of phenobarbital (weak acid) clearance is greater in alkaline urine than in acidic urine because there is a greater degree of ionisation so the drug is more likely to be absorbed in the urine.
148 of 149
State the drugs in order of lowest to highest clearance value.
Warfarin, Digitoxin, Diazepam, Ampicillin, Nifedipine, Lidocaine, Propranolol, Imipramine.
149 of 149

Other cards in this set

Card 2

Front

What does insulin do?

Back

Increases glucose uptake by Glut-4 in adipose tissue and muscle, inhibits glycogen breakdown so makes glycogen, synthesises glycogen, anabolic hormone, has alpha and beta subunits

Card 3

Front

Describe the insulin receptor?

Back

Preview of the front of card 3

Card 4

Front

What is diabetes mellitus and what can it cause?

Back

Preview of the front of card 4

Card 5

Front

What are the two forms of DM?

Back

Preview of the front of card 5
View more cards

Comments

No comments have yet been made

Similar Other resources:

See all Other resources »See all Biomedical Pharmacology resources »