General Pharmacology Principles Flashcards

1
Q

What does ADME stand for and give a breif description of each letter.

A

A- absorption- from the site of administration to systemic ciruculation (defined as major vessels adn well perfused organs)

Most often from GI tract (orally administered drugs must pass through the liver before reaching systemic circulation)

D- distribution- of the drug from systemic ciruclation to tissues (target and non-target) and back again

Drug not bound to plasma proteins (free drug) is disstributed into tissues - where it is either bound to tissues or back to circulation

Blood and lymph-pigott

M- metabolism -elimination of the drug from the body via metabolism

Hepatic metabolism

E- excretion -elimination of the drug from the body via excretion

Renal or biliary exretion of drug or its metabolites. Excreted drugs can be passively reabsorbed by kidneys following enterohepatic circulation (bile)

  • these movements are dynamic, occuring simaltaneously
  • net effect determines PDC at any time during the dosing interval (following adminstration of a fixed dose)

Boothe pg 2 ,5

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Define Pharmacokinetics

A

1 : the study of the bodily absorption, distribution, metabolism, and excretion of drugs

2 : the characteristic interactions of a drug and the body in terms of its absorption, distribution, metabolism, and excretion

Merriam-Webster Dictionary

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe the Plasma Drug Concentration Vs Time Curve

A
  • movement of drug through the body are characterized by plotting drug concentrations measured after adminstration of a known dose against timeon semilogarithmic paper.
  • Fig 1-3 (A and B)

Boothe pg 3,4,6,7

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Discuss oral absorption

A
  • Orally adminnistered drugs reach systemic circulation after absorption from the SI
  • rate and extent of drug absorption from GIT depends on many factors (which often passive diffusion):
  • GI pH (which favors absorption of weak acids)
  • Surface area (which favors absorption in the SI >stomach)
  • Motility (which favors mixing drugs thus increasing concentration of diffusable drug)
  • Permeability and thickness of mucusal epithelium
  • intesitnal blood flow (which maintains the concentration gradient across teh mucosal epithelium- most improtant for drugs with rapid transfer)

Bioavailability

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Define bioavailability and discuss

A
  • The percentage of an administered dose that reaches systemic circulation = bioavailability
  • Determined by measuring the area under a PDC vs time curve (AUC) after non-IV adminstration and comparing this number with the AUC after IV adminstration of the same dose.
  • if the AUC is the same for both then bioavailability is 100%
  • Bioavailability is used to predict efficacy of different routes of administration (or different forumlations)
  • Factors which effect absorption also effect bioavailability
  • Bioavailability is also decreased if the drug is metabolized by: intestinal cells, microbes or liver
  • Hepatic metabolism can have a profound effect on bioavailability and PDC (first pass metabolism)- so you can have great absorption and poor bioavailabitlity (so administer the drug by a route other than PO to account for high first pass metabolism)

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Explain first-pass metabolism

A
  • Occurs as drug absorbed from the GIT (stomach to lower colon) enters the portal vein and flows to liver, drug is then characterized by high extractin are remomved rapidly from the blood before entering systemic circulation
  • plasma concentrations of the drug may not reach therapeutic concentrations unless dose is increased to account for first pass metabolism.
  • Hepatic metabolism can have a profound effect on bioavailability and PDC (first pass metabolism)- so you can have great absorption and poor bioavailabitlity (so administer the drug by a route other than PO to account for high first pass metabolism)
  • The amount removed by the liver vareis with species and age and disease status

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Define and discuss drug distribution

A
  • Distribution from the central (blood) compartment to peripheral tissues, major factords which determine distribution include:
  • Lipid solubility (ability to penetrate cell membrane)
  • Plasma or tissue bound (degree to which it is)
  • Regional (organ) blood flow

Vd= volume of distribution= the amount of tissue to which a drug is distrubuted. It directly influences PDC

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Define bioequivalence and mean absorption time (MAT)

A

-“a drug whose rate of absorption is slower maybe completely absorbed, but the PDC may not reach smae magitude or peak as another preparation or route because the absorption occurs very slowly (absorption rate constant Ka)”

MAT- likely to be the parameter reported for assessing drug absorption from the site of adminsitration

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Define Volume of Distrubution and what give example of what causes it to increase or decrease?

A

Vd= volume of distribution= the amount of tissue to which a drug is distrubuted. It directly influences PDC

  • Maximal PDC is determined after distribution equilibrium and before excretion
  • Reported in L/kg
  • Increased by: disease associated with fluid retention or obesity
  • Decreased by: dehydration or weight loss

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Discuss protein binding in relation to drug distribution?

A
  • Plasma protein binding renders a drug more water soluable, thus facilitating its movement in circulation, however it cannot be distrubuted from the plasma into tissues (or tissues back to plasma).
  • Protein bound drugs are:
  • not pharacologically active
  • cannot be renally excreted
  • re more slow to metabolize by the liver
  • Displacement from plasma proteins also increased Vd because the drug is more likely to enter tissues when freed
  • Weakly acidic drugs thend to bind to albumin whereas weakly basic drugs tent to bind to alpha1-glycoproteins
  • Displacement of drug from protein binding sites become significant only if the drug is greater than 80% protein bound.
  • Hightly protein bound drugs do not distribute into tissues, but remain in systemic circulation (the Vd of the unbound portion of drug can be very high)

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Discuss drug distribution with respect to water vs lipid soluable drugs

A
  • Water-soluable drugs tend to be distrubuted to ECF with Vd 0.1-0.3L/kg
  • Lipid-soluable drugs tend to be distrubuted to ECF and ICF, b/c they cross cell membranes with large Vd >0.6L/kg
  • Regardless of soluability, drugs not bound to plasma proteins are able to pass unimpeded through fenestrated capillaries into ECF

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

The capillaries of which oragans are not fenestrated, thus requring drug to pass through the capillary endothelium in order to penetrate the target tissue?

A
  1. brain
  2. CSF
  3. Eye
  4. testis
  5. prostate

-lipid-soluabe drugs are more likely to penetrate the endothelial barrier

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Drug excretion is a combination of what factors?

A

Elimation by hepatic metabolism

Renal excretion

or both

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Discuss phase I metabolism

A
  • chemically changes drug so that it is (usually) more water soluable and more susceptably to phase II metabolism
  • recations include: oxidation/hydrolysis/reduction
  • Performed by enzymes referred to as cytochrome P450 enzymes
  • Phase I metabolites are usually inactive, but can be equally, more or less active or toxic than parent coupound

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are cytochrome P450 enzymes?

A
  • Enzymes responsible for the majority of phase 1 metabolism
  • large superfamily
  • found other places than the liver (lungs, skin, kidney)

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Discuss phase II metabolism

A
  • AKA conjucation, occurs when large water-soluable molecule is chemically added to parent or metabolite
  • Glucuronidation most common reaction (sulfonaton and acetylation less common)
  • with rare exceptions phase II metabolites are inactive
  • most metabolites are eliminated through the urine

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Summary of hepatic metabolism and elimination (Fig 1-8)

A

fig 1-8 boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Discuss renal elimination of drugs

A
  • renal elimination is most improtant/common route of drug elimination (>biliary)
  • rate of renal excretion:
  1. active tubular secretion in proximal tubule is a rapid process and no limited by protein binding (depends on RBF, drug pKa)
  2. Glomerular filtration is passive process, protein bound drugs cannot be filtered (depends on RBF, protein binding, MW)
  3. Un-ionized drugs that are sufficiently lipid soluable maybe passively resorbed (depends on drug concentration, MW, lipid soluability, drug pKa, urine pH). Reabsorption slows renal excretion.

-kidney also capable of metabolizing

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Discuss biliary exretion of drugs

A
  • very slow and clinically less improtant
  • characteristics which determine biliary excretion”
  • chemical structure
  • polarity
  • MW *major determinant
  • more contact with GI and flora and more likely to cause adverse GI SE
  • enterohepatic circulation

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Describe enterohepatic circulation

A
  • enterohepatic circulation can prolong the elimination half-life of a drug
  • After oral or parenteral administration, drug is conjucated in the liver and eliminated in the bile. -Drug travels the duodenum to the lower SI/colon. -Bacterial degredation results in deconjugation of the drug which can then be re-absorbed.

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

How are drugs concentrated in the urinary system?

A
  • Urinaryy pH can be therapeutically altered to change excretion rate of drug (weakly acidic drugs are more likely to be reabsorbed in acidic urine, but are trapped and excreted in alkaline urine)
  • Also drugs that are renally excreted can concentrate up to 300-fold in the urinary bladder (not kidney) compared to the plasma concentrations (ex-amoxicillin)
  • Also can use other drugs (ex probenecid) to compete with drugs during the active transport phase of excretion to inhibit their excretion (ex imipenem) to prolong theraputic PDC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Discuss drug elimination

A
  • rate of drug elimination (kel - is the slope frome the PDC vs time curve) describes the fraction of drug in the body irreversibly eliminated per unit time (time -1)

Boothe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Define and discuss Elimination Half-Life

A
  • Elimination half-life of a drug is the time necessary for half the drug to be eliminated from the body
  • is one of the most useful parameters for determining an appropriate dosing interval
  • Elimination half-life and dosing interval determines time to steady state and the amount of fluctuation in PDC during dosing intervals

Boothe

24
Q

Discuss first order vs zero order in the context of the elimintion rate constant

A
  • First order (constant fraction)
    ex) kel of 0.25 h -1 suggests that 25% of the drug remaining in the animals body is eliminated each hours
  • Zero order (constant amount)
    ex) 25mg decrease each hour
  • first order elimination might be considered protective mechanism to ensure rapid elimination of potentially toxic substances

Boothe

25
Q

Define and discuss drug clearance (how does this differ from drug elimination?)

A
  • Plasma clearance (Cl) is the volume of plasma irreversibly cleared of drug per unit time and representes the sume total of organ clearance
  • different from elimination becaue it is a volume per unit time, not a rate constant
  • Clearance of a drug representes the fraction (Kel) of the Vd of a drug that is cleared per unit time

Cl=Vd x Kel

-Cl can only be determined aftre IV adminstration

Boothe

26
Q

Define Pharmacodynamics and pharmacodynamic drug interactions

A

a branch of pharmacology dealing with the reactions between drugs and living systems

Merriam-Webster dictonary

pharmacodynamic drug interactions occur when one drug directly alters the chemical or physiologic response to another drug (variable response- additive vs synergistic vs decreae vs antagonistic)

Boothe

27
Q

How many half-lifes must elapse before 97% of the drug is eliminated? or how many half-lives must elapse following a fixed dosing regime before steady state is reached?

A

5

-Steady state is relevant issue only for drugs that accumulate- must be administered at a dosing interval that is shorter then the drug elimination half-life

Boothe

28
Q

How does renal disease affect drug ADME?

A
  • increased sensitivity to the drug d/t:
  • uremia-induced alterations in tissue receptors
  • Disease induced changes in pharmacokinetics and thus increased/decreased PDC
  • There is also changes in RBF and thus GFR and active tubular secretion
  • Factors which displace drugs from protein-bound sites (which may increase drug excretion):
  • hypoalbuminemia
  • competition for protein-binding sites d/t accumulation of uremic toxins
  • changes in conformation and thus binding affinity (due eto increased concentration of inflammatory proteins)
  • decreased binding of acidic drugs
  • normal to increased binding of basc drugs
  • excretion most likely to be decreasd d/t decreased:
  • nephron mass
  • RBF
  • Tubular function

Boothe

29
Q

How does hepatic disease affect ADME?

A
  • Efficacy of hepatic elimination is determined by hepatic clearance and hepatic extraction ratio, which depend on:
  • hepatic blood flow
  • extent of drug protein binding
  • intrinsic hepatic clearance
  • hepatic uptake (rate-limiting)
  • intracellular transport
  • metabolism

+/-biliary excretion

  • only unbound drugs can be extracted by the liver
  • hepatic bloodflow is reduced in chronic liver diseass/PSS
  • which can markedly increase the systemic bioavailability of drugs whose oral doses are genernally increased to account for first-pass metabolism
  • Cholestasis decreases the cytochrome P450 thus affecting elemination (those excreted in bile and not in bile)
  • Decreased protein binding (because of hypoalbuminemia) may increase hepatic clearance which compensates for reduced hepatic metabolism
  • Changes in protein binding will affect drug distrubution

Boothe

30
Q

How does cardiac disease affect ADME?

A
  • Effects drug distrubution b/c
    1) renal sodium/water retention
  • changes the size of body compartments
    2) increased pulmonary/systemic venous pressures
    3) increased sympathetic nervous system outputs
  • redistribution of blood flow (heart/brain recieve more, skeletal [big compartment] receives less)

=increase in PDC to brain/heart

  • Ascites can account for ~30% of body weight
  • if drug not in ascites increases PDC by 30%
  • if drug in ascites increases half-life
  • As CO declines, blood flow to liver/kidneys declines and there is decreased clearance of drug
  • in kidney intrarenal redistribution of blood (due to sympathetically meidated changes) causes blood from cortex to juxtaglomerular tubules to increase likelihood of tubular reabsorption (prolongs drug half life)
  • Rate of absorption impaired for parenterally administered drugs- redistribution of blood flow away from muscle/skin

Boothe

31
Q

Recommendations for adminsitering drugs to a patient with cardiac disease?

A
  1. critical drugs shoud be adminsitered IV b/c all other routes of absoprtion are limited
  2. drugs which are toxic (especially to brain/heart) should not be given rapidly IV (give 10-30min)
  3. high drug concentrations resulting form redistribution should be compensated for by decreased loading doses
  4. Maintenance doses of selected drugs cleared by liver, kidney or bothe should probably be lowered (although by how much is difficult to predict)

Boothe

32
Q

How does thyroid disease affect drug diposition?

A
  • both hyper and hypo thyroidism can affect drug disposition (unpredictably)
  • may increase or decrease cytochorom P450 enzymes or their cofactors

Boothe

33
Q

How does age (old) affect ADME with respect to different organ systems?

A
  • Aging is accompanied by a perminent loss of 30% of body cells with parallel decrease in oxygen consumption and changes in region blood flow rates
  • CO decreases
  • CBF decreases
  • reduced lung volume
  • atrophy of gastric mucosa and motility
  • decreased hepatic function/blood flow
  • decreased RBF/GFR

Boothe

34
Q

How does body weight and composition affect ADME?

A
  • increased proportion of body fat = decreased total body wtaer and cell mass
  • change in proportion of intracellular to extracellular fluids

In Fat Animal:

  • water-soluable drugs don’t enter fat = higher PDC
  • lipid-soluable drugs enter fat = increase Vd, decrease PDC, increase elimination half-life

Boothe

35
Q

What is the age breakdown for pediatric patients?

A

Pediatric generally refers to first 12 weeks of life

Neonatal (0-2 weeks)

Infant (2-6 weeks)

Pediatric (6-12 weeks)

Boothe

36
Q

How does age (pediactric) affect ADME?

A

Table 2-14 Boothe

37
Q

What are the 4 principle protein targets with which drugs can interact?

A

1) enzymes
2) membrane carriers
3) ion channels
4) receptors ** what further flashcards refer to**

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

38
Q

Define receptor and give 4 subdivisions

A

loosely- ‘molecule that recognizes specifically a second small molecule (ligand) whose binding brings about the regulation of a cellular process..in the unbound state a receptor is functionally silent’

1) ligand-gated ion channels
2) Tyrosine-kinase-coupled
3) Intracellular steroid
4) G-protein coupled (GPCR)

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

39
Q

Define Agonist (full, partial)

A

Agonist is a drug that binds to a receptor and produces a functional response

A Full Agonist is an agonist that produces the maximum response capable in that system, anything which produces a lower response is called a Partial Agonist. (buprenorphine- ex partial)

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

40
Q

What is the difference between efficacy and potency?

A

Efficacy is the ability to produce a response (or intrinsic activity).

Potency is the dose range over which a response is produced.

Not the same, not interchangeable

Not the same as affinity either (Which can have varying degrees of efficacy and ptoency)

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

Potency (strength) refers to the amount of drug (usually expressed in milligrams) needed to produce an effect, such as relief of pain or reduction of blood pressure. For instance, if 5 milligrams of drug A relieves pain as effectively as 10 milligrams of drug B, drug A is twice as potent as drug B.

Efficacy is a drug’s capacity to produce an effect (such as lowering blood pressure). For example, the diuretic furosemide eliminates much more salt and water through urine than does the diuretic hydrochlorothiazide. Thus, furosemide has greater efficacy than hydrochlorothiazide.

Merek Manual

41
Q

What are spare receptors?

A

Spare receptors are not pooled or hidden- they are simply surplus to requirements.

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

42
Q

Define antagonist (competative, non-competative) and types of non-competative (active site vs allosteric)

A

Antagonist block the effects of agnoists.

1) Competative- reversible, surmountable
- Can be overcome by increasing the amount of the agonist (share the same binding sites)
- reduce potency
2) Non-competative - irreversible, insurmountable.

Cannot be overcome by increasing the amount of agnoist (binding sites are different)

  • reduce efficacy
    a) Non-competative active-site antagonist

binds covalently or with very high affinity (irreversible)

b) Non-competative allosteric antagonist prevents the receptor from being activated (even if the agonist is already bound to the active site)

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

Principles of Pharmacology. Golan.

43
Q

Define Inverse Agnoist

A

Inverse agonist inhibits activity and is said to display negative efficacy (so the receptor can be active in the absence of an agonist)

Drugs and Receptors. Journal of Anesthesia, Crictical Care and Pain. Vol 4, Number 6, 2004

44
Q

What is ion trapping?

A
  • Ion trapping is the reason why basic (alkaline) drugs are secreted into an acidic pH environment, and acidic drugs are excreted into an alkaline pH environment.
  • In cell biology, ion trapping is the build-up of a higher concentration of a chemical across a cell membrane due to the pKa value of the chemical and difference of pH across the cell membrane
  • Does not require any energy or enzymes (not active movement of molecules)
  • Cells have a more acidic pH inside the cell than outside (gastric mucosal cells being an exception). Therefore, basic drugs (like bupivacaine, pyrimethamine) are more charged inside the cell than outside. The cell membrane is permeable to non-ionized (fat-soluble) molecules; ionized (water-soluble) molecules cannot cross it easily. Once a non-charged molecule of a basic chemical crosses the cell membrane to enter the cell, it becomes charged due to gaining a hydrogen ion because of the lower pH inside the cell, and thus becomes unable to cross back. Because transmembrane equilibrium must be maintained, another unionized molecule must diffuse into the cell to repeat the process. Thus its concentration inside the cell increases many times that of the outside. The non-charged molecules of the drug remain in equal concentration on either side of the cell membrane.

Wikipedia

45
Q

Theraputic Window Chart

A

Therapeutic window is a range of doses that produces therapeutic response without causing any significant adverse effect in patients.

Generally therapeutic window is a ratio between minimum effective concentrations (MEC) to the minimum toxic concentration (MTC). The levels of drug should always be between MEC and MTC in order to provide risk free therapeutic effects. If any drug crosses MTC then it will surely elicit toxic effects and if drug is unable to surpass MEC then it will cause therapeutic failure. MEC is also called as minimum inhibitory concentration (MIC).

Medimoon

46
Q

Theraputic index chart

A

Therapeutic index (TI) describes a relationship between the doses of a drug that causes lethal or toxic effects with the dose that causes therapeutic effects. It is also called as therapeutic ratio.

Mathematically you can calculate TI by following way;

Therapeutic Index: LD50/ED50

or

Therapeutic Index: TD50/ED50

Where

LD50 is the minimum amount of drug that causes adverse effects in 50% of the population. LD50 could also be replaced with Toxic dose (TD50)

ED50 is the quantity of a drug that can produce desired therapeutic effects in 50% of the population. Such types of studies are usually conducted in animal models

Ideally any drug that requires more amount to produce toxic or adverse effects in 50% of population will have wider therapeutic index and vice versa. Drugs having wider therapeutic index are safer in comparison to those having low therapeutic index because minor modification in the dose of such drugs (aspirin, acetaminophen) will not produce toxic effects.

Medimoon

47
Q

Define tachyphylaxis

A

Rapid appearance of progressive decrease in response to a given dose after repetitive administration of a pharmacologically or physiologically active substance

Medical Dictionary

Due to down regulation of receptors that may occur as a resilt of continual stimulation by agonist

Merck

Repeated administration of the smae dose of a drug resultss in a reduced effect of the drug over time.

Golan

**Ketamine can delay the onset of tachyphylaxis to an opioid-pigott

48
Q

Define desensitized vs inactivation with resept to drug receptor interactions

A

Desensitization: cellular response to repeated stimulation/repeated receptor binding results in less and less cellular effects (decreased ability of a receptor to respond to stimulation by a drug or ligand)

**pseudophed- pigott

Inactivation: complete lack of response to receptor/ligand binding (loss of ability of a receptor to respond to stimulation by a drug or ligand)

Principles of Pharmacology. Golan.

49
Q

What is the difference between graded dose response relationships and quantal dose response relationships?

A

Graded concetnration-response curve used for cpontinuos responses; relates the intensity yof a response to the size of the dose.

Quantal concentration response curve used for an all-or-none response, relates to a log distribution

Merck

50
Q

Hypothetical dose response curve (chart) and example dose response cureves (charts)

A
51
Q

Define refractory vs down regulation with respect to drug receptor interactions?

A

Refractory: after a receptor is stimulated a period of time is required before the next drug-receptor interaction can produce an effect

Down-regulation: repeated or persistent drug-receptor interaction results in removal of the receptor from sites wherre subsequent drug-recetor intereactions could take place.

**hormones- pigott

Golan

52
Q

Define the principle of induced fit.

A

The principle of induced fit suggests that drug-receptor binding can have profound effects of the confirmation of the receptor. Inducing conformational change in the receptor can improve the quality of binding and such its action

Receptors exisit in multiple conformational states (open/active, closed/inactive or desensitized) and that binding of a drug stabalizes one or more of these conformations.

Golan

53
Q

Define suicide substrate and give example.

A

Suicide substrate is a competitive inhibitor that is converted to an irreversible inhibitor at the active site of the enzyme.

Penicillins act as suicide substrates of glycopepetide transpeptidase in bacteria. The irreversibly bind (covalently) to the active site and perminently inactive the receptor.

Medical dictionary

54
Q

What is the difference between pharmacodynamics and pharmacokinetics?

A

Pharmacodynamics is the study of how a drug modifies an organism, whereas pharmacokinetics is the study of how the organism modifies a drug. Pharmacodynamics is sometimes abbreviated as PD and pharmacokinetics as PK, especially in combined reference (for example, when speaking of PK/PD models).

**PD (quantified), PK (qualitative)- pigott

Wiki

55
Q

What are the 3 major binding forces in drug-receptor interactions?

A

1) covalent - very strong. Irreversible
2) Electrostatic - weaker than covalent (strength variable). Due to hydrogen bonding

van der Waal- induced dipole interaction (weak)

3) Hydrophobic - weak. Due to molecules wanting to avoid an aqueous environment (more then their attraction to each other)

*range of degree of how tightly bound- pigott

Katzung, B. G. Basic Principles: Introduction in Basic and Clinical Pharmacology, (Katzung, B. G., ed) Appleton-Lange, 1998, p.1-4.

56
Q

What drug properties favor drug entry into the CNS?

A

The ideal compound to treat CNS infections is of small molecular size, is moderately lipophilic, has a low level of plasma protein binding, has a volume of distribution of around 1 liter/kg, and is not a strong ligand of an efflux pump at the blood-brain or blood-CSF barrier.

Clin Microbiol Rev. 2010 Oct; 23(4): 858–883.

doi: 10.1128/CMR.00007-10