• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/258

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

258 Cards in this Set

  • Front
  • Back
2 general requirements for FDA approval to market a drug product?
1. demonstrated safety & efficacy
2. demonstrated process control & validation
what are the 2 main stages of research and development
pre-clinical and clinical
T/F: the pre-clinical stage of research and development has the substages of drug discovery, biological characterization, preformulation, & initial formulations
True
what is the method of drug discovery?
most today result from carefully designed rational research programs
put the following steps of drug discovery in order?
lead, target, candidate, hit
target-----hit-----lead----candidate
this step of drug discovery is the identification and screening
hit
_________is the preclinical and then clinical drug development of drug discovery
candidate
identification and validation is which step in drug discovery
target
T/F: Lead is the identification and validation step of drug discovery
False Lead is the identification and optimization step of drug discovery
In which stage of research and development do you spend the most money?
clinical
the biological characterization of drug is its potential for use as a therapeutic agent and is charcterized by examinations of its
a. pharmacology
b. toxicology
c. disposition (what happens when u take it)
d. pharmacokinetics (how fast does it work)
e. all of the above
E all of the above
preformulation consists of what?
physiocochemical properties
analytical method development
In essence ________ ________ is the proess of turning an active compound into a form and strength sutiable for human use.
dosage formulation
once the physicochemical properties are understood, initial dosage forms aredeveloped for use in clinical trials; this is what stage of pre-clinical?
initial formulations
a comprehensive application delineating all aspects of the pre-clinical studies and the plan for clinical studies
investigational new drug application
the main purposes of investigational new drug (IND) application
1. to protect the rights & safety of the human subjects
2. to ensure that teh investigational plan is sound
FDA Review of IND
1. 30 day safety review
2. review by experts
what happens when IND is reviewed by experts?
the application is forwarded to a specific division within the FDA, where it is reviewed by experts within the field
what is the automatic time period during which teh drug cannot be administered to humans?
30 day safety review
For FDA drug classification system, drugs are classified by what 2 things?
1. chemical type
2. therapeutic potential
What are 3 phases of clinical research & development
phases 1,2,& 3
what is the purpose of phase 1?
Phase 2/?
phase 3?
phase 1: mainly safety
phase 2: mainly effectiveness, but also short term safety
phase 3: safety, effectiveness, & dosage
patients in this phase of testing are usually healthy volunteers?
phase 1
in which phase or phases are patients tested are those that are suffering from the indicated condition
phases 2 & 3
Number of patients used for each phase of teh clinical part of research and development?
phase 1: 20-100
phase 2: up to several hundred
phase 3: several hundred to several thousand
what is the length of testing for each phase of the clinical trials?
phase 1: several month
phase 2: several months to 2 years
phase 3: 1 to 4 years
what is the % of drugs completing each phase of the clinical trials?
phase 1 :67%
phase 2: 45%
phase 3: 5-10%
during _______ ________, the final formulation is completed as well as the packaging and labeling.
clinical studies
The new drug application has what kind of data?
pre-clinical & clinical data
what summaries are found in the new drug application?
1.summaries of human pharmakinetics
2. bioavailability
3. microbiology (if it's an antibiotic)
what are 4 points addressed in the new drug application?
1. a presentation of pre-clinical & clinical data
2. summaries of human pharmacokinetics, bioavailability, microbiology
3. discussion of the benefits and risks of the drug's usage
4. complete proposed product labeling (including dosage)
Describe new drug application
integrates all data
typically 100K pages or more and takes on average a year to rvu.
New Drug application is comprised of what 3 things?
1. the application
2. the inspection
3. the decision
The inspection for the new drug must comply with what?
1. The quality standards contained in the application
2. The quality standards specified in the FDA's Current Good Manufacturing Practice Standards
The decision for the new drug application consists of the complete response letter. What will the letter contain?
It will either contain an approval stating that the application is approved or it will outline deficiencies and may recommend actions needed to be taken for approval.
What is the phase of clinical studies that has post marketing?
phase 4
What happens in phase 4?
following approval, the drug product is continually monitored in clinical investigations which adds to the understanding of the drug's effects.
The 3 steps in post marketing?
1. phase 4 clinical studies
2. adverse reaction reporting
3. comprehensive annual reports are required.
what happens in adverse rxn reporting?
The drug's sponsor is required to rvu and report to the FDA all adverse effects reported to them by all sources such as MDs, postmarketing studies, pharmacists, etc.
In adverse rxn reporting, who reports to who?
The drug's sponsor reports to the FDA
what are orphan dz?
rare dzs or conditions that affects fewer than 200,000 people in USA
what is the problem with drugs and orphan dz?
there is no reasonable expectation that costs of research and development for the dz indicated can be recovered by sales of the product.
What is the purpose of developing IND for orphan drugs?
the purpose is to provide incentives to develop drugs where there is an insufficient market incentive by:

1. giving financial support for clinical trials
2. 7 years of exclusive marketing rights
What 4 variations are there in the drug development process?
1. IND for orphan drugs
2. Treatment IND
3. Abbreviated new drug application
4. Supplemental new drug application
This variation of the drug development process omits nonclinical laboratory studies and clinical studies except those pertaining to bioavailability
abbreviated new drug application which is used for generic drugs
Define supplemental new drug application?
required for certain types of changes like in method of synthesis, formulation, analytical standards, container, manufaturing facilities...etc
Define bioavalibality?
fraction of an administered dose that reaches the systemic circulation
Treatment IND is given for what reason?
for patients with life threatening or severely debilitation illnesses, where no satisfactory alternative exists.
Give examples of orphan dzs?
cystic fibrosis and chronic lymphocytic leukemia
Give examples of life threatening or severely debilitating illnesses that go thru treatment IND
advanced AIDS
Alzheimer's dz
Advanced Parkinson's dz
what is the purpose of treatment IND?
to facilitate getting the drugs to these patients while in clincial trials.
Give an example of treatment IND & explain?
"treatment protocol" where the IND is filed during phase 1 or 2 to allow treatment for the ill patients
The stage of drug development during which the physical & chemical properties of a drug substance are evaluated towards the formulation of a stable, safe, and effective dosage form.
Preformulation
what part of the drug is evaluated during preformulation?
the physical and chemical properties
what is the purpose of preformulation?
to form a stable, safe, and effective dosage form
the manner in which a drug is formulated is crucial for what?
its bioavailability and acceptability by the patient
solution, suspension, tablet, capsule, suppository represents what part of the drug?
dosage form
during preformulation, 4 points are considered by the physical pharmacist?
1. drug is suitable for dosage formulation, for teh desired route of administration
2. drug has sufficient water solubility
3. drug has sufficient lipid solubility
4. drug is sufficiently stable
Name the four drug properties that we are interested in?
1. physical properties
2. dissolution
3. partitioning
4. stability
most drugs are in what state?
solid state
Most solid drugs are what shape?
crystalline
Describe the arrangement of the crystalline form of the solid drug?
has a repeating or periodic arrangement of the individual moleucles that creates teh crystal lattice
T/F: Different molecular arrangements are possible within a solid. A particular arrangement defines the extrnal structure
False: a particular arrangement defines the INTERNAL structure
The internal structure determines what?
the crystal habit
Define habit in a crystal solid drug?
It's the description of the outer appearance of a crystal
How do you get different habits of crystalline form?
Different habits can result from minor changes in the synthetic conditions
Which solid form, crystalline or amorphous will dissolve faster?
amorphous
This type of solid form is without distince crystalline structure?
amorphous form
What is the arrangement of the amorphous form?
has no repeating or periodic arrangement
This form has random molecules with no crystal lattice, looser arrangement and in slight motion
Amorphous form
5 things that are affected by the form of the solid drug?
1. Flow properties
2. mechanical properties
3. bioavailability
4. melting point
5. solid state stability
Give an example of solid state stability
Hygroscopicity which is water uptake unto a solid or liquid and which can change the stability of the drug
Define polymorphism?
capacity for appearing in many forms
Depending on crystallization conditions, some drug solids may have at least ________ different molecular arrangements, each giving distinct ___________ species, or polymorphs
two
crystalline
The different properties of polymorphs?
1. intermolecular interactions
2. solubility
define matastable?
being in an unstable & transient but relatively long lived state of a chemical or physical system
Give an example of a molecule with weak molecular interactions
amorphous solids b/c they're metastable
Which polymorph would be more soluble?
metastable polymorphs more soluble than their more stable counterparts b/c of weaker interactions
A drug that may crystallize as a combo of pure drug and the solvent of crytallization is called what?

Give ex?
solvate

hydrate where water is part of the crystal
changes in hydrate form can affect what?
solubility
How do hydrates affect solubility? explain
hydrates are generally less soluble thatn their anhydrous counterparts.
This is because of the energetics of dissolution (the water and drug are holding on to each other more tightly)
how do hydrates affect bioavailability?
since there is less dissolution of the drug from the water, there is less bioavilability of the drug
Particle size of a solid drug can affect what 3 things?
1. dissolution rate
2. particle settling in suspensions
3. particle flow
The smaller the particle the ________ the dissolution
faster
The smaller the particle the _________ the surface area
greater
How did dissolution rate affect griseofulvin as a drug
micronization reduced the dose needed by 50% b/c the faster the dissolution the great the bioavailability
How did dissolution rate affect nitofurantoin?
crystals of the drug were too small and dissolution was too fast. The result was was nausea.
what are some factors that determine particle flow as a solid drug property?
1. particle cohesiveness
2. particle density
3. particle shape
4. particle size
What are 3 variables affected by particle flow?
1. mixing & pouring
2. speed of processing during manufacture
3. settling and stratification during handling, mixing, and storage
Define stratification?
process of arranging in layers
Particle cohesivenss of particle flow is affected by what 2 things?

how does each work in affecting particle cohesiveness?
electrostatic charge
adsorbed moisture

Electrostatic charge can affect particle interactions
Adsorbed moisture can make the powder sticky
A drug must be in _________ to be absorbed and to cause an _________.
solution
effect
a homeogenous mixture of 2 or more substances
solution
define a solute
the molecular, atomic, or ionic component of a sol'n that is dissolved by the solvent
the component of a sol'n that dissolves the solute
solvent
what are the 4 sol'ns that we need to know & give an example of each
1. gas/liquid carbonated h2o, holothane
2. liquid/liquid miscible liquids (eg alcohol in H2O)
3. solid/solid drug in polymer
4. solid/liquid drug in water
define miscible liquids
liquids that completely dissolve in each other in all proportions
this process involves the relocation of a solute molecule from an environment where it is surrounded by identical molecules into a cavity in a liquid where it becomes surrounded by non-identical molecules, with which it may interact to varying degrees
process of dissolution
each step of the dissolution process involves what?
energy
T/F: the overall dissolution process can be endothermic or exothermic
True
tell the difference btwn endothermic & exothermic
endo: heat is absorbed
exo: heat is given off or evolved
what are the steps in the dissolution of a crystalline solute
1.removal of solute molecule
2. creation of a hole in the SOLVENT
3. placement of SOLUTE in the hole
***** See picture in notes under drug dissolution******
the concentration of a solute in a saturated solution at a certain temperature
solubility
How is solubility expressed?
the # of mL of solvent needed to dissolve 1gm of solute
Relate the solubility of a sol'n to how much solvent is needed for 1 gm of solute
the more mL of solvent needed for each gm of solute, the less soluble the sol'n will be.
ex: <1ml of solvent for 1gm of solute means the sol'n is very soluble whereas >10,000mL of solvent for 1gm of solute means the sol'n is very insoluble
besides # of mL solvent/gm of solute, how else is solubility expressed?
molality
molarity
percentage
4 factors governing the solubility of a solid in a water
1. nature of the solid
2. nature of the solute
3. nature of the solvent
4. Temperature
T/F: metastable polymorph is more soluble than its more stable counterpart
True
the 2 natures of the solid in sol'n w/ water
polymorph
pseudoopolymorph
define pseudopolymorph
anhydrous crystals are generally more soluble than hydrates & therefore have better bioavailibility
give an example of a pseudopolymorph drug
ampicillin
2 things must be considered for a solute's solubility in water
1. molecular structure of the solute
2. ionization state of the solute
what defines the molecular structure of the solute's solubility?
size, shape, and substituent groups can all be important for solubility
what happens in a molecular structure of a solute that has hydrophilic substituents?
most can form H+ bonds with H2O.
the effect of hydrophilic substituents on water solubility can be dramactic. give an ex of this
phenol is 100x more soluble than benzene b/c that alcohol group in phenol increases its solubility dramatically
Again with regard with molecular structure of a solute, how does size and shape compare with the substituent grp in affecting the solubility?
Give ex?
the bulk of a molecule can be more important than the substituent.
ex of this is estradiol where the corticosteroid ring structure & 2 hydroxyl grps have a solubility of 5mcg/ml
__________ __________is one of the most important physiocochemical properties determined during preformulation since it affects both water and lipid solubility
the ionization state of the solute
the ionization state of the solute affects what 4 things?
1. dosage formulation
2. absorption
3. distribution
4. excretion
T/F: most drugs have ionizable groups where they are either weak bases, weak acids, or amphoteric
True
which part of the drug is generally the most water soluble form?
the ionized form
The degree of ionization is a fxn of medium pH and can be measured by ______.
pKa or dissociation constant
define pKa?
it's the pH at which the drug is 50% ionized
be familiar with the henderson hasselbalch equation for acid & base
ACID: pH= pKa + log [A-]/[HA]

BASE: pH=pKa + log [B]/[BH+]
99.9% of the drug will be ionized at 3 pH units below (for a ______) or above (for an ________) the drug's _______.
base
acid
pKa
relate ionization of a drug with its pH?
for a weak acid drug, it will be unionized in acidic pH. As the pH increases, it will become ionized & therefore its solubility will increase
be familiar with modified henderson hasselbalch equations for acid & base?J
ACID: pH=pKa + log [S-So]/[So]

BASE: pH = pKa + log [So]/ [S-So]
what does S & So stand for?
S is the overall solubility of the drug
So is the solubility of the unionized form (S= So+[ ] of ionized form)
3 things that pertain to the nature of the solvent in affecting solubility?
1. pH of the solvent
2. solvent polarity
3. presence of solubilizing agents
how does the solvent pH affect the solubility
it affects the drug's ionization state
how does the solvent polarity affect the solubility?
changing the solvent's polarity to better match the drug's polarity is often used as a method to improve solubility (like dissolves like)
how is the polarity of a solvent measured?
it's measured by its dielectric constant, w/c is the ability of a solvent to separate oppositely charged ions.
name 5 types of solvents
1. H2O
2. glycerin
3. propylene glycol
4. ethanol
5. PEG 400
Some solvents can also be used as __________ to help solublize drugs
co-solvents
Chloroform is a type of solvent. why is it not a good co-solvent?
b/c it's not miscible in H2O
give a couple of examples of solubilizing agents?
surfactants, cyclodextrins
What is the fxn of co-solvents? what is the exception?
When you use co-solvents, you want to decrease the polarity of the solvent.
The exception would be in inhalers where you want to INCREASE polarity
How does temperature affect solubility?
since dissolution is usually an endothermic process where energy is needed to break bonds in a solid, greater heat (higher temps) would generally increase the solubility of the solid
what are some common methods for altering the solubility? (4)
1. altering the solid
2. altering the solute
3. altering the solvent
4. adding solubilizing agents
how can u alter the solubility of a solid
1. create the desired solid form
2. form a solid sol'n or despersion
how can u improve the drug's solubility by altering the solid?
a drug can be dissolved or finely dispersed in a water soluble solid. This can be done by separating the drug where all the crystals are separated and then mixing them with something hydrophilic
what 2 approaches can alter the solute?
creating salts
creating prodrugs
of all the ways of affecting solubility, what is a very common form?
salt formation where you alter the solute; 50% of drugs are done this way
which types of drugs can theoretially be transformed into a salt?
every drug that has acid or base properties
what is the general process of salt formation
an ionized drug forms an ionic interaction with a counterion in a solvent containing a strong acid or base
in salt formation, what happens to salt?
the salt gets crystalized from the solvent
how does salt formation affect solubility?
it can either decrease or increase drug's solubility
what are 2 reasons one would use salt formation to decrease drug solubility?
stability
taste
how does stability of drug relate to a decrease in solubility when salt formation occurs?
why is this?
a solid drug (ie in suspension) is more chemically stable than drug molecules in solution. this is b/c chemical breakdown generally occurs to drug molecules in sol'n
how does salt formation to decrease drug solubility affect taste?
why is this?
a decrease in drug solubility can help mask a bad taste; this is b/c drugs must be in sol'n (therefore soluble) in order to interact w/ taste bud chemoR
give an ex of a drug salt that decreases drug solubility & increase stability
propoxyphene napsylate is less solube than prooxyphene hydrochloride & thus more stable under aqueous conditions of manufacture
give an ex of a drug salt that decreases drug solubility & mask taste?
same drug as the one for stability: proposyphene napsylate is less bitter than propoxyphene hydrochloride b/c its less soluble
2 features of why one would want salt formation of a drug to increase solubility
1. salt formation can improve drug's water solubility significantly

2. a very low intrinsic solubility (less than 1mg/ml) indicates the need for a salt form for the drug.
how does salt formation of a drug increase solubility?
b/c of the greater degree of ionic dissociation of salt molecules, there is a greater degree of interaction with water molecules
give 2 ex of drugs where their salt formation is more soluble & in what form are they used?
1. phenytoin is very slightly soluble in water therefore used an oral suspension while phenytoin sodium is more soluble in water & therefore used for injection

2. ampicillin is slightly soluble in water & is used as an oral suspension while ampicillin sodium is very soluble in water & therefore used for injection
T/F: the solubility of the ionized drug needs salt formation b/c it's less soluble than it's unionized form
False: the unionized drug needs a salt formation b/c it's less soluble than its ionized form
what are some examples of salts for basic drugs?
the following anions:
1. hydrochloride
2. sulfate
3. tartrate
4. maleate
5. citrate
6. mesylate
7. succinate
what are the salts used for acidic drugs?
cationic
1. sodium
2. potassium
3. calcium
give 4 drug names that use the hydrochloride salt to form a salt drug?
1. pseudoephedrine hydrochloride
2. diphenhydramine "
3. propoxyphene "
4. clindamycin "
give a drug name that uses the sulfate salt to form a salt drug?
terbutaline sulfate
name a drug that uses the salt tartrate to form a salt drug?
metoprolol tartrate
maleate salt can form what drug?
chlorpheniramine maleate
citrate salt can form what 2 drugs?
tamoxifen citrate & fntanyl citrate
mesylate can form what drug?
doxazosin mesylate
succinate can form what drug?
metoprolol succinate
sodium can form what 2 drugs?
phenytoin sodium & ganciclovir sodium
potassium can form what drug?
clavulanate potassium
calcium can form what drug?
leucovorin calcium
what are 7 properties of pharmaceutical salts we need to look at?
1. pH of sol'ns of drug salts
2. taste (palatability)
3. toxicity
4. lower % of active drug
5. hygroscopicity
6. polymorphism
7. corrosiveness
relate pH of salt to its formed sol'n?
--salts prepared from weak bases yield more acidic solutions
--salts prepared from weak acids yield more alkaline solutions
give 2 ex of drug salts with their pH?
1. minocycline HCl IV sol'n has pH of 2-2.8
2. ganciclovir Na IV sol'n has pH of 11.
give an ex of salt drug & taste effect?
potassium salt drugs tend to have a metallic taste
be familiar with drug salt toxicity and % activity of a drug?
--some salts are more toxic than others
-- sometimes more of a drug is needed when combined with a salt b/c less of the drug is active when it's combined with a salt
a synthetic drivative of a drug that is transformed in vivo to liberate an active drug molecule
prodrug
a prodrug approach is used to do what?
alter the solute (remember a solute can be altered by adding a salt or by using a prodrug)
how are most prodrugs formed?
they are created with an ester linkage that is cleaved in vivo with the help of esterases
what are some pharmacolgical purposes for synthesizing a prodrug?
1. biostability/ prolonged action
2. targeting (targeting brain-- lipophilic to cross BBB)
3. solubility
what are some pharmaceutical purposes for synthesizing a prodrug?
1. taste
2. stability
3. solubility
give an example of how a drug decreases solubility?
becomes more lipophilic which could improve absorption, distribution and other drug properties
For erythromycin, its prodrug EES is given to decrease solubility, tell 3 reasons why?
1. better tasting- drug is in suspension
2. bypasses gastric degradation-- erythromycin degrades rapidly in acidic conditions
3. better absorption--more lipohilic
1 A prodrug can also increase solubility, what would be a good reason for this?
2 How is it most commonly done?
3 give examples of drugs
1. reason: to create a more water soluble prodrug for inje.ction
2. a phosphate group is added to create a phosphate ester derivative. in vivo phosphate ester linkages are cleaved by phophatases.
3. clindamycin phophate, fosphenytoin sodium, dexamethasone sodium phosphate
T/F: a phosphate ester is the same as a phosphate salt
False: it is not the same.
Give an ex of a phosphate salt
disopyramide phosphate
For some of the more poorly water soluble drugs, the drug's __________ rate will be the most important determinant of its oral _____________.
dissolution
bioavailability
What can be the rate limiting step for absorption for a poorly water soluble drug
it would be the drug's dissolution rate
The dissolution of a solid in a liquid basically occurs in 2 consecutive stages, what are they?
1. an interfacial rxn that liberates solute molecules from the solid phase
2. diffusion of the liberated molecules away from the solid into the bulk liquid
what is the static boundary layer?
the area which is very highly [ ] w/ drugs & also known as the stagnant layer.
The overall rate of mass transport of the solute from the solid in a liquid will be what?
it will be determined by the slowest stage, which is usually diffusion through a static boundary layer
what are the factors affecting dissolution rates (Noyes-Whitney)
A: total surface area of the undissolved solid
Cs: drug solubility
C: concentration of solute in the bulk liquid
k: dissolution rate constant
How does "A" or the total SA of the undissolved solid affect dissolution rate
--size of particles: as size decreases, SA increases
--dispersibility of teh powder in the dissolution medium: if the particles clump, they will inhibit H2O acces & therefore dissolution will be slower
How does "C" or [ ] of solute in the bulk liquid affect dissolution?
volume of the dissolution medium: increase volume, decrease [ ] for ex, taking med w/ a glass of water

any process that removes dissolved solute from the dissolution medium; this is absorption of the drug
how does "k" of dissolution rate constant affect dissolution?
thickness of the stagnant boundary layer w/c can be decreased by agitation or shaking

diffusion coefficient of solute in the dissolution medium which decreases by an increase in liquid viscosity so the more viscous the sol'n the lower the dissolution
**** if you know something won't dissolve in syrup, dissolve it in H2O first then mix it w/ more viscous sol'n
define partition:
to move as a molecule from 1 phase to another
ex: between polar and nonpolar phases
what is the importance of partitioning btn polar & nonpolar phases?
1. traversing the lipid bylayer
2. other relevant partitioning phenomena
drugs that traverse a partiular membrane by passive diffusion must __________ into it.
partition
The drug must have what properties to cross membrane?
it should have both some aqueous and lipid solubility
besides the lipid bylayer, name 5 other partitions that drugs could go through?
1. into fat
2. into microorganisms
3. into plastic
4. btwn oil & water: seen in emulsions
5. btwn chromatographic phases: for drug analysis or separating drugs analytically
Fxn of partition coefficient?
during preformulation, the ability of a drug to partition btwn 2 poorly miscible (polar & nonpolar) phases is determined by measuring the partition coefficient of the drug
If a drug is more lipophilic, how would it affect P (partition coefficient)
it would increase it
what is the partition coefficient equation?
P= [Co]/[Cw]
Co is the drug's [ ] in an organic (nonpolar) phase
Cw: drug's [ ] in the aqueous (polar) phase.
What is one of the most important methods for determining a drug's lipophilicity?
measuring a drug's partition coefficient
what factors determine P?
choice of organic phase
effect of ionization
which organic solvent best mimics membrane properties?
octanol which is the most common organic solvent used
what are the 3 oragnic solvents mentioned that determine P?
octanol: best mimics membrane properties
butanol: mimics buccal membrane
chloroform: mimics BBB
For ionizable drugs, what has the higher P? what does say about polarity of the drug?
unionized has higher P which means the drug is less polar
where is P generally studied?
it's studied over a range of pHs similar to what is found in the GI tract, as a means of estimating where absorption of the drug might be greatest.
where might be the absorption of the drug be the greatest?
it's more likely absorbed in regions w/ pHs favoring the unionized form
what are some factors to consider for GI absorption?
1.ionized form of the drug can partition in some cases. an ex of this is very lipophilic corticosteroids
2. ionization is an equilbrium process: when unionized drug is absorbed, some remaining ionized drug becomes unionized to maintain the equilibrium
3. the SA of the absorption site is often more important. SA is greatest in SI-far greater than in the stomach therefore most drugs and nutrients get absorbed there.
the extent to which a drug product retains, within specified limits, and throughout its period of storage & use the same properties & characteristics that it possessed at the time of its manufacture
drug stability
what is the drug product
finished dosage form that contains an active drug + inactive ingredient
what is the shelf life?
period btwn date of manufacture of a drug product & the exp date.
what are the properties to be considered relevant to stability of a drug?
1. chemical properties
2. physical properties
3. microbiolgical properties
what are the drug's chemical properties & what affect can changes in these properties do?
it's the chemical integrity where the drug must stay intact.
if there is changes in these properties, therapeutics can be affected, toxicity, and organoleptic properties can also be affected
what are some examples where changes in the chemical property of the drug led to toxicity?
1. tetracycline can epimeriz and dehydrate to potentially fatal nephrotoxic cpd
2. penicillins can decompose in acidic sol'ns to possibly allergenic cpds.
one of 2 moleucles that differ only in the spatial arrangement around a single carbon atom
epimerize
what causes fanconi's syndrome
nephrotoxicity from tetracycline epimerization. rare syndrome
what does organoleptic properties mean?
susceptible to sensory impressions such as sight, sound, & taste which is a big one
give 2 ex of organoleptic properties?
1. epinepherine can oxidize to a therapeutically active, but intensely colored product
2. erythromycin can dehydrate to a therapeutically active, but worse tasting cpd.
how do physical properties affect stability of a drug?
this is also critical b/c crystal structure changes & leads to polymorphism
changes in physical structures can affect what?
therapeutics & organoleptic properties
ex of therapeutics that can be affected with physical properties changes
blood levels can change from dissolution changes
ex of organoleptic properties that can be affected with physical properties changes?
dissolution in suspensions where drug in sol'n elicits taste
what are the microbioligical properties that can affect stability
microbial invasion of a drug product is consdiered a stability probelm which can be unappealing and even deadly
what are the 6 mechanisms of chemical degradation?
1. hydrolysis
2. oxidation
3. photolysis
4. dehydration
5. isomerization
6. polymerization
what is the most common mechanisms of chemical degradation?
hydrolysis & also oxidation
define hydrolysis
cleavage of a molecule by reaction w/ H2O
what are some ex of functional groups susceptible to hydrolytic cleavage?
1. esters like in aspirin, procaine, cocaine, & physostigimine
2. amides like in dibucain & chloramphenicol
3. lactams like in penicillins, cephalosporins, chlordiazepoxide
4. lactones like in pilocarpine, spironolactone
****know general structure of each class see notes under chemical degradation mechanisms
define oxidation
refers to the removal of electrons
a type of oxidation that involves the uncatalyzed oxidation of a substrate by molecular O2
autoxidation
this type of oxidation is a slow process
autoxidation b/c O2 is not very reactive
molecular oxygen is a ________ __________ & has 2 unpaired electrons with parallel spins
triplet diradical
what are some other oxygen species that might be more reactive than molecular oxygen
superoxide O2-
hydrogen peroxide H2O2
hyroxyl radical OH
which reactive O2 is formed with the aid of a metal catalyst
Hyroxyl radical which is very reactive
free radical mechanism of oxidation involves what 3 things?
1. initiation
2 propagation
3. termination
This formation of substrate free radicals necessary for the propagation of the chain?
initiation
what is the formula for initiation?
RH + In -----> R + InH
the 3 features of initiation
1. involves hydrogen abstraction
2. can occur through autooxidation by molecular O2 or w/ other chain initiating radical
3. chain initiating radicals are generated by the action of light, heat transition metals (iron & copper) or reactive cpds (peroxides)
what happens in propagation & what is the rxn
free radicals grow in #
R + O-O ------> R-O-O
R-O-O + RH -------> R-O-O-H + R
free radicals combine to form chemically inert products
termination of free radicals
what are the rxns for termination?
2R-O-O ------> inert products
R + R-O-O -----> inert products
2R -----> inert products
what are some functional groups that are susceptible to oxidation & drug ex of each
1. phenols -----> morphine
2. catechols ------> epinephrine
3. thols
4. polyunsaturated hydrocarbons
this type of chemical degradation is due to chemicl breakdown caused by lite usually by complex mechanisms
photolysis
what is the most common photolytic mechanism
photoxidation
define potoxidation
include certain hydrolysis + dehydration rxns
Give example of a drug that can undrgo photolysis
sodium nitroprusside in aqueous sol'n which is used in IV infusions. Shelf life in the dark is 1 yr. Once exposed to light, its shelf life becomes 4 hours!
a chemical degradation where a water molecule is eliminated from the chemical structure?
dehydration
give an ex of dehydration?
erythromycin where deydration occurs under acidic conditions to yield a less active product
what happens in a physical dehydration degradation?
removal of water from a crystal hydrate (this is a type of polymorphism). This process can cause a change in solubility
give an ex of some drugs that can undergo physical dehydration degradation?
theophyllin hydrate & ampicillin trihydrate
conversion of a drug to its optical or geometric isomer
isomerization
define optical
isomerism about an asymmetric carbon atom
synonym for cis-trans isomerism
geometric isomer
give an example of a drug that undergoes isomerization?
amphotercin B
what is the significance of isomerization?
1. 50-60% of drugs are chiral cpds
2. enantiomers can have different bilogical properties w/ ex of albuterol vs levlbuterol
3. drugs marketed as pure enantiomer may racemize w/ ex of thalidomide which undergoes base catalyzed raceization in aqueous media
1 of a pair of molecular entities which are mirror images of each other & non super impossable
enantiomers
the partial conversion of 1 enantiomer into another
racemize
geometric isomers can differ in their physiochemical properties, what are they?
solubility
stability
compatibility w/ other cpds
this type of chemical degradation has 2 or more drug molecules that combine to form a complex molecule
polymerization
give an ex of polymerization? side effect
concentrated solutions of aminopenicillins
ampicillin & amoxicillin can form dimers, w/c are thought to be allergenic