1/33
Looks like no tags are added yet.
Name | Mastery | Learn | Test | Matching | Spaced |
---|
No study sessions yet.
Which specific toxicities are mechanistic studies used to determine if potential drugs might cause?
membrane integrity
mitochondrial toxicity (MitoTox)
oxidative stress
inflammation
How do mutation assays for genotoxicity work?
Ames test first → secondary follow-up tests performed in mammalian cells as needed
HPRT assay
Mouse lymphoma TK assay (MLA)
What are the two “in vivo” toxicity assays?
acute and chronic toxicity
acute toxicity
single dose studies over a broad concentration range to identify dose-limiting toxicities and specific organ toxicities
maximum tolerated dose (MTD)
highest dose that doesn’t cause unacceptable side effects
chronic toxicity
lower dose levels tested over longer period of time; usually daily administration for 14-28 days but dose and length can depend
For both acute and chronic tests:
animals are sacrificed and their tissues are studied for damage; often involves at least two different species (rodent + non-rodent); tested through at least two routes of administration
lethal dose 50 (LD50)
dose required to kill 50% of animal group
effective dose 50 (ED50)
dose required to produce desired effect in 50% of animals
therapeutic index
calculated as the ratio of LD50 divided by ED50
safety pharmacology
series of “in vitro” and “in vivo” studies designed to identify any undesired effects of potential drug on key organs
What tests would be run on the cardiovascular system for safety pharmacology?
blood pressure
heart rate
ECGs
ventricular vs pulmonary pressure
echocardiography
cardiac ion channel assessments
What tests would be run on the respiratory system for safety pharmacology?
respiratory rate
hemoglobin oxygen saturation
tidal and minute volumes
What tests would be run on the CNS for safety pharmacology?
functional observation battery
modified Irwin screen
body temperature
large animal neurological evaluations
Why do we want to reduce reliance on animal models for data?
don’t always do the best job of predicting human efficacy/toxicity
expensive and time consuming
can’t replicate individual genetics or specific human disease parameters
ethical concerns
organ-on-a-chip
microfluidic devices lined with living human cells that simulate the structure and function of human organs
single or multiple organs
specific stimuli to determine effects of certain conditions
Why do we use organ-on-a-chip?
to understand metabolism and toxicity (liver), cardiotoxicity, and lung absorption/toxicity
3D cell culture models
spheroids or organoids; readily mimics natural environment of an organ and can be used to test both efficacy and toxicity
physiologically based pharmacokinetic (PBPK) models
math-based models systems designed to predict ADME/PK properties of a small molecule
utilize known info about human body and parameters of drug to predict ADME/PK
chemistry, manufacturing and controls (CMC)
series of studies to ensure that final drug product maintains the same structure, quality, and consistency across each batch prepared prior to dosing humans
API analytical characterization studies
clearly demonstrate that the API is the correct structure and identify any impurities during the manufacturing process
analysis of formulated dosage form
identifies what the components and stability is of the final dosage form
manufacturing process and development and validation
ensures that the process complies with good manufacturing practices (GMP), is scalable, and will consistently produce the drug
API structural characterization
full suite of analytical techniques (NMR/IR/MS) to clearly demonstrate the structure of the API
impurity profile
list of any organic/inorganic impurities generated from either the API synthesis or generated from the formulated drug
synthetic/process route
helps identify potential impurities while also ensuring safety and reproducibility
solid form ID
process used and final identification of API solid form for dosing
Name the solid forms that an API can make:
crystals (polymorphs)
non-crystals (amorphous solids)
salts (charged)
hydrates
formulated drug analysis
provides full characterization of the API, excipients, contain-closure system, and stability studies
container-closure system
combo of packaging components for final drug; primary (vials/bottles) and secondary (boxes/labels)
real-time stability
how stable the formulated drug is over time for the recommended shelf life under ideal conditions
accelerated stability
elevate normal conditions to determine potential stability issues (temperature/humidity)
forced degradation
a measure of stability and byproducts associated with harsh conditions (high heat/light/pH)
good manufacturing practices (GMP)
specific parameters enforced by FDA to guarantee safety/quality of drugs through consistent production across different batches