• 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

1/45

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;

45 Cards in this Set

  • Front
  • Back
Title: Baby Elephant
Artist: Dave White
Pharmacology
study of the effect of drugs on the function of living systems
Toxicology
study of the effect of poisons on the function of living systems
Chemical agents that cause toxicity
1. drugs
2. insecticides/herbicides
3. plant toxins
4. animal toxins
5. chemical weapons
6. radioactive elements
Paracelsus
-'grandfather of toxicology'
-"all things are poison and nothing is w/out poison, only the dose permits something not to be poisonous"
-the dose makes the poison!
Adverse Drug Reactions (ADRs)
-noxious or unintended responses occurring at therapeutic doses (bad reaction to normal dose)
~5% of all acute hospital admissions
-Type A vs. Type B
Type A ADRs
effects:
-related to known pharmacology; undesirable
-common, dose-related
-predictable
examples:
-hemorrhage w/anticoagulants
-respiratory depression w/opioids
-sedation w/anxiolytic & older antihistamine
drugs
Type B ADRs
effects:
-unrelated to known pharmacology
-rare
-unpredictable
-often idiosyncratic
examples:
-anaphylaxis w/penicillin
-allergic liver damage by halothane
-bone marrow suppression by chloramphenicol
-individual allergy/genetic basis
Pharmacokinetics
-the effects of the body of poison (related to ADME)
-with this info, it's possible to predict concentration
of toxin that reaches site of injury & resulting
damage
ADME - Absorption
ingestion: mercury & dioxin in fish
pesticides in produce
salmonella (dairy), botulinum (meat) toxins

inhalation: asbestos, nerve gases
ADME - Distribution
via blood stream
ADME - Metabolism
-Phase 1 by cytochrome p450 (oxidation, reduction,
hydrolysis - make molecules more polar)
-Phase 2 conjugation to allow excretion in urine &
bile
-Detoxification
-Toxification
Detoxification
compound rendered less toxic
Toxification
relatively inert compound converted into toxin
ADME - Excretion
Toxins not excreted may be stored in:
-bone (eg. lead)
-fat (eg. DDE a metabolite of pesticide DDT
dichlorodiphenyl trichloroenthane)
-the toxin may be released slowly into body
Molecular Mechanisms of Toxicology
1. Allergic responses
2. Receptor, ion channel, & enzyme-mediated toxicity
3. Biochemical pathways
4. Organ-directed toxicity --> liver & kidney
5. Mutagenesis & carcinogenesis
6. Teratogens
Molecular Mechanisms of Toxicology: Allergic Responses
-common form of ADR, w/different time course to pharmacological effects
-4 basic clinical syndromes - types I, II, III, IV
-TYPE I hypersensitivity reactions
-can trigger anaphylactic shock!
-TYPE II antibody-mediated cytotoxic hypersensitivity
-Type III immune complex-mediated hypersensitivity
-involve haematological reactions (those
pertaining to bl. cells & bl. forming organs)
-Type IV delayed-type hypersensitivity
Molecular Mechanisms of Toxicology: Receptor, ion channel & enzyme-mediated toxicity
-4 major superfamilies' of receptors
-enzymes: metabolic & catabolic pathways
-carriers: uptake/transport systems
-others: proteins involved in vesicle release
-Animal toxins block ion-conduction
-a-bungarotoxin on nicotinic acetylcholine
receptor (nAChR)
-bind irreversibly & competitively to nicotinic
acetylcholine receptor found at neuromuscular
junction - paralysis, resp. failure, death
-Voltage-gated K+ channels block by dendrotoxins
-prolong duration of action potentials &
increase acetylcholine release at neuro-
muscular junction - muscle hyperexcitability,
& convulsive symptoms
4 Major Superfamilies' of Receptors
1. ligand-gated ion channels
-ionotropic receptors
-voltage-gated ion channels
2. GPCRs - G protein coupled receptors
(metabotropic receptors)
3. enzyme-linked receptors (tyrosine kinase activity)
4. nuclear receptors (regulate gene transcription)
Molecular Mechanisms of Toxicology: Biochemical Pathways
-cyanide inhibits mitochondrial cytochrome c oxidase to prevent cellular respiration
-carbon monoxide: displaces oxygen from hemoglobin causing hypoxia
Molecular Mechanisms of Toxicology: organ-directed toxicity
Hepatic: -necrosis --> acetaminophen poisoning
-hepatic inflammation (hepatitis):
halothane covalently binds to liver
proteins to trigger autoimmune reaction
-chronic liver damage (cirrhosis)
-long-term ethanol abuse causes cellular
toxicity & inflammation
Nephrotoxicity:
-changes in glomerular filtration rate
(GFR) --> largely due to bl. flow altering drugs
(NSAIDs)
-allergic nephritis --> reaction to NSAIDs &
antibiotics
-chronic nephritis --> long-term NSAID &
acetaminophen use
Molecular Mechanisms of Toxicology: Mutagenesis & Carcinogenesis
-mutagens cause changes to cell DNA that are
passed on when cell divides - if this produces a
neoplastic cell the agent is termed a carcinogen
-2 major classes of gene involved in carcinogenesis
-proto-oncogenes
-tumor-suppressor genes
Proto-oncogenes
promote cell cycle progression
-eg. constitutive activity of growth factor tyrosine-
kinase receptors can cause neoplastic
transformation
Tumor-suppressor genes
inhibit cell cycle progression
-mutations in tumor suppression gene product
p53 (prevalent in smokers)
-p53 = cellular policeman
Molecular Mechanisms of Toxicology: Teratogens
-Teratogenesis: creation of birth defects during fetal
development
-Teratogens: substances that induce birth defects
Pre-Clinical Drug Testing
-in vitro cytogenetic evaluation of chromosome
damage in response to drug
-carcinogenicity --> chronic drug dosing - look for
tumors
-repro (teratogenicity) testing:
preg. females from one rodent species & one
non-rodent species does
w/drug at diff. organogenesis stages - look for
birth defects
-preliminary toxicity testing
-therapeutic index
Preliminary Toxicity Testing
-max. non-toxic dose (given for 28 days to 2 species)
-animals examined post-mortem & tissue damage
accessed
-lethal dose LD50 - dose of drug which kills 50% of
treated animals w/in specified time
-NOAEL
-LOAEL
NOAEL
No Observed Adverse Effects Level
-highest concentration that does not have toxic
response
-determine NOAEL & convert to 'Human Equivalent
Dose"
-adjust for anticipated exposure in humans
-adjust for inter-species difference in affinity &
potency
-apply >10 fold safety factor
LOAEL
Lowest Observed Adverse Effects Level
-lowest concentration that produces a toxic response
Therapeutic Index
ratio of dose of drug the produces unwanted (toxic) effect to that producing a wanted (therapeutic) effect
Why do we need Toxicity Testing?
-Elixir Sulfanilamide disaster of 1937 - most
consequential mass poisonings of 20th century
-sulfanilamide diluted in diethylene glycol to give a
red Elixir Sulfanilamide --> over 100 patients died
-under existing drug regulations, toxicity testing was
not required
-in reaction: US Congress passed 1938 Federal Food,
Drug, & Cosmetics Act - required proof of safety
before releasing new drugs
Clinical Trials
1. NIH
2. study participant recruitment:
3. ethical norms of clinical trials
4. protection of human subjects
5. 9 keys points
6. vulnerable populations
Clinical Trials: NIH definiton
prospective biomedical or behavioral research study of human subjects that is designed to answer specific questions about biomedical or behavioral interventions
-drugs, treatments, devices, & new ways of using
the three
Clinical Trials: Study Participant Recruitment
1. identify eligible participants
2. explain study
3. provide informed consent
4. reassess eligibility
5. assign to groups (treatment vs. placebo)
6. participants should be told:
-may have side effects (adverse effects)
-time commitment
-benefits & risks
-may withdraw at any time
-enrollment 100% voluntary
Clinical Trials: Ethical Norms
sound study designs take in account
-randomization of sharing of risks
-proper use of placebo
-processes to monitor safety of rx/tx
-competent investigators
-informed consent
-equitable selection of participants
-compensation for study related injuries
Clinical Trials: Protection of Human Subjects
-rely on integrity of investigator but outside groups
also have oversight
-participants' rights protected by Institutional Review
Boards (IRBs)
-IRB = "any board, committee, or other group
formally designated by an institution to review,
approve the initiation of, and conduct periodic
review or biomedical research involving human
subjects"
Clinical Trials: Key Points
1. voluntary informed consent
2. experiment must be for good of society & results
not obtainable by other means
3. experiment based upon prior animal studies
4. physical & mental suffering & injury avoided
5. no expectation of death/disabling injury occurring
6. risk vs. benefit
7. protect subjects against injury, disability, death
8. only scientifically qualified persons involved
9. subject can terminate his/her involvement
Why/Why Not Participate In Study?
-Participate: give back to society, exhausted all other
treatments, health care services, payment &
incentives, support
-Not participate: mistrust of studies, don't want to
be 'guinea pig', do not meet criteria, cannot give up
time for study visits, barriers (language, distance)
Stages of Drug Development: Phase I
-tests new biomedical intervention in small group of people (20-80) for first time to evaluate safety
-dose range
-side effects in humans
Stages of Drug Development: Phase 2
-studies biomedical or behavioral intervention in larger group of people (several hundred) to determine efficacy and further evaluate safety
Stages of Drug Development: Phase 3
-investigates the efficacy of the biomedical or behavioral intervention in large groups of humans (100's to 1,000's)
-comparing intervention to other standard or
experimental interventions
-monitor adverse effects
-collect info that will allow intervention to be used
safely
Stages of Drug Development: Phase 4
-studies are conducted after intervention has been
marketed
-designed to monitor effectiveness of approves
intervention in the general population and to
collect information about any adverse effects
associated with widespread use
TGN1412 Disaster
highlighted need for accurate toxicity testing:
-TGN1412 is monoclonal antibody (MAB) designed to
bind CD28 protein to activate leukocytes
-TGN1412 could fight leukemia by triggering cytokine
release
-animal studies of TGN1412 indicated no toxicity
-6 volunteers were given 1:500 dilutions of doses
used in animal studies at 30 min. intervals according
to agreed protocols
-w/in minutes of 6th volunteer receiving dose,
serious side effects occurred: severe headache,
fever & pain --> brief coma, kidney failure, head
swelling
Potential Flaws in TGN1412 Study
-lack of biological knowledge (of how CD28 works)
-use of healthy volunteers w/intact immune
response could trigger a 'cytokine storm'
-TGN1412 works differently between species
-dose regime too short (given too frequently)
-testing should have been staggered over several
days
-blister test: expose small amount of skin to drug to
check adverse reaction prior to whole body
exposure
Summary: Treatment & Prevention of Toxicity
1. preclinical toxicity testing vital part of drug
development
2. new compounds must be assessed in particular
for mutagenic, carcinogenic & teratogenic potential
3. preliminary toxicity testing typically uses LD50,
NOAEL, & LOAEL values
4. LD50 experiments are not perfect
5 Prevention of toxicity is based on knowledge of
molecular mechanisms of toxin action