• 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

1/81

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;

81 Cards in this Set

  • Front
  • Back
Origin of HIVs
- cross-species transmission on SIV/HIV genomic sequence comparison
- HIV-1 from chimpanzees (SIVcpz)
- HIV-2 from sooty mangabeys (SIVsm)
HIV-1 genetic subtypes
- aka clades
- Groups M, N, O, P represent 4 independent primate-human transmission events
- Group M further classified into clades (A, B, C...); West-central Africa
- Group N: Cameroon
- Group O: West-central Africa
- Group P: Cameroon (?)
Nucleocapsid contains
- 2 copies of ssRNA - each strand a result of provirus
- viral enzymes (proteins that facilitate chemical reactions) - reverse transcriptase, integrase, protease
HIV virus classification
- retrovirus
- lentivirus subfamily
"Simple" retrovirus genome
- 7000-11000 ribonucleotides long
- encodes 3-4 viral genes: gag, pro, pol, env
Gag gene
- encodes capsid proteins
Pro gene
- encodes protease
Pol gene
- encodes reverse transcriptase and integrase
Env gene
- encodes envelope (spike) proteins
LTR (Long Terminal Repeat)
- proviral sequence created by integration of provirus
- 5' LTR contains promoter region where cellular RNA polymerase binds to begin transcription of viral RNA; 3' LTR contains transcription termination sequences
Virion structure
- enveloped virus
- capsid [p24]
- matrix [p17]
- spikes [gp120 + gp41]
HIV attachment
- host cells must display 2 receptors: CD4 molecule + one of 2 chemokine receptors (CCR5 or CXCR4)
1. gp120 initiates attachment to CD4 receptor
2. binding to chemokineR changes shape of gp120; reveals gp41
- gp41 inserts into cell membrane, triggers membrane fusion
Chemokine receptors
- small proteins synthesized and release by certain cells that communicate/activate other immune system cells
Genetic mutation in CCR5 coreceptor
- 1996: CCR5 coreceptor identified, CCR5 deletion mutant identified [CCR5Δ32] - Koup & Landau found mutant CCR5 allele in 2 homosexual, high-risk HIV- men
- genetic testing revealed mutation in Caucasian Americans (spec., Western European origin); <1% homozygous [both alleles mutated], ~20% heterozygous [one mutant allele, one wild-type (i.e., functional) allele]
Allele
- variant form of a gene
Mutation
- change in nucleotide sequence within genome
- change in the code, deletion of nucleotide(s), insertion of new nucleotide(s)
- can occur spontaneously (no outside influence)
- can be 'induced' by influences outside the cell (chemicals, radiation)
Effect of mutation
- no effect
- deleterious effect
- advantageous effect
Potential effect of mutation on protein
- change in DNA nucleotide sequence within a gene encoding a protein may:
- have no effect on amino acid sequence
- result in a different amino acid (may or may not change protein function)
- result in premature termination of protein synthesis which likely will produce a non-functional protein
Cellular targes for HIV infection
- T-helper cells: CD4 and CCR5 (or CXCR4)
- Macrophage: CD4, CCR5
Inside an HIV-infected cell
- after partial uncoating, RT (carried inside the infecting virus capsid) begins synthesizing a dsDNA copy of HIV genome; process occurs in pre-integration complex (PIC); RT does not 'proofread'; RT can 'switch' between template RNA strands
- dsDNA genome transported across nuclear membrane in a complex containing viral integrase
- integration of proviral DNA by action fo viral integrase & cellular DNA repair enzymes; IN chemically 'breaks' bond in each of the strands of chromosomal DNA; positions viral dsDNA into chromosome break; cellular DNA enzymes 'repair' the break, restoring integrity of host cell chromosome
- expression of proviral DNA by cellular RNA polymerase; host cell RNA polymerase binds to promoter region in proviral LTR; RNA polymerases do not 'proofread'; HIV-1 genome encodes 15 viral proteins
Structural genes
- gag (capsid & matrix proteins)
- pol (RT, RNase, IN, protease)
- env (gp120, gp41)
Regulatory genes
- tat (transcriptional activator)
- rev (nuclear export of viral RNAs)
Accessory genes
- vif, vpr, vpu, nef
-Static drugs
- stop replication
-Cidal drugs
- kill microbe
Chemoprophylaxis
- attempt to prevent disease
- administer pre-exposure
Chemotherapy
- attempt to treat disease
- administer after exposure (most often, symptomatic patient)
Reverse transcriptase
- copies ssRNA into strand of DNA, then replicates DNA into its complement (reverse transcription)
Integrase
- breaks chemical bonds that makes up strands of chromosomes
Protease
- left over from when virus was released from its previous host cell
Unique features of retrovirus replication
- only way to kill viral DNA in the cell is to kill the cell itself
- viral info can be in a cell but not active
- cellular enzyme copies provirus DNA
Tat
- regulatory gene
- transcriptional activator
Rev
- regulatory gene
- nuclear export of viral RNAs
Accessory genes
- vif, vpr, vpu, nef
From nonproductive to productive infection
- activation of T-cell triggers HIV replications
Synthetic drugs
- manufactured from individual chemicals (chemists study reactions, components)
Antibiotics
- chemical compounds made by living microorganisms (usually by soil microorganisms)
Semi-synthetic antibiotics
- comes from naturally occurring compounds from microorganisms at its base, like antibiotics
- these naturally-occurring compounds are altered in a lab
Broad spectrum
- used to treat a variety of infections
- usually antibiotics
Narrow spectrum
- used to treat specific disease
Selective toxicity
- kill the pathogen, not the host
- least amount of damage to diseased person, most amount of damage to pathogen
In vivo drug stability
- how stable is the drug in the body
In vivo drug distribution
- where does the drug go in the body
- can it go where it needs to go
Drug interactions
- with other prescription drugs
- with OTC drugs
- with herbal ("natural") drugs
Antagonism
- one drug nullifies another's effect
Synergism
- two drugs work better together than separately
Greatest selectivity found in antibacterial drugs
- easiest to see differences between prokaryotic cells and human cells
- can stop synthesis of peptidoglycan/repairing cell wall (beta-lactam antibiotics - penicillins & vancomycin)
- can target assembly of ribosomes - different ribosomes from humans (have 50S & 30S; humans have 60S & 40S) - chloramphenicol & tetracyclines
Selective toxicity in fungi & protozoa
- much more problematic; must kill a eukaryotic pathogen inside a eukaryotic organism
- drugs with fewest side effects target ergosterol in plasma membranes of fungi
- cyst stage in protozoans not susceptible to drugs (metabolically 'dormant') - pathogen must be actively growing in order to treat it
- pathogens hard to treat once they are inside cells of the body
Antiviral synthetic drugs
- nucleotide must have a phosphate group
- deoxyguanosine
- ex. acyclovir: when acyclovir triphosphate is added into a growing DNA strand, no additional nucleotides can be added. No -OH for DNA polymerase to add on to; stops synthesis of DNA strand
Physician's Desk Reference
- source for information on prescription, OTC, and herbal drugs
- also has basic information on a wide range of human diseases and conditions
Microbial drug resistance
- mutations passed on to progeny (allows them to continue replicating) - bacteria, eukaryotic, viruses
Drug resistance in bacteria through...
- conjugation: direct contact between cells (DNA transferred)
- transduction: DNA carried into cell by a virus
- transformation: extracellular DNA taken up by cell
Mechanisms of drug resistance
- drug inactivation: inactivation of drug like penicillin by penicillinase, an enzyme that cleaves a portion of the molecule and renders it inactive
- decreased permeability: the receptor that transports the drug is altered, so that the drug cannot enter the cell
- activation of drug pumps: specialized membrane proteins are activated and continually pump the drug out of the cell
- use of alternate metabolic pathway: the drug has blocked the usual metabolic pathway, so the microbe circumvents it by using an alternate, unblocked pathway that achieves the required outcome
4 sectors of the pharmaceutical industry
- research intensive pharmaceutical firms (Pfizer, Lilly, Merck, Glaxo Smith Kline)
- Generic drug manufacturing firms (Sandoz, Teva)
- biotechnology firms (Amgen)
- drug delivery firms (Alza, Inhale)
Pre-clinical drug development
- lab and animal tests to establish pharmacology and toxicology in animal models
- pharmacokinetics (ADME) - provides data for dosing, drug formulation
- pharmacodynamics - drug's positive and adverse effects in animals
- toxicology - collect date on drug's safety in both high and low doses, including 24-month carcinogenicity of the product, impact on reproductive health
- human tissue cultures are sometimes used
Goal of pre-clinical drug work
- to provide data to support an Investigational New Drug Application (IND)
- IND provides evidence that human testing of the drug should be allowed
- FDA has 30 days to request more info, deny, or approve IND
Contents of an IND
- results of pre-clinical studies
- drug's chemical structure and how it is thought to work/mechanism of action
- listing of adverse effects
- manufacturing info
- detailed clinical testing plan which lists how the drug will be tested, patient types, and where the testing will take place and by whom
Clinical development
- process of testing a drug in humans
- goal = show the drug is safe and effective in treating specific conditions in certain patients
- prior to 1962 only the safety of the drug needed to be proven, but with the passage of 1962 FDA amendments effectiveness is required
Phase 1 Trials
- establishes safety of administration in humans and a range of safe doses
- trials conducted on young healthy male humans, who are put in a carefully controlled environment & are under observation for hours or day
- # of subjects 20 to 100
Phase 2 trials
- first clinical test of drug on people with the disease
- 100-500 patients
- determines if drug actually works ("proof of concept")
- helps establish dosing (min & max), safety (adverse effects), and efficacy testing, or endpoints
Phase 3 trials
- large scale randomized, blinded, placebo controlled trial (sometimes comparative product used instead of placebo)
- can take 3-6 years & cost millions
- 1000-5000 patients with targeted disease or condition
- goal = collect efficacy info about product to support New Drug Application to FDA (collect end-oint effectiveness data)
- many times conducted by contract research organizations (CROs)
Placebo group
- group of patients who are receiving the inactive, fake, or "sugar pill," but do not know it.
Randomization
- subjects are randomly assigned to either the placebo or test group
- being assigned to the test group is by chance
Single blinded
- subjects do not know if they are receiving the placebo or test drug, however the investigator knows
Double blinded
- subjects and caregiver or investigator do not know who is receiving test drug
- most common study
NDA (New Drug Application)
- submitted to the FDA for review following completion of phase 3 & when company believes that sufficient efficacy and safety data were collected
- non-biological agents - NDA cent to FDA's Center for Drug Evaluation and Research (CDER)
- biologicals: a BLA (Biological Application) is submitted to FDA's Center for Biological Evaluation and Research (CBER)
Draft package insert included in NDA
- proposed package insert
- specifies: indications, explains dosages and dosing administration, includes key statement as to safety, risks, mechanism of action, adverse effects
FDA classifies NDAs
- puts NDA into 2 categories based on: novelty of active ingredient, clinical improvement
- if product satisfies an unmet need or represents a significant therapeutic advancement, FDA may grant a priority review which takes 6 months or less
- if it doesn't fit the above, then the drug fits the normal time frame of about 12 to 15 months for FDA review
Phase 4
- "post-marketing surveillance" or "post-approval" studies - after drug has been approved and product is on the market
- not all products undergo phase 4 studies
- used to monitor adverse effects & check on safety of drug product
Patents
- granted anywhere along line of drug development, but normally occur early
- patent life is 20 years after patent is filed and approved
- pharmaceutical companies file for drug patent once they think they have an innovative molecule; normally this occurs before preclinical studies
Exclusivity
- exclusive marketing rights granted by FDA upon approval of the drug
- can run concurrently with patent time
Exclusivity times
- orphan drugs: 7 years
- new chemical entity: 5 years
- pediatric exclusivity: 6 months added to existing exclusivity or patent
- patent challenge: 180 days (ANDAs only)
sterilization
- process in which the end result is a sterile product - all infectious microorganisms that can replicate have been eliminated from that material
- only absolute term
disinfection
- inanimate objects
- adding or using a chemical that will kill microorganisms but does not guarantee total elimination of microbes
sanitization
- inanimate objects
- any process prescribed by a state health department designed to reduce the number of microbes on/in a specific material
antisepsis/asepsis
- living tissue
- chemical safe enough to use on living tissue that reduces number of microbes
pasteurization
- treating liquids - applying heat to kill microbes within that product thereby lessening the likelihood that there are harmful microbes in that product
scale of most to least resistant microorganisms
- bacterial endospores, protozoan cysts, gram-negative bacteria, viruses without envelopes, gram-positive bacteria, viruses with lipid envelopes
factors affecting microbial inactivation
- number of microbes
- nature of microbe
- inhibition vs. death
- pH
- temperature (generally, cold doesn't kill)
- presence of organic matter (it competes for the chemical that is supposed to be killing the microbe)
approaches to killing microbes
- physical agents: temperature (heat kills, cold storage preserves, heat changes shape of protein); desiccation (drying); radiation (ionizing = high energy, non-ionizing (UV) = lower energy)
- mechanical removal: filtration (membrane filtration removes microbes, used for liquids & gases)
- chemical methods: soaps, alcohols, halogens, hydrogen peroxide
microbes & hands
- normal microflora (what is already on your body) vs. transient microflor (what you pick up during the day)
- ease of microbial transfer: from non-porous surface (no liquid absorbed), from porous or absorbant surface - lower ability for transfer of microbes to you, from finger to lip
- hand washing: highly effective in preventing microbial speed