• 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

1/133

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;

133 Cards in this Set

  • Front
  • Back
Microbes can be ___,____or____.
bacteria, fungi, or viral.
Defined as the invasion of pathogens OR the reaction of tissues to pathogen invasion.
Define Infection
Name three factors that determine the infection from a pathogen. TQ.
Virulence of pathogen, number, and/or host resistence.
The ability of a pathogen to be immune to anti-infective (either natural or reSquired).
Resistance
The range of activity of a drug (can be narrow, intermediate, or broad).
Spectrum
A combination of two drugs produces more effect than would be expected if their individual effects would be added.
Synergism
A combinatino of two drugs produces less effect than either drug alone.
Antagonism
What are the six components that one must consider when determining patient-drug-pathogen interaction.
Pharmokinetics (patient-drug), pharmodynamics (drug on patient), immunity (patient on pathogen), sepsis (pathogen on patient), resisance (pathogen on drug), selective toxicity (drug on pathogen).
Absorption, metabolism and excretion are all part of what?
Pharmokinetics (what the patient does to the drug)
What determines enzyme metabolism of a drug?
Chemical structure.
What diminishes the usefulness of drugs?
Toxic metabolites.
Useful anti-infectives must have what?
selective toxicity and favorable pharmacokinetics.
What is the ideal pharmodynamic (drug on patient) response of an anti-infective?
No response. "kill microbes, keep patient safe"
What is the target of anti-infective drugs.
The microbial cell, NOT the host.
True or False, antibacterial drugs are usually safe, but can become toxic from inapproriate use (e.g. dose, allergy).
True
Name three resistance mechanisms (pathogen on drug).
mutation, adaption, and gene transfer.
Defined as drug that exhibits toxicity toward microbe while having little or no effect on host.
Selective toxicity (drug on pathogen)
What two mechanisms determine selective toxicity?
1. blocking pathways essential for growth or survival.
2. Blocking a reaction vital for both host and microbe, but due to the difference in components (or affinity) involved, effect is exerted predominately on the microbe.
Static vs Cidal
Static is inhibition of growth or replication. (inhibitory)
Cidal is death of organism (lethal)
Defined as the appearance of a new infection during treatment of the primary infection-as a result of treatment.
Superinfection
What is the role of PBP in cell wall synthesis?
Penicillin binding proteins (a protein innate to many microbes) assists in the assemby, maintenance and regulation of bacterial cell walls.
How do beta-lactam drugs (penicillin) work?
They bind to the penicillin binding protein (pbp) membrane which results in inhibition of enzymes that form crosslinks in cell wall. This action weakens the cell wall so it can't withstand osmotic pressure=cell lysis and death (bacteriocidal). "beta-lactam cock block PBP"
How many groups of penicilllin are there? What group is penicillinase resistant?
Four groups. Group 2 (anti-staphylococcal) are penicillinase resistant. Group 2 is used to treat strep and staphloycocci, but not MRSA.
What is penicillinase?
An enzyme made by bacteria to degrade the beta-lactam ring of antibiotics making them useless.
What group of penicllin is used to treate systemic infections?
Group 4 or extended spectrum penicillins
Describe the spectrum covereage of cephalosporins?
1st generation (more gram postive; used to treat skin infections/prophylaxis)-2nd (same)-3rd more negative & hospital infections-4th mostly negative, pseudomonas and anerobic infections, tx meningitis and crosses the BBB.
What drugs could be used to treat meningitis?
4th generation cephalopsporin,
What is the greatest danger associated with penicillin use?
Allergic reaction
Name four ways in which bacteria can become beta-lactam resistance?
1. inactivation of beta-lactam ring, 2. alteration of PBPs (MRSA), 3. reduction of antibiotic access to PBP's (gram -) and 4. elaboration of efflux mechanisms (gram -).
What are two ways to fight bacterial resistance to beta-lactam?
1. Group 2 penicillin and some cephalosporins are resistant to beta-lactamases. 2. Use a beta-lactamse inhibitor like (clavulconic acid) with the abx, e.g. Augmentin
Name four drugs that are cell metabolism inhibitors (protein synthesis)?
M-TAC: macrolides, tetracycline, aminoglycosides, clinidamycin.
How do macrolides and clindamycin work?
50s inhibitors (ribosome): interferes with protein synthesis by inhibiting the enzyme peptidyl transferase: bacteroSTATIC.
How does tetracyclin work?
30s inhibitor: interfere with synthesis of bacterial proteins by binding to bacteria directly: bacteroSTATIC.
How do aminoglycosides work?
They are cell metabolism inhibitors. Specifically, they inhibit 30s (a ribosome subunit) and therefore disrupt initiation of protein synthesis: bacteroCIDAL.
These cell metabolism inhibitor is stored in dentine and enamel of unerupted teeth.
Tetracylcine
Can clindamycin be used for meningitis?
No, can't get into CSF
What cell metabolism inhibitor is most toxic?
Aminioglycoside, limits their use. Toxic to CN 8=auditory and vestibular disturbances.
This cell metabolism inhibitor interferes with other abx actrion, decreases effectivenes of PO BC, and increases serum levels of drugs metabolized in the liver.
Macrolides.
Tetracyclin can not be given with what drugs because it decreases the antibiotics absorption?
Calcium supplements and vitamins.
Name two cell metabolism inhibitors that do so by inhibiting Folic Acid?
Sulfonamides and trimethoprim
How do sulfonamides and trimethorprim inhibit cell metabolism?
These are antibacterial agents that inhibit folic acid synthesis via competitive inhibition=bacterioSTATIC. Of note, bacteria can't use preformed folic acid, humans can.
True or False: trimethoprim can cross into CSF during times of inflammation?
True
This antibiotic in high concentrations can cause crystal formation leading to kidney obstruction.
Sulfonamides
This antibiotic competes for sites that bind bilirubin. In newborns, unbound bilirubin is deposited into the basal ganglia which can cause kernicterus (toxic encephalopathy).
Sulfonamide
This drug interferes with folic acid and use in pregnancy may affect the fetus.
Trimethoprim
Name three cell replication inhibitors.
Quinolone, fluoroquinolones, and nitrofurans (not really a cell inhibitor).
This drug class inhibits cell replication by inhibiting DNA gyrase actitivy thereby preventing DNA unwinding.
Quinolones
This drug class inhibits cell replication by targeting topoisomerase IV (an enzyme responsible for seperating daughter cells after division.
Fluoroquinolones
True or False, DNA gyrase and topoisomerase are specific for micro-organisms.
True
True or False quinolones and fluoroquinolones are bacteriostatic but can become bacteriocidal when concentration increased.
True
True or False: Metal ions reduce absorption of quinolones.
True
What is the unique mechanism of Nitrofurans?
Metabolites (the active part of drug) of nitrofurans effect DNA/RNA synthesis and protein synthesis of bacteria. Selectively toxic. And important: bacteriostatic and bacteriocidal depending on dose.
True or False resistance is almost never seen in nitrofurans?
True
Why does nitrofuran have a fast onset of action?
Because it has rapid metabolism.
Can pregnant women take nitrofan? Why?
No-b/c interferes with RBC enzyme systems in babies
This is a anti-tuberculosis drug that only works on dividing cells.
Isoniazid
What is the MOA of isoniazid?
Blocks mycobacterium cell wall. When used alone can develop resistance.
What is the MOA of rifampin?
Blocks RNA synthesis and does not target mammalian polymerases. It is active against several bacteria and some viruses.
Why are isoniazd and rifampin used together? Whats the risk?
Alone can lead to resistance of TB strands. Increased risk for hepatitis.
Define mycoses.
The infection and resulting disease caused by fungi
Allergic disease, mushroom poisoning, and mycotoxins are all examples of what?
Adverse affects of fungi
What are the two clinical groupings of fungi?
Skin and deep mycoses
True or False: dimorphic systemic mycoses can happen to anyone.
True. Fungi can overcome physiological defenses by changing morphological form. Usually occurs in lungs.
Human mycoses is generally related to what two things?
Immunological status and environmental exposure.
Name two causes for increased human mycoses
More travel, greater age, disease, sicker pts=greater immunocompromise.
What is the main difference between animal and fungal cell membrane?
Animalls cell made of cholesterol, Fungi cell membrane made from ergosterol.
Name the four structural classes of anti-fungals.
Membrane disrupting agents, ergosterol synthesis inhibitors, glucan synthesis inhibitors, and antimetabolites and misc.
Name a class of antifungals that disrupt the membrane.
Polyenes (amphotericin B and Nystatin)
How do polyenes work? What type of microbe do they attack? Name two.
Bind to sterols (preferably ergosterol, but not exclusively) and disrupt the osmotic intergrity of the cell membrane (ions leak from inside to outside of cell)-FungiCIDAL. Amphotericin B and Nystatin.
This drug can be used to treat meningits but must be given intrathecally.
Amphotericin B a polyene
This drug has poor oral absorption but is often used to treat oral canidiasis.
Amphotericin B
What is the major limitation to Amphotericin B
Neprhotoxicity, b/c it also binds to cholesterol in human cells and killing them
What drugs increases the risk of neprhotoxicity?
Cyclosporine and aminoglycoside
The use of Amphotericin B will increase the accummulation of renaly cleared drugs, name two.
Flucytosine and fluconazole.
Azoles and Allylamines both do what?
Both are ergosterol synthesis pathway inhibitors but address it in two different ways making them fungistatic or fungicidal
Name two broad classes of azoles.
Imidazoles (older) and triazoles (newer)
What is the main difference between imidazoles and triazoles?
Metabolized more slowly and have less effect on human sterol synthesis.
Explain the MOA of azoles.
Inhibit Lanosterol demethylase (a CYP 450) and impair the synthesis of ergosterol=fungiSTATIC
This antifungal (an azole) can be used to treat meningitis
Fluconazole
True or False: Because azoles are bacterostatic, resistance can develop
True
True or False Azoles have numerous and dangerous interactions.
True: any other drug that uses the cyp 450 can cause major interactions.
What drug changes the pH of stomach and inhibits absorption of azoles
H2 antagonists/PPI; need an acidic environment for absorption
Explain the MOA of Allylamines.
Inhibit squalene epoxidase which blocks the entire pathway of ergosterol synthesis=fungiCIDAL. Note: ergosterol is needed for synthesis of the cell membrane in fungi.
What is the difference of azoles and allylamines if the both block the ergosterol pathway?
Azoles blocks Lanosterol Demethylase (later in pathway of ergosterol synthesis) and allaylamines block Squalene Epoxidase at the beginning of pathway.
Terbunafine (lamictal) is what?
An allylamine used to treat dermatophytosis
Explain the MOA of echinocandins: glucan synthase inhibitors.
Echinocandins block glucan syntase, an enzyme that is important in cell wall synthesis.
Echinocandis are often used in what type of therapy?
Salvage therapy for invasive aspergillus when nothing else works.
Explain the MOA of antimetabolites (a structural class of antifungals)
Can either block fungal DNA and protein synthesis ( as in flucytosine) or block fungal mitosis (as in griseofulvin)
What is an important complication of animetabolite use?
bone marrow suppression
This antimetaboite can be used to treat fungal meningits in AIDs patients, >50% absorped into CSF
Flucytosine
This antimetabolite is deposited in keratin precuror cells where it persists, providing prolonged fungal resistance.
Griseofulvin
Outline the different virus types.
DNA or RNA, enveloped or non-enveloped, double strand or single strand.
Name the four paths of viral entry.
1. translocation (naked). 2. Injection of genetic material (naked). 3. Receptor mediated endocytosis (naked and enveloped). 4. Membrane fusion (enveloped).
What form of viral entry uses a cage?
Receptor mediated
The drugs to treat herpes do so by what mechanism?
Inhibition of viral DNA polymerase.
Drugs to treat influenza do so by what mechanism?
Inhibit M2 protein which is the protein needed to take the virus out of its protective cage so that it can get into the nucleus.
Enfuviritide (T-20) is what type of drug?
A fusion or entry inhibitor. Used as an antiretroviral drug that inhibits entry of virus into CD4 cells.
What is enfuviritide (T-20) made from? It is used in what type of patients?
HIV envelope. Multi-drug resistant HIV
Explain the MOA of enfuviritide.
Docks to receptor site where the HIV particle would normally dock and therefore inhibits entry.
What is important to remember about enfuviritide (t-20)
Does not work on already affected cells.
Name three adverse affects of enfuviritide (t-20)
1. nodule or cyst development
2. Hardening of skin
3. peripheral neuropathy
Name two drugs that are specific for treatment of influenza A
Amatidine and Rimatidine
Name the complications of amantidine and rimantidine
no to pregnancy of lactation. high doses may produce cardiac arrythmias, HALLUCINATIONS, and suicidal ideation
What two drugs decrease rimantidine plasma levels?
Aspirin and acetaminophen
What is the MOA of anit-herpetic drugs?
DNA synthesis inhibitors by inhibiting DNA polymerase and DNA replication
How do antiherpetic drugs tx herpes?
Dont cure, but decrease shedding, decrease pain (except in shingles), increase speed of healing.
Acyclovir and Valcyclovir (prodrug) MOA is what?
Affinity for viral thymidine kinase which converts antiherpetic drugs to acyclovir monophosphatase which gets incorporated in to virus and inhibits virus replication.
What are the two ways that acyclovir (valcycloir) inhibit virus replciation?
competively inhibit incorporation of dGTP and act as a chain terminator because it lacks 3 prime hydroxy.
True or False: Acyclovir is specific for viral cells b/c is has an affinity of thymidine kinase (only found in virus).
True
Why is acyclovir given as prodrug?
Valcylovir is the prodrug for acylclovir and is completely converted to acyclovir by first-pass metabolism. Bioavailability is 3-5 times higher.
Penciclovir/famciclovir are what? What is their MOA?
Anti-herpatics. Competitive inhibitor of viral DNA polymerase via specifity to viral TK. Tx HSV and VSV infected cells
Ganciclovir and Valganciclovir (prodrug) are what?
Anti-herpetic. MOA: competitive inhibitor of viral DNA polymerase. Unique in that is treats ALL herpetic; especially CMV
Ganiciclovir/Valganciclovir are used to treat what?
Treatment of CMV retinitis in immunocompromised pts and prophylactic use.
This antiviral has a specificity for viral TK and ihibits viral DNA polymerase AND cellular DNA polymerase making it more toxic.
Trifluridine
Trifluridine is administered how and used to treat what?
Topical to tx ocular herpes
Cidofir
A nucleotide analog that does not require viral TK or other viral kinases; its activated by HOST cell kinases. Used against MUTANT viruses.
Foscarnet
Not a purine or pyrimidine analog. Inhibits viral DNA and RNA polymerases (tx. hiv, hsv, and cmv) Used for tx of viruses resistant to nucelosides/nucleotide analogs.
Nucleoside and nucelotide transcriptase inhibitors (NRTI) specificity is what?
Blocks HIV replication and the infection of new cells, but they have little effect on cells that are already infected. ``
Nucleoside and nucelotide transcriptase inhibitors (NRTI) MOA is what?
competitive inhibitor of reverse transcriptase.
Nucleoside and nucleotide transcriptase inhibitors (NRTI) can do what?
All can produce potentially fatal syndrome called lactic acidosis and severe hepatomegaly.
Nonnuceosdide inhibitors (NNRTIs) MOA?
Inhibit viral reverse transcriptase by inducing a conformational change in enzyme that causes enzyme inactivation.
Efavirenz
A non-nucleoside reverse transcriptase inhibitor (NNRTI); can't give in pregnancy
Nevirapine
NNRTI; adults and children but monitor hepatotoxicity
Delavirdine
NNRTI; adults and adolescents over 16
What drugs do you need to be wary of when giving NNRTI?
Other drugs that are metabolized for CYP 450
What is the MOA of protease inhibitors?
Inhibit HIV protease activity (which is required for production of mature infectious virus).
Who should you avoid using protease inhibitors in?
Can cause increase bleeding in hemophiliacs.
What is HAART therapy?
Highly active anti-retroviral therapy.
What consists of combo therapy in the treatment of HIV
RT + protease inhibitors. more effecious at lower doses and reduced toxicity.
Neuraminidase Inhibitors work on viruses?
Block viral release (influenza)
Zanamivir & Oseltamivir
Both are neuraminidase inhibitors, reduce influenza by 1-2 days
Zanamivir (relenza) works on what type of influenza? What can it do to lungs?
This drug is a neuraminidase inhibitor (blocks viral release). Used to tx Influenza A & B , poor PO bioavailability so given in inhaler form, may cause bronchospasms
True or False: Osteltamivir (tamiflu) oral bioavailability: 70%; GI distress. Used for prophylactic uses.
True