• 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

1/280

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;

280 Cards in this Set

  • Front
  • Back
What are 5 important nosocomial pathogens?
- staphylococcus aureus
- acinetobacter baumannii
- klebsiella pneumonia
- pseudomonas aeruginosa
- enterococcus faecium
What percentage of staphylococcus aureus is resistant to methicillin?
60%
What are two examples of antibiotics which exhibit concentration-dependent killing?
- aminoglycosides
- fluoroquinilones
What are two examples of antibiotics which exhibit time-dependent killing?
- beta-lactams
- vancomycin
What is an example of a superinfection?
- pseudomembraneous colitis due to growth of Clostridium difficile
- often after clindamycin
What are three general mechanisms of antibiotic resistance?
1) mutation and selection
2) uptake of extracellular DNA from related bacteria and recombination
3) plasmid mediated acquisition of R-factors
In which bacteria is mutation and selection often the mechanism of antibiotic resistance?
mycobacterium tuberculosis
In which 3 bacteria are recombination often the mechanism of antibiotic resistance?
- Haemophilus
- Neisseria
- Streptococcus
In which 4 bacteria are plasmid-borne R-factors often the mechanism of antibiotic resistance?
- Klebsiella
- Pseudomonas
- E. coli
- Staphylococcus
What are the 3 most common bacterial pathogens causing acute respiratory infections?
- streptococcus pneumonia
- haemophilus influenza
- mycoplasma pneumonia
What classes of drugs inhibit nucleic acid synthesis? (4)
- sulfonamides
- trimethoprim
- fluoroquinolones
- quinolones
What is sulfonamide MOA?
- analog of PABA
- inhibits folic acid synthesis
What is trimethoprim MOA?
- analog of dihydrofolate
- inhibits DHFR
Which two nucleic acid inhibitors are often combined and why?
- TMP/SMZ (trimethoprim and sulfamethoxazole)
- they inhibit two distinct steps in the same pathway so they are synergistic
- SMZ inhibits earlier step in pathway
Which two nucleic acid inhibitors are never combined and why?
- nalidixic acid and nitrofurantoin
- antagonistic effect
What is therapeutic use of sulfonamides?
- sulfadiazine + pyrimethamine for treating toxoplasmosis
What is therapeutic use TMP/SMZ? (4)
- UTIs
- RTIs caused by H. influenza and S. pneumonia
- Shigella enteritis
- PCP in AIDS patients
What are 3 major toxicities with sulfonamides?
- blood dyscrasias
- skin rashes
- kernicterus in newborns (from displacing protein-bound bilirubin which deposits in basal ganglia)
What is important drug interaction with sulfonamides and precaution must be taken?
- they inhibit P450-mediated metabolism of warfarin
- must monitor INR
In addition to sulfonamide toxicities, what are additional toxicities with TMP/SMZ? (2)
- megaloblastosis, leukopenia, and thrombocytopenia in patients with folic acid deficiencies (alcs, homeless, malnourished)
- rash, fever, and hepatitis in AIDS patients treated for PCP
What are 3 mechanisms of resistance to sulfonamides?
- **synthesis of altered dihydropteroate synthase which has lower affinity for sulfonamide
- overproduction of PABA
- reduced uptake of sulfonamide
What are 2 mechanisms of resistance to TMP/SMZ?
- overproduction of DHFR
- expression of altered DHFR with reduced affinity for trimethoprim
What is the MOA of quinolones and fluoroquinolones?
- inhibit two enzymes in DNA replication:
- DNA gyrase (gyrA)
- topoisomerase IV (parC)
Which two enzymes are inhibited by quinolones and fluoroquinolones?
- DNA gyrase (gyrA)
- topoisomerase IV (parC)
What are the common fluoroquinolone drugs? (3)
- ciprofloxacin
- ofloxacin
- levofloxacin
What drugs end in "floxacin"?
fluoroquinolones
What is normal function of DNA gyrase (gyrA) and topoisomerase IV (parC) and which antibiotics inhibit these?
- relieves positive supercoiling to allow DNA replication to continue
- inhibited by quinolones and fluoroquinolones
How do fluoroquinolones show selective toxicity?
- inhibit bacterial DNA gyrase at much lower concentrations than the mammalian enzyme
What are therapeutic uses of fluoroquinolones? (4)
"broad spectrum, oral administration"
- UTIs (inc Pseudomonas aeruginosa)
- enteritis (Salmonella, Shigella, E. coli, Campylobacter)
- vibrio cholerae infections
- respiratory, bone, soft tissue infections
What are major toxicities of fluoroquinolones?
- nausea and GI distress
- CNS effects
- damages growing cartilage (contraindicated in kids and pregnant women)
In which patients are use of fluoroquinolones contraindicated?
- kids (< 18) and pregnant women
- damages growing cartilage
What is important drug interaction of fluoroquinolones?
- inhibits P450-mediated degradation of theophylline (asthma drug) and caffeine
- can lead to seizures
What are 2 mechanisms of resistance to fluoroquinolones?
- alterations In DNA gyrase (gyrA) and topoisomerase IV (parC) lower affinity for drug
- decreased permeability of the drug (mutations in porins)
What kind of drug is nalidixic acid and what is its use?
- older quinolone
- used to treat uncomplicated UTIs
What kind of drug is nitrofurantoin and what is it used for?
- prodrug that is reduced by bacterial enzymes into reactive species that destroys DNA
- used to treat uncomplicated UTIs
What are the main 2 drug types that inhibit cell wall synthesis?
- beta lactams
- vancomycin
What are the 4 types of beta-lactams?
- penicillins
- cephalosporins
- monobactams
- carbapenems
Which of the beta-lactams have the widest spectrum of activity?
carbapenems
What 2 processes of cell wall synthesis are inhibited by beta-lactams?
- transpeptidation
- carboxypeptidation
How do beta-lactams work?
- they are analogs of the acyl-D-ala-D-ala C terminus of the peptide chain
- when it binds the PBP, the bond is stable so the enzyme is inhibited and cell wall synthesis can't proceed
What is the most important determinant of a beta-lactams effectiveness against an organism?
- the "affinity" for any one of the essential PBPs
- high affinity = low concentration of antibiotic = high acylation rate for the PBP
Compare ampicillin and penicillin G against E. coli
- they have equal affinity for PBPs
- ampicillin is more polar and diffuses through porin channels more easily so it has much lower MIC
What 2 properties define intrinsic resistance of an organism to a beta-lactam?
- affinity of drug for PBPs
- ability of drug to reach the PBPs in the periplasm of gram(-) bacteria
What 4 mechanisms define extrinsic resistance to beta-lactams?
- production of beta-lactamase that hydrolyzes anti-biotic
- decrease in outer membrane permeability
- overexpression of efflux pump that pumps drug out of periplasm
- mutations in essential PBPs that decrease rate of inactivation by antibiotic
What is meant by "extrinsic resistance" of an organism?
- resistance existing above and beyond that found in a wild-type strain
- compared to intrinsic resistance which would be typical of wild-type
What is similar and different between PBPs and beta-lactamases?
SAME: serine in beta-lactamase and PBP attacks beta-lactam bond, forming an acyl-enzyme complex
DIFF: beta-lactamases rapidly hydrolyze which inactivates the drug
In what 2 ways is beta-lactamase mediated?
- chromosomally: inducible
- plasmid: found on R factors and transmitted among strains
How do beta-lactamases differ between gram(+) and gram(-) bacteria?
gram(+): b-Ls are extreted into the medium
gram(-): b-Ls are retained in periplasm
Are gram(+) or gram(-) organisms more susceptible to beta-lactamases?
- gram(-) because the beta-lactamase is retained in the periplasm where it is needed
What drugs were developed to be resistant to beta-lactamse? (5)
- methicillin (no longer used)
- oxacillin
- nafcillin
- 2nd and 3rd generation cephalosporins
- carbapenems
Which beta-lactamase does not form an acyl-enzyme complex with antibiotics and what are 2 problems from this?
metallo-beta-lactamse (NDM-1)
- hydrolyzes most beta-lactams
- not inhibited by clavulanic acid
What bacteria often has mutations in the promoter region to increase expression of efflux pumps?
Neisseria gonorrhoeae
Which bacteria (4) commonly confer resistance to beta-lactams through alterations in PBPs?
- Staph. aureus
- Strep. pneumoniase
- Haemophilus influenza
- Neisseria gonorrhoeae
What is PRSP and what drugs are used to treat it?
- penicillin-resistant Strep. pneumoniae
- contains mutations in all of the essential PBPs
- treated with vancomycin, linezolid, or streptogramins
What is relationship between PBP acylation rate and MIC of a beta-lactam?
the PBP with a higher acylation rate has a lower MIC
In what 2 ways does Neisseria become resistant to penicillin?
- plasmid-mediated production of a beta-lactamase
- chromosomal mutations in endogenous genes
What chromosomal mutation occurs in Neisseria strains which are penicillin or ceftriaxone resistant?
PBP2
Mutations of PBP2 in Neisseria gonorrhoeae cause resistance to what 2 drugs?
- peniccilin
- ceftriaxone
What drugs are used to treat MRSA? (3)
- vancomycin
- linezolid
- streptogramins (dalfoprisin/quinupristin)
How did MRSA develop resistance?
- acquired new PBP (PBP2a) from an animal pathogen (Staph. fleurettii)
- PBP2a shows very low rate of acylation with almost all beta-lactams
What are therapeutic uses of PenG and PenV? (4)
"mainly gram(+)"
- neisseria meningitidis (ceftriaxone is preferred)
- strep. pneumoniae
- enterococci (with aminoglycoside)
- syphillis
What is therapeutic use of penicillinase-resistant penicillins?
- staphylococcus aureus (NOT MRSA)
What are therapeutic uses of ampicillin and amoxicillin?
"wider gram(-) coverage than PenG and PenV"
- E. coli
- H. influenzae, Pen-sensitive strep. pneumo (URI, otitis media)
- listeria
What are therapeutic uses of ticarcillin, pipericillin, and mezlocillin?
"difficult to treat gram(-) bacteria"
- proteus mirabilis
- pseudomonas aeruginosa
- serratia marcescens
- klebsiella pneumoniae
What are the broadest spectrum beta-lactams? (2)
"carbapenems"
- imipenem
- meropenem
What are the therapeutic uses of imipenem and meropenem?
- serious infections of unknown origin
- mixed infections
What drug is combined with imipenem and why?
- cilistatin (renal dipeptidase inhibitor)
- imipenem is hydrolyzed and inactivated by a renal dipeptidase
What is cilistatin and what is it's use with antibiotics?
- renal dipeptidase inhibitor
- used with imipenem which is normally hydrolyzed by imipenem
What are two beta-lactamase inhibitors?
- clavulanic acid
- sulbactam
What are clavulanic acid and sulbactam?
beta-lactamase inhibitors
What two drugs combine a beta-lactam with beta-lactamase inhibitors?
- Augmentin (amoxicillin + clavulanate)
- Unasyn (Ampicillin + sulbactam)
What drugs make up Augmentin?
amoxicillin and clavulanate
What drugs make up Unasyn?
ampicillin and sulbactam
What is MOA of beta-lactamase inhibitors?
- they are beta-lactams
- react with beta-lactamase to form acyl-enzyme complex and inhibit the enzyme
What is the 1st generation cephalosporin and what is its therapeutic use?
- cephalexin
- mostly gram(+) coverage
- treats community-acquired skin infections
What is 2nd generation cephalosporin and what is it's therapeutic use?
- cefuroxime
- increased gram(-) coverage
- treats community-acquired pneumonia caused by H. influenzae or Klebsiella pneumonia
What is cephalexin and what does it treat?
- 1st generation cephalosporin
- mostly gram(+) coverage
- treats community-acquired skin infections
What is cefuroxime and what does it treat?
- 2nd generation cephalosporin
- increased gram(-) coverage
- treats community-acquired pneumonia caused by H. influenzae or Klebsiella pneumonia
What are two 3rd generation cephalosporins and what do they treat?
- ceftriaxone and cefotaxime
- penetrate CNS, useful for meningitis by gram(-)
- also treatment of choice for gonorrhea
What is treatment of choice for gonorrhea?
- ceftriaxone (3rd generation cephalosporin)
What is ceftriaxone and cefotaxime and what do they treat?
- 3rd generation cephalosporins
- penetrate CNS, useful for meningitis by gram(-)
- also treatment of choice for gonorrhea
What are two important properties about beta-lactams regarding how they are processed by the body?
- not metabolized
- excreted in urine
How are most beta-lactams processed and what are the exceptions?
- excreted in the urine (not metabolized)
- nafcillin and cefoperazone are excreted in bile
How fast are most beta-lactams eliminated and what is the exception?
- rapidly eliminated (30-120 mins) by glomerular filtration
- exception is ceftriaxone which has 8 hr half life
What is most common side effect of penicillins?
- hypersensitivity (rash, fever, bronchospasm, anaphylaxis)
- least toxic antibiotics bc humans have no cell wall
Which penicillin can be used if a patient has had a previous allergic reaction?
- aztreonam (a monobactam)
What are three main concerns with cephalosporins?
- hypersensitivity
- intense local pain upon IM injection
- some cause bleeding disorders
What is a unique potential toxicity of cefotetan, cefoperazone, and cefamandole)?
- bleeding disorders
- side chain interferes with prothrombin formation
What cephalosporins can cause bleeding disorders as well as disulfiram-like reaction when used with alcohol? (3)
- cefotetan
- cefoperazone
- cefamandole
Which cephalosporins carry a high risk of superinfections?
2nd and 3rd generation
What is a unique risk with 2nd and 3rd generation cephalosporins?
superinfections
What are therapeutic uses for vancomycin?
"bactericidal for gram(+) - doesn't penetrate outer membrane of gram(-)"
- infections caused by MRSA, Enterococcus faecium/faecalis, and penicillin-resistant-strep pneumo
- pseudomembraneous colitis caused by C. diff
What drug is used to treat pseudomembraneous colitis caused by Clostridium difficile and what property makes this work?
- oral vanvomycin
- not absorbed from GI tract
How is vancomycin normally given and what is the exception?
- parenterally bc it isn't absorbed by GI tract
- exception is oral vancomycin in pseudomembraneous colitis due to C. diff
What is MOA of vancomycin?
- binds tightly to acyl-D-ala-D-ala C terminus of peptide chain to prevent glycan polymerization and crosslinking of the peptide
In what organism is vancomycin resistance often observed?
Enterococci faecium
What are 2 key observations of vancomycin resistance?
- plasmid-mediated
- induced directly by vancomycin
What 3 enzymes mediate vancomycin resistance?
- VanX
- VanH
- VanA
What is the mechanism of vancomycin resistance? (i.e. what is the product of VanX, VanH, and VanA)
- D-ala-D-lac instead of D-ala-D-ala
- vancomycin does not bind to acyl-D-ala-d-lac with high affinity, but cell wall cross-linking is not affected by substitution of D-ala for D-lac
What enzyme is assocatiated with VRSA and how does it occur?
- vanA
- obtained genese by horizontal transfer of a transposon
When is VISA seen?
after prolonged exposure to vancomycin (25 days to 18 weeks)
What 2 drugs make up the bactericidal protein synthesis inhibitors?
- aminoglycosides (except spectinomycin)
- streptogramins
What 5 drugs make up the bacteriostatic protein synthesis inhibitors?
- tetracyclines
- chloramphenicol
- macrolides
- clindamycin
- linezolid
How do aminoglycosides and linezolid inhibit protein synthesis?
- block formation of the initiation complex
Which drugs inhibit protein synthesis by blocking formation of the initiation complex? (2)
- aminoglycosides
- linezolid
How do chloramphenicol, tetracycline, and clindamycin inhibit protein synthesis?
- block aminoacyl-tRNA binding
Which drugs inhibit protein synthesis by blocking aminoacyl-tRNA binding? (3)
- chloramphenicol
- tetracycline
- clindamycin
Which drugs block protein synthesis by blocking translocation of the new peptidyl-tRNA back to the P site? (2)
- macrolides
- spectinomycin
How do macrolides and spectinomycin inhibit protein synthesis?
- block translocation of the new peptidyl-tRNA back to the P site
What are aminoglycosides active against as single agents?
- only gram(-) bacteria (except staphylococci)
What inhibits penetration of aminoglycosides through porins and binding to sites on cytoplasmic membrane?
- divalent cations (Ca2+ and Mg2+)
What drug is inhibited by divalent cations (Ca2+ and Mg2+)?
aminoglycosides
What is necessary for aminoglycosides to reach binding sites in gram(+) bacteria?
- beta-lactams
What is important to remember about oxygen and aminoglycosides?
- uptake of drug requires oxygen
- thus, aminoglycosides are ineffective against strict anearobes
What is an example of synergism with aminoglycosides?
addition of a beta-lactam
What is an example of synergism with beta-lactams?
addition of aminoglycoside
What are therapeutic uses of aminoglycosides?
- moderate to severe aerobic gram(-) infections
- almost always combined with a beta-lactam
What is a precaution of using aminoglycosides and beta-lactams?
- premixing the two drugs inactivates them (forms a complex)
What are main toxicities of aminoglycosides and how bad is it? (2)
- ototoxicity
- neprotoxicity
**severe; limits their use
How are aminoglycosides administered and what are their distribution limits?
- parenteral admin (usually IM)
- excluded from CNS and ocular fluids
What most often causes resistance to aminoglycosides and how is this resistance mediated?
- modification of the antibiotics by enzymes (adenylation, acetylation, phosphorylation)
- plasmid-mediated and thus transmissable
Which aminoglycoside is least susceptible to enzymatic inactivation?
amikacin
What are the therapeutic uses of tetracyclines? (4)
- Rocky mtn spotted fever (Rickettsia)
- cholera (vibrio choerae)
- lyme disease (Borrelia)
- brucellosis (Brucella)
Which 4 bacteria can be targeted by tetracyclines?
- rickettsia
- vibrio cholerae
- borrelia
- brucella
Which 4 diseases can be treated with tetracyclines?
- rocky mtn spotted fever
- cholera
- lyme disease
- brucellosis
Which 3 drugs are tetracylines and what is their specturm?
- tetracycline, doxycycline, minocycline
- "broad-spectrum" but rarely used d/t resistance and toxicities
How are tetracyclines administered and what is their distribution?
- oral or IV
- distribute well except CNS
What are main toxicities of tetracyclines?
- GI irriatation (oral admin)
- phototoxicity (rashes on skin in sun)
- hepatic tox
- renal tox
- deposition in bones and teeth
- fanconi syndrome
What drug is associated with Fanconi syndrome and what are its effects?
- caused by ingestion of outdated tetracycline
- nausea, vomiting, polyuria, proteinuria
What antibiotics are contraindicated in pregnancy?
- fluoroquinolones
- tetracyclines
In what patients are tetracyclines contraindicated?
- pregnant women and children < 8 yrs
- causes deposition in bone and teeth
What is a newer tetracycline which has expanded their use and spectrum of action?
tigecycline
What are therapeutic uses of tigecycline?
- MRSA
- penicilin-resistant strep pneumoniae
- many gram(-) bacteria (Citrobacter, Klebsiella, Shigella)
- many anaerobes (Bacteroides)
What are the mechanisms of resistance to tetracyclines?
- **production of an efflux pump
- production of TetM protein that protects ribosome from inhibition by tetracycline
- mutations in ribosomal structural proteins
What is spectrum and general use of chloramphenicol?
- "broad spectrum" but toxicities limit use
- reserved for treatment of serious infections when other drugs are contraindicated
What are therapeutic uses of chloramphenicol? (2)
- bacterial meningitis (H. influenzae or N. meningitidis) in patients with penicilin allergy
- Rocky mtn spotted fever (Rickettsia) when tetracycline is contraindicated
What are major toxicities of chloramphenicol? (3)
- aplastic anemia (irreversible and fatal)
- dose-related, reversible bone marrow depression
- gray-baby syndrome (circulatory collapse)
What is main resistance mechanism to chloramphenicol and how is it mediated?
- inactivated by acetyl transferase
- this enzyme is often found of multi-drug resistant plasmids
What drugs end in "cycline"?
tetracyclines
What drugs end in "thromycin"?
macrolides
What drugs have antagonistic effects with macrolides? (2)
- chloramphenicol
- clindamycin
What 3 drugs are macrolides?
- erythromicin
- clarithromycin
- azithromycin
What are therapeutic uses of macrolides? (4)
- Legionnaire's disease (Legionnella)
- mycoplasma pneumoniae
- chlamydia trachomatis
- whooping cough (Bordetella pertussis)
How are azithromycin adn clarithromycin distributed?
- accumulate in phagocytes and are released at site of infection
What are main toxicities of macrolides?
- generally well-tolerated
- GI distress and hepatotoxicity can occur
What are major drug interactions of macrolides?
- erythromycin and clarithromycin potentiate effects of corticosteroids, cyclosporins, digoxin, and warfarin
Which macrolides potentiate effects of corticosteroids, cyclosporine, digoxin, and warfarin?
- erythromycin and clarithromycin
- NOT azithromycin
Which macrolide does not inhibit P450 enzymes?
- azithromycin
What are 4 mechanisms of resistance to macrolides?
- decrease in permeability
- efflux pump
- modification of ribosome target site
- hydrolysis by an esterase
What are therapeutic uses of clindamycin?
- severe anaerobic infections caused by Bacteroides fragilis
- alternative for toxoplasmosis and PCP
What drug is given with clindamycin to treat toxoplasmosis?
- pyrimethamine
What drug is given with clindamycin to treat PCP?
- primaquine
How has use of clindamycin changed recently?
- use was limited due to toxicities, but now expanding to treat opportunisitic infections associated with AIDS
What are main toxicities with clindamycin? (4)
- GI distress (found in feces up to 2 weeks)
- skin rashes
- hepatotoxicity
- superinfection by C. diff (--> pseudomembraneous colitis)
What drugs can be used to treat C. diff? (2)
- metronidazole
- oral vancomycin
What are 3 mechanisms of resistance to clindamycin?
- **production of methylase that modifies binding site
- direct inactivation of drug
- decreased permeability
What drugs end in "pristin"?
streptogramins
- dalfopristin
- quinupristin
What are the streptogramins?
- dalfopristin
- quinupristin
**always used together (synergistic)
What is the MOA of streptogramins?
bind the 50S subunit and cause comformational change which blocks peptide elongation
What are the therapeutic uses of streptogramins? (3)
- vancomycin-resistant Enterococci faecium (but NOT E. faecalis)
- penicillin-resistant Strep. pneumoniae
- MRSA
What are the main 2 toxicities of streptogramins?
- pain at injection site / phlebitis
- myalgias / arthralgias
What are important drug interactions of streptogramins?
- inhibit P450 enzymes
- increase concentration of cyclosporine and Ca channel blockers
What are the 2 mechanisms of resistance to streptogramins?
- methylase that blocks binding of rRNA
- acetyltransferase that inactivates antibiotic
What is MOA of linezolid?
- binds 50S subunit and prevents formation of 70S initiation complex
What are therapeutic uses of linezolid? (4)
- VRE
- pen-resistant Strep pneumo
- MRSA
- group A and B strep
What is main resistance mechanism to linezolid?
- mutations in rRNA that decrease affinity of drug
What are main toxicities related to linezolid?
- bradycardia with concurrent use of beta-blocker
- thrombocytopenia
- superinfections
What are important drug interactions with linezolid?
- a reversible MAOI
- avoid food high in tyramine and can NOT be given with MAOIs, bupropion, tricyclic antidepressants, SSRIs, or St. John's Wort
In what kind of organisms can metronidazole be used and what is its MOA?
- taken up only in anaerobic bacteria and parasites
- converted to active metabolite that disrupts DNA
What are the therapeutic uses of metronidazole? (3)
- pseudomembraneous colitis from C. diff
- bacteroides fragilis
- parasitic infections (trichomoniasis, giardia, amebiasis)
Which antibiotic can be used to treat anaerobic bacteria and parasites?
metronidazole
What are main toxicities associated with metronidazole?
- nausea, headache
- disulfram-like reaction with EtOH
- not recommended in pregnant women
What is main resistance mechanism of metronidazole?
- decrease or loss of enzymes involved in activation of antibiotic
What are therapeutic uses of daptomycin? (4)
- MRSA
- VISA
- VRSA
- VRE
What happens to daptomycin if used for respiratory infections?
- inactivated by pulmonary surfactant
Which drug is inactivated by pulmonary surfactants?
daptomycin - therefore not used for respiratory infections
What is MOA of daptomycin?
- binds to cell membrane of gram(+) bacteria to cause membrane depolarizaiton, loss of membrane potential, and cell death
What are main 2 toxicities of daptomycin?
- skeletal muscle damage
- peripheral neuropathy at high concentration
What is primary mechanism of resistance to daptomycin?
- remodeling of the phospholipid membrane and diversion from division of septum where it exerts its effects
What 3 characteristics of tuberculosis make it difficult to treat?
- mycobacteria are often intracellular (in macrophages)
- very slow growing organism
- tubercular nodules are poorly perfused
What defines MDR-TB?
- resistance to two of the first-line drugs: isoniazid and rifampin
What defines XDR-TB?
- resistance to two first line drugs: isoniazid and rifampin
- PLUS, resistance to any fluoroquinolones
- PLUS, at least 1 of 3 second line aminoglycosides (amikacin, kanamycin, capreomycin)
What are 3 injectable second-line aminoglycosides for TB treatment?
- amikacin
- kanamycin
- capreomycin
What are the 4 first line TB drugs?
- isoniazid
- rifampin
- pyrazinamide
- ethambutol
What are the 5 second line TB drugs?
- streptomycin
- d-cycloserine
- ethionamide
- ciprofloxacin
- amikacin
What is MOA of isoniazid?
- prodrug activated by KatG
- active form inhibits InhA which is required for synthesis of mycolic acid, a critical component of mycobacterial cell wall
What is a special feature of isoniazid which makes it a mainstay in the TB regimen?
- penetrates cells and is active against intracellular organisms
How is isoniazid metabolized?
- acetylation
What 2 situations can cause increased toxicity to isoniazid?
- slow acetylation
- renal insufficiency
What are the 2 main toxicities in isoniazid?
- **hepatotoxicity (can be fatal)
- peripheral and CNS neuropathy
What are important drug interactions of isoniazid?
- inhibits CYP3A4 (a P450)
- decreases metabolism of phenytoin and diazepam
What are the 2 primary mechanisms of resistance to isoniazid and what level of resistance do they confer?
- overproduction of the InhA gene product (low resistance)
- mutation or deletion of KatG gene (high resistance)
What is MOA of rifampin?
- inhibits DNA-dependent RNA plymerase
What are therapeutic uses of rifampin? (2)
- **first-line TB drug
- meningitis prophylaxis from exposure to N. meningitidis and H. influenzae
What is main toxicity associated with rifampin?
hepatotoxicity
What are main drug interactions of rifampin?
- induces 4 of 7 P450 enzymes
- this increases elimination of other drugs
- can cancel contraceptive effects of oral steroids
What is MOA of ethambutol?
inhibits the synthesis of arabinogalactan, an essential component of mycobacterial cell walls
What is therapeutic use of ehtambutol?
- first-line TB drug
What is main toxicity associated with ethambutol?
- optic neuritis with red-green discrimination (resolves with removal of drug)
What are main 2 mechanisms of resistance to ethambutol?
- overexpression of enzyme that synthesizes arabinogalactan
- mutations that lower affinity of enzyme for ethambutol
What is therapeutic use of pyrazinamide?
- first-line TB drug
What is MOA of pyrazinamide?
- converted to pyrazinoic acid (active form)
- inhibits fatty acid syntase 1
What are main 2 toxicities associated with pyrazinamide?
- hepatotoxicity
- hyperuricemia --> gout
What is main mechanism of resistance to pyrazinamide and how fast does it occur?
- mutations in pyrazinamidase that blocks conversion to active form
- develops rapidly during single therapy
What is treatment for mycobacterium avium complex (MAC)?
- ehtambutol + azithromycin or clarithromycin
What is used for prophylaxis against MAC in AIDS patients with low T-cell counts?
rifabutin
What are 4 general types of antifungals?
- amphotericin B
- flucytosine
- triazoles
- caspofungin
Which two antifungals are used synergistically?
- amphotericin B (AMB)
- flucytosine
Which antifungals are useful with less side effects than AMB and what 3 drugs are in this class?
"triazoles"
- fluconazole
- itraconazole
- voriconazole
What is a new antifungal useful for treatment of invasive aspergillosis and candidiasis?
caspofungin
What is caspofungin used for?
- invasive aspergillosis
- candidiasis
What is MOA of amphotericin B?
- forms a transmembrane pore by forming complex with ergosterol (component of fungal membranes)
- leakage of ions and metabolites leads to cell death
What are challenges that arise from MOA of amphotericin B?
- also forms complex with cholesterol in mammalian cells
- has 10-fold selectivity for ergosterol but still has low therapeutic index
What are therapeutic uses of amphotericin B? (3)
- Cryptococcus meningitis
- Candida infections
- Coccidioides meningitis (intrathecal)
How is amphotericin B packaged to decrease toxicity?
in a liposomal preparation
What are main toxicities with amphotericin B? (3)
"one of most toxic agents in use"
- **potent nephrotoxin
- hypokalemia and hypomagnesemia
- hypersensitiviy
What drugs must be used cautiously with amphotericin B?
- other nephrotoxic drugs
- aminoglycosides and cyclosporin)
What is important about pharmacokinetics of amphotericin B?
- given by slow IV infusion
- rapid sequesteration in tissues, slowly released
- long half-life (15 days)
What is main mechanism of resistance to amphotericin B and how often does it occur?
- decreased levels of ergosterol or alteration of its structure
- development of resistance during therapy is rare
What is MOA of flycytosine (5-fluorocystosine)?
- taken up by fungal cell and converted to FdUMP
- FdUMP forms complex that blocks DNA synthesis
When is flucytosine used?
- combined with AMB to treat Candida and Cryptococcus meningitis
What are main toxicities of flucytosine? (4)
- bone marrow depression
- hair loss
- GI distress
- hepatotoxicity
What is main mechanism of resistance to flycytosine?
- mutations in cytosine permease or cytosine deaminase
What is the MOA of triazoles?
- block ergosterol synthesis by inhibiting 14-demethylase, an enzyme necessary for fungal cell wall synthesis
What are therapeutic uses for triazoles? (4)
- blastomycosis
- histoplasmosis
- candidiasis
- aspergillosis
What is an alternative to AMB + flucytosine for treating coccidiodal and cryptococcal meningitis?
fluconazole (a triazole)
Which triazole can penetrate the CNS?
fluconazole
What are main toxicities of triazoles?
- vomiting, diarrhea, rashes
- impaired hepatic function
- teratogenic
In what patients are triazoles contraindicated?
pregnant women (they are teratogenic)
What are important drug interactions of triazoles?
- inhibit P450
- potentiate warfarin, cyclosporine, digoxin
What are main 2 mechanisms of resistance to triazoles?
- overproduction or alteration of 14-demethylase
- expression of multidrug efflux pump
What is MOA of caspofungin?
- inhibits synthesis of glucans (essential part of fungal cell wall) by blocking glucan synthase
What are therapeutic uses of caspofungin?
- invasive candidiasis and aspergillosis
What are main toxicities of caspofungin? (2)
- phlebitis at injection site
- embryotoxic (contraindicated in pregnant women)
What antifungals are contraindicated in pregnant women?
- triazoles
- caspofungin
What antifungals are safe to give in pregnant women?
- AMB
- flucytosine
What antivirals are used to treat HSV and VZV? (3)
- acyclovir
- valcyclovier
- famcyclovir
What are acyclovir, valcyclover, and famcyclovir used to treat?
- HSV
- VZV
What antivirals are used to treat cytomegalovirus? (2)
- ganciclovir
- valganciclovir
What are ganciclovir and valganciclovir used to treat?
- cytomegalovirus
What are 4 drug types used to treat HIV and other retroviruses?
- nucleoside/tide reverse transcriptase inhibitors (NRTIs)
- non-NRTIs
- protease inhibitors
- fusion inhibitors
What 3 drugs are used to treat influenza virus?
- zanamivir
- oseltamivir
- adamantadine
What are zanamivir, oseltamivir, and adamantadine used to treat?
- influenza virus
What is a key difference in RNA viruses after attachment to host membrane?
- RNA viruses internalize into endosomes which then become acidified
How does acyclovir and valacyclovir select for viruses?
- catalyzed by viral thymidine kinase but not host cell Tkase (100-fold selectivity)
What is general MOA of acyclovir, valacyclovir, ganciclovir, and valganciclovir?
- nucleoside analogues that inhibit viral DNA polymerase
What is therapeutic use of oral acyclovir and valacyclovir? (1)
- genital herpes
What is use of IV acyclovir and valacyclovir? (3)
- herpes simplex encephalitis
- neonatal HSV
- serious HSV and VZV infections
What is primary mechanism of resistance to acyclovir and valacyclovir?
- alteration in viral tyrosine kinase or viral DNA polymerase
Who is at risk for complicated CMV infections? (2)
- unborn babies
- immunocompromised patients
In immunocompromised patients, reactivation of latent infection can cause what? (5)
- CMV retinitis
- colitis
- CNS disease
- esophagitis
- pneumonia
What are therapeutic uses of ganciclovir and valganciclovir?
- CMV retinitis
- CMV prophylaxis in transplant patients
Which of the two agents used to treat CMV is preferred?
- valganciclovir over ganciclovir
- it has 100x greater activity
What are side effects of ganciclovir and valganciclovir?
- myelosuppression
- nausea, diarrhea, fever, rash, headache
What are 2 mechanisms of resistance to ganciclovir and valganciclovir?
- decrease in formation of triphosphate form
- mutations in DNA polymerase
How are the "ciclovirs" activated?
- must be phosphorylated to the active triphosphate form
What is general MOA of foscarnet and its therapeutic use?
- inhibits viral DNA polymerase at different site than ciclovirs
- treats CMV infections
What is main way foscarnet is different than the ciclovirs?
does not require phosphorylation or activation
What are main 2 therapeutic uses of foscarnet?
- CMV retinitis
- acyclovir-resistant HSV and VZV
What are 3 main toxicities of foscarnet?
- nephrotoxicity
- electrolyte imbalances
- HA, seizures, hallucinations
What is main mechanism of resistance to foscarnet?
- point mutation in viral DNA polymerase
What is MOA of amantadine?
- blocks M2 proton channel that acidifies the virus and promotes uncoating of viral RNA
What is therapeutic effect of amantadine, zanamivir, and oseltamivir?
- prevents 70-90% of illness when initiated before exposure
- reduces symptoms by 1-2 days if initiated after onset of illness
What are side effects of amantadine?
- GI distress
- CNS effects
What is main resistance mechanism of amantadine and how common is it?
- point mutation in M2 proton channel
- fairly widespread
What is MOA of zanamivir and oseltamivir?
- sialic acid derivatives that act as neuramidase inhibitors
- block release of progeny influenza virus
How are zanamivir and oseltamivir administered?
zan: inhalation
osel: orally
What strains show resistance to oseltamivir and where is mutation?
- H5N1 (avian)
- H1N1 (swine)
- single mutation in neuramidase
What is HAART and what are the 4 classes of drugs used?
"highly active anti-retroviral therapy"
- NRTIs (nucloetide/side reverse transcriptase inhibitors)
- NNRTIs (non-NRTIs)
- INSTIs (integrase strand transfer inhibitors)
- PIs (protease inhibitors)
What is enfuviritide and what does it treat?
- fusion inhibitor
- treats HIV
What is Maraviroc and what does it treat?
- CCR5 antagonist that binds to co-receptor and blocks viral binding to host cell
- treats HIV
What are tenofovir, emtricitabine, and efavirenz and what do they treat?
- NRTIs and NNRTIs
- block viral reverse transcriptase
- treat HIV
What is raltegravir and what does it treat?
- integrase inhibitor; inhibits integration into host DNA
- treats HIV
What are atazanavir, darunavir, and ritonzvir and what do they treat?
- protease inhibitors; block processing of long peptide product produced by translation
- treats HIV
What are the NRTIs and their MOA?
- tenofovir and emtricitabine
- competitively inhibit HIV reverse transcriptase and cauase chain termination when incorporated into viral DNA
What is an NNRTI and its MOA?
- efavirenz
- inhibits HIV reverse transcriptase
What are 3 protease inhibitors and their MOA?
- atazanavir, darunavir, ritonavir
- block cleavage which results in immature, non-infectious particles
What is a integrase inhibitor and its MOA?
- raltegravir
- targets integrase that integrates viral DNA into genome (only used incombination with other drugs)
What is a fusion inhibitor and its MOA?
- enfuviritide
- prevents conformational changes required for fusion with host membrane
What is CCR5 antagonist and how is its MOA unique?
- maraviroc
- by targeting a cellular protein instead of a viral protein, resistance would be minimized