• 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

1/124

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;

124 Cards in this Set

  • Front
  • Back
bioavailability of AGs?
Aminoglycosides are not absorbed orally (F < 3%) and must be administered either intravenously or intramuscularly for treatment of systemic infections
how must AG be administered?
AG must be administered either intravenously or intramuscularly for treatment of systemic infections
how are AGs typically administered?
Rapid (bolus) intravenous doses can be given but are not generally recommended; doses are generally given via slow IV infusion over 30-60 minutes
ways AGs can be admin besides IV?
Aminoglycosides may also be administered by the intrathecal, intraventricular, subconjunctival, and intravitreal routes for treatment of infections in systemic sites with poor antibiotic penetration
Are AGs hydrophilic/hydrophobic?
Hydrophilic, ionically charged compounds which tend to be restricted in distribution primarily to intravascular and interstitial fluids, i.e. blood, ascitic fluid, etc.
which tissues is AG penetration poor?
Penetration into pulmonary secretions/tissues, CNS, and avascular sites (e.g. diabetic foot ulcers) is relatively poor

*need high doses to penetrate
how well are AGs distributed? What's the apparent Vd?
Relatively small apparent volume of distribution (Vd) = 0.20 - 0.30 L/kg based on IBW; a Vd of 0.25 L/kg is very commonly used in clinical practice

*found mainly in extracellular fluids
AG Vd commonly used in practice?
0.25 L/kg
which weight is used when calculating Vd of AGs?
(L/Kg)
Kg's are in IBW
which disease states have altered Vd for AG dosing???
Spinal cord injury
Liver disease associated with low albumin and/or ascites
Neonates
Burns (initial phases)
Acute and chronic renal failure
Severe edema
Peritonitis
Critical illness (ICU patients)
how is Vd altered in the following patients:
Spinal cord injury
Liver disease associated with low albumin and/or ascites
Neonates
Burns (initial phases)
Acute and chronic renal failure
Severe edema
Peritonitis
Critical illness (ICU patients)
all have increased Vd
what is the estimated Vd in ICU pts?
0.35 L/kg
what is the estimated Vd in neonates?
0.4-0.5 L/kg
what is the Vd of AGs into adipose tissue?
Vd into adipose tissue is only approximately 0.1 L/kg due to its relatively avascular nature
How do you calculate Vd for obesity or significant from IBW (TBW >20-30% more than IBW)?
2 methods:

Vd = [0.25 L/kg x (IBW)] + [0.1 L/kg x (TBW - IBW)]

**Most common method:
Vd = 0.25 L/kg x [IBW +
0.4(TBW - IBW)]
= 0.25 L/kg x ADW
how are AGs eliiminated?
Aminoglycosides are normally not metabolized, and elimination is >98% through glomerular filtration and renal excretion of unchanged drug
how does AG clearance related to CrCl?
total body clearance (CL) = CLR = calculated CrCL
T1/2 of AGs
t½ = normally 2-3 hours, highly variable due to changes in Vd and CL
how is AG clearance limited in hemodialysis patietns?
Approximately 30-40% of total body stores are eliminated with each session of hemodialysis; limited clearance with peritoneal dialysis
AG patients with altered CL?
Cystic fibrosis
Spinal cord injury
Neonates
Burns
Renal dysfunction/failure
Dialysis
How is AG CL altered in the following patients?
Cystic fibrosis
Spinal cord injury
Neonates
Burns
Renal dysfunction/failure
Dialysis
Cystic fibrosis (increase)
Spinal cord injury (increase)
Neonates (decrease initiallly, then increase after 1 yo)
Burns (increase)
Renal dysfunction/failure (decrease)
Dialysis (decrease)

*when decrease CL, T1/2 increases
Cockroft gault CrCl equation
(140-age) x weight(IBW)
-----------------------------------
72 x Scr

multiply by 0.85 in women
risk factors for AG nephrotoxicity
- daily dose
*- cumulative dose
*- duration of therapy (especially >10 days)
*- dehydration
- hypokalemia
*- previous aminoglycoside therapy
- female sex
- liver disease (cirrhosis, biliary disease)
*- concurrent nephrotoxins
*- Cmin concentrations > 2 mg/L
*- advanced age
*- previous underlying renal dysfunction
- sepsis
- hemorrhage
at what Cmin are patients at risk for nephrotoxicity?
Cmin > 2 mg/L
Decision process for considerations in AG nephrotoxicity
Clinical management of aminoglycoside-induced renal dysfunction depends on the relative risk vs. benefit of continuing aminoglycoside therapy

In many patients, the need for continuing aminoglycosides will necessitate adjusting the daily dose and continuing therapy

In other patients, the aminoglycosides may be safely discontinued without adversely affecting clinical outcomes

The decision of whether to continue or discontinue therapy in the face of aminoglycoside-related toxicity is made on purely clinical grounds and is entirely patient-specific.
consequences of AG ototoxicity?
Cochlear damage begins in the base of cochlea and moves toward the apex with progressive loss of hair cells; once lost, no regeneration of these cells occurs
Hearing loss is initially in the high-frequency range and becomes progressively worse in lower frequencies as well; vestibular damage may also result in dizziness, vertigo, loss of balance
Ototoxicity is less well correlated with serum concentrations, although prolonged Cmin concentrations of > 2 mg/L may place patients at increased risk
Cmax therapeutic ranges from gentamicin and tobramycin:
severe systemic infections; pneumonia)
8 - 10 mg/L (severe systemic infections, pneumonia)
Cmax therapeutic ranges from gentamicin and tobramycin:
(less severe systemic infections)
6 - 8 mg/L (less severe systemic infections)
Cmax therapeutic ranges from gentamicin and tobramycin:
(UTI)
4 - 6 mg/L (urinary tract infections)
Cmax therapeutic ranges for amikacin:
severe systemic infections; pneumonia
25 - 30 mg/L (severe systemic infections, pneumonia)
Cmax therapeutic ranges for amikacin:
other infections
20 - 25 mg/L (other infections)
True or False???

Peak = Cmax
False
Cmax is a very defined parameter at the top of the conc. vs. time curve

peak varies, and is the measurement that occurs when samples are obtained for analysis (below the Cmax ont he conc. vs. time curve)
what parameters do you need to know to do PK calculations?
male/female
Scr
weight
age
height
dx being treated
IBW calculation: males
50 + 2.3 x (inches>60)
IBW calculation: females
45.5 + 2.3 x (inches >60)
which weight do you use when administering AG loading dose?
ADW or TBW
calculate AG loading dose
approx. 2 mg/kg

(ADW or TBW)
how to calculate a maintenance dose for AGs?
calculate CrCl (cockroft-gault)

drug CL = CrCl

estimate Vd: 0.25 L/kg x IBW or 0.25 x (ADW)

estimate eliminatin rate constant: k=Cl/Vd

estimate T1/2= 0.693/k

choose desired Cmax and Cmin(1.0 is easy) serum conc.

choose the duration of IV infusion (30-60 mins)
therapeutic Cmin for gentamicin and tobramycin?
< 2 ml
therapeutic Cmin for amikacin?
<10 ml
calculate AG dosing interval:
Tmax =
[ln(desiredCmax/desiredCmin)/k] + Tin
calculate AG dosing interval using Fish common sense method
T = (3 x T1/2) + Tin
round up to the next most conventient dosage interval (8,12,24,48)
AG maintenance dose calculation
Dose (mg) = (desired Cmax x CL x tin) x [1 – e^ -kτ/1 – e^ -ktin]
Desc. vancomycin (VAN) absorption
Oral absorption of this agent is extremely poor (F < 5%)

*therapeutic concentrations are only achieved after oral dosing in patients with severely impaired renal function who are unable to excrete the small amounts of drug absorbed after multiple doses
which microbe is good to treat due to lack of VAN absorption?
Clostridium difficile colitis

b/c lack of absorption produces extremely high intraluminal conc. of ABs in small doses
how should VAN be administered?
IV infusion over 60 mins for normal doses
why is IM admin. CI for VAN?
intramuscular administration contraindicated due to extreme pain and potential for muscle necrosis
what compartment model does VAN follow?
Vancomycin distribution may be characterized by a two- or three-compartment model

Vd of the central compartment is approximately 0.2 - 0.6 L/kg, with an alpha distribution half-life of approximately 20 minutes
Vd of the central compartment of VAN?
Vd of the central compartment is approximately 0.2 - 0.6 L/kg, with an alpha distribution half-life of approximately 20 minutes
steady state Vd of VAN?
Steady-state Vd is approximately 0.5 - 0.9 L/kg (TBW) with a terminal half-life of 6 - 8 hours in patients with good renal function
protein binding of VAN?
Vancomycin is approximately 50% protein-bound in patients with normal renal function
tissue penetration of VAN?
Penetration of vancomycin is adequate for most tissues/fluids, but distribution into the CSF is poor and intrathecal or intraventricular administration is often recommended for CNS infections
VAN metabolism in normal renal fxn?
Metabolism of vancomycin is very minimal (5%) in patients with normal renal function
Unclear whether metabolism occurs in the liver or in extra-hepatic sites
primary form of VAN elimination?
Elimination of vancomycin is primarily by glomerular filtration and excretion of unchanged drug (apparently little or no tubular secretion)
total body clearance of VAN?
Total body clearance (CLs)
= 0.7 x CrCl
half life of VAN
normally 6-8 hrs, but highly dependent on renal fxn
T1/2 in ESRD can reach 140-160 hrs
how is VAN cleared by hemo/peritoneal dialysis?
poorly cleared, but some newer methods are more effecient
ESRD on dialysis, the dose can last up to a week
high flux dialysis machin, the dose only last 2-3 days(dont use VAN)
what VAN nephrotoxicity originallky caused by?
A high incidence of nephrotoxicity originally related to vancomycin has since been attributed to the presence of nephrotoxic impurities contained within the original product formulations
These impurities have been largely removed and the nephrotoxic potential of vancomycin is now generally felt to be quite low
factors that put pt. at higher risk for nephrotoxicity due to VAN?
Combination therapy with aminoglycosides

Other concurrent nephrotoxins
Increased age

Previous underlying renal dysfunction

***Prolonged, elevated Cmin concentrations ( > 15 mg/L)

Doses > 4 grams/day

Patients with these risk factors may warrant closer clinical monitoring for signs of early toxicity (elevated creatinine)
most important factor regarding VAN nephrotoxicity
***Prolonged, elevated Cmin concentrations ( > 15 mg/L)
VAN otottoxicity
Characteristically manifests clinically as tinnitus and/or hearing loss in the high-frequency range
[VAN] associated with ototoxicity
Ototoxicity has primarily been reported in patients with Cmax serum concentrations > 80 mg/L

*Since the minimum bactericidal concentration (MBC) of vancomycin for staphylococci and streptococci is usually < 2 - 4 mg/L, there is little to justify concentrations this high
Traditionally VAN dosing
Vancomycin has traditionally been dosed to achieve Cmax = 30-40 mg/L and Cmin = 5-15 mg/L

*doesnt make sense to dose to a certain Cmax b/c VAN is TIME DEPENDENT
VAN clinical response/toxicities related to conc.
Vancomycin clinical response, toxicities actually poorly correlated with serum concentrations

Data relating outcomes to Cmax mostly anecdotal and difficult to reproduce
how quickly do VAN antimicrobial effects occur?
Antimicrobial effects of vancomycin occur relatively slowly (“slowly bactericidal”)
standard VAN dosing
Standard doses of vancomycin (~30 mg/kg/day, or 1 gram every 12 hours) usually achieve concentrations that provide favorable % Time>MIC
Typical MICs of usual pathogens are < 4 mcg/ml
monitoring VAN conc.
Monitoring of serum concentrations is not routinely indicated for patients with normal renal function or mild-moderate insufficiency
which is better sign of antimicrobial activity for VAN:
AUC/MIC or %T>MIC
More recent data suggest that AUC24/MIC ratios may actually be better predictors of vancomycin microbiological and clinical activity than %T>MIC
AUC24/MIC ratios  350-400 have been shown in several studies to be associated with better clinical outcomes in patients with pneumonias and other severe infections
Higher Cmin concentrations are associated with increased AUC24/MIC ratios
therapeutic ranges of VAN:
higher trough
More recently, the use of higher trough concentrations has been advocated for the treatment of more severe infections (e.g., pneumonia, CNS infections, sepsis).
why are higher [VAN] used for severe infections?
Reasons for increased concentrations include recognition of vancomycin’s poor distribution into lung tissues, increasing MICs of many staphylococcal and enterococcal strains, and little risk of increased toxicity.
Trough concentrations of 15-20 mg/L are now often recommended (or even higher, e.g. 20-25 mg/L) in patients who are failing vancomycin therapy or have very severe infections
Some evidence for increased risk for nephrotoxicity with trough levels this high, also with doses >4 gms/day
what is recommended [VAN] trough level for pts failing therapy or with severe infections?
Trough concentrations of 15-20 mg/L are now often recommended (or even higher, e.g. 20-25 mg/L) in patients who are failing vancomycin therapy or have very severe infections
Some evidence for increased risk for nephrotoxicity with trough levels this high, also with doses >4 gms/day
what are the 3 positions for when VAN conc. should be routinely monitored?
Position #1:
Vancomycin concentrations should be routinely monitored due to the risk of potentially serious clinical failures and adverse effects which may be associated with serum concentrations outside of the therapeutic range

In addition, routine monitoring of concentrations is an accepted standard of medical practice

Position #2
Routine monitoring of vancomycin serum concentrations is unwarranted due to lack of hard clinical data relating serum concentrations to either efficacy or toxicity Monitoring should be reserved for high-risk patients, e.g.:

Patients with significant renal dysfunction

Patients receiving concurrent therapy with known nephrotoxins

Critically ill patients in whom significant variation in pharmacokinetic parameters is likely to exist

Patients with severe infections in whom treatment failure would have disastrous consequences

Position #3
Vancomycin serum concentrations should NEVER be monitored in ANY patient, under ANY circumstances
what is the best position to take for routine measurement of VAN conc.?
Routine monitoring of vancomycin serum concentrations is unwarranted due to lack of hard clinical data relating serum concentrations to either efficacy or toxicity Monitoring should be reserved for high-risk patients, e.g.:

Patients with significant renal dysfunction

Patients receiving concurrent therapy with known nephrotoxins

Critically ill patients in whom significant variation in pharmacokinetic parameters is likely to exist

Patients with severe infections in whom treatment failure would have disastrous consequences
high risk patients that require routine monitoring of VAN conc?
Patients with significant renal dysfunction

Patients receiving concurrent therapy with known nephrotoxins

Critically ill patients in whom significant variation in pharmacokinetic parameters is likely to exist

Patients with severe infections in whom treatment failure would have disastrous consequences
initiate VAN therapy:
loading dose?
Loading doses of vancomycin are not typically recommended due to the very high peak concentrations produced with standard dosing regimens
initiate VAN therapy:
calculate a maintenance dose
The method for doing this is virtually identical to that used for the aminoglycosides; same steps, same equations. Remember to use kinetic parameters specific for vancomycin:
a.Estimate Vd = 0.7 L/kg(based on TBW; may also use ADW)

b.Estimate CL = 0.7 x CrCL (remember, CrCL is based on IBW or ADW)

c.Estimate kel

d.Estimate t½

e. Choose desired Cmax and Cmin = anywhere within the accepted "therapeutic range“ (typically a Cmax of 30 mg/L and Cmin of 10 mg/L)

f. Choose duration of infusion, tin = usually approx. 60 minutes

g. Calculate dosing interval, t = traditionally, every 12, 24, 48, etc. hours

h. Calculate dose

i. Double-check Cmin
ἀγαθόs
good
concentration sampling strategies for VAN
Same considerations as with the aminoglycosides, except vancomycin peak concentrations are usually drawn 60 minutes after the end of the infusion instead of 30 minutes as with aminoglycosides
VAN dosage regimen serum concentrations
a.Calculate k
b.Calculate CL from patient-specific serum concentrations
c.Calculate t½
d.Calculate Vd
e.Choose Cmax and Cmin
f.Choose duration of infusion, tin
g.Calculate dosing interval, t
h.Calculate dose
i.Double-check Cmin
basic tetracycline structure?
All tetracyclines are derivatives of the basic naphthacene structure
(4 benzene rings)
what do diff. substitutions on naphthacene structure do?
Different substitutions significantly alter the agents’ antibacterial activities and pharmacokinetic properties
short acting tetracyclines?
tetraxycline, oxytetracycline
intermediate acting tetracyclines
demeclocycline
long acting tetracyclines
doxycycline, minocycline
importance of tetracycline stereochemistry?
5 stereocenters
only one stereocenter, C6-OH, can be altered without destroying the activity of the drug
If any other stereocenter is modified, the activity of the drug is destroyed
reactivity chemistry of tetracyclines
Tetracyclines are chemically very reactive compounds

Chelation of di- and trivalent cations is clinically relevant
MOA tetracyclines
In Gram-negative bacteria, passive transport through porin channels is followed by energy-dependent active transport across inner cytoplasmic membrane
Drug permeation in Gram-positive bacteria is not well characterized
Drugs reversibly bind primarily to the 30S ribosomal subunit
Block binding of the aminoacyl-transfer RNA to the acceptor site on the messenger RNA-ribosome complex
Inhibition of bacterial protein synthesis

Characterized as bacteriostatic

**not O2 dependent, works MOA works in anaerobes too
tetracyclines cidal or static?
inhibition of protein synthesis
=== bacteriostatic

*AGs are the only cidal protein synthesis inhibitors
tetracycline resistance
Decreased accumulation of drug due to decreased influx, or acquisition of an energy-dependent efflux mechanism

Mutations of ribosomal binding site also common

Resistance often chromosomal and constitutive, but may also be plasmid-mediated
Plasmid-mediated resistance is usually inducible
Bacteria become resistant after exposure to drug

Resistance to one tetracycline usually implies resistance to all
-->Exception is minocycline
tetracycline absorption
Good oral absorption from the GI tract
Bioavailability:
Tetracycline, oxytetracycline, demeclocycline = 60-80%
Doxycycline, minocycline = 90-100%

Absorption substantially altered by presence of food and/or dairy products
Tetracycline bioavailability decreased by >50%
Doxy, minocycline reduced by up to 20-30%

Very effective chelators of di- and trivalent cations
Dairy products, aluminum hydroxide gels, calcium, magnesium, iron salts
decrease in tetracycline bioavailability when taken with food/dairy products
Tetracycline bioavailability decreased by >50%
Doxy, minocycline reduced by up to 20-30%
tetracycline distribution
Vd is quite large (1.5-2.5 L/kg)
Distribute well into nearly all tissues, although CNS penetration is less than other sites
Inflammation of meninges is not required for drug entry into CSF
Vd tetracyclines
large (1.5-2.5 L/kg)
tetracycline excretion
Most tetracyclines excreted as unchanged drug in the urine or feces, little to no hepatic metabolism
Tetracycline = >60-70% renal excretion as unchanged drug
Demeclocycline = 45-55% renal, 30% fecal
Doxycycline = 30-40% renal as unchanged drug
Minocycline = 10-20% renal excretion as unchanged drug, seems to uniquely undergo hepatic metabolism
Biliary excretion with enterohepatic recycling common
tetracycline absorption
Good oral absorption from the GI tract
Bioavailability:
Tetracycline, oxytetracycline, demeclocycline = 60-80%
Doxycycline, minocycline = 90-100%

Absorption substantially altered by presence of food and/or dairy products
Tetracycline bioavailability decreased by >50%
Doxy, minocycline reduced by up to 20-30%

Very effective chelators of di- and trivalent cations
Dairy products, aluminum hydroxide gels, calcium, magnesium, iron salts
decrease in tetracycline bioavailability when taken with food/dairy products
Tetracycline bioavailability decreased by >50%
Doxy, minocycline reduced by up to 20-30%
tetracycline distribution
Vd is quite large (1.5-2.5 L/kg)
Distribute well into nearly all tissues, although CNS penetration is less than other sites
Inflammation of meninges is not required for drug entry into CSF
Vd tetracyclines
large (1.5-2.5 L/kg)
tetracycline excretion
Most tetracyclines excreted as unchanged drug in the urine or feces, little to no hepatic metabolism
Tetracycline = >60-70% renal excretion as unchanged drug
Demeclocycline = 45-55% renal, 30% fecal
Doxycycline = 30-40% renal as unchanged drug
Minocycline = 10-20% renal excretion as unchanged drug, seems to uniquely undergo hepatic metabolism
Biliary excretion with enterohepatic recycling common
which tetracyclines need renal dosing adjustments?
Tetracycline = >60-70% renal excretion as unchanged drug

Demeclocycline = 45-55% renal, 30% fecal

Doxycycline = 30-40% renal as unchanged drug
spectrum of activity for tetracyclines
Gram + aerobes: intrinsic activity against streptococci and staphylococci, including some strains of MRSA and VRE
Best activity against MRSA: Minocycline > Doxycycline >> others

Gram - aerobes: active against wide range of bacteria but limited by PK considerations
urinary concentrations sufficient for treating UTI due to E. coli and other enteric bacteria
Systemic use usually reserved for treatment of “unusual” infections, e.g. tularemia, plague

Anaerobes: Originally active against many anaerobes, but resistance now limits use

Atypicals: Good activity, especially against Chlamydia

Other: H. pylori, rickettsial pathogens (e.g. Rocky Mountain Spotted Fever, Lyme Disease, Q fever)
adverse effects of tetracyclines
Hypersensitivity reactions (rare) = anaphylaxis, urticaria, periorbital edema, fixed drug eruptions, rashes

Phototoxicity = most frequent with demeclocycline, least frequent with minocycline
Occurs within minutes to hours after sun exposure

Deposition in bone and teeth = chelation
Tetracyclines cause discoloration of teeth
Could cause temporary stunting of growth
Should not be used during pregnancy or in children <8 years of age

Gastric discomfort = irritation of gastric mucosa

Hepatotoxicity = most common with tetracycline, esp. in pregnant women and those with hepatic or renal dysfunction

CNS = vestibular symptoms with minocycline (30-90%)

Superinfections = overgrowth of Candida

Thrombophlebitis with IV administration
drug interactions with tetracyclines
Drugs or food containing di- or trivalent cations
Aluminum, calcium, iron, magnesium
Antacids, iron preparations including multivitamins
Should be administered 2-3 hours before or 2 hours after antibiotic
Warfarin (increase bleed risk)
Oral contraceptives
what is the result of tetracycline deposition in bone/teeth?
chelation (w/ Ca2+)
Tetracyclines cause discoloration of teeth
Could cause temporary stunting of growth
Should not be used during pregnancy or in children <8 years of age
what is absolute CI to tetracyclines due to tetracycline depositon in bone/teeth?
pregnancy
children < 8 yo
which tetracycline causes vestibular symptoms?
minocycline (30-90%)
clinical uses of tetracyclines
Considered to be very broad-spectrum agents, but resistance often limits clinical use for “routine” bacterial infections
Alternative therapy for a wide variety of bacterial, chlamydial, mycoplasmal, and rickettsial infections
Minocycline becoming important in treatment of community-acquired infections caused by MRSA
Doxycycline is usually the preferred agent within the class for most infections
Well tolerated
Improved compliance (BID dosing)
May be administered intravenously
Demeclocycline rarely used for infectious indications
why were glycylcyclines developed?
drug resistance
glycylcyclines are not affected by major mechs. of tetracycline resistance: ribosomal modification, active drug efflux
tigecycline spectrum of activity
Highly active against Gram-positive bacteria
Staphylococcus aureus (including MRSA)
S. epidermidis (including MRSE)
Most streptococci
Enterococci (including VRE)

Excellent activity against many Gram-negative organisms, including nosocomial strains
Enterobacteriaceae, Citrobacter, Acinetobacter, Stenotrophomonas
Poor activity against Pseudomonas aeruginosa

Excellent activity against clinically relevant anaerobes
Bacteroides fragilis, other Bacteroides group, Fusobacterum, Clostridium, Peptostreptococcus
tigecycline PK
Mean Cmax at steady state = 0.87 μg/mL after 30-minute infusion
Mean T1/2 = 42 hours
Extensively distributed into most tissues
60% eliminated through biliary/fecal excretion, 40% renal
No adjustments needed in renal impairment, or Child-Pugh class A and B hepatic impairment
can tigecycline be used for blood stream infections?
NO, Cmax at steady state is only 0.87 micg/ml after 30 min infusion...extensive tissue distribution, doesnt stay in serum
most common SE tigecycline
Generally well tolerated; GI adverse effects common

Nausea = 30% (usually during infusion),
vomiting = 20%
Most GI complaints mild in nature, subside after first 1-2 days
Discontinuation rates due to GI adverse effects only ~1%
clinical uses of tigecycline
Very broad spectrum of activity
Includes many important multidrug-resistant Gram-positive and Gram-negative pathogens
Aerobic and anaerobic activity well suited to treatment of mixed infections
Demonstrated clinical efficacy in complicated skin/skin structure and intra-abdominal infections
Generally well tolerated, no significant toxicities
Lack of P. aeruginosa activity is problematic

*not 1st line for anything, alternative drug
colistimethate/colistin (polymixin E)
colistimethate is a prodrug rapidly hydrolyzed to colistin
MOA colistin
Large cyclic polypeptide with MW ~ 1750
After IV or IM administration, colistimethate rapidly hydrolyzed to the pharmacologically active colistin base

Colistin acts as a cationic detergent
Disrupts & damages bacterial cytoplasmic membrane
Causes leakage of intracellular components and eventual cell death
Classified as a bactericidal drug
is colistin cidal or static?
bactericidal
PK colistin
PK/PD properties of colistin not well described
Usually characterized as concentration-dependent
Absorption <2-3% after oral administration, must be given parenterally for treatment of systemic infections
Distribution into most tissues is considered to be good (except CNS)
Often administered as aerosolized drug for adjunctive treatment of pulmonary infections
Eliminated approx. 65% to 75% as unchanged drug in the urine
Plasma half-life ~ 4-8 hours, but may be increased to > 48-72 hours in severe renal impairment
colistin elimination
Eliminated approx. 65% to 75% as unchanged drug in the urine
plasma T1/2 colistin
Plasma half-life ~ 4-8 hours, but may be increased to > 48-72 hours in severe renal impairment
how must colistin be given?
parentally for systemic infections b/c absorption <2-3% after oral admin.
SEs colistin
Nephrotoxicity
May occur in up to 30-35% of patients
Particularly problematic in patients receiving other nephrotoxins, or those with previous renal impairment or disease

CNS toxicity
Facial and peripheral paresthesias & numbness, dizziness, vertigo, slurring of speech

Generalized rash and pruritis

Gastrointestinal disturbances

Hypersensitivity reactions

Superinfections, e.g. Clostridium difficile
what is the most problematic SE of colistin?
Nephrotoxicity (30-35%)
spectrum of activity for colistin?
Activity limited to primarily "Gram-negative aerobes"
Enterobacteriaceae, Pseudomonas aeruginosa, Acinetobacter species

Resistance to colistin is quite unusual and activity is retained against organisms that are resistant to multiple other antibiotics
“MDR” = multidrug-resistant
clincial use of colistin?
Clinical use of colistin is limited to infections caused by MDR Gram-negative bacteria that:
Are not susceptible to other drugs, and/or
Have already failed therapy with other drugs

*works when nothing else does...drug of last resort b/c of toxicity