• 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

1/256

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;

256 Cards in this Set

  • Front
  • Back
MIC =
Lowest concentration of antibiotic required to inhibit bacterial growth
Determined in vitro by several different techniques
Used as a measure of antibiotic potency, i.e. the lower the MIC the more potent the antibiotic
Commonly used for comparison of activities of various antibiotics
MBC=
minimum bactericidal concentration
Lowest concentration of antibiotic required to kill bacteria
what MIC is most potent and what one is most pharmacologically active?

MIC= 0.12, 1, 0.25, 2
0.12 is the most potent, but it may not be the most effective so cannot determine what one is the most pharmacologically active
Gatifloxacin vs. Standard Therapy or Levofloxacin for Community-Acquired Pneumonia
916 patients in three separate trials randomized to receive one of four antimicrobial regimens
Studies included either outpatient or inpatient treatment x 7-14 days

Gatifloxacin 400 mg QD IV/PO
- 427 / 446 (96%)

Clarithromycin 500 mg PO BID
- 175 / 188 (93%)

Ceftriaxone 1-2 g IV QD + ECN or Clarithromycin
- 95 / 106 (90%)

Levofloxacin 500 mg IV/PO QD
- 165 / 176 (94%)

there is no clinical of statistical difference between cure rates
Factors Influencing Clinical Outcome and antibiotices
BUG: Virulence factors
Intrinsic susceptibility
Resistance mechanisms

DRUG: MOA
In vitro MICs
PK properties

HOST: Genetic determinants
Underlying illnesses
- Altered PK/PD
Antimicrobial Pharmacodynamics
Correlates the drug concentration with the clinical effect (i.e., bacterial killing) (or inhibition)
Uses surrogate markers such as minimum inhibitory concentration (MIC)
Indexes serum concentration (PD property) (pharmacokinetic profile) to MIC to predict clinical outcomes
The pharmacodynamic relationship is different for different antimicrobial classes

what the drug is doing to the bacteria which is related to drug concentration
Optimizing Antimicrobial Therapy
antibiotic--->(PK properties)---> concentration at site of infection and Pathogen MIC/MBC--->(PD properties) host factors/bacterial killing all determine clinical cure
what property of a drug is more important to determine the concentration of the drug at the site of infection PK or PD
PK
what property governes what a drug does in the patient PK or PD
PK
what a graph looks like for an antibiotic that is time dependent
woman, ninja (n.)
مُنَقَبَة
list of time dependent antibiotics
penicillins
cephalosporins
monobactams
carbapenems
macrolides
clindamycin
oxazolidiones
azithromycin
vancomycin
list of concentration dependent antibiotics
aminoglycosides
fluoroquinolones
metronidazole
ketolides
daptomycin
telavancin
with time dependent antibiotics what is influencing the efficacy
time>MIC

how long the drug concentration is above the MIC
with concentration dependent antibiotics what is influencing the efficacy
Peak/MIC

cmax
PD Parameters Predictive of Outcome (time dependent examples and therapeutic goal)
b-lactams
Macrolides
Vancomycin


Optimise duration
of exposure
PD Parameters Predictive of Outcome (concentraion dependent examples and therapeutic goal)
Cmax/MIC
Quinolones
Aminoglycosides

Optimise magnitude
of exposure

(side not quinolones are also dependent on the AUC/MIC not just cmax and MIC)
If the total daily dose of a drug is 4 Gm/day, what is the best way to administer the drug in order to optimize dosing from a pharmacodynamic perspective?
If the drug is concentration-dependent, administer as a 4 Gm QD regimen

If the drug is time-dependent, administer as a 1 Gm QID regimen
Effect of Changing Dose or Dosing Interval on Pharmacodynamic Parameters
Effect of Changing Dose or Dosing Interval on Pharmacodynamic Parameters
Effect of Changing Dose or Dosing Interval on Pharmacodynamic Parameters
Optimizing Pharmacodynamics of Antimicrobials

Concentration-Dependent Drugs:
Give relatively large doses less frequently to maximize Cmax and/or AUC

relative to MIC
Optimizing Pharmacodynamics of Antimicrobials
Time-Dependent Drugs
Give smaller doses more frequently to maintain higher Css, ave
Time-dependent bactericidal activity
e.g., b-lactams and Macrolides
Rate and extent of bacterial killing does not increase with increasing drug concentration
Clinical Goal: Maximize drug duration, i.e., time serum level remains above the MIC (Time > MIC)
Time above MIC is expressed as a percentage relative to the dosing interval (τ)
6 hrs above MIC with τ = 8 hrs Time > MIC = 75%
what is time above MIC expressed in
Time above MIC is expressed as a percentage relative to the dosing interval (τ)
6 hrs above MIC with τ = 8 hrs Time > MIC = 75%
what will a continuous infusion result in
100% T>MIC

which is not necessarily better, clinical benefits tend to peak at 50% T>MIC
when a antibiotic is time dependent and the MIC50 and cmax perameters are given can you determine what drug is best
nope because it is time dependent
The “In vitro – in vivo Paradox of antibiotics
Patients infected with “resistant” pathogens may often be effectively treated despite the apparently poor in vitro activity of the drug being used


this is due to the fact that tissue concentrations are different and the patient may have good immune funtion which are not taken into account in vitro
are antibiotic breakpoints based on plasma or tissue concentrations
plasma because done in vitro, so if the site of infection is in the tissues the effectiveness of the drug is hard to determine
Fluoroquinolone Pharmacodynamics Cmax/MIC ratio
> 10-12 correlated with favorable clinical & microbiological response
Fluoroquinolone Pharmacodynamics AUC/MIC ratio against gram - infections
> 125-250 associated with increased likelihood of clinical efficacy and decreased resistance in Gram-negative infections
Fluoroquinolone Pharmacodynamics AUC/MIC ratio against gram + infections
AUC/MIC > 30-50 associated with optimal in vitro activity and decreased resistance in Gram-positive infections
goal of fluoroquinolones with gram + and - infections
Goal:
AUC:MIC > 125 gram – organisms
AUC:MIC > 30-50 gram + organisms
if you have a choice of cipro, levo, gati or moxi whos AUC ratios are all will above the MIC except cipro to treat MSSA does it matter which one you choose
the only one that would not be efficacios is cipro all the others would work equally as well
why are none of the fluoroquinolones used againts MRSA
because none of their AUC ratios are above the AUC/MIC point where they are effective

MRSA is resistant
what 3 fluoroquinolones are effective against s. peumoniae and why
gati, levo and moxi because the ration of AUC.MIC is well above the point to be efficacious

whereas cipro is not
Fluoroquinolone Pharmacodynamics AUC/MIC ratio against gram + infections
AUC/MIC > 30-50 associated with optimal in vitro activity and decreased resistance in Gram-positive infections
Fluoroquinolone Pharmacodynamics AUC/MIC ratio against gram + infections
AUC/MIC > 30-50 associated with optimal in vitro activity and decreased resistance in Gram-positive infections
goal of fluoroquinolones with gram + and - infections
Goal:
AUC:MIC > 125 gram – organisms
AUC:MIC > 30-50 gram + organisms
goal of fluoroquinolones with gram + and - infections
Goal:
AUC:MIC > 125 gram – organisms
AUC:MIC > 30-50 gram + organisms
if you have a choice of cipro, levo, gati or moxi whos AUC ratios are all will above the MIC except cipro to treat MSSA does it matter which one you choose
the only one that would not be efficacious is cipro all the others would work equally as well
if you have a choice of cipro, levo, gati or moxi whos AUC ratios are all well above the MIC except cipro to treat MSSA does it matter which one you choose
the only one that would not be efficacios is cipro all the others would work equally as well
why are none of the fluoroquinolones used againts MRSA
because none of their AUC ratios are above the AUC/MIC point where they are effective

MRSA is resistant
why are none of the fluoroquinolones used againts MRSA
because none of their AUC ratios are above the AUC/MIC point where they are effective

MRSA is resistant
what 3 fluoroquinolones are effective against s. peumoniae and why
gati, levo and moxi because the ration of AUC.MIC is well above the point to be efficacious

whereas cipro is not
what 3 fluoroquinolones are effective against s. peumoniae and why
gati, levo and moxi because the ration of AUC.MIC is well above the point to be efficacious

whereas cipro is not
Aminoglycoside PK/PD Relationships in Pseudomonas Bacteremia (graph)
when the cruve platues there is not better efficacy in the drug just more of a risk of toxicities
Aminoglycoside PK/PD Relationships in Pseudomonas Bacteremia
when the cruve platues there is no added benefit it puts the patient ar more of a risk of toxicities
Aminoglycoside Pharmacodynamics
Maximum bactericidal effects and improved clinical response correlated with Cmax 8-10 x MIC
Achieving high Cmax:MIC ratios also correlated with decreased selection of resistant bacteria
Uptake of drugs into renal tubular cells and endolymph of ear most efficient with low, sustained concentrations
High, transient concentrations may minimize this uptake
Probably due to saturable active transport processes
what Cmax should be reached fro AG to be efficacious
Cmax 8-10 x MIC
Achieving high Cmax:MIC ratios also correlated with decreased selection of resistant bacteria
is the uptake of AG saturable
yes
Uptake of drugs into renal tubular cells and endolymph of ear most efficient with low, sustained concentrations
High, transient concentrations may minimize this uptake
Probably due to saturable active transport processes
Aminoglycoside PK/PD Relationships in Pseudomonas Bacteremia
ther is no added benefits after the cruve has platued it just puts the patien at higher risk of toxicities
traditional vs. extended dosing of AG
traditional dosing: the drug was given multiple times throughout the day never reaching concetrations of 8-10 times the cmax

now the drug is given QD as a big dose in order to reach higher concentrations, initally clinitians were afraid of toxic affects on the kidney so did not want high concentrations

However, the process of uptake into the kidneys is saturable so even though higher doses are given all at once the kidneys are not at higher risk of nephrotoxicities
Pharmacodynamic Considerations in Aminoglycoside Dosing
Use of “once daily” or extended-interval dosing regimens provides:
More optimal pharmacodynamic parameters, enhanced bactericidal activity
Efficacy similar to divided-dosing regimens
Potentially decreased toxicities relative to divided-dosing regimens
Decreased reliance on serum concentrations and pharmacokinetic monitoring
Potentially decreased cost of therapy

taking advantage of the PK/PD properties
why are infusion times extended
so the time above MIC increase, therefore bugs that were previously resistant are now suscetable
Continuous Infusions of -lactams for ICU Pulmonary Infections
“Presently, there is insufficient evidence to warrant the replacement of intermittent infusion by continuous infusion -lactams as the new standard for all patient populations. Undoubtedly, continuous infusions can be used to maximize -lactam pharmacodynamics; however, it is still unclear whether continuous infusions will improve patient outcomes, reduce antibiotic resistance, minimize toxicities related to peak concentrations, or decrease healthcare costs.”
Piperacillin/tazobactam for P. aeruginosa Infections: Extended vs. Intermittent Infusions
Cohort study of patients receiving extended 4-hr infusions (N = 102) vs. standard intermittent infusions (N = 92)
18 gm/day extended vs. 12-18 gm/day intermittent
Overall results showed no significant differences between groups in mortality or median LOS
Extended infusion group showed significantly lower mortality (P = 0.04) and median LOS (P = 0.02) in subset of patients with APACHE II score ≥17 receiving extended infusion vs. intermittent infusions
No significant differences in patients with APACHE II < 17 (the higher the score the sicker)

overall, no difference in use of extended infusions, however
in sicker patients extended infusions improved outcomes
ager, agri
field, territory
Clinical Efficacy of Doripenem 500 mg Q8H (4-hr infusion) vs. Imipenem 2-3 gm/day (0.5-1hr infusion) in Ventilator-Associated Pneumonia
overall there was not much of a difference in outcomes, but when looking at the sicker patients they had better outcomes with extended infusion times
Continuous Infusions of Time-Dependent Antibiotics for Gram-Positive Infections
No formal recommendations for use of continuous infusion therapy
May consider for treatment of Gram-positive infections in patients who:
Have not shown adequate clinical or microbiological response to conventional dosing regimens
Have infections in which drug penetration to site of infection may play a role in treatment failure
Clinical situations in which continuous infusions may be considered:
Endocarditis caused by MRSA, coagulase-negative staphylococci, enterococci
Gram-positive meningitis
Septic arthritis, osteomyelitis
Gram-positive pneumonia, pulmonary abscess
Optimal doses, duration unknown
What Does Pharmacodynamics Do?
Correlates drug activity in vitro with pharmacokinetic profile of individual agents
Relates pharmacologic features of drugs to predicted and/or observed clinical outcomes
Aids clinicians in making decisions regarding selection of agents that are most likely to yield favorable clinical results
Aids clinicians in selecting most favorable dosing strategies
What are Appropriate Pharmacodynamic Targets?
Cephalosporins, penicillins
T > MIC of ≥40-50% of dosing interval
Carbapenems
Bacteristatic: T > MIC of 30% of dosing interval (not high enough)
Bactericidal: T > MIC of ≥40-50% of dosing interval
Fluoroquinolones
Gram-positive (S. pneumoniae): AUC/MIC >30
Gram-negative: AUC/MIC >125
Aminoglycosides
Cmax/MIC >8-10
What Does Pharmacodynamics NOT Do?
Does not provide completely accurate predictions of clinical success for any given agent in a specific patient
Does not guarantee optimal therapy under all circumstances
Does not provide easily obtainable information that allows point-of-care decision making for individual patients

using population estimates, making predicitons for everyone therefore not individualizing the treatments
Limitations of Applying Pharmacodynamics in the Clinical Setting
Pharmacodynamics still an evolving discipline
Most data supporting PK/PD applications derived from in vitro and in vivo animal models
Most clinical data retrospective and/or modeled
Prospective data generated in less severely ill populations, not ICU patients
PK data used in studies often not derived from critically ill, may not properly reflect variability
Nearly all data obtained from only a couple of organisms (e.g. S. pneumoniae, P. aeruginosa) & simply assumed that PD relationships are similar for all other organisms
is protein binding taken into account when dealing with PD properties of antibiotics
no
Calculate dosing interval, τ = may be done in one of two ways
Tmax ={ [ln(desired Cmax/desireed Cmin)]/ k} + tin

tin is the infusion time

- OR, the "Fish common sense method":
τ = (3 * t½) + tin, round up to the next most convenient dosage interval (8, 12, 24, 48 hours, etc)
C=
Co(e^-kt)
i.FINALLY, calculate a maintenance dose:
dose (mg)= (desired Cmax* CL* tin)* [(1- e^-kt)/1-e^ktin)}
double check your work by
At this point you should also take the time to double-check your Cmin to be sure that the calculations were done correctly:
Cmin= Cmax * e^-kt

"t" is the amount of time elapsed between Cmax and Cmin
Note that Cmax occurs at the end of the infusion, not when the infusion is started

t= dosing interval-infusion time
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

what is your first step?
calculate IBW and if needed calculate ABW
1.Calculate IBW= 45.5 kg + 2.3 kg * (5 inches)= 57.0 kg

Calculate AD= IBW + 0.4 * (TBW - IBW)
= 57.0 kg + 0.4 * (84.1 kg – 57.0 kg)
= 57.0 kg + 11.0 kg
= 68.0 kg
if start with a loading dose of 2 mg/kg what Cmax does it result in
Cmax of about 6
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.
Calculate the loading dose
2. Calculate a loading dose:

185 pounds = 84.1 kg

Dose = 2 mg/kg * 84.1 kg = 168 mg

(Round up to 170 mg so the IV room pharmacist don't hate you when they get the order)

use TBW because it is only one dose, so be aggressive

can also find the loading dose with the other equation this is the easy way
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

calculate a maintence dose start by....
estimate CrCL= [(140-age)(IBW or ABW(kg))]/72*Scr

=[(140 - 48) * 68/ 72*2.0]*0.85= 37 mL/min
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

after calculating CrCL for the maintence dose what is the next step?
CL (L/hr)
= CrCL (mL/min) * 60 min/hr * 1L/1000mL
= 2.22 L/hr
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

after calculating CrCL and CL for the maintence dose what is the next step?
estimate Vd
= o.25 L/kg * ADW or IBW
= o.25 L/kg * 68 kg
= 17.0 L
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

after calculating CrCL, CL and Vd for the maintence dose what is the next step?
estimate K=CL/Vd

= (2.22 L/hr)/(17L) = 0.130 hr^-1
EXAMPLE
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

after calculating CrCL, CL, Vd and k for the maintence dose what is the next step?
estimate half life= 0.693/k
= 0.693/ 0.130^-1 = 5.3 hrs
why do you not use TBW when calculating Vd for the maintence dose
because you want to be more carefull at this point you are not just giving one dose
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.

choose a desired Cmax and Cmin serum concetrations
Since this could be a moderately severe systemic infection, we choose between either 6 - 8 mg/L or 8 - 10 mg/L as a desired Cmax concentration
A nice mid-point to aim for is 8 mg/L (Notice that there is room for clinical judgment in making this determination.)
Since clinical pharmacokinetics is not an exact science and there is significant room for error, we don't want to cut the Cmin too low
A desired Cmin of approximately 1.0 mg/L is a reasonable place to start (if the patient has more risk factors would go lower around 0.5)

Choose duration of the IV infusion (tin) = 30 minutes (0.5 hr)
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides.
After determining the maintence dose and Cmax and Cmin Calculate dosing interval, τ
tmax= ln[(desired Cmax/desired Cmin)/k] +tin

= [(ln (8/10)/0.130^hr-1)+ 0.5 hr
T= 16.5 hours--> round up to 24 hours

or the "Fish common sense method"
T = (3* t1/2) + tin= (3*5.3 hrs)+0.5 hrs
= 16.4 hours--> round up to 24 hours (producing a Cmin of 0.3-0.4)

if round down to Q12 hours Cmin increases to about 2 because that is close to 1 half life
MIC =
Lowest concentration of antibiotic required to inhibit bacterial growth
Determined in vitro by several different techniques
Used as a measure of antibiotic potency, i.e. the lower the MIC the more potent the antibiotic
Commonly used for comparison of activities of various antibiotics

measures pharmocologic activity
MBC=
Lowest concentration of antibiotic required to inhibit bacterial growth
Determined in vitro by several different techniques
Used as a measure of antibiotic potency, i.e. the lower the MIC the more potent the antibiotic
Commonly used for comparison of activities of various antibiotics
if a drug has a MIC of 0.12 or 0.5 to a particular antibiotic what one is the most potent
the one with 0.12

it is the most pharmacologically active bu tmay not be the most effective
factors influencing clinical outcomes
host: genetics, underlying illness, altered PK/PD

bug: virulence factors, intrinsic susceptablility, resistance mechanisms

drug: MOA, in vitro MIC, PK properties
Antimicrobial Pharmacodynamics
Correlates the drug concentration with the clinical effect (i.e., bacterial killing or inhibition)
Uses surrogate markers such as minimum inhibitory concentration (MIC)
Indexes serum concentration (pharmacokinetic profile) to MIC to predict clinical outcomes
The pharmacodynamic relationship is different for different antimicrobial classes

what drugs do to bacteria which is related to drug concentrations
Limitations of Applying Pharmacodynamics in the Clinical Setting
Pharmacodynamics still an evolving discipline
Most data supporting PK/PD applications derived from in vitro and in vivo animal models
Most clinical data retrospective and/or modeled
Prospective data generated in less severely ill populations, not ICU patients
PK data used in studies often not derived from critically ill, may not properly reflect variability
Nearly all data obtained from only a couple of organisms (e.g. S. pneumoniae, P. aeruginosa) & simply assumed that PD relationships are similar for all other organisms

These points should all make sense based on what we discussed in class. In the second bullet, by “modeled” I mean that many of the studies supporting PK/PD applications came from computer models where MICs and PK parameters are simulated and outcomes predicted based on the model rather than from actual clinical data. This is obviously not the ideal way to validate PK/PD applications in the real world when clinical outcomes of patients are at stake. The third bullet makes the point that, since PK parameters may be very different & highly variable in seriously ill patients compared to normal volunteers, the PK data used in deriving pharmacodynamic parameters & clinical decisions needs to be from the appropriate populations where the results of your predictions will actually be applied.
are PD predictions based on blood concentrations or tissue
The point to be made here is simply that, as stated several times in class, current PD estimates and predictions are mostly based on drug concentrations in blood rather than what’s going on in the tissues at the site of infection. The pharmacokinetic behavior of drugs is obviously quite dynamic and quite complex. Even something as basic as protein binding of drugs is difficult to adequately account for with our current understanding of antibiotic pharmacodynamics – another reason why PD is still an evolving science.
β-Lactam Concentrations vs. MIC Breakpoints in the average patient
In “normal” patients with “average” pharmacokinetics, standard dosing regimens for many drugs are already adequate to provide good pharmacodynamic performance, as in the case with all of these beta-lactams. Standard doses achieve concentrations that are far in excess of pathogen MICs and provide excellent %T>MIC. But this is assuming that all drugs exhibit “average” PK properties in all patients.
Antimicrobial Pharmacokinetic Alterations in Critically Ill Patients
Critically ill patients, e.g. those in the hospital and/or ICU, often exhibit PK parameters that are very different from those found in less-sick patients. Here are a few examples of the differences in PK parameters that can be seen in various populations. We’re obviously not always treating “average” patients and alterations in PK parameters need to be accounted for in order to appropriately determined PD properties.
Application of Appropriate PK and MIC Data is Critical to Clinical Decision-Making
Important limitations of PK data in severely ill populations
PK data often not available
Specific drugs and regimens
Specific patient populations (e.g. medical vs. surgical pts, organ failure)
Great variability in antimicrobial PK, particularly in severely ill populations
MICs not always available for individual patients
Pathogens not identified in many infections
Empiric treatment for entire duration of therapy is common
MICs not routinely determined or reported by many clinical laboratories
Immune status, other comorbidities may effect PK/PD relationships and host response but are not easily accounted for
Immune status, other comorbidities may effect PK/PD relationships and host response but are not easily accounted for
another limitation of PD at this point in time is that we also have no way of taking into account patient-specific factors that may influence response to infections. Remember, for instance, that antibiotics often just suppress infections while the immune system catches up. Immunocompromised patients may respond very differently to our antibiotic regimens; the current state-of-the-art in pharmacodynamics can’t account for that.
Clinical Application of Pharmacodynamics
High, aggressive dosing (within reason) should be routinely used for systemic infections
The more severe the infection, the higher the doses that should be used
Consider PK variability in critically ill patients
Consider consequences of treatment failure with inadequate initial treatment
Risk-versus-benefit considerations important
Potential for increased efficacy
Potential for increased toxicity
Consider consequences of treatment failure with PD
with some infections you just can’t afford to take a lot of chances because if the patient fails due to under-dosing or incorrect dosing, there may be no second chance to get it correct. In a patient with endocarditis, you don’t want their heart valves getting chewed off by the bacteria because you didn’t dose them aggressively enough to meet PK/PD goals. High, aggressive dosing is the only way to make up for the PK variability that may be present, but you can’t overdo it either. You still have to be careful with potentially toxic drugs. If you don’t think you can achieve the desired PK/PD goals without putting the patient at risk for excessive toxicity in the process – pick a different drug.
Pharmacodynamic Dosing Principles
Attempt to choose drugs and dosing regimens that yield optimal pharmacodynamic relationships
Consider not only the appropriate drug, but also:
Appropriate dose
Appropriate dosing interval
Appropriate route
Appropriate application of pharmacodynamics has the potential to enhance bacterial eradication and decrease risk for development of resistance
Using the wrong drug, or using the correct drug but given at too-low doses and/or improper intervals, tends to do the opposite
M.B. is a 48 year old female admitted to the hospital with abdominal trauma following a motor vehicle accident. She is to be started empirically on metronidazole and gentamicin for suspected peritonitis. The physician asks you to calculate a dose of aminoglycoside to begin her treatment. What do you recommend? M.B. weighs 185 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 2.0 mg/dL. Assume that your hospital uses 30 minute infusion times for aminoglycosides

calculate a maintenance dose
k= 0.130hr^-1
desired Cmax= 8 mg/L
CL = 2.22 L/hr
tin= 0.5 hr
(8 mg/L * 2.22L/hr * 0.5hr) [ 1-e^-(0.130/hr* (24 hrs)/1-e^-(0.130/hr)*(0.5 hrs)
= 8.8890.955/0.0634)

= 135 mg Q 24 hrs
Double-check the calculated trough concentration to make sure that the math came out right:
Cmin =Cmax * e ^-k * t
"t" is the amount of time elapsed between Cmax and Cmin
Note that Cmax occurs at the end of the infusion, not when the IV bag is hung on the pole.
"t" to be used in calculating Cmin is then:
τ - tin = 24 hrs - 0.5 hr = 23.5 hours.
Cmin = 8.0 mg/L * e^ -(0.130/hr) * (23.5 hrs)
Cmin = 0.4 mg/L
FINAL ANSWER = 170 mg IV loading dose, followed by 135 mg IV every 24 hrs
when does cmax occur for AG dosing
Note that Cmax occurs at the end of the infusion, not when the IV bag is hung on the pole.
Cmax and Cmin concentrations are first ideally drawn for AG when....
either at the time of the first dose; or when the serum concentrations have achieved steady state, i.e. after the regimen has been continued for a minimum of 3.3 half-lives of the drug (at which time ~90% of steady-state concentrations have been reached)
In actual practice, Cmax and Cmin AG concentrations are commonly first obtained at
the time of the third maintenance dose
In our previous example, this would be 48 hours after the first maintenance dose, at which time the drug should certainly be at steady state
Samples should be drawn based on the calculated t½ and _____ based on the number of doses administered if you want serum concentrations which are able to be interpreted correctly
NOT
Blood drawn for AG Cmax concentration determination should be obtained...
30 minutes after the END OF THE INFUSION
AG Cmin blood samples should be drawn
as close to the beginning of the next dose as possible
how many half lives does it take to get to SS
5
drawing levels of AG is based on what?
half lives
T1/2=
0.693 *Vd/CL
t=
dosing interval - infusion time
(T- tin)
Concentration Sampling Strategies
“Sawchuck-Zaske Method”
Following the first dose of drug (often the loading dose), concentrations are obtained immediately after the end of the infusion + approximately midway through the dosing interval
Useful when accurate information regarding kinetic parameters are needed rapidly, e.g. critically ill patients
Provides accurate information regarding both Vd and elimination rate (k). Vd and k are calculated by the following formula:
(because starting from a concentraion of 0)

Vd= (dose/tin/k)* [1-e^-k*tin/Cmax-(min*e^-k*tin)]

where t = time between Cmax and midpoint serum samples
disadvantages of the “Sawchuck-Zaske Method”
have to be there when the first dose is given (not convient)
what is considered the most accurate measurement of Vd
“Sawchuck-Zaske Method”
Considered most accurate method for determination of Vd because if the patient has not previously received any drug, the Cmin is known to be 0, and the term (Cmin *e-ktin) also simply becomes 0
"Dose-Peak-Trough" method for AG
Concentrations are obtained immediately after the end of the infusion and just prior to the next dose

Gives an accurate estimate of elimination rate but is less accurate for calculating Vd
A disadvantage is that one must wait the entire dosing interval to obtain the second sample and the laboratory results will also be delayed accordingly
Vd and k are calculated by the same equations as in the Sawchuck-Zaske method
"Trough-Dose-Peak" method for AG
A sample is obtained just prior to the dose, the dose is administered, and a second sample is obtained after the end of the infusion

taking two leves from the same dose which is ok because at SS and the trough should be the same as the other dose because at SS

Also gives a more accurate estimation of elimination rate than of Vd
An advantage is that both peak and trough concentrations are obtained in a more timely, convenient manner
Most common method used in actual clinical practice and should be the one that you are most comfortable with
Working with Serum Concentrations Rule #1: If it's not broken, don't try to fix it!
There's usually no reason to modify regimens if:
Concentrations look acceptable
Patient is responding to therapy
No suspicion of actual or impending toxicity

have to look at the levels drawn: do they represent a tru Cmax and Cmin (don't make assumptions
calulate k with measured peak levels
k= ln[Cpeak/Ctrough]/t
are the peak values true cmax and cmin values
no they are the measured values
To correctly choose the correct value for "t" for AG, several different time intervals must be considered:
τ = entire dosing interval, from start of one dose until the next dose
tin = duration of the infusion
tinpk = time between end of infusion and when peak level is drawn
tpktr = time elapsed between when peak is drawn until trough is drawn
tend = time between when trough is drawn until the next dose is given
τ=tin + tin-->pk + tpk-->tr + tend
If the peak concentration is drawn directly at the end of the infusion for AGs
Cpeak = Cmax and tinpk = 0. Likewise, if the trough concentration is drawn immediately before the next dose, Ctrough = Cmin and tend = 0.
what periodof time are you using for t in Ag dosing to figure out T
between Cpeak and Ctrough
Tpk-->tr
example of finding T for AG:
dose was given at 12 pm
Peak taken at 3 pm
tin= 1 hr
trough takne at 11 am
dosing interval 200 mg Q24hrs
24-1-2-1=20

T=24
tin=1
tin--pk=2
tend=1
"t"=
(AG dosing)
T-tin-tin-->pk-tend
example of finding T for AG:
dose was given at 12 pm
Peak taken at 3 pm
tin= 1 hr
trough takne at 11 am
dosing interval 200 mg Q24hrs

"the easy way"
use the difference between trough and peak
24-4=20
does the tin account for elimination
nope
example of finding T for AG:
Dose given at 8am
tin= 0.5 hrs
peak taken at 12 pm
trough taken at 6:30 am
200mg Q 8 hours
8-0.5-3.5-1.5= 2.5

the easy way
8-5.5=2.5
If the "Trough-Dose-Peak" method of sampling is being used, there is a much easier and less confusing way of calculating "t":
ttr-->pk = time between when the trough sample is drawn until the peak sample is drawn
To calculate k:
"t" = τ - ttr-->pk
when using the patients actual serum concentrations how do you find cmax
cmax= cpeak/e^-kt

In this equation,
"t" = tin-->pk , which is the period of time between when Cmax actually occurred and when the peak level was drawn and measured.
when using the patients actual serum concentraions how do you calculate CL
Cl= [dose/tin/cmax]* [1-e^-ktin/10e^-kT)

Dose =dose being used in the dosing regimen from which concentrations are being measured
tin =infusion time actually used with the dose from which the concentrations are being measured
Cmax =value for Cmax which was just calculated in step #4 above (may use Cpeak IF Cpeak = Cmax)
τ =dosing frequency being used in the dosing regimen from which concentrations are being measured

the dose and interval are the ones the patient was receiving when the samples were taken
when using the patients actual serum concentraions how do you calculate Vd
Vd= Cl/k

Now that we have PATIENT-SPECIFIC values for k, CL, and Vd we can proceed to design a new dosing regimen in the same manner as we did previously with our empiric dosing recommendations.

compare with the expected values calculated
what is the highest Vd obtainable with Ag
about 1 L/kg
once all the pateint specific PK values have been calculated for AG choose a desires Cmax and Cmin and calculate the dosing interval
Choose duration of the IV infusion (tin) = usually either 30 minutes or 60 minutes (0.5 or 1 hour)

Calculate dosing interval, τ = may be done in one of two ways:
Tmax={ ln[desired cmzx/desired cmin]/k}+tin

or
τ = (3 * t½) + tin, round up to the next most convenient dosage interval (8, 12, 24, 48 hours, etc.)
If the serum concentrations measured were fairly good for AG....
make minor adjustments, the same dosing frequency which was previously being used in the patient can be kept
If the serum concentrations were very unfavorable for AG
large dosing adjustments will be necessary, and especially if the measured Cmin concentration was > 2 mg/L, the calculations for τ should be done and a new τ used (if necessary)
what is the desired Cmin for AG
< 2 mg/L
M.B., the 48 year old female from the previous example, was begun on the antibiotic regimen which you previously calculated and recommended (gentamicin 135 mg every 24 hours). Serum concentrations are obtained around the third maintenance dose and the following data is reported:

Start of dose =08:00
End of dose =08:30
Trough level = 1.1 mg/L, obtained at 07:45
Peak level = 5.4 mg/L, obtained at 10:00
what needs to be changed??
M.B. continues to be febrile to 38.9 degreees, with a WBC of 24,500 and a differential of 78% segs and 16% bands. What do you recommend? M.B. now weighs 193 pounds, is 5 feet 5 inches tall, and has a serum creatinine of 1.8 mg/dL.
Rule #1: If it's not broken, don't fix it! In this case, trough concentration is good, but the peak concentration is much lower than we predicted and the patient continues to show signs of active infection. The concentrations need to be examined more closely and the regimen will probably need to be modified.
what happens if a patient is retaining fluid for Ag
changes Vd so retaining more drug
what happens to the half life of AG if kidney funciton declines
the half life increases because the elimination rate constant (k) is less
what does the elimination rate constant (k) represent
the fraction of drug eliminated every hour
Summary of “Traditional” AG Dosing
No concentrations obtained (emperic dosing
estimated Vd
estimated Cl
estimated k
estimated T1/2
(all obtained from AG pop parameters)

Concetrations obtained for patients previous does
calculatek
calculated T1/2
calculate Cl
calculate Vd
(all obtained from pateint specific serum concentratins, not estimates

both of the above lead to:
choose Cmax and Cmin
choose duration in infusion, tin
calculate dosing interval
calculate
dose
double check Cmin
the Ag population PK parameters are...
average values
graph comparing traditional and Conventional Ag dosing
Efficacy Considerations
for QD dosing and AG
Concentration-dependent bactericidal activity = Cmax:MIC ratio
Post-antibiotic effect (PAE) = suppression of bacterial growth after concentrations of an antibiotic fall below the MIC
Adaptive resistance = if bacteria are not exposed to sufficient aminoglycoside concentrations, they can down-regulate uptake of the drug and become refractory to further exposure
do you worry about cmins with QD dosing of AG
no because getting to non-detectable concentrations of drug, so toxicities are less of a concern
Toxicity considerations: not necessarily related to peak concentrations for AG
Nephrotoxicity = related to saturable intracellular accumulation of drug in the renal cortex. Less frequent administration has been shown in animal models and some human studies to result in lower renal cortical drug concentrations.
Ototoxicity = In rat models, less frequent administration was shown to result in less uptake of drug into the inner ear tissues.
Post-antibiotic effect (PAE)
suppression of bacterial growth after concentrations of an antibiotic fall below the MIC

AG penetrate bacterial cells at high concentrations and inhibti protein synthesis, which the bacteria take a while to overcome. So even when concentrations of AG or low there are still pharmocological effects

it is much more dependent on the cmax/mic ration
Potential Advantages of Extended Interval Aminoglycoside Dosing
More certainty peak drug concentrations will be high enough (much more dependable)
Enhanced bactericidal activity
Decreased adaptive resistance
Potentially decreased nephrotoxicity
Potentially decreased ototoxicity
Decreased reliance on drug monitoring
Decreased costs
Convenience
Potential Disadvantages of Extended Interval Aminoglycoside Dosing
Comparatively little clinical experience (professional discomfort)
Lack of data in certain patient populations (ICU, renal dysfunction, transplants, immunocompromised, children, pregnancy)
Monitoring methods/therapeutic (is there a certain Cmax we should aim for? is there a maximun cmax? Do we need to obtain a certain cmin?)
Controversies Surrounding Extended Interval Aminoglycoside Dosing
Dosing
What is the appropriate mg/kg dose?
How should adjustments for renal function be made: decreased dose or increased interval, or both?

Appropriate patients
Caution is recommended in the following types of patients for QD dosing of AG
Moderate-Severe renal impairment
Neutropenic infections
Sepsis/life-threatening infections
Endocarditis
Infections caused by Staphylococcus or Enterococcus in which aminoglycoside is used in combination regimens
Patients expected to have altered aminoglycoside pharmacokinetics:
Critically ill
Severe burns
Pregnant/postpartum
Cystic fibrosis
Pediatrics/neonates
how should QD dosing of AG be monitored
There is not a universally accepted answer to the question
Therefore, number and timing of sampling for drug concentrations varies widely with practice styles

a lot of it depends on the population
The Hartford Nomogram what dose of AG is administer
Administer initial dose of 7 mg/kg gentamicin or tobramycin

based on IBW it excludes obese patients
The Hartford Nomogram based on estimated renal function
Creatinine clearance
>60 ml/min dosed Q24hrs
40-60 ml/mon Q36hrs
20-39 mL/min Q48 hrs
<20 mL/min Q48hrs

Obtain “midpoint” serum concentration between 6 and 14 hours after start of infusion, and plot on the nomogram to determine dosage interval adjustment
is the hartford nongram used with amikacin
nope
the hartforn nonogram (pic)
Potential Limitations of the Hartford Nomogram
Assumes that a 7 mg/kg dose will always be given and that a 1-hour infusion time will always be used
Nomogram was designed to provide a Cmax of 20 mg/L (to achieve a peak:MIC ratio of 10 for Pseudomonas aeruginosa at Hartford Hospital) and a drug free interval of 4 hours at the end of the dosing interval
Based on volume of distribution of 0.3 L/kg.
What about patients with altered volume of distribution?
What about patients with changing renal function?
Does not allow for wide inter-patient variability in drug clearance
Patients expected to have abnormal aminoglycoside pharmacokinetics (ascites, burn >20% BSA, pregnant, ESRD) were excluded from the study that validated the Nomogram
Patients with enterococcal endocarditis were also excluded.
what is the Vd that the hartforn nonogram is based on
0.3 l/kg

a lot of patients do not have this Vd
what patients are excluded from the hartforn nonogram
Patients expected to have abnormal aminoglycoside pharmacokinetics (ascites, burn >20% BSA, pregnant, ESRD) were excluded from the study that validated the Nomogram
Patients with enterococcal endocarditis were also excluded.
measuring peak only with qd dosing of ag
Measuring the peak concentration facilitates assessment of adequacy of the dose amount (i.e. Cmax), but reveals little about the risk of toxicity (i.e. Cmin)
Not very useful in most situations

doesn't tell anyting about drug elimination or risk of toxicity
measuring peak and trough with qd dosing of ag
May consider monitoring peak and trough within first three doses when aminoglycoside pharmacokinetics are expected to be altered
However, runs the risk that trough concentrations will be undetectable and that the levels will therefore be useless
measuring peak and midpoint with qd dosing of Ag
Because trough concentrations are often undetectable, obtaining peak and midpoint instead of peak and trough avoids undetectable trough level and allows calculation of pharmacokinetic parameters
Preferred in patients with deteriorating clinical status

this gives 2 data points to work with and is good for the complicated pt because able to make adjustments
measuring the trough only with qd dosing of Ag
Less aggressive. Allows assessment of drug accumulation/drug toxicity. Assumes Cmax is high enough because of large doses given
When undetectable, may raise question as to adequacy of dose because duration of drug-free interval remains unknown
May consider this method in patients expected to have normal aminoglycoside pharmacokinetics
Check within first three doses and repeat every four to five days. If patient has worsening renal function or signs of nephrotoxicity, check trough more often

good for the uncomplicated patient because it is very limited and you are not able to make adjustments because only one data point

saving your booty by documenting if the elimination of the drug is what it is expected tobe or if the patient is at risk of toxocty

looking at the elimination of the drug and potential toxicities

troughs can be non-detectable
image of vanco
vanco histroy
Classed as a glycopeptide antibiotic
Produced by bacteria, Nocardia orientalis, isolated from soil samples obtained in Indonesia and India (1956)
Complex tricyclic glycopeptide having a molecular weight of nearly 1,500
Extensively used clinically since the late 1950s
Has been remarkably overused without significant resistance – until recently
what makes vanco a glycopeptide
the petide bonds throughout the molecule
Vancomycin: Spectrum of Activity
Narrow-spectrum bactericidal antibiotic
Activity against ONLY Gram-positive organisms
Good activity against Staphylococcus aureus (including MRSA), S. epidermidis and other coagulase-negative staphylococci, and streptococci
Active against most Enterococcus species
Activity includes most Gram-positive anaerobes including Clostridium spp.
No activity against Gram-negative bacteria because the large molecular weight prevents it from penetrating the outer membrane
is vanco cidal or static
cidal
vanco activity against gram +
Activity against ONLY Gram-positive organisms
Good activity against Staphylococcus aureus (including MRSA), S. epidermidis and other coagulase-negative staphylococci, and streptococci
Active against most Enterococcus species
Activity includes most Gram-positive anaerobes including Clostridium spp.
vanco activity againt gram -
No activity against Gram-negative bacteria because the large molecular weight prevents it from penetrating the outer membrane
vanco MOA
Inhibits cell wall synthesis by high-affinity binding to cell wall precursor
binds with high affinity to the D-alanyl-D-alanine portion of the cell wall precursor
Vancomycin also inhibits synthesis of cell wall-associated phospholipids
Mechanism of action does not involve PBPs

Vancomycin is bactericidal, but not as rapid as β-lactam antibiotics (“slowly cidal”)
is vanco MOA enzymatic
nope it is no-enzymatic it binds directly with high affinity to the D-alanyl-D-alanine portion of the cell wall precursor and interfers with the transpeptidase MOA
is vanco time or concentration dependent
time
is daptomycin time or concentration dependent
concetration
pic of vanco moa
Vancomycin binds with high affinity to D-alanyl-D-alanine terminus of cell wall precursor units, inhibits release of building block unit from carrier (D-alanine), thus prevents peptidoglycan synthesis
Vancomycin: Mechanism of Resistance
Several mechanisms of resistance have been identified:
Most important mechanism involves modification of cell-wall precursors that will no longer bind vancomycin
Change of D-alanyl-D-alanine to D-alanyl-D-lactate → binding of vancomycin is prevented but cell wall synthesis is able to proceed
Mechanism present in vancomycin-resistant enterococci (VRE), particularly E. faecium (VREF)
Plasmid-mediated mechanisms also thought to involve interference of transport/entrance of vancomycin into bacteria (less common)
is vanco absorbed after PO admin
nope
Vancomycin very poorly absorbed after oral administration
Must be administered parenterally for treatment of systemic infections
However, oral route can be used to treat Clostridium difficile colitis because infection usually localized to colonic mucosa in the lumen (so do not need systemic absorption)
is vanco distributed into the CNS
Distribution into the CSF is limited
how is vanco eliminated
>70% of parenteral doses excreted by glomerular filtration
Remainder of dose eliminated via unknown routes
half life of vanco
T1/2 = approx. 4-6 hours in normal renal function
Vancomycin: Adverse Effects “Red man” or “Red neck” syndrome
Infusion-related reactions
“Red man” or “Red neck” syndrome
Pruritis and flushing of face, neck, and upper extremities and trunk
Rash
Hypotension, tachycardia
True incidence unknown, but studies suggest range of 0-35%
Occurs 10-20 minutes after start of infusion
Mediated by drug-induced histamine release
Managed by slowing rate of infusion, (60 min, 90 min, and 2 hr infusion times, can also dilute in a lot of fluids)

when turn of the infusion this reaction disapates
prior administration of antihistamines

histamine mediated infusion reaction

interaction with the mast cells
nephrotoxicity and vanco
Incidence <5% when administered alone
Incidence increased by concomitant use of aminoglycosides or other nephrotoxic drugs (this is not an unusaul combo)
Recent data also suggest daily doses >4 grams may increase risk

not as prevalent as Ag
ototoxicty and vanco
Very unusual
Tinnitis, vertigo, hearing loss

can be reversible
hypersensitivity and vanco
Range from rashes (common) to anaphylaxis (rare)
Vancomycin: Adverse Effects
Nephrotoxicity
Incidence <5% when administered alone
Incidence increased by concomitant use of aminoglycosides or other nephrotoxic drugs
Recent data also suggest daily doses 4 grams may increase risk
Ototoxicity
Very unusual
Tinnitis, vertigo, hearing loss
Hypersensitivity reactions
Range from rashes (common) to anaphylaxis (rare)
Neutropenia, thrombocytopenia (rare) (monitor CBCs)
Phlebitis and pain at IV administration site (common)
Vancomycin: Drug Interactions
Additive/synergistic nephrotoxicity when given concomitantly with other nephrotoxic agents
Aminoglycosides
Amphotericin B
Cisplatin
Cyclosporine
Mycophenolate
Etc. etc.
Vancomycin: Clinical
Uses
Remains a very important drug for treating variety of serious Gram-positive infections
Particularly useful in patients with severe β-lactam allergy
Usual preferred agent when MRSA or methicillin-resistant S. epidermidis (MRSE) documented or suspected as cause of infection
Also important in treatment of systemic Enterococcus infections, Gram-positive anaerobic infections (e.g. Clostridium)
Used in oral treatment for C. difficile-associated colitis
what drug is you rfirst choince for the treatment of MRSA
VANCO
what is structurally different between vanco and telavancin
telavacin has a lipophilic tail making it more active at the bacterial cell membrane than vanco

Membrane anchoring
- Improved potency
- Rapid cidality
- Activity vs. VISA
telavacin hs
Favorable PK-ADME properties
- Long half-life
- Renal clearance
telavancin class
Classed as a “Lipoglycopeptide”
Similar to vancomycin but not exactly
Inhibits cell wall synthesis; also affects bacterial cell membrane function
is telavacin static or cidal
Bactericidal activity
Much more rapidly cidal than vancomycin
is telavancin time or concentration dependent
concentration
bacterial resistance and telavancin
Little resistance observed in vitro or in clinical studies
half life of telavancin
Dosed once daily (T1/2 = 7-11 hours)
what formulation does telavancin come in
IV only
do you need to dose adjust for renal dysfunction with telavancin
yep
what it type of bacteria is telavancin active against
Excellent activity against Gram-positive organisms including MRSA, MRSE, VRE
pic of moa of vanco
Cell Wall Synthesis in Gram-positive Bacteria
pic of telavancin MOA (inhibition of cell wall syntesis)
through transglycosylation the long side chanin inserts into the cell membrane interacting with lipid II
televancin pic disrupting bacterail cell membrane
creates false porins so the cell depolarizes and the cell membrane function is disrupted
is televancin selectove for bacterial cell membranes
no but we do not have lipid II so it creates less membrane dysfunctionin in us
most common Se of telavacin
altered taste (metallic)
foamy urine
nephrotoxicity
Telavancin: Renal Adverse Events
Renal adverse events were 3.4% in clinical studies
Occurred primarily in patients with risk factors:
Preexisting renal disease, diabetes mellitus, congestive heart failure, hypertension
Also more frequent in patients with concomitant medications known to affect renal function
e.g., NSAIDs, ACE Inhibitors, loop diuretics
Improving or recovered in the majority of patients in both treatment groups by the end of the cSSSI study
Monitoring of serum creatinine is recommended in all patients receiving telavancin
pregnancy cat of telavancin
Pregnancy category C
Reduced fetal weights and increased rate of limb & digit malformations in rats, rabbits, and pigs
No human data
Risk Evaluation and Mitigation Strategy (REMS) program in place due to pregnancy risk
Mandated by FDA as condition of approval & marketing
Requires “medication guide” be provided to patients
“Dear Health Care Provider” letters to targeted providers
Serum pregnancy test recommended prior to drug use
Pregnancy registry to monitor outcomes in women exposed during pregnancy (1-888-658-4228)
telavacin and coagulation tests
Certain tests are affected by telavancin through interactions with in vitro testing methods:
Prothrombin time (PT)
International normalized ratio (INR)
Activated partial thromboplastin time (aPTT)
Activated clotting time (ACT)
Coagulation-based factor Xa tests
Blood samples should be obtained as close as possible before administration of next dose of telavancin
Tests which are not affected:
Thrombin time (TT)
Whole blood (Lee-White) clotting time
Ex vivo platelet aggregation
Functional (chromogenic) factor X and Xa assays
Bleeding time
D-dimer, fibrin degradation products


not effecting the liver and does not bind to proteins
doesn't cause bleeding
just interferes with the test

this is a concentrated effect
how can you avoid the affects telavacin has on coagualation tests
collect a blood sample at the through to minimize this interaction

it is concentration related
telavancin efficacy and clinical role
Excellent clinical efficacy shown in skin/soft tissue infections, pneumonia
FDA approved in late 2009 for complicated skin/soft tissue infections
Clinical role has yet to be defined, although efficacy in pneumonia is potentially very useful

not a first choice drug at this time
cyclic lipopetide pic
Daptomycin (Cubicin™)
Cyclic lipopeptides
Naturally occurring compounds derived from Streptomycin roseosporus, first discovered in 1980s
Novel group of cyclic amino acid compounds
Large molecules, molecular weight >1,600
Contain both hydrophillic and lipophilic properties
Daptomycin = first cyclic lipopepetide approved for use
Clinical trials conducted in early 1990s but stopped due to toxicity concerns
Toxicity problems resolved, FDA approved 2003
Daptomycin: Spectrum of Activity
Narrow-spectrum agent active against only Gram-positive organisms
Very rapidly bactericidal
Concentration-dependent rate of killing
Highly active against:
Staphylococcus aureus (including MRSA), S. epidermidis (including MRSE), and other staphylococci
Streptococci
Enterococcus species, including VRE
Good activity against Gram-positive anaerobes
No clinically relevant activity against Gram-negative bacteria
is daptomycin cidal or static
very rapidly cidal
what is daptomycin highly active against
Staphylococcus aureus (including MRSA), S. epidermidis (including MRSE), and other staphylococci
Streptococci
Enterococcus species, including VRE
Daptomycin Mechanism of Action
Binds to Gram-positive bacterial cell membrane
Calcium-dependent insertion of lipid tail
Rapidly depolarizes the cell membrane
Rapid efflux of potassium
Destroys ion-concentration gradient across membrane

Cell death (this helps explain the rapid cidal effects)
Multiple failures in biosystems, DNA, RNA, protein synthesis
(not directly linked to membrane effects)
when a patient is hypocalcemic does daptomycin work
no, because the activity when is forms porins and disrups the concentration gradients in the bacterial membrane is Ca dependent
Cyclic Lipopeptides: Resistance
Although resistance can be induced in the laboratory setting, in vivo resistance is rare
Mechanisms of resistance are therefore not well characterized
Resistance may involve alteration in cell membrane potential leading to reduced drug binding
No cross-resistance observed with other antibiotics (unique MOA)
Daptomycin Pharmacokinetics
Not absorbed after oral administration
Must be administered parenterally
Vd is relatively small, reflecting limited distributed throughout body
Does not penetrate well into respiratory tissues, rapidly inactivated by surfactant
>80% renally cleared, minimal known metabolism
Dosage adjustment required for renal impairment
T1/2 = 8 hours (QD dosing)
can daptomycin be used in respiratory infections such as pneumonia
no it Does not penetrate well into respiratory tissues, rapidly inactivated by surfactant
what inactivates daptomycin in the respiratory tract
surfactants (rapidly)
Daptomycin Adverse Effects
Considered to be a safe, well tolerated drug
GI: constipation, nausea, vomiting, diarrhea (3-6%)
CNS: Headache, insomnia, dizziness (2-5%)
Rash (4%)
Elevations in creatine phosphokinase (CPK) (created problems in early clinical trails)
Occurs in <3% of patients, usually asymptomatic
Muscle weakness, myalgias observed in early studies
Elevation of hepatic transaminases (3%) (irrelevant)
Injection site reactions
Daptomycin Drug Interactions
No effect on cytochrome P450 system in vitro
Only pertinent drug-drug interaction is with the statins
Increased risk of musculoskeletal toxicity, myopathy
Risk is quite low, but patients should be carefully monitored during daptomycin therapy
this is not an absolute CI and may be hard to avoid so just closely monitor
Daptomycin: Clinical Use
Good activity against MRSA, MRSE, other Gram-positive organisms
Clinically effective against VRE
Considered good alternative to vancomycin
Better tolerated than Synercid™, easy to administer
Bactericidal, while linezolid is static
Alternative agent for treating variety of serious Gram-positive infections including skin/soft tissue, bone and joint, bloodstream, endocarditis infections
Not useful for respiratory tract infections
Useful in patients with severe β-lactam allergy
Exact clinical role has yet to be defined
daptomycin spectrum of activity
Good activity against MRSA, MRSE, other Gram-positive organisms
Clinically effective against VRE
Considered good alternative to vancomycin
Better tolerated than Synercid™, easy to administer
Bactericidal, while linezolid is static
structure of oxazolidinones
The Oxazolidinones
Synthetic class of antibiotics
In 1978, series of agents were found to be effective against plant pathogens
Further manipulations in molecule led to development of new agents for use in humans
Orally absorbed
Displayed activity against a variety of streptococci and staphylococci, comparable to vancomycin
One agent approved for use in the U.S.
Linezolid (Zyvox™)
Approved in 2000 for treatment of a variety of gram-positive infections
what is the only drug active against multiple resistance gram + bacteria that is PO
Linezolid
Linezolid: Spectrum of Activity
Narrow-spectrum
Bacteriostatic, not cidal
Primarily active against only Gram-positive organisms
Good activity against Staphylococcus aureus (including MRSA), S. epidermidis and other coagulase-negative staphylococci, and streptococci
Active against Enterococcus species, including E. faecalis as well as vancomycin-resistant E. faecium (VREF)
Activity includes many Gram-positive anaerobes
No clinically relevant activity against Gram-negative bacteria
Some activity against atypical bacteria
is linezolid static or cidal
static
what is linezolids primary activity
Primarily active against only Gram-positive organisms
Good activity against Staphylococcus aureus (including MRSA), S. epidermidis and other coagulase-negative staphylococci, and streptococci
Active against Enterococcus species, including E. faecalis as well as vancomycin-resistant E. faecium (VREF)
Activity includes many Gram-positive anaerobes
is linezolid active against gram -
no
Linezolid: Mechanism of Action
Inhibits an early step in bacterial protein synthesis
Prevents the formation of the tRNAfMet-mRNA-30S (or 50S) subunit ternary complex
prevents formation of 70S ribosome complex that initiates protein synthesis
Does not block the elongation or termination steps of translation
Linezolid appears to bind to both 50S and 30S ribosomal subunits
Unique mechanism seems to preclude cross-resistance with other existing antibiotics
pic of linezolid MOA
Oxazolidinones: Mechanisms of Resistance
Resistance usually related to specific point mutations in domain V of the 50S ribosomal subunit
Domain V very highly conserved region of the ribosome
Integral part of peptidyl transferase center
Resistance relatively unusual in clinical practice, but cases occasionally reported
Usually in VRE, but recently in MRSA as well
Usually associated with prolonged courses of therapy (often >21 days)
Other risk factors include indwelling prosthetic devices, undrained abscesses, prior linezolid therapy
Absorption of linezolid PO
Linezolid very well absorbed after oral administration
Bioavailability approaches 100%
Food slightly decreases rate but not extent of absorption
Only agent available both IV and PO for treatment of multidrug-resistant Gram-positive infections

with or wothout food
how is linezolid metabolized
Metabolized by non-enzymatic oxidation to aminoethoxyacetic acid and hydroxyethyl glycine
~80% of drug is eliminated in urine  30% active drug and 50% metabolites
No metabolism by, or interaction with, CYP 450
Dosage adjustment required for neither renal nor hepatic dysfunction
T1/2 of linezolid
T1/2 = 5-7 hours

BID
Vd of linezolid
Vd = 40-50 L, well distributed throughout body
are PO and IV doses of linezolid interchangable
yep
because oral absorption is that good
Linezolid: Adverse Effects
Considered to be a safe & well tolerated drug in most patients
GI: nausea, vomiting, diarrhea (3-6%)
Headache (2-4%)
Elevation of hepatic transaminases (3-7%)
Reversible bone marrow suppression (1-5%)
Thrombocytopenia, leukopenia, anemia
Usually occurs late in therapy (>10-14 days)
Most common in severely ill patients
Taste alterations, tongue discoloration (2-3%)
Injection site reactions
what is the most important SE of linezolid and what do you do if it happens
overall bone marrow suppression
Reversible bone marrow suppression (1-5%)
Thrombocytopenia, leukopenia, anemia
Usually occurs late in therapy (>10-14 days)
Most common in severely ill patients

if happens switch drugs do not treat through
Linezolid: Drug Interactions
No effect on cytochrome P450 system
Reversible monoamine oxidase inhibitor
Mild, reversible, and competitive inhibition of both MAO-A and MAO-B
Potential interaction with adrenergic and serotonergic agents
In Phase I studies, mild to moderate changes in blood pressure seen in normal healthy volunteers given concomitant pseudoephedrine or phenylpropanolamine
Serotonin syndrome has been reported in patients receiving SSRIs and linezolid, but uncommon
Linezolid: Clinical Uses
Good activity against MRSA, MRSE, other Gram-positive organisms
Clinically effective, widely used for VRE
Considered good alternative to vancomycin
Better tolerated than Synercid™, easy to administer
Availability in oral form fills important role in outpatient treatment
Alternative agent for treating variety of serious Gram-positive infections including respiratory tract, skin/soft tissue, bone and joint infections
Useful in patients with severe β-lactam allergy
what are the 2 antibiotics in the streptogramins combo product
Quinupristin
Streptogramin B
and
Dalfopristin
Streptogramin A
Streptogramins Antibiotics
Macromolecular antibiotics produced by Streptomyces pristinaespiralis
Members of the MLS (macrolide-lincosamide-streptogramin) group of antibiotics since they share a common mechanism of action
Act at ribosome to inhibit protein synthesis
Streptogramins consist of two types, A and B, which are both macrocyclic lactone rings
Type A are cyclopeptides with a MW of approx. 500
Type B are cyclic hexadepsipeptides with MW of approx. 800
quinupristin (tpye B0 structure (pic)
dalfopristin (type a) structure (pic)
dalfopristin (type a) structure (pic)
Quinupristin/Dalfopristin (Synercid™)
First streptogramin antibiotics in U.S.
novel antibiotic consisting of two components
Synercid™ approved for use in 1999
Combination of quinupristin (type B) and dalfopristin (type A) in a 30:70 w/w ratio
Streptogramins A and B inhibit bacterial cell growth by blocking bacterial protein synthesis
Separately, A and B components are bacteriostatic, temporarily halting protein synthesis
Combined, A and B components are bactericidal, acting synergistically to permanently inhibit protein synthesis
Quinupristin/Dalfopristin (Synercid™) are they cidal or static when alone
static and cidal when together
Quinupristin/Dalfopristin (Synercid™) are they cidal or static when together
cidal
Synercid™: Spectrum of Activity
Narrow-spectrum bactericidal antibiotic
Primarily active against only Gram-positive organisms
Good activity against Staphylococcus aureus (including MRSA), S. epidermidis and other coagulase-negative staphylococci, and streptococci
Active against certain Enterococcus species, including vancomycin-resistant E. faecium (VREF)
Not active against E. faecalis
Activity includes many Gram-positive anaerobes
No clinically relevant activity against Gram-negative bacteria
Some activity against atypical bacteria
Synercid™: Primarily active against
Good activity against Staphylococcus aureus (including MRSA), S. epidermidis and other coagulase-negative staphylococci, and streptococci
Active against certain Enterococcus species, including vancomycin-resistant E. faecium (VREF)
Not active against E. faecalis
Activity includes many Gram-positive anaerobes
Type A streptogramin (Dalfopristin)
MOA
binds to the 50S ribosome to prevent peptide chain elongation (early phase)
inhibits protein elongation by interfering with the binding site of peptidyl transferase, the enzyme that facilitates protein synthesis by transporting amino acids to the ribosome
produces conformational changes in the 50S ribosome, increasing its affinity for the type B streptogramin
Type B streptogramin (Quinupristin)
MOA
binds to the 50S ribosome at L24, a protein that is part of the polypeptide exit channel
Stable drug-ribosome complex constricts the ribosomal exit channels
prevents further polypeptide chain formation and results in early chain termination with release of incomplete peptide chains
Interruption of protein synthesis eventually results in cell death
how does A streptogramin (dalfopristin) increase the the binding affinity of B streptogramin (quinupristin)
produces conformational changes in the 50S ribosome, increasing its affinity for the type B streptogramin
Mechanisms of Bacterial Resistance to Quinupristin/Dalfopristin
Most important is plasmid-mediated target modification
Causes resistance to quinupristin by methylation of common binding sites
Infrequently, resistance mediated by drug-modifying enzymes or efflux (dalfopristin)
Quinupristin hydrolase
Dalfopristin acetylase
Synercid™: Adverse Effects
Phlebitis
Incidence up to 75% of patients
Can be very severe, including thrombophlebitis (clot fromation at site of inflammation)
Necessitates use of central venous catheter for long-term administration

Arthralgia, myalgia (very painful and do not become tolerant too)
3%–12% of patients
May be very severe, often requiring analgesics (APAP, NSAIDs, opiates) to maintain therapy
Diluting drug in large volumes for infusion may help
Elevated hepatic enzymes, increased bilirubin
Other: headache, nausea, vomiting, diarrhea, rash
how is synercid administered
Necessitates use of central venous catheter for long-term administration

very necrotic od small vessels
Synercid™: Drug Interactions
CYP 3A4 significantly inhibited by Synercid
Dose reductions necessary for drugs metabolized via CYP 3A4 pathway
CYP 3A4 substrates with associated QTc prolongation should be avoided
Quinupristin/dalfopristin is not itself metabolized by cytochrome P450 enzymes
No alterations in Synercid pharmacokinetics by other CYP 3A4-metabolized drugs
No interactions with drugs metabolized by other CYP 450 pathways
what are synercids affects on the p450 system
CYP 3A4 significantly inhibited by Synercid
Dose reductions necessary for drugs metabolized via CYP 3A4 pathway
CYP 3A4 substrates with associated QTc prolongation should be avoided
Synercid™: Clinical Uses
Good activity against MRSA, MRSE, other Gram-positive organisms
First antibiotic in U.S. to be clinically effective against VREF
Lack of activity against E. faecalis problematic
Overall clinical usefulness limited by administration issues, toxicities, drug interactions
Not as widely used as other agents, e.g. vancomycin, linezolid, daptomycin
Alternative agent for treating variety of serious Gram-positive infections
Useful in patients with severe β-lactam allergy

became obsolete when linezolid came onto the market