You are on page 1of 32

Basic

Pharmacology

1
1
The author LORENZO JOSÉ
FRAILE SAUCE

D
 egree in Veterinary Medicine and PhD in Pharmacology
from the University of Zaragoza, Spain.

H
 e worked as a full-time swine and bovine practitioner for
eight years.

F rom 2004 to 2010, he worked as a senior researcher at CReSA (Center of Animal


Health Research) working in the fields of epidemiology, pharmacology and
immunology, focusing his work on bacterial and viral swine and bovine diseases.

From 2010 to now, he has been working as a professor at the University of Lérida
(Spain). He has been actively collaborating in field trials with different pig and
2 bovine production companies.

Throughout his career, he has published more than fifty papers in peer-reviewed
journals.

In collaboration with Ana I. de Prado


Graduated in Veterinary Medicine from the University of León
(Spain). She holds a Master’s degree in Management and
Consultancy on Dairy Cattle Farms from the Autonomous
University of Barcelona and a Master’s degree in Research in
Medicine and Animal Health from the University of Santiago de Compostela (Spain).

After working as a practitioner and milk quality advisor, she joined the pharmaceutical
industry in 2002, working for different companies as a marketing and technical advisor
for vets and farmers, carrying out different projects and field trials.

She currently works for Ceva Animal Health as a Global Technical Manager in ruminants.
Basic Pharmacology
1. Aims and objectives ............................................................................................... 4
2. General concepts: what are we going to talk about? .......................................... 5
3. Pharmacokinetics
Pharmacokinetic parameters: ............................................................................... 8
3.1. Plasma clearance ............................................................................. 9
3.2. Volume of distribution ................................................................... 10
3.3. Half life ............................................................................................ 11
3.4. Area under the curve ...................................................................... 12
3.5. CMax ................................................................................................... 15 3

4. Pharmacodynamics
Pharmacodynamic parameters: ......................................................................... 16
4.1. Minimum inhibitory concentration ............................................... 17
4.2. Minimum bactericidal concentration ........................................... 19

5. Clinical efficacy and its relationship with pharmacokinetic and


pharmacodynamic parameters .......................................................................... 21
5.1. What do we use PK-PD models for? ............................................. 21
5.2. Using PK-PD: designing an administration schedule ................. 21
5.3. PK-PD and antimicrobials, their classification ............................ 24
5.4. Using different antimicrobials at the same time ....................... 26

6. Withdrawal period, a critical consideration to ensure food safety ................ 28


7. List of references ................................................................................................ 29
Basic Pharmacology

Basic pharmacology
Veterinary surgeons study pharmacology as part of their degree but often the
basics are forgotten over time, yet we use pharmacology every day without thinking
about the details.

The principles of antimicrobial therapy are based on a three way therapeutic


relationship between the bacteria that cause the infections, the animal and the drug
which is administered to treat the infection. The choice of an antimicrobial treatment
and the design of a dose regime depend both on the knowledge the clinician has
about the disease and on the pharmacokinetics (PK) and pharmacodynamics
(PD) of the drugs chosen to treat it. PK/PD models are helpful to decide these
administration schedules. PK/PD models also allow us to classify antimicrobials and
which helps us understand their mode of action. Withdrawal periods are also critical
to help ensure food safety.

4
1.
Aims and objectives
Veterinarians prescribe and use medicines on a
daily basis and so the main aim of this booklet
is to provide a solid grounding in the basic
concepts of pharmacology by:

E xplaining pharmacological concepts from


a practical point of view.

 efining pharmacokinetic and


D
pharmacodynamic parameters and their
interpretation.

 nderstanding the importance and use of


U
PK/PD models.

C onsidering how antimicrobials are used in


daily practice.

1
Basic Pharmacology

2.General concepts
The principles of antimicrobial therapy are based on a three way therapeutic relationship
between the bacteria causing the infection, the affected animal and the drug which is
administered to treat the infection.

5
Pharmacokinetics Infection
Imsmpon
re
city

unese
i
Tox

Susceptibility

Pharmacodynamics

1
Basic Pharmacology

The main objective of antimicrobial therapy is to ensure that the infected area is supplied with
a high enough concentration of the drug which remains effective and lasts for a long enough
period of time in order to achieve both clinical and bacteriological cure, whilst minimising the
appearance of undesired side effects.

Remains effective

High enough
Antimicrobial concentration Lasts for a sufficient
Ensuring period of time
therapy of active
ingredients
Main objective
Minimises undesired
effects

Examples of undesired effects:


6
Unwanted side effects in the animal being treated.

Development of microbial resistance to the drug being administered.

In livestock, the presence of drug residues in edible tissues at levels higher


than those allowed (Lees, 2002).

Key considerations for veterinary surgeons before applying any therapeutic regime are:

The drug to be used


The correct dose to be administered
The frequency of administration
The route of administration
The length of treatment required to reach MIC

1
Basic Pharmacology

Treatment success
The choice of an antimicrobial treatment and the design of a dosage depends on the available
information of the causal microorganism (ideally this will be based on a culture and sensitivity
analysis but clinical experience is also a valuable diagnostic tool), the effect of the drug on
the microorganism (pharmacodynamics, susceptibilily), the effect of the drug on the animal
being treated (toxicity), the presence of the drug in this particular animal (pharmacokinetics)
combined with other considerations such as the emergence of antimicrobial resistance, animal
welfare and the cost/benefit ratio of the treatment (Goodmand and Gillman, 2006).

Microorganism
information

Other Treatment
considerations success Pharmacodynamics

Pharmacokinetics Toxicity

1
Basic Pharmacology

3.
Pharmacokinetics: what does the animal do to the drug?
Pharmacokinetics (PK) is the science that describes, by means of mathematical concepts, the
kinetics of molecules when they are introduced to an organism. It defines the pattern of ADME
processes (absorption, distribution, metabolism and excretion) that control the presence of
these substances (Rowland, 1995; Baggot, 2001, Martín-Jiménez, 2002).

The process of a substance entering the


Absorption blood circulation.

The dispersion or dissemination of


Distribution substances throughout the fluids and
tissues of the organism.
ADME
processes The recognition by the organism that a
Metabolism foreign substance is present and the
irreversible transformation of parent
compounds into daughter metabolites
that could be easily excreted.

The removal of the drugs from the


Excretion organism.
8

What do we use pharmacokinetic studies for?


We can use PK parameters to make predictions about the concentration of antimicrobials for
specific doses and thus design the administration schedule.

This is possible because the kinetics of the drugs is linear:

The speed at which an antimicrobial is eliminated from the organism


is directly proportional to the amount which remains within it.

Pharmacological products (especially antimicrobials) which fail to show a linear behaviour in


their kinetics are not usually used (Prescott, 2000).

1
Basic Pharmacology

Observe in this example the evolution of the concentrations of an antibiotic administered at a


dose of 1.25 mg/kg of live weight by intravenous and intramuscular route in lactating cows.

1
Concentration, μg/ml

0.1

0.04
0 240 480 720 960 1,200 1,440
Minutes
Levels in serum Levels in serum Levels in milk Levels in milk
after IV injection after IM injection after IV injection after IM injection

The graph above shows that: 9


The antibiotic administered intravenously reaches the highest concentration in
serum but this concentration is maintained for the shortest period of time.
T he antibiotic administered intramuscularly reaches the maximum concentration
later than the IV administered one.
T he antibiotic concentration in milk is higher than the serum concentration at the
same kinetic times for the IV and IM route of administration throughout the time.
T he antibiotic administered by both administration routes appears approximately in
milk at the same kinetic time and it disappears approximately at the same time too.

3.1. Plasma clearance (CL)


Plasma clearance is the most important pharmacokinetic parameter because it is
one of the three which determine the maintenance dose (Toutain, 2004a):

1
Basic Pharmacology

CL x Css
Maintenance dose =
F

Maintenance dose: quantity of antimicrobial administered per unit of time.

CSS: steady blood Concentration of the drug (steady state) that allows the desired effect.
– In the case of an antimicrobial, the clinical response may be an improvement of the symptoms
associated with the infection.

F: bioavailability. This parameter ranges from 0 to 1. The bioavailable dose of a product is the real
amount of drug that reaches the systemic circulation (it is therefore “available” to perform its action)
when a formulation is administered by extravascular route.
– That is to say, if a drug is administered orally and its bioavailability is 1, this means that it is
completely absorbed.

CL: Plasma clearance. the volume of blood that is cleared of the target substance per unit of
time and per kilogram of live weight (its units are ml/min/kg).

I n the case of antibacterial therapy, CL is the direct measurement of the ability of an


animal to eliminate an antimicrobial.
10

3.2. Volume of distribution (Vd)


The volume of distribution is defined as the constant of proportionality between the amount of an
antimicrobial in the tissues of an organism and concentration in plasma.

Antimicrobial in the organism


Vd =
Concentration in plasma

A high volume of distribution does not necessarily imply


This parameter is not directly that the drug is widely spread throughout the organism,
related to the distribution of the since it could show tropism for a specific organ.
drug in specific physiological
The volume of distribution of a drug can be much higher
compartments.
than the total body water volume (Toutain, 2004b).

1
Basic Pharmacology

Generally speaking, a high volume of distribution is a positive characteristic for any drug because
it implies that it could be used to treat infections in many parts of the body. As an example,
the following table shows the volume of distribution of known antimicrobials in cattle and the
potential clinical consequences.

Examples of volume of distribution

Antimicrobial Volume of distribution (l/kg) Comments

Only feasible to treat extracellular


Gentamicin (aminoglycoside) 0.19 infections.
Low penetration in lung tissue
Higher tissue penetration
Ampicillin (β-lactams) 0.44 than aminoglycosides
but less than macrolides

Tylosin (macrolide) 1.33 High penetration in lung tissue

Florfenicol (amphenicol) 0.76 Wide tissue distribution

3.3. Half-life (t½)


11
The half-life is the time required, after the intravenous administration of a drug, for the
concentration in plasma to be reduced to half once the distribution equilibrium has been
reached.

Timeline
Concentration
in plasma

Distribution
equilibrium
Drug
administration
Is reduced
to half

1
Basic Pharmacology

This implies that the calculation of half life can only be carried out when the decrease of the
concentration in plasma is due solely to the elimination process (Toutain, 2004c).
From a practical point of view, the relevance of this parameter is limited.

So, for a drug with a half-life of 12 hours, a steady


It is important to point out that, state will be reached after the second or third day of
after the administration of administration (30-60 hours).
multiple doses of a drug, a steady
state is reached after 3 to 5 For this reason, for drugs with a very long half life
half-lives have elapsed. (> 24 hours), it could be useful to use a loading dose to
achieve the steady state as quick as possible.

For example:

The steady state will be reached


quickly in a multiple dose
Half-life of penicillin administration
is short in cattle
12
Using a loading
dose for these
Half-life of some sulphonamides drugs could be
is long in cattle reasonable

Although it is not
normally used under
field conditions
3.4. Area under the curve (AUC)
AUC is a direct measure of the drug exposure in an animal after administration (antimicrobial in
this case) and it is directly related to the administered dose and the clearance (CL).
Thus, the lower the clearance the higher is the AUC:

Administered dose Administered dose


CL= AUC=
AUC CL

1
Basic Pharmacology

The area under the curve is a plot of the change of concentration over time.
Concentration

IV administration

Oral administration
Time

The prediction of the concentration You cannot assume that concentration in tissue is the
in blood of a drug after its same as concentration in plasma at any specific time.
administration can be calculated
by means of pharmacokinetic It is important to know the concentration in tissue
equations but this procedure does 13
because it is the concentration of antimicrobial in the
not give the concentrations present
in the other tissues of the organism target organ.
at the same kinetic time.

Tissue kinetic study allows the calculation of the partition coefficient (P):

Ctss Ctss: the concentration of the antimicrobial in tissue at a steady state.


P=
Cpss Cpss: the concentration of the antimicrobial in plasma at a steady state.

The value of P gives information about the degree of accumulation of


the antimicrobials in the tissues of interest and it is very useful to
evaluate the potential effectiveness of the antibacterial treatments
in specific organs or tissues (Sánchez-Rubio, 1999).

From a practical perspective, the higher the P value, the better from a clinical point of view.

1
Basic Pharmacology

Study example: these graphs show the results of mean concentration of ceftiofur and its
metabolites in plasma, lochia and uterine tissues, after the subcutaneous administration of
ceftiofur (hydrochloride) at a dosage of 1 mg/kg in dairy cows [mean ± SD (n= 4 cows)]. SD:
standard deviation.
4 4
Plasma Lochia

3 3
Concentration, μg/ml

Concentration, μg/ml
2 2

1 1

0 0
0 4 8 12 16 120 24 0 4 8 12 16 120 24
Post-treatment time (hours) Post-treatment time (hours)

4 4
Caruncles Endometrium

3 3
Concentration, μg/g

Concentration, μg/g

2 2

14 1 1

0 0
0 4 8 12 16 120 24 0 4 8 12 16 120 24
Post-treatment time (hours) Post-treatment time (hours)

These graphs show that:


T he concentration achieved in plasma is notably higher than in other tissues and maximum
serum concentration is obtained 2 hours following administration (well before that seen in
other tissues).
T he antibiotic levels in the endometrium are consistently higher than the caruncles & lochia
and closely mirror the serum concentrations.
I n the caruncle, the maxium concentration is reached later than in the lochia, but the
concentration is relatively constant throughout.
At 24 hours post-treatment, the lowest concentration is observed in lochia.
It is important that the drug concentration in each tissue is not linked directly to the clinical
efficacy of any antimicrobial treatment because it is necessary to know the MIC of the
antimicrobial against the causal bacteria.

1
Basic Pharmacology

3.5. Maximum concentration (Cmax)


50
This is the maximum concentration
observed after the administration of a 40
medicinal product to an animal by any

Concentration, (μg/ml)
Cmax
route of administration. 30

The graph opposite shows the maximum


20
concentration of an antibiotic after
intramuscular administration.
10

0
0 1 2 3 4
Time (hours)
Routes of administration
The duration of treatment of most infections is usually between 3-7 or 5-7 days to effect a clinical
cure. From a practical point of view, the pharmacokinetic profile observed depends not only on
the route of administration but also on the particular medicinal product used. Thus, two medicinal
products that contain the same drug, at the same dose and administered by the same route
(intramuscular) could have different pharmacokinetic profiles.

The usual routes of administration of prescription medicines in cattle are intramuscular, 15


subcuticular and intravenous. Oral medication has a place, but it is more commonly used for
calves. In dairy cows the oral route is usually reserved for intra-ruminal metabolic drenches. Intra
mammary administration is used in dairy cows for the treatment of mastitis and also during the
dry cow period, although this is increasingly being replaced with teat sealants.

1
Basic Pharmacology

4.
Pharmacodynamics: what is the effect of the medicine on the animal?

The aim of antimicrobial therapy is to eliminate the microorganism responsible for disease from
the host and therefore reach a cure. However, antimicrobial drugs cannot completely eliminate a
pathogen without support from the the host’s immune system.

The fundamental objective of antimicrobial therapy is to help the


natural defence mechanisms to overcome the infectious agent
(Prescott, 2000).

Pharmacodynamics describes the relationship between the concentration of an antimicrobial in


the organism over time and the intensity and duration of its pharmacological effects.

Time course of the


concentration
Pharmaco-
16
Antimicrobial dynamics
(PD)
Intensity and duration of
the pharmacological effect

The effect usually measured is not, in fact, the destruction of the


microorganism but the inhibition of bacterial growth.

Growth inhibition measurement is carried out by in vitro


microbiological techniques which allow clinicians to test
the susceptibility of the microorganism to one or several
antimicrobials.

Ideally the clinician would have access to the usual susceptibility


pattern and the licensed antimicrobials available for treatment.

Example of in vitro sensitivity testing. A clear area


around the antibiotic disc shows elimination of the
bacteria and therefore susceptibility. No clear area
suggests there is little bacterial susceptibility and the
antibiotic may not be an appropriate choice in this case.

1
Basic Pharmacology

Susceptible

Qualitative Intermediate
techniques

Microorganism Resistant
susceptibility

MIC
Quantitative
techniques
MBC

4.1. Minimum inhibitory concentration (MIC)


The MIC is the lowest antimicrobial concentration that inhibits in vitro the growth of the target
bacteria in specific incubation conditions (Sánchez-Rubio, 1999; Mckellar, 2004).

These conditions are not the same as those in which bacteria grow in vivo (blood, extracellular
fluid, intracellular environment, urine, milk or in the presence of pus or debris). This explains why 17
sometimes the data obtained in vitro does not always match what occurs in vivo.

In spite of its limitations, the MIC is the most used pharmacodynamic parameter
when it comes to antimicrobials (Mouton, 2005).

Not all the strains of a bacterium have the same MIC. For this reason, two criteria are used to
evaluate the susceptibility of a bacterial species to an antimicrobial. Namely, the MIC50 and the
MIC90, which are defined as the lowest concentrations of antimicrobial that inhibit growth of 50%
and 90% of the total population of the target bacteria.

From a practical point of view, taking into account the MIC90 is more useful because this
parameter covers most of the pharmacodynamic variations observed at population level. In the
next table we can observe the comparative MIC and MPC1 values for 285 M. haemolytica strains
collected from cattle.

1. Mutant prevention concentration (MPC): the lowest drug concentration that blocks the growth of mutant bacterial sub-populations.

1
Basic Pharmacology

MIC/MPC distribution values (μg/ml)


Drug
≤ 0.008 0.016 0.031 0.063 0.125 0.25 0.5 1 2 4 8 ≥16 ≥32 MIC50/MIC90

MIC distribution

Ceftiofura 17 22 2 0.016/0.016

Enrofloxacin 31 114 16 39 85 0.016/0.125

Florfenicol 5 120 160 2/2

Tilmicosin 3 64 56 82 38 42 2/8

Tulathromycin 2 15 53 98 117 1/2

MPC distribution MPC50/MPC90

Ceftiofura 2 1 19 15 4 1/2

Enrofloxacin 4 31 59 59 49 65 18 0.25/1

Florfenicol 8 64 142 55 16 4/8

Tilmicosin 1 60 58 87 79 16/≥32

Tulathromycin 3 61 138 77 6 4/8

a
Testing against 41 isolates.

Observations from the table


18
T he calculation of MIC50 and MIC90 allows the comparison of antimicrobials for clinical efficacy.
The calculation of MPC50 and MPC90 allows for similar comparison but related to the development
of antimicrobial resistance.

T he lower the MIC value, the better from a clinical point of view. However, considering
pharmacokinetic parameters, it is also necessary to associate the MIC value directly
(pharmacodynamic parameter) with clinical efficacy.

T he rank order (from the lowest to the highest) of MIC90 values was ceftiofur > enrofloxacin >
florfenicol = tulathromycin > tilmicocosin.

T he rank order of antibacterial potency (from the lowest to the highest) based on MPC90 values
was enrofloxacin > ceftiofur > tulathromycin> florfenicol>tilmicosin.

This shows importance of MPC values to help optimize therapy and reduce resistance selection.

1
Basic Pharmacology

4.2. Minimum bactericidal concentration (MBC)


The minimum bactericidal concentration is defined as the lowest concentration of an
antimicrobial required to reduce the initial bacterial population by 99.99% after 24 hours of
incubation at 37 ºC with a standard number of inoculums.

MBC
It reduces the initial bacterial
population by 99.99%

It has been demonstrated that the MIC The techniques to quantify the MBC are
and MBC for bactericidal antimicrobials very complicated.
are very similar.

The MIC is used as a key 19


The MIC provides a good idea pharmacodynamic parameter
of the bactericidal activity. in antibiotherapy.

The analysis of the susceptibility of a bacterium to an antimicrobial carried out


in vitro can underestimate the activity in vivo because of the post-antimicrobial
effect (PAE) and post-antimicrobial leukocyte enhancement effect (PALE).

The post-antimicrobial effect (PAE) can be defined as the growth suppression of a


microorganism as a consequence of being exposed to an antimicrobial, even when the drug is no
longer at the infection site.

I t can be quantified as the difference between the times needed by two populations of the
same bacteria, one having never been exposed to an antimicrobial while the other having
already been exposed, to multiply their population by 10.

T he intensity of the effect depends on the microorganism, the drug, the concentration reached
and the exposure time.

1
Basic Pharmacology

The PAE is greater in vivo than in vitro (Mouton, 2005).

T he PAE can be very important as it can explain the efficacy shown by concentration-
dependant antimicrobials when administered at very long dosing intervals.

Greatest PAE
Gram-positive and
Gram-negative bacteria Gram-positive bacteria gram-negative bacteria

Macrolides β-lactams Carbapenems

Fluoroquinolones –

Aminoglycosides –

The post-antimicrobial leukocyte enhancement effect (PALE) can be defined as the highest
susceptibility to phagocytosis shown by bacteria after exposure to an antimicrobial.

The drugs with a greater PAE tend to have a greater PALE.

 bviously, this effect is not taken into account in in vitro studies, but may be significant when
O
20 it comes to explaining the efficacy of some administration schedules in the treatment of
certain diseases.

For instance, there are new medicinal products that are administered in one single administration
(quinolones or macrolides), the efficacy of these products could be explained not only by their
persistence in the body for long periods of time (one week for example in the case of macrolides)
but also according to their PALE on the bacteria. This effect helps to explain why one antimicrobial
could be efficacious to control a bacterial infection for a longer period of time although there are no
detectable concentrations in the target tissue after its administration.

1
Basic Pharmacology

5.
Clinical efficacy and its relationship with pharmacokinetic
and pharmacodynamic parameters

5.1. What do we use PK/PD models for?


A PK/PD model is a mathematical description providing clinically relevant information about the
relationship between the pharmacokinetics of a drug and its pharmacological effect.

Nowadays, both microbiologists and specialists in infectious diseases believe that PK/PD
models can be an appropriate tool for the design of optimum administration schedules. Clinical
efficacy studies cannot be replaced by these models, although they should compliment an
administration schedule.

21

5.2. Using PK/PD to design an administration schedule


Although the most useful pharmacokinetic parameters when establishing an administration
schedule are the AUC, the maximum drug concentration achieved (Cmax) and the time at which
the concentrations in plasma exceed a previously defined pharmacodynamic threshold, the most
used pharmacodynamic parameter in the case of antimicrobials, as has already been explained
in detail, is the MIC.

The relationship between the concentration of the antimicrobial in


various tissues of the animal under treatment and the effect on the
target bacteria is not simple.

1
Basic Pharmacology

The efficacy of the treatment being applied could depend on:

Reaching a concentration in plasma several times higher than the MIC of the pathogen (Cmax/MIC).

 aintaining a concentration in plasma above the MIC for a prolonged period (T>MIC)
M
(Craig, 1998).

A combination of the exposure time to the antimicrobial and the concentration reached (AUC/
MIC).

If these objectives are not reached, the probability of the emergence of resistant
strains after the treatment increases (Blondeau, 2001).

The PK/PD parameters which have been studied in depth are AUC/MIC, Cmax/MIC and T>MIC.

Antimicrobial drugs have been classified as concentration-dependent where increasing


concentrations at the site of infection improves bacterial kill, or time-dependent where exceeding
the MIC for a prolonged percentage of the inter-dosing interval (T>CMI) correlates with improved
efficacy.

Ratios of 100-125 for AUC0-24/MIC and 10 for Cmax/MIC have been recommended to achieve high clinical
22
efficacy for concentration-dependent antimicrobial drugs such as enrofloxacin and marbofloxacin,
and exceeding MIC by 1-5 times for between 40 and 100% of the inter-dosing interval is appropriate for
most time-dependent agents, like amoxicillin or classical macrolides (Mckellar, 2004).

1
Basic Pharmacology

In PK/PD models, average pharmacokinetic parameters and MIC90 of the population of animals
and bacteria are used to compare the PK/PD parameters obtained with the threshold values to
predict clinical efficacy.

The figure below shows the pharmacokinetic profile of an antimicrobial after intramuscular
administration and different PK/PD parameters.

Cmax/MIC
Aminoglycosides
Fluoroquinolones

Fluoroquinolones
AUC>MIC β-lactams
Concentration

MIC
T>MIC
PAE 23

Threshold values that could be


associated with clinical efficacy:
β-lactams • AUC0-24/MIC ≥ 125.
• Cmax/MIC ≥ 10.
Time • T >MIC= 40-100%.

For example, in the particular case This objective can be reached if the Css is
of β-lactams, the most appropriate higher than 5 x MIC.
dosage regime is the one which
maintains a Css above the MIC for For this reason, the dosage regime can be
most of the dosing interval. crucial in selecting resistant strains.

1
Basic Pharmacology

5.3. PK/PD and the classification of antimicrobials


Antimicrobials which are normally used in cattle medicine can be divided into three groups,
taking into account the PK/PD parameters and their correlation with the clinical efficacy of the
treatments (Mckellar, 2004):

Antimicrobial classification depending on PK/PD models

Concentration-dependent antimicrobials Time-dependent antimicrobials Co-dependent antimicrobials

Time-dependent antimicrobials are Co-dependent antimicrobials


Concentration-dependent
those whose effect on microorganisms are those whose effect on
antimicrobials are those whose
depends on the exposure time to the microorganisms depends equally
effect on microorganisms depends
drug rather than on its concentration, on their concentration as
on their concentration:
as long as it exceeds the MIC: on the exposure time:
• Aminoglycosides.
• ββ-lactams. • New macrolides (azalides).
• Fluoroquinolones. • Fluoroquinolones for anaerobic germs.
• Some macrolides.

In their case, the Cmax/MIC ratio is the In their case, the T>MIC is the In their case, the PK/PD index which
PK/PD index which best PK/PD index which best correlates best correlates with clinical efficacy
correlates with clinical efficacy. with clinical efficacy. is usually the AUC/MIC ratio.

24 This is a feature of both the antimicrobial and the classification of the bacteria.

For example, marbofloxacin presents:


A concentration-dependent activity for some gram-negative microorganisms
(e.g. Pseudomonas spp.).
A co-dependent activity for other gram-negative bacteria (e.g. Pasteurella multocida).
A time-dependent activity for gram-positive bacteria (e.g. Staphylococci).

T his shows that in order to select the correct antimicrobial regime, the prescriber needs a good
knowledge of both the microorganism to be treated and the drug to be administered.

The information provided in the previous table helps us understand the different administration
schedules of commonly used in cattle antibiotics. There are some antibiotics that require
multiple administrations or have a long acting presentations (ß-lactams, for instance), and
others are administered in one-shot presentations (quinolones for example).
The rationale behind this difference is based on how the PK/PD parameters can be achieved in
order to be clinically effective.

1
Basic Pharmacology

Although it is not possible to generalise in all cases, the PK/PD parameters can be examined in
relation to their capacity to predict the therapeutic success of the antimicrobials:

PK/PD parameters associated with clinical success using antimicrobials

Parameter Antimicrobial

Cmax/MIC ratio Aminoglycosides and fluoroquinolones.

Aminoglycosides, fluoroquinolones, new macrolides (e.g. tulathromycin)


AUC/MIC ratio
and tetracyclines.
Penicillins, cephalosporins, classical macrolides (e.g. tilmicosin),
T>MIC
pleuromutilins (e.g. tiamulin) and lincosamides.

Finally, another way of classifying antimicrobials is by looking at their bacteriostatic or


bactericidal effect.

A ntimicrobials with a bacteriostatic effect are those which only inhibit bacterial growth, in such
a way that the clinical and bacteriological overcome of the infection depends on the animal’s
immune system (e.g. tetracyclines and “classical” macrolides).

 actericidal antimicrobials are able to destroy the target bacteria (e.g. β-lactams and
B 25
fluoroquinolones).

1
Basic Pharmacology

Amphenicols Florfenicol
Macrolides2 Tilmicosin
Pleuromutilins Tiamulin
Mainly bacteriostatic Lincosamides Lincomycin
Tetracyclines Doxicycline
Sulphonamides Sulphametazine
Diaminopiridines Trimethoprim

ACTION Mainly time-dependent


bactericidal
Penicillins
Cephalosporins
Amoxicillin
Ceftiofur

Mainly concentration-
dependent bactericidal Aminoglycosides Streptomycin
with very significant Fluoroquinolones Enrofloxacin
postantimicrobial action

2. This is true for all macrolides. New macrolides must be classified in a case by case scenario.

5.4. Using a combination of antibiotics at the same time


Ideally a single antimicrobial would be selected to treat an infection for maximum clinical efficacy
and to reduce the probability of generating antimicrobial resistance. However, in some cases, we
26 need to combine different antimicrobials.

It is important to remember that some combinations could be antagonistic, synergistic or


indifferent if the antimicrobials are acting on the same bacteria.

Antagonistic combination: the combined effect of two antimicrobials is lower than the effect of
the antimicrobials administered separately. This combination must be avoided.

Antimicrobial ββ-lactams Aminoglycosides Polypeptidic Quinolones

Macrolides
Pleuromutilins
Tetracyclines
Sulphonamides Antagonistic combination
Diaminopiridines
(Trimethoprim)
Amphenicols

Aminoglycosides and polypeptidic combination is also antagonic.

1
Basic Pharmacology

Synergistic combination: the combined effect of two antimicrobials is greater than the effect of
those antimicrobials administered separately.

Antimicrobial β-lactams Aminoglycosides Polypeptidic

β-lactams NA Synergistic Synergistic

Aminolycosides Synergistic NA Antagonistic

Polypeptidic Synergistic Antagonistic NA

NA: non-applicable. Sulphonamides and trimethoprim combination is also synergistic.

Indifferent combination: Do not use a combination of antibiotics unless there is synergy.

Any other kind of antimicrobial combination is indifferent. For instance:

Quinolones and all other bactericidal antimicrobials.

A ny combination of bacteriostatic antimicrobials, with the exception of sulphonamides and


trimethoprim.

27

1
Basic Pharmacology

6.
Withdrawal period, a critical aspect to ensure food safety
When selecting an antimicrobial for use in cattle or other food producing animals, it is important
to ensure the level of drug residues present in edible tissues are below those established by the
European legislation maximum residue limits (MRLs).

The withdrawal period is a specific set period of time, after the last dose of the veterinary
medicine has been administered, that must elapse before an animal or foodstuffs from an
animal can enter the food chain. To determine the withdrawal period, regulatory authorities must
employ a scientific process that includes establishing the Maximum Residue Limit (MRL) for that
medicine.

The MRL is the maximum concentration of residue accepted by the European Union (EU) in a food
product (e.g. meat, milk) obtained from an animal that has received a veterinary medicine.
Concentration

Absorption
Distribution

28 Depletion phase
(elimination only)

Ct < MRL
MRL

Treatment Withdrawal period Food production


Ct: concentration in tissues.

For some antimicrobials both the parent drug and any active metabolites are taken into
consideration and must all be below the MRL at slaughter. This is specific for each drug.

Specific analytical methods have also been developed to ensure the levels of antimicrobials or
their active metabolites are below the established levels in the milk tank.

1
Basic Pharmacology

7. List of references

Baggot J.D., The physiological basis of veterinary clinical pharmacology. Blackwell Sciencie Ltd., Oxford, 2001.

Blondeau, et al. Mutant prevention concentrations of fluorquinolones for clinical isolates of Streptococcus pneumoniae.
Antimicrobial Agents Chemotherapy 2001; 45: 433-438.

Craig W.A. Pharmacokinetic/pharmacodynamic parameters: Rationale for antibacterial dosing of mice and men. Clinical
infectious diseases 1998; 26: 1-12.

Goodman & Gilman’s. The Pharmacological Basis of Therapeutics. Authors: Brunton, Laurence; Lazo, John ; Parker, Keith.
McGraw-Hill education 2006; 11th Edition.

Lees, et al. Principios de antibioterapia. En Farmacología y terapéutica veterinaria. Botana L.M, Landoni F., Martín-
Jiménez, T. Ed. Mcgraw-Hill interamericana 2002.

Martín-Jiménez, T. Farmacocinetica I: absorción y distribución. En Farmacología y terapéutica veterinaria. Botana L.M,


Landoni F., Martín-Jiménez, T. Ed. Mcgraw-Hill interamericana 2002.

Mckellar, Q.A. et al. Pharmacokinetic/pharmacodynamic relationship of antimicrobial drugs used in veterinary medicine.
Journal Veterinary Pharmacology Therapeutics 2004; 27: 503-514.

Mouton J.W., Vinks A.A. Pharmacokinetic/pharmacodynamic modelling of antibacterials in vitro and in vivo using
bacterial growth and kill kinetics. The minimum inhibitory concentration versus stationary concentration. Clinical
pharmacokinetics 2005; 44(2): 201-210.
29
Prescott, et al. Antimicrobial therapy in veterinary medicine. 3. ed. Iowa State University Press, Ames, Iowa 2000.

Rowland, M., Tozer, T., Clinical pharmacokinetics. Concepts and applications. Williams and Wilkins, Media PA 1995.

Sánchez-Rubio, A., Sánchez Recio, MM. Basis of anti-infective therapy. Pharmacokinetic-pharmacodynamic criteria and
methodology for dual dosage individualisation. Clinical Pharmacokinetics 1999; 37(4): 289-304.

Toutain, et al. Plasma clearance. Journal Veterinary Pharmacology Therapeutics 2004a; 27: 414-425.

Toutain, et al. Volumes of distribution. Journal Veterinary Pharmacology Therapeutics 2004b ; 27: 441-453.

Toutain, et al. Plasma terminal half-life. Journal Veterinary Pharmacology Therapeutics 2004c; 27: 427-439.

1
Glossary

ADME: absorption, distribution, metabolism, excretion. MIC90: lowest concentration of antimicrobial that inhibit growth
of 90% of the total population of the target bacteria.
Antagonistic combination: the combined effect of two
antimicrobials is lower than the effect of the antimicrobials Mutant prevention concentration (MPC): lowest drug
administered separately. concentration that blocks the growth of mutant bacterial
sub-populations.
Area under the curve (AUC): the area under the curve in a plot
of concentration in plasma versus time. Partition coefficient (P): ratio of concentrations in tissue and
plasma at steady state.
Bactericidal antimicrobials: those which are able to destroy
the target bacteria. Pharmacodynamics (PD): science that describes the
relationship between the time-course of the concentration
Bacteriostatic antimicrobials: those which only inhibit of a drug in the organism and the intensity and duration of
bacterial growth. its pharmacological effects.

Co-dependent antimicrobials: those whose effect on Pharmacokinetics (PK): science that describes the kinetics of
microorganisms depends equally on their concentration as drug molecules when they are introduced in the organism
on the exposure time. and defines the pattern of ADME processes.

Concentration-dependent antimicrobials: those whose effect Plasma clearance (CL): volume of blood that is cleared of drug
on microorganisms depends on their concentration. per unit of time and per kilogram of live weight.

Half-life (t½): time required for the concentration in plasma Post-antimicrobial effect (PAE): growth suppression of a
to be reduced to half once the distribution equilibrium has microorganism as a consequence of being exposed to an
been reached. antimicrobial, whilst the drug is no longer at the infected
area.
Indifferent combination: the combined effect of two
antimicrobials is similar to the observed effect for each Post-antimicrobial leukocyte enhancement effect (PALE):
antimicrobial administered separately. highest susceptibility to phagocytosis shown by bacteria
after exposure to an antimicrobial.
Maximum concentration (Cmax): maximum concentration
observed after the administration of a drug to an animal by Synergistic combination: the combined effect of two
any route of administration. antimicrobials is greater than the effect of those
antimicrobials administered separately.
Maximum residue limits (MRL): maximum concentration
of residue accepted by the European legislation in a food Time-dependent antimicrobials: those whose effect on
product obtained from an animal that has received a microorganisms depends on the exposure time.
veterinary medicine.
Volume of distribution (Vd): constant of proportionality
Minimum bactericidal concentration (MBC): lowest between the concentration in plasma and the amount of a
concentration of antimicrobial able to reduce the initial drug in the organism.
bacterial population by 99.99% after 24 hours of incubation
at 37 ºC with a standard amount of inoculums. Withdrawal period: interval between the last administration
of a veterinary medicinal product to animals under normal
Minimum inhibitory concentration (MIC): lowest antimicrobial conditions of use and the production of foodstuff from
concentration that inhibits in vitro the growth of the target such animals to ensure that such foodstuffs do not contain
bacteria in specific conditions of incubation. residues in quantities in excess of the maximum residue
limits laid down.
MIC50: lowest concentration of antimicrobial that inhibit growth
of 50% of the total population of the target bacteria.
August 2018

Use medicines responsibly (www.noah.co.uk/responsible)

Ceva Animal Health Ltd


Unit 3, Anglo Office Park, White Lion Road, Amersham, Bucks HP7 9FB
Tel: 01494 781510 www.ceva.co.uk

You might also like