Pharmacology/Toxicology: 4 Flashcards

1
Q

Administration rate of potassium

How much potassium is in RL?

A

(in form of potassium chloride)

<0.5mEq/kg/HOUR

in Liège: Braun KCl 14.9% amp.20ml –> 2mEq/ml K+

for a 500kg horse: 250mEq/HOUR –> 125ml/HOUR -> 6amp/HOUR

Attention if adding to RL: already 20mEq in a 5L bag

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

fluid composition of Lactated Ringer’s Solution

difference to Plasmalyte and Normosol?

A

LRS: Na 130 K 4 Cl 109 Ca 3 pH 6.5

Plasma Lyte A: Na 140 K 5 Cl 98 Mg 3 pH 7.4

Normosol - R: Na 140 K 5 Cl 98 Mg 3 pH 6.6

LRS contains lactate (metabolized by liver and kidney)

whereas the other two contain acetate and gluconate (by a number of tissues)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Mechanism of action of Misoprostol

side effects

A

= synthetic analogue of PGE1 (5µg/kg PO BID)

= Cytotec 200-mcg in Liège: 12comprimés BID pour cheval de 500kg

Prostaglandin E2 and prostacyclin (PGI2) are the major PGs synthetized by the gastric mucosa

–> bind to EP3 Receptors on parietal cells and stimulate the Gi pathway –>

  • decreasing intracellular cyclic AMP and gastric acid secretion
  • ups mucin and bicarbonate secretion and
  • increase mucosal blood flow

side effects: ABORTION, abdomial cramping, diarrhea, sweating

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

dose of heparin for sepsis and types of heparin

A

heparin 20-80IU/kg IV or SC every 6-12hrs

high molecular weight heparin causes anemia by inducing aggregation of red blood cells, which is undesirable in sepsis

low molecular weight heparins appears to be less likely to have that effect

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Mechanism of action of benzimidazoles

A

f.e. fenbendazole

inhibition of microtubule polymerization by by binding to beta tubulin, also reduction of energy metabolism

-> flaccid paralysis

negligable side effects

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Mechanism of action of macrocyclic lactones

Side effects?

A

fe eavermectins: immobilization of affected organisms by inducing tonic paralysis of the musculature by activating ligand gated Cl channels (only present in invertebraes) causing Cl influx and hyperpolarization

also stimulates synptic excretion of GABA in the mamalian brain –> open chloride channels –> membrane hyperpolarisation

–> causes ataxia and blindness in overdose or potentially foals <4months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Mechanism of action of Biosponge

A

Biosponge = Di Tri Octahedral Smectite, binding Clostridum toxins in vitro

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

difference between ionized and non ionized drugs?

A
  • ionized drug is hydrophilic and poorly lipid soluble.
  • nonionized drug is lipophilic and can cross biologic membranes.

depends on the environment

f.e. Weak acids such as penicillins and cephalosporins are highly ionized in plasma and therefore do not penetrate into the mammary gland very well, so these are most effective when administered by local infusion into the udder, where the extremely high local concentrations negate local pH effects.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Difference between Pharmako-kinetics and -dynamics in antimicrobials

A

Pharmacokinetics is what the body does to a drug; the processes of absorption, distribution to the various organs and tissues, metabolism, and elimination.
Pharmacodynamics is what the antimicrobial does to the bacteria.
It describes the drug action and responses of the bacteria.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Define MIC and MBC

A

MIC: The lowest drug concentration that inhibits bacterial growth. Often expressed as the concentration that inhibits 50% (MIC50) or 90% (MIC90) of the bacterial growth.

=> based on safely achievable plasma concentrations. This does not necessarily take into account extremely high concentrations of antimicrobials achieved in organs and fluids of excretion (kidney, urine, bile) or with local administration of high drug concentrations (e.g., ophthalmic ointments).

f.e. Macrolide antimicrobials are characterized by negligible plasma concentrations despite very high lung and intracellular concentrations

Minimum bactericidal concentration (MBC): The lowest drug concentration that kills 99.9% of bacteria.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

What are breakpoints? What is the problem in equine medicine?

A

A breakpoint is a chosen concentration (mg/L) of an antibiotic which defines whether a species of bacteria is susceptible or resistant to the antibiotic.

If the MIC is less than or equal to the susceptibility breakpoint the bacteria is considered susceptible to the antibiotic. If the MIC is greater than this value the bacteria is considered intermediate or resistant to the antibiotic.

Problem: only ceftiofur, ampicillin, amikacin, and gentamicin have CLSI-approved breakpoints for equine pathogens - the rest is derived from human medicine

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

What is the postantibiotic effect?

A

for some ABs; bacterial growth remains suppressed for a period after drug concentration has decreased below the MIC.

This postantibiotic effect (PAE) may be the reason that dosage regimens that fail to maintain drug concentration above the MIC are still efficacious. The PAE depends on the antimicrobial and the bacterial pathogen.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Classify the antibiotic classes into bactericidal and bacteriostatic

A

BACTERICIDAL

  • Aminoglycosides
  • β-lactams
  • Fluoroquinolones
  • Trimethoprim/sulfonamides

BACTERIOSTATIC

  • Chloramphenicol
  • Macrolides
  • Sulfonamides
  • Tetracyclines
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Classify the antibiotic classes into concentration- or time-dependant

A

CONCENTRATION-DEPENDENT ANTIMICROBIALS
Aminoglycosides
Fluoroquinolones
Metronidazole
TIME-DEPENDENT ANTIMICROBIALS
Cephalosporins
Chloramphenicol
Macrolides
Penicillins
Sulfonamides
Tetracyclines

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Name drugs with a postantibiotic effect of over 3hours

A

Gram positive: Fluoroquinolones, Macrolides, Chloramphenicol, Tetracycline

Gram negative: Fluoroquinolones, Aminoglycosides

Anaerobes: Metronidazole

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Combination of Chloramphenicol and Macrolide

A

= antagonistic combination both
these classes of drugs competitively bind to the same binding
site on bacterial ribosomes, thus effectively battling each other
for the site of action.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

β-lactam antibiotics

  • include …
  • activity against
  • time/dose? static/cidial?
  • binds to
  • distribution
  • excretion
  • half life
A
  • include the penicillins, cephalosporins, and carbapenems.
  • excellent activity against most gram-positive bacteria and very few associated side effects.
  • bactericidal and time dependent.
  • low plasma protein binding,
  • distribute well to the ECF in most tissues, but do not distribute well to protected sites, such as the central nervous system, the eye…
  • excreted renally
  • short half-life and require frequent dosing.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Mechanism of action of β-lactam antibiotics. Spectrum is defined by..?

A

β-Lactam antibiotics bind irreversably on penicillin-binding proteins (PBPs) responsible for building the bacterial cell wall. Therefore they are active only against rapidly multiplying organisms in which the binding of penicillin within the
cell wall interferes with production of cell wall peptidoglycans and results in cell lysis in a hypo-osmotic or iso-osmotic environment.
Differences in the spectrum and activity bc of relative affinity for different PBPs.

To bind to the PBPs, the β-lactam antibiotic must first diffuse through the bacterial cell wall. Gram-negative organisms have an additional lipopolysaccharide layer that decreases antibiotic penetration. => gram-positive bacteria more susceptible

poor penetratation in mammalian cells => ineffective for intracellular pathogens.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Name the resistance mechanisms against B-Lactams

A
  • failure of the antibiotic to penetrate the outer bacterial cell layers
  • alteration of PBPs that decrease the affinity of the PBP for the antibiotic. f.e. methicillin-resistant staphylococci (alterations of the PBPs to a low-affinity PBP2a - by gene mecA)
  • production of β-lactamase enzymes => hydrolyze the cyclic amide bond of the β-lactam ring and inactivate the antibiotic. f.e. Staphylococcal β-lactamases are produced by coagulase-positive Staphylococcus spp. The synthesis of these enzymes is plasmid encoded, and the enzymes are exocellular.
    • => add inhibitors such as clavulanic acid and sulbactam
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

BENZYLPENICILLIN

  • Spectrum
  • ionized/non-ionized
  • absorption
  • excretion
  • adverse effects
A

= Penicillin G

  • Aerobic bacteria: β-hemolytic streptococci, β-lactamase–negative staphylococci, Actinomyces spp., some Bacillus anthracis, Corynebacterium
    spp. Most species of anaerobes susceptible, excluding β-lactamase–producing
    Bacteroides spp
  • Because penicillin is a weak acid with a pKa of 2.7, it is highly ionized in plasma.
  • depending on product: Na/K Peni IV, Procain Peni IM, Benzathine penicillin G is the least soluble; very slowly absorbed, producing sustained but subtherapeutic plasma concentrations => not recommended in horses
  • renal: glomerular filtration and active renal tubular secretion.
  • immune-mediated hemolytic anemia (type II hypersensitivity) and anaphylaxis (type I hypersensitivity)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Difference between aminopenicillins and benzylpenicillin?

A

The aminopenicillins (zB Amoxicillin) are able to penetrate the outer layer of gram-negative bacteria better than benzylpenicillins (penicillin G)

therefore they have activity against many of the gram-negative bacteria (E. coli, Salmonella, Pasteurella spp.) as well as gram-positive bacteria.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Name anti-pseudomonal penicillins

A

Pseudomonas = gram neg

Carbenicillin, ticarcillin, and piperacillin

TICARCILLIN licensed intrauterin in horses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Aminoglycosides

  • Examples
  • Spectrum
  • ionized/non-ionized
  • -cidal/-static? concentration/time?
  • ineffective under which circumstances?
A
  • streptomycin, neomycin, gentamicin, amikacin, tobramycin,
  • aerobic gramnegative bacteria and staphylococci.
  • highly ionized at physiologic pHs; ; binds to and is inactivated by the nucleic acid material released by decaying white blood cells. (ineffective in the
    acidic, hyperosmolar, anaerobic environment of abscesses)
  • bactericidal and concentration dependent.
  • ineffective against anaerobic bacteria (penetration into the bacteria requires an oxygen-dependent transport mechanism) and against intracellular bac (f.e. Salmonella) and in puss
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Mechanism of action of aminoglycosides

A

Susceptible, _aerobic gram-negative bacteria actively pump the aminoglycoside
into the cell (_oxygen-dependent interaction between the antibiotic cations and the negatively charged ions of the bacterial membrane lipopolysaccharides)

=> bind to the 30S ribosomal subunit and cause a misreading of the genetic code, interrupting normal bacterial protein synthesis.

=> changes the cell membrane permeability, resulting in additional antibiotic uptake, further cell disruption, and ultimately cell death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

Mechanisms of aminoglycoside resistance

A

Plasmid-mediated resistance transferable between bacteria.

containing phosphotransferases, acetyltransferases, and adenyltransferases acting
internally to alter the aminoglycoside and prevent it from binding to ribosomes. Amikacin is least susceptible to enzyme inactivation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

Distribution of aminoglycosides

Elimination

A
  • polar antibiotics; therefore distribution is limited to the ECF space:

penetrates into synovial, perilymph, pleural, peritoneal, and pericardial fluid.

not present in CSF, respiratory secretions, and ocular fluids. Gentamicin does not cross the placenta of late-term mares;
predominant site of drug accumulation is the renal cortex in most species.

  • eliminated in urine by glomerular filtration

The administration of therapeutic fluids, colic surgery, or peritoneal lavage does
not significantly change the pharmacokinetics of concurrently administered gentamicin.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q
A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Mechanism of nephrotoxicity of aminoglycosides.

Risk reduction through supplementation of …

A

filtration through the glomerulus => renal tubule

=> carrier-mediated pinocytosis (with phospholipids) and translocatation into cytoplasmic vacuoles of renal tubular cells => fusion with lysosomes => sequestration => overloaded lysosomes swell and rupture.

=> Lysosomal enzymes, phospholipids, and the aminoglycoside are released into the cytosol of the proximal tubular cell, disrupting other organelles and causing cell death.

  • supplementation of Calcium can reduce the risk of nephrotox
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

Gentamicin dose

A
  1. 6mg/kg IV SID
    foals: may need double bc ECS larger!
30
Q

Early diagnosis of aminoglycoside induced nephrotoxicity

A
  1. increase in urine gamma glutamyl transferase (UGGT) enzyme and an increase in the UGGT:urine creatinine (UCr) ratio. The UGGT:UCr may increase to 2 to 3 times baseline within 3 days of a nephrotoxic dose.
  2. development of proteinuria is the next best indicator of nephrotoxicity and is easily determined in a practice setting.
  3. Elevations in serum urea nitrogen and Cr confirm nephrotoxicity but are not seen for 7 days after significant renal damage has occurred.
31
Q

Amphenicols

  • examples
  • static/cidic?
  • mechanism of action
  • spectrum
  • mechanisms of resistance
A
  • f.e. Chloramphenicol
  • bacteriostatic antibiotic
  • binds to 50 S ribosomal subunits >>> inhibition of peptidyl transferase and thereby preventing protein formation.
  • very wide spectrum of activity, including streptococci, staphylococci (including many MRSA), anaerobes, Haemophilus, Salmonella, Pasteurella, Mycoplasma, and Brucella spp
  • plasmid-mediated bacterial production of acetylase enzymes.
32
Q

Chloramphenicol

  • Bioavailability
  • Adverse effects
  • Drug interactions
  • elimination
  • distribution
A
  • 80% after oral administration in foals, only 40% in adults
  • drug = protein synthesis inhibitor => doserelated anemia and pancytopenia are associated with chronic therapy (longer than 14 days), causing a decrease in protein synthesis in the bone marrow. ATTENTION when handling drug - can cause aplastic anemia in humans!
  • should not be administered concurrently with penicillins, macrolides, aminoglycosides, or fluoroquinolones.
    (CHPC may antagonize the activity of penicillins or aminoglycosides, and they act on the same ribosomal site as the macrolides.)
  • renal excretion of parent drug and by hepatic glucuronide conjugation and elimination in feces.
  • penetration into abcesses excellent
33
Q

Sulfonamides

and

potentiated sulfonamides

-> mechanisms of action

A

sulfonamides are a group of organic compounds with chemotherapeutic activity (hence they are antimicrobials, not antibiotics).

=> inhibit the bacterial enzyme dihydropteroate synthetase (DPS) in the folic acid pathway, >> blocks bacterial nucleic acid synthesis. Sulfonamides substitute for PABA (=wichtiger wuchsstoff zur bildung von folsäure)

potentiated: combination of a sulfonamide and pyrimidine potentiator

=> The addition of TMP or PYM to a sulfonamide creates a bactericidal combination.

TMP inhibits bacterial folic acid synthesis at the next step in the folic acid sequence, inhibiting the conversion of dihydrofolic acid to tetrahydrofolic acid by inhibiting dihydrofolate reductase.

34
Q

Distribution and elimination of potentiated sulfonamides

Adverse effects

Drug interactions?

A
  • well distributed into peritoneal fluid, CSF, synovial fluid, and urine.
  • renal elimination
  • Crystalluria, hematuria, and renal tubular obstruction can result from poorly soluble sulfonamides (but normally dose of sulfonamides are too low in combination with TMP to cause crystallisation), Anemia, thrombocytopenia, and even pancytopenia after long-term, high-dose administration (EPM) bc of folate deficiency due to inhibition of folate production by intestinal bacteria
  • concurrent use with detomidine is contraindicated because it appears that the potentiated sulfonamide sensitizes the myocardium and results in cardiac dysrhythmias and hypotension that may be fatal.

The procaine in procaine penicillin G is a PABA analog and may reduce efficacy if used concurrently with potentiated sulfonamides.

35
Q

Tetracyclines

  • f.e.
  • mechanism of action
  • static/cidal?
A
  • f.e. Oxytetracycline (OTC), doxycycline (DXC), and minocycline (MNC)
  • bind to the 30S ribosomal subunit and interfere with bacterial protein synthesis. Drug entry into the bacteria is by an energy dependent
    mechanism. Mammalian cells do not possess the tetracycline transport mechanism.
  • bacteriostatic at usual therapeutic concentrations but bactericidal at high
    concentrations.
36
Q

Tetracyclines

  • spectrum
  • additional actions?
A
  • broad-spectrum in activity: effective against gram-positive and gramnegative bacteria, most anaerobes, intracellular pathogens (Lawsonia, Rhodococcus) as well as Chlamydia, Mycoplasma, and Rickettsia spp. and some protozoa (Anaplasma spp.),
  • limited activity against staphylococci, not active against enterococci. Pseudomonas, E.coli, Klebsiella, and Proteus are usually resistant.

=> additional actions: f.e. DXC and MNC: anticollagenolytic activity through inhibition of matrix metalloproteinases (MMPs), antiinflammatory activity, and ability to enhance corneal repair;

37
Q

Tetracyclines

  • Distribution
  • Elimination
  • Side effects
  • Drug interference
A
  • distributes well in all tissues except CSF; excellent distribution of OTC in urine, doxycycline intracellular++ and intrauterine++

OTC is eliminated in urine unchanged primarily by glomerular filtration. Unmetabolized drug is also _eliminated with bile into the GI trac_t and may undergo enterohepatic recirculation, prolonging its effects.

DXC is primarily excreted into the feces by way of nonbiliary routes in an inactive form. Therefore DXC does not accumulate in patients with renal insufficiency.

  • OTC = nephrotoxic at prolonged treatment (not single dose); Rapid IV administration of OTC results in hypotension and collapse. This is attributed (intravascular chelation of calcium, decreased blood pressure from the drug vehicle, or both. Pretreatment with IV calcium borogluconate prevents collapse.
  • Do not use in foals with rifampicin (unexplained hemolytic anemia and/or high liver values after concurrent long term treatment)
38
Q

Macrolides

  • f.e.
  • mechanisms of action
  • static/cidal?
  • base/acid?
A

Because of their adverse GI effects, these drugs are typically contraindicated in adult horses; however, erythromycin, clarithromycin, and azithromycin are commonly used in foals.

bind to the 50S ribosomal subunit in a manner similar to that of CHPC, and they
interfere with protein synthesis.

bacteriostatic but may be bactericidal at high concentrations.

weak base => works optimally at an alkaline pH; therefore they have reduced activity in acidic environments (e.g., pus, abscesses) but may be clinically effective because of high concentrations due to ion trapping.

39
Q

Spectrum of Macrolides

A

Macrolides are not effective against gram-negative bacteria, except some strains of Pasteurella and Haemophilus.18

Azithromycin is more active than the macrolides against gram-negative bacteria and anaerobes.

Susceptible bacteria include:
staphylococci, streptococci, Campylobacter jejuni, Clostridium
spp., Rhodococcus equi, Lawsonia intracellularis, Mycoplasma
spp., and Chlamydia spp

40
Q

Distribution of macrolides

Elimination

Resistance

A

The macrolides concentrate in leukocytes, making them very effective against intracellular pathogens such as R. equi

Erythromycin is extensively metabolized, with much of the parent drug and active metabolite excreted into the bile.

Unstable in gastric acid! Use coated

bacterial resistance develops quickly after repeated exposure.

Mechanisms: decreased drug entry into bacteria, inability to bind to the bacterial 50S ribosomal subunit, and plasmid-mediated production of esterases.

41
Q

Macrolides and rifampicin?

A

Clarithromycin is also frequently administered in combination with rifampin due to synergistic effects against R. equi. However, coadministration of rifampin reduces clarithromycin bioavailability by as much as 90%, resulting in plasma
concentrations of clarithromycin that are below the MIC90 of R. equi.

42
Q

Fluorchinolones

  • pe
  • mechanism of action
  • time/conc? static/cidal?
  • spectrum
A
  • ciprofloxacin, enrofloxacin, marbofloxacin and levofloxacin,…
  • inhibit bacterial deoxyribonucleic acid (DNA) gyraseand inhibit the DNA resealing, resulting in abnormal spatial DNA configuration, which leads
    to DNA degradation by exonucleases
  • concentration dependent, bactericidal
  • most gram-negative bacteria; some grampositive bacteria; and Mycoplasma, Chlamydia, and Rickettsia spp. particularly effective against the enteric gramnegative pathogens, including some strains resistant to aminoglycosides and cephalosporins. not active against anaerobic
    bacteria.
43
Q

Fluorchinolones

  • distribution
  • elimination
  • side effects
  • dose
A
  • concentrate within phagocytic cells, lipophilic, diffuse well into tissues Extremely high concentrations are achieved in the kidney, urine, liver, and bile.
  • renal elimination
  • age dependant cartilage damage with subsequent arthropathy. (not recommended for use in pregnant humans or animals, but seems to have little effect on the developing fetus). increased seizure activity bc of γ-aminobutyric acid (GABA) receptor antagonism
  • ONCE DAILY USE, enro: 7.5-10mg/kg
44
Q

Rifampicin =

  • Mechanism of action
  • static/cidal?
  • spectrum
A
  • rifamycins are antibiotics produced from Streptomyces mediterranei.
  • Rifampin inhibits DNA-dependent RNA polymerase in susceptible organisms, suppressing RNA synthesis.
  • bacteriostatic or bactericidal, depending on the susceptibility of the bacteria and the concentration of the drug.
  • effective against a variety of mycobacterium species and S. aureus, Haemophilus, and R. equi. Rifampin is considered especially active in the treatment of staphylococcal and rhodococcal infections and in the eradication of pathogens located in difficult-to-reach target areas, such as
    inside phagocytic cells.
45
Q

Rifampicin

  • Distribution
  • Elimination
  • Side effects
A
  • Rifampin is highly lipophilic and penetrates most tissues, as well as milk, bone, abscesses, and the central nervous system.

penetrates puss bc highly acidic

  • deacetylated in the liver
  • stains everything red - urine, tears, sweat,…
46
Q

Mechanism of action and spectrum of metronidazole

Alternative use?

A
  • rapidly taken up by bacteria, where it is metabolized by a reduction process to cytotoxic derivatives (short-lived free radical compounds). These cytotoxic compounds damage DNA and other critical intracellular macromolecules. Aerobic bacteria lack the reductive pathway necessary to produce the radical compounds
  • highly effective against anaerobic bacteria, including Bacteroides fragilis (penicillin resistant), Fusobacterium, and Clostridium spp. Metronidazole-resistant C. difficile may cause diarrhea in foals. Metronidazole has good activity against protozoa, including Giardia and Trichomonas spp.
  • antiinflammatory effects in human beings, particularly in the GI tract, and has been used for the treatment of chronic inflammatory bowel diseases.
47
Q

Metronidazole

  • Distribution
  • Absorption
  • Elimination
  • Side effects
A
  • Metronidazole is lipophilic and widely distributed in tissues. It penetrates bone, abscesses, and the central nervous system.
  • Rapidly absorbed PO, per rectum: 30% absorption
  • liver
  • anorexia and salivation
48
Q

Name the most potent antibiotic against Bacteroides fragilis

A

Metronidazole

49
Q

What does COX do?

Where do the isoforms come from?

What is the influence on renal function and the GI tract?

A

the enzyme cyclooxygenase (COX) converts arachidonic acid to prostaglandins, thromboxane, and prostacyclin

COX-1 is considered to be the constitutively expressed form and is necessary for normal homeostatic mechanisms in the body, COX-1 is found in platelets, the kidneys, and the GI tract;

COX-2 is the inducible form produced in response to injury. identified in fibroblasts, chondrocytes, endothelial cells, macrophages, and mesangial cells. COX-2 is induced by exposure to various cytokines, mitogens, and endotoxin and is upregulated at sites of inflammation

both COX-1 and COX-2 are involved in mucosal defense in the GI tract. and in normal renal function, insofar as prostaglandins affect renal circulation through vasodilation, renin secretion, and sodium and water excretion

50
Q

How do selective COX-2 inhibitors work?

5 examples

How is the activity against each isomer of COX determined in vitro?

A

There is a valine-leucine substitution in COX-2 that is not present in COX-1. This creates a side pocket in the tertiary structure of the molecule, and it is this side pocket that COX-2 inhibitors preferentially bind to.

The selectivity of the drug for COX-2 versus COX-1 therefore depends on the
affinity a drug has for this site.

  • etodolac, meloxicam, deracoxib, carprofen, and firocoxib.
  • COX-1: determined by measurement of thromboxane B2, the stable metabolite of TXA2, which is thought to be specific for COX-1 production in
    platelets. For
  • COX-2: macrophages are stimulated with lipopolysaccharide (LPS), and production of prostaglandin E2 (PGE2) is measured.
51
Q

What is the concept of low-dose use of flunixin?

At which point does it become ineffective?

A

The NSAIDs are commonly used in horses to attenuate the prostaglandin-mediated effects of endotoxin.

Flunixin meglumine administered at doses as low as one fourth the label dose will block prostaglandin production when administered before endotoxin exposure without obscuring the signs of colic pain, and with a lesser risk of adverse effects.

However, at low doses it does not alter endotoxin-induced leukopenia, and with ongoing inflammation there is little benefit in using the low dose over the recommended dose.

Flunixin and phenylbutazone significantly inhibit movement of polymorphonuclear and mononuclear cells and antagonize the effects of endotoxin on bowel motility.

52
Q

Analgesic mechanism of NSAIDs - how does it work?

A

The NSAIDs act as analgesics by inhibiting COX and preventing the production of prostaglandins that sensitize the afferent nociceptors at peripheral sites of inflammation.

However, increasing evidence suggests that some NSAIDs have a central mechanism of action at the level of the spinal cord for analgesia unrelated to COX inhibition. This action is synergistic with opioids and β2-receptor–adrenergic drugs.

53
Q

Time to onset and duration of analgesia and anti-inflammatory effects of NSAIDs?

A

time to onset and duration of analgesia of NSAIDs does not correlate well with
their antiinflammatory properties.

Because the analgesic effect has a more rapid onset and shorter duration of action than the antiinflammatory action, dosage regimens for effective analgesia may need to be different than those for antiinflammatory effects, or adjunct analgesics may need to be used.

54
Q

Why are doses of NSAIDs so different in different species?

A

= chiral drug

Most chiral drugs are formulated as racemic mixtures, containing equal amounts of each enantiomer, because pure enantiomeric compounds are difficult and expensive to manufacture.
All of the propionic acid NSAIDs (i.e., ketoprofen, carprofen, vedoprofen, naproxen) are chiral compounds, and, except for naproxen, they are formulated as racemic mixtures. After administration, some enantiomers undergo chiral inversion, as hepatic enzymes convert one form of the enantiomer to the other form. When chiral inversion of the propionic acid derivatives occurs, it is almost invariably unidirectional, from R to S. The degree of chiral inversion varies among species and cannot be predicted from one species to another, making extrapolating dosages for NSAIDs extremely hazardous.

55
Q

Elimination of NSAIDs?

pH and distribution of NSAIDs

A

Most NSAIDs undergo hepatic metabolism either through oxidation or glucuronide conjugation before being eliminated in the urine

weak acids and highly bound to plasma proteins such as albumin. Therefore well absorbed from the stomach, and most of the drug in the plasma is protein bound => predominantly distributed in the ECF, and only low concentrations
of NSAIDs are found in normal tissues and joint fluid.

In damaged tissues and joints, however, total (bound and unbound) NSAID concentrations increase to therapeutic levels because of increased blood flow, vascular permeability, and acute phase protein penetration into sites of inflammation.

56
Q

Concurrent use of corticosteroids and NSAIDs?

A

Concurrent use of corticosteroids is generally contraindicated because corticosteroids increase the secretion of gastric acid, pepsin, and trypsin; alter the structure of gastric mucin; and decrease mucosal cell proliferation. This action is synergistic with NSAID-induced GI mucosal damage, but the specific risks of concurrent NSAID and steroid administration are unknown.

57
Q

Adverse effects of NSAIDs

A
  • renal papillar necrosis
  • ulceration of the GI tract
  • inhibition of platelet aggregation by preventing thromboxane production via the COX-1 pathway. Recovery of platelet function is dependent on the pharmacokinetics of the NSAID and the mechanism of COX inhibition. Aspirin
    permanently modifies COX, so platelet function is restored only by the production of new platelets
58
Q

Important effect (+mechanism) of Aspirin besides anti-inflammatory or analgesic effect?

A

Aspirin is the most effective NSAID for antiplatelet therapy.
Aspirin irreversibly acetylates the COX present in platelets, which inhibits the formation of thromboxane A2, which is responsible for vasoconstriction and platelet aggregation.
Because platelets do not contain a nucleus, they are incapable of synthesizing additional COX; therefore platelet thromboxane production will be inhibited for the life of any circulating platelets.

Antiplatelet therapy may be beneficial in the management of equine laminitis, disseminated intravascular coagulation, and equine verminous arteritis. A precise
antiplatelet dose has not been established, but a dose of 12 mg/kg prolongs bleeding time for 48 hours.

59
Q

Aspirin

  • pH
  • absorption
  • metabolism and elimination
  • distribution
A

sodium salicylate = weak acid, it is best absorbed in the acidic environment of the upper GI tract, although overall bioavailability is low via the oral route (6%). Absorption is higher when given via the rectal route (17%). (available only in oral forms)

During absorption aspirin is partially hydrolyzed to salicylic acid and distributed throughout the body.

The highest concentrations are attained in the liver, heart, lungs, renal cortex, and plasma.

Aspirin is hepatically metabolized by glycine and glucuronide conjugation. Salicylates and their metabolites are rapidly excreted in urine by way of glomerular filtration and active tubular excretion,

60
Q

Advantages and disadvantages of carprofen

A

+ longer elimination half-life in horses than most other NSAIDs, at 21 hours for the R enantiomer and 17 hours for the S enantiomer following IV administration.

  • produces only modest reductions in the concentrations of ecosanoids.

+ other beneficial antiinflammatory effects, including inhibition of matrix metalloproteinases in cartilage explants and inhibition of the activation of NfκB-dependent genes such as inducible nitric oxide synthase

61
Q
A
62
Q

Diclofenac

A

single topical application of diclofenac cream produced measurable concentrations
of diclofenac in transudate within 6 hours and significantly attenuated carrageenan-induced local production of PGE2.

almost no systemic effect, can be used together with systemic NSAIDs

63
Q

Firocoxib

  • onset of action?
  • dose?
A

Firocoxib = NSAID in the coxib class, highly COX-2 selective

The onset of antiinflammatory action is within 2 hours, peak response occurs between 12 and 16 hours, and duration of action is 36 hours. Analgesic effects
have a more rapid onset and shorter duration.

0.3mg/kg loading dose, afterwards 0.1mg/kg, available on IV and PO

64
Q

Flunixin

  • bioavailability
  • plasma elimination halflife, inflammatory exsudate half life
  • protein binding?
  • onset of action
  • elimination
A
  • rapidly absorbed after oral administration, with a bioavailability of 86%. Absorption is delayed by feeding.
  • Plasma elimination half-life is 1 to 2 hours. (13 in foals!!!), elimination half-life in inflammatory exudate is 16 hours
  • highly protein bound (86%) but high volume of distribution.
  • onset of antiinflammatory action within 2 hours, peak response occurs between 12 and 16 hours, and duration of action is 36 hours. Analgesic effects have a more rapid onset and shorter duration.
  • Only 14% of a dose is excreted in urine, but otherwise little is known about the metabolism of flunixin.
65
Q

Efficiacy of NSAIDs for prevention of endotoxemia

A

Flunixin is more effective than phenylbutazone in preventing the clinical signs of endotoxemia but appears equivalent to ketoprofen

66
Q

Effect of flunixin with ischemic GI disease

A

Flunixin treatment of horses with ischemic GI disease may cause prolonged permeability defects in recovering mucosa

67
Q

Advantages of Ketoprofen against other NSAIDs

A

+ ketoprofen is clinically similar to flunixin meglumine in efficacy.

- In an experimentally induced synovitis model, phenylbutazone was more effective in reducing lameness and synovial fluid prostaglandin concentrations.

+ In horses with chronic laminitis, ketoprofen was more effective than phenylbutazone
at relieving pain but only at a higher-than-label dose
(3.63 mg/kg).

+ In comparative toxicity studies in both horses and donkeys, ketoprofen at the label dose had less potential for toxicity than flunixin meglumine or phenylbutazone

68
Q

Selectivity of meloxicam?

Oral Bioavailability?

Comparison to other NSAIDs?

A

slightly COX-2 selective in the horse with an IC50 COX-1: IC50 COX-2 of 3.8.

Oral bioavailability is nearly complete, and absorption is not affected by feeding

Direct comparison of flunixin and meloxicam in postoperative cases of small intestinal strangulating obstructions showed that meloxicam was as effective as flunixin when comparing most major clinical variables, a_lthough more horses in the
meloxicam group showed gross signs of pain_. Additionally, meloxicam treatment did not impair mucosal recovery after ischemic injury in a small intestinal strangulation model.

suitable for pain after orthopedic surgery

69
Q

Clinical use of piroxicam?

A

NSAID of the oxicam group

used for treatment of squamous cell carcinoma (SCC). Some tumors, particularly SCC, have been shown to produce COX-2. Treatment with piroxicam has produced long-term remissions of SCC in the bladder, urethra, and periocular structures.

Adverse effects, even at the low doses used (0.2 mg/kg PO q24hr), have been noted, and include diarrhea and abdominal pain. It is not known at this time whether or not meloxicam would have a similar effect, with a higher safety profile.

70
Q

Plasma protein binding of PBZ?

Metabolism and elimination?

Mechanism of toxicity during overdosing?

Therapeutic efficacy for how long?

PBZ and reproductive system?

A

Plasma protein binding in horses is greater than 99%.

metabolized in the liver to oxyphenbutazone, an active metabolite that is eliminated more slowly from the body than phenylbutazone. Oxyphenbutazone inhibits the metabolism of phenylbutazone => capacity of the liver to metabolize phenylbutazone becomes overwhelmed at relatively low drug doses, resulting in dosedependent kinetics. The elimination half-life increases with increasing dose rates and increasing age. The elimination half-life from exudate is 24 hours.

Therapeutic efficacy lasts for more than 24 hours as a result of the irreversible binding of phenylbutazone to COX, slow elimination from inflamed tissues, and long elimination half-life of oxyphenbutazone. Therefore high or frequent doses of phenylbutazone result in disproportionately increasing plasma concentrations, which easily result in toxicity.

Phenylbutazone and its metabolite cross the placenta and are excreted in milk.
Less than 2% is excreted in the urine as unchanged drug.

71
Q

Azepromazin: mechanism of action and dose

A

Alpha adrenergic antagonist

0.04-0.07mg/kg