The sulfonamides are the oldest group of antibiotics used therapeutically. All sulfonamides that are currently used were derived from the first clinically relevant sulfonamide, sulfanilamide, itself a derivative of the azo dye prontosil. The discovery of its efficacy in vivo but not in vitro indicated that metabolism by the host was necessary for efficacy and contributed to the understanding of the role of drug metabolism in bioactivation. Once the metabolite was identified as the active drug, a number of manufacturers produced hundreds of different sulfanomide antimicrobial preparations. The FDA had not yet been empowered by Congress to evaluate drug safety, resulting in the lack of safety limitations. Among the vehicles in which drugs were prepared was a product containing ethylene glycol. The subsequent death of more than 100 persons, including children, ingesting the product contributed to congressional approval of the Food Drug and Cosmetic Act of 1938. It was this act that empowered the FDA to evaluate drugs for safety before marketing.
The sulfonamides were the first group of commercially available antimicrobials used systemically.246 Their use was somewhat curtailed by the advent of the penicillins, only to increase again in the 1970s with their combination with the diaminopyridine trimethoprim. Not surprisingly, long-term use of these drugs has contributed to the development of resistance that has limited their clinical use.246 However, a decline in their use, in part because of concerns regarding drug allergies, probably has contributed to a decline in resistance. Sulfonamides generally are used in combination with diaminopyrimidines for treatment of bacterial infections, with use of sulfonamides as sole agents generally limited to treatment of coccidiosis.
As derivatives of sulfanilamide, all sulfonamides have the same nucleus. Functional groups have been added to produce compounds with varying physical, chemical, pharmacologic, and antibacterial properties, but the active amine is in position 4 and any substitutions at this position must be freed in vivo (Figure 7-13). Although amphoteric, sulfonamides generally behave as weak organic acids and are much more soluble in an alkaline than in an acidic environment. Those of therapeutic interest have pKa values between 4.8 and 8.6. Water-soluble sodium or disodium salts are used for parenteral administration. Such solutions are highly alkaline, somewhat unstable, and readily precipitate out with the addition of polyionic electrolytes. In a mixture of sulfonamides (e.g., the sulfapyrimidine group), each component drug exhibits its own solubility; therefore, a combination of sulfonamides is more water soluble than a single drug at the same total concentration. This is the basis of triple sulfonamide mixtures used clinically (primarily in large animals). The N-4 acetylated sulfonamides, except for the sulfapyrimidine group (sulfadiazine), are less water soluble than their nonacetylated forms. Highly insoluble sulfonamides are retained in the lumen of the gastrointestinal tract for prolonged periods and are known as “gut-active” sulfonamides. Most sulfonamides used clinically for treatment of bacterial infections are “potentiated.” The “potentiator” of sulfonamides is a diaminopyrimadine; examples include trimethoprim, ormetoprim, and pyrimethamine (the latter being the preferred drug for toxoplasmosis) (see Figure 7-13).
Figure 7-13 `The mechanism of action of the sulfonamides and the diaminopyrimidines. By itself, either type of drug is bacteriostatic, but the two-point sequential inhibition of folic acid synthesis results in bactericidal effects. The progenitor of the sulfonamides is sulfanilamide (inset). As such, all are arylamines. Metabolism of the arylamine to a hydroxyalamine and nitroso compound contributes to the toxicities, including drug allergies, associated with sulfonamides.
Folic acid is an essential bacterial substrate necessary for protein and nucleic acid metabolism. Bacterial synthesis of folic acid is accomplished in several sequential steps (see Figure 7-13). The sulfonamides are structurally similar to PABA and act as competitive substrates (antimetabolites) for the synthetase enzyme. Among the many sulfonamides used clinically are sulfadiazine, sulfamethoxazole, sulfachlorpyridazine, sulfadimethoxine, and sulfasalazine; sulfisoxazole is the model drug upon which C&S testing is based.
Because folate metabolism is required for many cellular functions, bacterial growth is inhibited; consequently, the antibacterial effects of sulfonamides as sole agents are bacteriostatic. The diaminopyrimidines trimethoprim and ormetoprim also impair folic acid synthesis but at a different point in the metabolic pathway. They prevent the conversion of dihydrofolate to tetrahydrofolate by inhibiting the reductase enzyme. By themselves, these drugs also are bacteriostatic. It is the combination of a sulfonamide antimicrobial with a diaminopyrimidine antimicrobial (“potentiated”) that results in subsequent two-point inhibition of bacterial folic acid synthesis and thus bactericidal rather than bacteriostatic activity (see Figure 7-13). 5,246 Mammalian cells are not affected by these drugs because they are dependent on dietary sources of folic acid; in contrast, microbes cannot use external sources of the substrates. Further, the affinity of bacterial enzymes for the drugs is much higher than the mammalian enzymes. The competitive nature of the mechanism of killing activity of potentiated sulfonamides leads to a time-dependent effect. High inoculums may require higher doses for efficacy.
The spectrum of activity of sulfonamides is considered broad, but efficacy is variable because of acquired resistance. However, a decline in their use during the last decades (due to concerns regarding allergies) appears to be associated with an increased in susceptibility for a number of organisms. The spectrum of combined products includes gram-positive, gram-negative, and anaerobic organisms. The sulfonamides exhibit good to moderate activity against E. coli; Enterobacter spp.; Klebsiella spp.; Proteus spp.; Pasteurella spp.; and anaerobic organisms such as Actinomyces, Bacteroides, Fusobacterium spp., and selected clostridia.247-249 The spectrum of these drugs does not include Serratia spp., P. aeruginosa, Rickettsia, or Mycoplasma spp. The sulfonamides exhibit good efficacy against Brucella spp., Actinomyces spp., and selected protozoal organisms such as Pneumocystis carinii and Cryptosporidium spp. Some Chlamydia spp. are susceptible to sulfonamides, whereas others are not. The difference appears to be based on whether the organism can obtain folic acid from the host.249a Mycoplasma organisms are not susceptible to sulfonamides. By itself, trimethoprim has a potency that is twentyfold to 200-fold less than that of sulfonamides.246 Potentiated sulfonamides are generally useful for uncomplicated infections of many body systems.
Inherent resistance to sulfonamides reflects, in part, the ability of the microbe to make use of host folic acid. Resistance to the sulfonamides and to trimethoprim or ormetoprim occurs relatively rapidly. Chromosomal resistance results in impaired drug penetration, reduced affinity of the enzyme for the substrate, or increased bacterial production of PABA. Plasmid-mediated resistance occurs rapidly because of altered drug penetration and decreased affinity of the enzyme for the substrate. Resistance to one sulfonamide generally results in resistance to all sulfonamides.246 The increasing emergence of resistance has sharply curtailed the use of these drugs. The role of trimethoprim/sulfonamide combinations for the critically ill patient or for chronic infections should be based on C&S information because of the incidence of resistance.
The sulfonamides are generally rapidly and completely absorbed after oral administration, although there are exceptions (see the discussion of structure and chemistry). Trimethoprim and ormetoprim are well absorbed after oral administration. Sulfasalazine is poorly absorbed as an intact molecule and is used primarily for gastrointestinal diseases. After oral administration sulfasalazine is partially absorbed in the small intestine. It undergoes enterohepatic circulation and ultimately is eliminated in the urine. Most of the drug (70%) is metabolized by colonic bacteria to its component parts: sulfapyridine and 5-aminosalicylic acid. Sulfapyridine is rapidly absorbed and subsequently eliminated in urine. The 5-aminosalicylic acid may provide the major therapeutic benefit for chronic inflammatory bowel disease.246
Solutions intended for parenteral administration must be buffered to prevent pain and irritation caused by the alkalinity of the compounds. Topical administration is not recommended because of the effects of these drugs on wound healing. An exception is made for silver sulfadiazine and mafenide, which are used primarily for burn patients in human.246 Sulfadiazine is combined with silver in a topical otic preparation approved for use in dogs. Protein binding of the sulfonamides varies from 15% to 99%. Examples include sulfadiazine at 30% to 50% bound, sulfadiamethoxine, at greater than 75%, and sulfasalazine up to 99% bound. Protein binding contributes to a relatively long half-life, allowing for convenient dosing intervals.
KEY POINT 7-32
The disposition of the sulfonamides, which are time dependent, is markedly variable among members of this drug class.
The tissue penetrability of the sulfonamides varies. All are distributed at least to extracellular fluid. Sulfamethoxazole (the model drug for susceptibility testing) is limited to interstitial fluid, whereas sulfadiazine is distributed to total body water.246 Sulfadiazine penetrates most body tissues extremely well, including the prostate.250 The penetration of these drugs varies with the sulfonamide component. Prostatic penetration is facilitated by a high pKa. Sulfadiazine (pKa 6.4) is among the best distributed sulfonamides but only achieved 11% of serum concentration in the prostate of dogs in one study (the original reference for this study could not be found). Drugs with a more basic pKa may appear to better penetrate the prostate, although this may reflect ion trapping in prostatic fluids. Sulfadiazine can attain therapeutic concentrations in CSF, particularly if given intravenously, and is the preferred sulfonamide for CNS infections.246 Trimethoprim achieves tissue concentrations four times higher than that in plasma. The combination of a sulfonamide with a diaminopyramidine at a ratio of 1:5 trimethoprim/sulfonamide results in a bactericidal effect and a tissue distribution ratio of 1:20 in most tissues.5 This ratio, however, is described in humans, and information in dogs or for sulfadimethoxine and ormetoprim does not appear to be available.
Sulfonamides that undergo hepatic metabolism are generally acetylated. All sulfonamide antimicrobials are arylamines. The dog lacks some genes that encode for N-acetyltransferases responsible for metabolism of arylamines.251 Thus metabolism in the dog may involve other pathways, facilitating the formation of potentially nitroso metabolites that are responsible for allergic or other idiosyncratic reactions (see the section on adverse reactions) (see Figure 7-13).252 Drugs are renally excreted as either the parent compound or the conjugated metabolite by either glomerular filtration or active tubular secretion. Both passive reabsorption and enterohepatic circulation can prolong the elimination half-life of selected sulfonamides.246 Acetylated metabolites of sulfonamides are often less soluble than the parent compounds, which increases the risk of renal damage should drug precipitate and form crystals. However, this is unlikely in dogs because of deficient acetylation. The risk is reduced in other species because of the use of combination products, which reduces the total amount of dose needed for efficacy. The elimination half-lives of the drugs vary with the sulfonamide component and among the species. The duration at which sulfonamides remain in the body leads to classification as short-acting (12 hours or less: sulfacetamide, sulfathiazole, and sulfisoxazole), intermediate-acting (12 to 24 hours: sulfadimethoxine, sulfisoxazole, sulfamethoxazole, sulfapyridine, sulfamethazine, and sulfadiazine), and long-acting (longer than 24 hours).246 In the dog, according to the package insert, sulfadimethoxine concentrations are 39 μg/mL 24 hours after dosing. Peak ormetoprim at 2 hours was 1.09 μg/mL in dogs but was 0.09 μg/mL at 24 hours, indicating a half-life of about 6 hours. It is not clear whether the differences in half-life between sulfadimethoxine and ormetoprim “match” in terms of ideal proportion throughout the labeled 24-hour dosing interval.
Reactions to sulfonamide antimicrobials reflect the greatest proportion of antimicrobial adversities in the dog.252 The adversities to sulfonamide antimicrobials but not other sulfonamides (e.g., nonsteroidal antiinflammatories, zonisamide, furosemide) probably reflect the basic structure of the sulfanilamide molecule, which is an arylamine, in which the amine group is directly attached to the benzene ring (see Figure 7-13). The susceptibility of dogs to sulfonamide toxicity may reflect the species’ deficiency in acetylation and specifically N-acetylation. The proposed mechanism of toxicity reflects shunting of the sulfanilamide arylamine to an oxidative phase I pathway (see Figure 7-13). Oxidation of the arylamines yields hydroxylamine, a metabolite that can be cytotoxic at high concentrations; the metabolite also is somewhat allergenic. Hydroxylamine can be further metabolized (often spontaneously) to a nitroso compound, which is somewhat cytotoxic but is more immunogenic than the hydroxyarylamine. The potential role of the arylamine as a cause of sulfonamide toxicity is supported by the lack of apparent toxicity by other sulfonamide drugs used in dogs, which, lacking a primary arylamine, are not converted to hydroxylamine. The likelihood of adversity may be related to the type of metabolites formed and the rate of acetylation. As such, the likelihood of toxicity occurring may vary among the sulfonamide antimicrobials. The mechanism of hypersensitivity may reflect haptenization of the metabolite and a subsequent T-cell response, although other mechanisms (e.g., humoral response or cytotoxicity) may contribute.252 Deficiencies in glutathione, ascorbic acid, or other radical scavengers may increase the risk of either type A or B reactions; the role of supplementation in preventing or treating adversities apparently has not been addressed scientifically but may be prudent. Controversy exists as to whether the parent sulfonamide might be immunogenic.252 The “potentiator” may also be responsible for some reactions; for example, trimethoprim has been associated with skin eruptions or hepatopathy in humans; further, use of sulfadiazine as the sole coccidiostat in dogs has not been associated with drug allergies.
KEY POINT 7-33
Adversity to the sulfonamides probably reflects their metabolism to toxic or allergenic metabolites.
With the exception of thyroid-gland suppression, sulfonamides, and sulfadiazine in particular, appear to be free of type A or I adverse drug reactions at doses higher than those used therapeutically. For example, suppression of the thyroid gland was the only adverse effect evident in dogs treated with sulfadiazine at 300 mg/kg a day for 20 days. Any sulfonamide, including antimicrobial drugs, may profoundly alter thyroid physiology at high doses (25 mg/kg twice daily). The sulfonamide is a reversible substrate inhibitor of thyroid peroxidase, preventing the iodination and coupling of tyrosine residues necessary for formation of thryoxine and thyronine.253 Whereas labled doses of sulfadiazine and trimethoprim do not appear to cause thyroid suppression at least for 4 weeks, clinical hypothyroidism has occurred in one dog treated with trimethoprim sulfadiazine at 48 mg/kg/day for 10 weeks. Experimentally induced suppression of thyroid hormone (T4) synthesis occurred in 57% of dogs treated for pyoderma at 60 mg/kg/day for 6 weeks. Decreased thyroid hormone synthesis generally will be clinically relevant by 3 weeks of therapy but may take 6 to 8 weeks or longer and will return to normal within 3 weeks after therapy is discontinued.
Aplastic anemia has been reported in dogs receiving 30 to 60 mg/kg of sulfadiazine a day,252 although the role of allergy versus folic acid deficiency was not documented. Because mammalian cells can use dietary folic acid, supplementation might be considered, particularly for patients that develop anemia (normocytic rather than megaloblastic)252 consistent with folic acid deficiency while receiving a sulfonamide. Cats appear to be more sensitive to the effects of trimethoprim/sulfonamide combinations. Doses of 300 mg/kg per day for 10 to 30 days orally resulted in lethargy, anorexia, anemia, leukopenia, and increased blood urea nitrogen. Before the advent of triple and potentiated sulfonamide preparations, crystalluria was a common type I side effect, with subsequent renal damage. Nonetheless, with high doses of any sulfonamide product, prudence dictates that the hydration status of the animal be normal, particulary if urinary pH is acidic.
Although the sulfonamides are generally safe drugs, the advent of hypersensitivity drug reactions (immunologic) has limited their use. Immune-mediated diseases of the skin, kidney, liver, and eye are not dose dependent.
Sulfonamide antimicrobial toxicity in animals has been reviewed.252,254 The incidence of systemic sulfonamide toxicity in dogs has been reported as 0.25%. In a study of dogs (n = 40), inclusion criteria included clinical signs consistent with a drug allergy and treatment with a sulfonamide antimicrobial for at least 5 days.254 The breeds most often represented were Golden Retrievers, Miniature Schnauzers, German Shepherd Dogs, Labrador Retrievers, and Samoyeds, with Miniature Schnauzers and Samoyeds being overrepresented. The lack of representation by Doberman Pinschers was suggested to reflect decreased treatment of this breed with sulfonamides. Ages ranged from 6 months to 14 years (mean 5.7 ± 3.2), and neutered female dogs were overrepresented (60%). Three sulfonamides were represented, with 64% of afflicted dogs receiving sulfamethoxazole, 23% sulfadimethoxine, and 13% sulfadiazine; either trimethoprim or ormetoprim also were administered. No information was available regarding the proportion of sulfonamides prescribed to dogs. The frequency of each drug being administered was not determined. Doses ranged from 23 to 81 mg/kg/day, and time of onset ranged from 5 to 36 (mean 12) days. The most common clinical signs and the proportion of animals afflicted were fever (55%), thrombocytopenia (54%), hepatopathy (28%), neutropenia (27%), keratitis sicca (25%), and hemolysis (22%). Facial palsy was an unusual clinical sign. Other clinical signs included arthropathy, uveitis, skin and mucosal lesions, proteinuria, facial palsy, hypothyroidism, pancreatitis, facial edema, and pneumonitis. Dogs with hepatopathy or thrombocytopenia had a significantly lower recovery rates.254 Dogs with hepatopathy also tended to have received the highest doses, suggesting that a toxic metabolite might be responsible and the adversity might be, in part, type A rather than type B (i.e., dose dependent and thus predictable). The fact that some animals developed adversities in as little as 5 days might also support a type A or idiosyncratic type B reaction, rather than allergy. Large breeds, with Doberman Pinschers overrepresented, appear to be at greater risk for developing arthropathy (as reviewed by Trepanier).252
Keratoconjunctivitis sicca is a more common side effect of sulfonamides in dogs, occurring in as many as 15% of animals receiving sulfonamides.254 It has been reported in dogs after treatment with sulfasalazine, sulfadiazine, and sulfamethoxazole. The reaction may reflect direct cytotoxicy to the lacrimal gland rather than an allergic reaction, but nonetheless, time of onset may be months to years after therapy is initiated. Female dogs may be at greater risk. Resolution of clinical signs is more likely if the inciting drug is discontinued early; otherwise, normal function may not recur once the drug is discontinued. Prognosis is more favorable for younger dogs receiving the drug for a short period of time.
The sulfonamides have been associated with a number of drug interactions in humans.2 Inhibition of elimination with subsequent prolonged or increased effects have been reported for oral hypoglycemic agents, dapsone when combined with trimethoprim, folate antagonists (increased risk of megaloblastic anemia), methanamine (increased risk of crystalluria), procainamide (decreased metabolism when combined with trimethoprim), and warfarin (increased anticoagulant activity with trimethoprim). In contrast, increased elimination has been reported for cyclosporine when combined with either sulfonamides or trimethoprim.2
Because of the advent of resistance, the use of sulfonamides is limited to uncomplicated infections of most body systems. The concentration in urine supports the use of potentiated sulfonamides for urinary tract infections. Trimethoprim/sulfonamide combinations are indicated for treatment of infections caused by susceptible bacteria in difficult-to-penetrate tissues such as the prostate and CNS.246 These drugs are among the drugs of choice for treating Nocardia and Actinomyces spp. Synergistic effects have been cited toward these organisms when used in combination with beta-lactam antibiotics.
Tetracyclines historically have been widely used, but development of resistance has largely curtailed empirical use in the last decade. However, the decline in use appears to have led to a decrease in resistance, and susceptibility increasingly is demonstrated through C&S data, potentially leading once again to more common use of these drugs.
Three naturally occurring tetracyclines are obtained from Streptomyces: chlortetracycline (the prototypic drug but no longer available in human-medicine preparations), oxytetracycline, and demethylchlortetracycline (see Figure 7-5). Several tetracyclines have been derived semisynthetically (tetracycline from chlortetracycline, rolitetracycline, methacycline, minocycline, doxycycline, lymecycline, and others). Elimination half-lives permit a further classification into short-acting (tetracycline, oxytetracycline, chlortetracycline), intermediate-acting (demethylchlortetracycline and methacycline), and long-acting (doxycycline and minocycline) formulations. All of the tetracycline derivatives are crystalline, yellowish, amphoteric substances that, in aqueous solution, form salts with both acids and bases. They characteristically fluoresce when exposed to ultraviolet light. The most common salt form is the hydrochloride, except for doxycycline, which also is available as doxycycline hyclate. The tetracyclines are stable as dry powders but not in aqueous solution, particularly at higher pH ranges (7-8.5). Preparations for parenteral administration must be carefully formulated, often in propylene glycol or polyvinyl pyrrolidone with additional dispersing agents, to provide stable solutions. Tetracyclines form poorly soluble chelates with bivalent and trivalent cations, particularly calcium, magnesium, aluminum, and iron. Doxycycline and minocycline exhibit the greatest liposolubility and better penetration of bacteria.
Tetracyclines bind bacterial ribosomes and impair protein synthesis (see Figure 7-6). Bacterial ribosomal activity was described in the section on aminoglycosides. The tetracyclines bind to the 16S portion of the 30S ribosomal subunits, preventing access of the amino-acyl tRNA to the acceptor site on the mRNA ribosome complex80 (see Figure 7-6). Because tRNA binding is prevented, amino acids cannot be added to the peptide chain, and protein synthesis is impaired. Tetracyclines are bacteriostatic in action and should not be used in the immunocompromised patient, whether disease or drug induced (i.e., glucocorticoids or anticancer drugs). Their effects are described with other bacteriostatic ribosomal inhibitors as time dependent but are probably related to AUC. The tetracyclines also inhibit matrix metalloproteinases, an action separate from their antibacterial properties.
Tetracyclines enter cells either through porins or active transport pumps.80 They are considered broad spectrum (see Table 7-2), being effective against gram-positive, gram–negative, anaerobic organisms, as well as cell wall–deficient and rickettsial organisms and others. Their spectrum includes gram-negative organisms, particularly Pasteurella spp., and often E. coli, Klebsiella, and Salmonella spp. P. aeruginosa is generally not included; although susceptibility may be indicated on C&S data, caution should be exercised when selecting tetracyclines. They generally are intrinsically more effective against gram-positive organisms (see Tables 7-3 and 7-4). As such, Staphylococcus and Streptococcus spp. generally are included in the spectrum. However, the broad general use of these drugs has led to resistance by many organisms and use against gram-positive organisms should be based on C&S testing. The spectrum of action also includes Chlamydia, Mycoplasma, Rickettsia, and Hemobartonella organisms. Spirochetes (Borrelia, Leptospirosis spp. also are generally susceptible, and several mycobacterial organisms are susceptible. Tetracyclines target Brucella spp.) although in human medicine generally they are combined with rifampin or gentamicin. Tetracyclines generally are effective toward actinomycosis and are generally considered more effective than chloramphenicol.80
Tigecycline is a glycylcycline, a class of drugs that are synthetic analogs of the tetracyclines. Specifically, it is a glycolamide derivative of minocycline. The spectrum of this class is similar to that of the tetracyclines; however, they often remain effective against strains that have developed resistance to tetracyclines through increased efflux transport mechanisms.80
Resistance to tetracyclines is plasmid mediated and inducible.80 Most resistance to tetracyclines results from either decreased influx or increased transport of the drug out of the microbial cell. Other mechanisms include altered binding site (which may reflect a mutation) and enzymatic destruction. Cross-resistance does not necessarily occur and depends on the mechanism. Drugs that minimize the impact of efflux pumps have been developed, including the glycylcyclines.255 These drugs also have a higher binding affinity than tetracyclines.
The oral absorption of tetracyclines is variable, with chlortetracycline being the least bioavailable, oxytetracycline more so, and doxycycline the most lipid soluble of the tetracyclines, being 100% bioavailable. Absorption is decreased in the presence of divalent and trivalent cations such as those present in milk products or antacids; exceptions occur for doxycycline and minocycline. Tetracyclines, particularly doxycycline, are widely distributed to most body tissues, and theoretically, inflammation need not be present for distribution into the brain80 (see Table 7-5). Drugs will distribute through the placenta into the fetus and into milk. Doxycycline is able to penetrate cell membranes and thus gain access to intracellular organisms. Doxycycline is 99% protein bound, which prolongs its elimination half-life; note that concentrations in body fluids (see Table 7-5) are likely to reflect unbound drug, whereas that in plasma may reflect bound drug, decreasing ratios. Tetracyclines, with the exception of lipophilic tetracyclines such as minocycline and doxycycline, do not penetrate the CSF. The latter drugs are thus preferred because of better tissue penetrability for treatment of infections caused by susceptible bacteria in difficult-to-penetrate tissues, reaching 30% to 40% of plasma concentrations. Minocycline is characterized by a larger Vd in people than is doxycycline, suggesting the potential of better tissue penetrability, but may also be more bound to bone or other tissues containing cations. Tetracyclines accumulate in reticuloendothelial cells.80 Tetracyclines are incorporated into forming bone and the enamel and dentin of teeth and cause discoloration of teeth upon eruption. The age at which this occurs in dogs and cats is not clear.
KEY POINT 7-34
Among the tetracyclines, doxycycline and minoclycline stand out for their lipid solubility and biliary excretion.
Doxycycline (PC 0.68 and pKa 3.09)57 was studied in the dog in both plasma and interstitial fluid (using ultrafiltration) after intravenous and constant-rate influsion (to allow establishment of steady-state concentrations). The drug is 91% bound to plasma proteins in dogs, resulting in a total AUC difference sixfold higher in plasma compared with interstitial fluid. Further, the interstitial fluid Cmax (of unbound drug) was only 0.14 μg/mL at steady-state conditions; in contrast, PDCs extrapolated from the terminal component of the elimination curve was 1.6 μg/mL. The concentration of interstitial fluid drug was equivalent to the concentration of unbound drug in plasma.57 Vd of unbound drug was 0.65 ± 0.08 L/kg; clearance was 1.66 ± 2.21 mL∗kg/min.
With the exception of doxycycline and minocycline, the tetracyclines are eliminated by both renal (approximately 60%) and biliary (40%) excretion. Presumably, minocycline is eliminated essentially in the bile, whereas the route of elimination of doxycycline is less obvious. In humans it is eliminated by both renal (41%) and biliary (59%) mechanisms. In dogs intestinal elimination of the unchanged drug appears to be the predominant route, with only about 16% of a given dose being excreted unchanged in the urine. Tetracyclines undergo enterohepatic circulation. Toxic concentrations may accumulate in patients with renal disease. Differences that justify use of minocycline instead of doxycycline are difficult to ascertain. Adverse reactions to minocycline may, however, be more likely.
The tetracyclines are available as intravenous, parenteral, and ocular preparations. Tetracyclines should not be given intramuscularly because of local tissue damage and irritation. For the same reason, tetracyclines are not indicated for topical treatment other than the eye.
Tetracyclines cause several adverse effects in small animals. Toxicity may be worsened in patients with renal disease because of decreased elimination. Gastrointestinal upset follows direct irritation of the gastrointestinal mucosa after oral administration; administration of doxycycline with food will reduce gastrointestinal side effects. Rarely, hepatotoxicity may occur. Rapid intravenous administration may result in collapse. Although the likelihood of this occurring in small animals is not clear, prudence dictates slow administration of a diluted solution (i.e., 1:10) when tetracyclines are administered intravenously. Although the mechanism is not certain, calcium binding may be important. Intravenous administration of tetracycline has caused anaphylactic shock in dogs. Diluting fluids should not contain calcium or other cations to which tetracylines might chelate. Hypersensitivity has also been reported in a dog after intramuscular administration of tetracycline. Minocycline may be more likely to cause allergic drug reactions in drugs. Lesions characterized by erythema of the skin and mucous membranes occurred in dogs after administration of most doses of minocycline. Anemia may also occur (10 mg/kg, administered intravenously). Brown to gray discoloration of teeth may occur because of chelation of tetracyclines in calcium deposits of dentin and, to a lesser degree, enamel. Tetracycline and oxytetracycline cause a yellow discoloration, whereas chlortetracycline produces a gray-brown discoloration; of all the tetracyclines, oxytetracycline causes the least tooth discoloration. Because chelation might occur in forming dentin as well as enamel, tetracyclines should be avoided from 3 weeks’ gestation to at least 1 month after birth. Among the lipid-soluble tetracyclines, doxycycline may be less likely to cause discoloration. In humans minocycline may stain teeth regardless of tooth development because of chelation with iron; the drug probably has not been used sufficiently in animals to determine whether a similar effect will occur. Other side effects caused by tetracyclines include drug fever (in cats), an antianabolic effect, and a Fanconi-like syndrome in the kidneys, with the latter more likely with expired or degraded tetracyclines.255
Doxycycline has been associated with esophageal erosions in cats (and humans).80 In a study of 30 cats, no orally administered tablets had passed in 30 seconds; only 40% had entered in 5 minutes. In contrast, 90% of tablets passed within 30 seconds when followed with 6 mL of water, with 100% passage at 90 seconds. For capsules only 17% had passed by 30 seconds, but 93% had passed by 60 seconds.256 The impact of esophageal damage is not unique to doxycline; other drugs are ulcerogenic because of local effects. Indeed, the cat has been used as a model to assess the ulcerogenic potential of orally administered drugs.257 For doxycycline, the risk may be decreased with use of the monohydrate salt. In the event that erosions do occur, among the treatments to consider would be pentoxifylline.258
Because of chelation with cations (magnesium, calcium, aluminum, and so on), tetracyclines should not be simultaneously administered with cation-containing drugs (e.g., antacids, sucralfate, buffered aspirin, calcium-containing supplements, fluids). Cholestyramine may also bind to tetracyclines. Tetracyclines, with the exception of doxycycline, should not be administered with food.
Because tetracyclines bind to the 30s ribosomal subunit, combination with antimicrobials that target the 50s subunit might be considered (e.g., the phenicols, macrolides, and lincosamides) with scientific support. One study indicates an in vitro synergistic effect of the combined use of doxycycline and azithtromycin against P. aeruginosa.259
The therapeutic indications for tetracyclines are many but have decreased in recent years because of the advent of resistance. Treatment of microbial infections is best based on C&S data. Doxycycline is the preferred tetracycline because of its ability to move intracellularly compared to other tetracyclines. Doxycycline generally is indicated among first-choice therapies for obligate intracellular organisms, including ehrlichiosis, Rocky Mountain spotted fever, chlamydiosis, mycoplasmosis, and hemobartenellosis. Doxycycline also has been used to treat canine brucellosis. Other potential indications include leptospirosis and Lyme disease.
Chloramphenicol has been widely used in the past, but the development of resistance and human toxicity to chloramphenicol have severely curtailed its use and commercial availability. Florfenicol is a commercially available thiamphenicol derivative approved for treatment of bovine respiratory diseases complex. A sulfonyl group replaces the aromatic ring nitro group that is otherwise associated with chloramphenicol’s irreversible bone marrow suppression in humans (see Figure 7-5). As chloramphenicol increasingly is difficult to obtain commercially, florfenicol may find a niche for use in small animals, particularly cats, in which the disposition is more predictable than with dogs.
Like tetracyclines, chloramphenicol and florfenicol bind bacterial ribosomes and impair protein synthesis (see Figure 7-6). However, binding occurs at the 50s subunit with inhibition of peptidyl transferase. Actions are bacteriostatic in action, and these drugs should not be used in immunocompromised patients. As with other bacteriostatic ribosomal inhibitors, the effects of chloramphenicol and florfenicol should be considered time dependent. As with tetracyclines, although host ribosomes do not bind as effectively as do bacterial ribosomes, some host ribosomal activity will be impaired. Binding sites for chloramphenicol are close to those for clindamycin, which it competitively inhibits.80 Chloramphenicol also inhibits mitochondrial protein synthesis in mammalian cells, with erythropoietic cells particularly sensitive.
Chloramphenicol is considered broad spectrum (see Table 7-1), being effective against gram-positive, gram-negative, and anaerobic organisms. P. aeruginosa is generally not included. The spectrum of action also includes Chlamydia, Mycoplasma, Rickettsia, and Hemobartonella organisms. As previously noted, tetracyclines are considered more effective than chloramphenicol for the latter organisms, but chloramphenicol tends to be more clinically effective for other organisms. The spectrum of activity of florfenicol is similar to chloramphenicol; although the anaerobic spectrum has not been described, it is assumed to be similar. The MIC for florfenicol of small animals generally reflects 1.0 to 8.0 μg/mL.
Resistance to chloramphenicol is caused by destruction (acetylation) of the drug by microbial enzymes. The fluorine ring of florfenicol may impair bacterial acetylation, and thus florfenicol is more resistant to bacterial deactivation;260 selected organisms resistant to chloramphenicol may be susceptible to florfenicol.2
Chloramphenicol is very well absorbed after oral administration in its crystalline form. Many of the originally-approved preparations are no longer available in the United States. The liquid form is less well absorbed, so much so that the palmitate form should not be used for cats because of variability in oral absorption. The chloramphenicol succinate ester is the water-soluble form intended for injection (see Table 7-1). The succinate must be hydrolyzed by plasma, hepatic, pulmonary, or renal esterases before activity. Chloramphenicol palmitate is a suspension for oral administration. Its ester is hydrolyzed by small intestinal lipases; the freed chloramphenicol is then orally absorbed. The freed chloramphenicol is among the most lipid soluble of the clinically used drugs and achieves moderate to high concentrations in most body tissues, including the CSF. It is, however, unlikely to achieve bactericidal concentrations in most tissues, including the CNS. Most of the drug is eliminated by hepatic metabolism. Glucuronidation is a major route of elimination of chloramphenicol. Cats eliminate chloramphenicol more slowly because of deficiencies in both phase I and phase II metabolism. Greater concentrations may occur in cat urine than in dog urine as a result.261 Pediatric patients also may not eliminate chloramphenicol as efficiently as young adult dogs.
KEY POINT 7-36
Although chloramphenicol is bacteriostatic, its excellent tissue distribution and relatively long half-life render it potential appealing choice in immunocompromised patients.
Chloramphenicol was studied after single oral dose as the commercially available Chloromycetin (50 mg/kg) in dogs.262 Although pharmacokinetics were not reported, the Cmax (μg/mL) at Tmax were, respectively, for Greyhounds with feeding, 21.6 ± 4.8 at 1.5 hours, or without feeding, 18.6 ± 6.7 at 3 hours; large dogs (22-26 kg), 20.0 ± 4.8 at 1.5 hours; and small dogs (11.4 to 15.5 kg), 27.5 ± 7.0 at 3 hours. Peak concentrations were notably higher in small dogs than large dogs. Half-life in Greyhounds was 3.2 hours in fasted dogs versus 1.9 hours in fed dogs; the elimination half-life (based on noncompartmental anaylsis of published data) in large dogs was 2.3 hours compared with 3.4 hours in small dogs. Average half-life among all groups was 2.7 ± 0.7 hours; mean residence time was 4.6 ± 0.67 hours. Neither oral bioavailability nor clearance was determined.
During approval for use in humans, chloramphenicol was studied in dogs.263 Chloramphenicol was measured on the basis of an analytic procedure that detected chloramphenicol and its metabolites; therefore the relevance of the data must be considered. Homogenate tissue concentrations were described after subcutaneous administration of 35 mg/kg for 2 dogs: at 1.5 and 3 hours, plasma concentrations were 21 and 13, respectively, yielding an elimination half-life of 2.9 hours. Concentrations in the brain and CSF at the same time were 15 and 8 μg/mL (brain) and 7 and 9 μg/mL (CSF), yielding a 3-hour plasma:tissue ratio of 0.7. A second study measured chloramphenicol using a bioassay. However, only a single dog was studied after oral administration of 150 mg/kg of the crystalline powder form. The Cmax was 45 μg/mL at 4 hours and approximately 15 μg/mL at 8 hours, yielding a disappearance half-life of 2.5 hours. This should extrapolate to a Cmax of approximately 14 μg/mL when 50 mg/kg is administered. Although 54% of the drug was eliminated in the urine, only 6.3% of the drug in the urine was pharmacologically active. Intravenous administration of 50 mg/kg yielded an initial plasma concentration of approximately 39 μg/mL and a concentration of approximately 5 μg/mL at 8 hours, yielding a half-life of about 1.5 hours.263
Chloramphenicol has been studied in cats (n = 5). Oral administration of the crystalline powder in capsules yielded Cmax (μg/mL) of 43 to 62 at 40 mg/kg, 25 to 42 at 20 mg/kg tid, and 8 to 25 at 50 mg bid.264 Cats were also dosed with succinate intravenously, intramuscularly, subcutaneously, or orally (crystalline powder in capsules).265 Concentrations at 30 minutes (Tmax for each route except oral) were, respectively (μg/mL) 19.5 ± 1.5 (intravenous), 18.6 ± 2.6 (intramuscular), 14.8 ± 2.9 (subcutaneous) and 9.8 ± 2.61 (oral) after administration of 20 mg/kg (n = 5). The mean half-life of all three routes was 4.4 ± 1.38; range was 3.3 hours for subcutaneous and 6.9 hours for intravenous. AUC for each route was similar (lowest at 55 ± 7 μg∗hr/mL for intravenous, highest at 67 ± 9 for subcutaneous). Finally, the bioavailability of the palmitate salt suspension is poor in cats, particularly in the fasted state.266 Peak concentrations of the crystalline form following 100 mg/cat was 25 ± 5 (fasted) or 31 ± 3 (fed) versus 6.5 ± 1.3 (fasted) and 16 ± 3 (fed) for the succinate form.
Florfenicol has been studied in dogs and cats.260,267 In dogs, although predictable PDCs (1.64 μg/mL) are achieved at 20 mg/kg after intramuscular administration, concentrations are unpredictable after subcutaneous administration. The drug appears to follow a “flip-flop” model, with the elimination half-life in dogs following intramuscular administration much longer (9 hours) compared with intravenous administration (<1 hour). A second study determined the oral bioavailability and described in more detail the PK of florfenicol (based on HPLC) in dogs (n = 6) after intravenous and oral administration (20 mg/kg).267 Florenicol clearance was 1.03 ± 0.49 l∗kg/hr, and the Vdss 1.45 ± 0.82 L/kg.The elimination half life is 1.11 + 0.94 hour after intravenous and 1.24 ± 0.64 after oral administration. Oral bioavailability was 95 ± 11%, with Cmax reaching 6.18 μg/mL at a Tmax of 0.94 hour. Florfenicol amine is a major metabolite of florfenicol, with a longer half-life in dogs (2.26 hours), but it has only 1/90 the activity of the parent compound, and its contribution to microbiological activity is considered negligible. Dogs showed no evidence of side effects after either intravenous or oral administration. The disposition of florfenicol by the intramuscular route appears to be more predicatable in cats than dogs, with Cmax (22 mg/kg) reaching 20 μg/mL after IM administration and 27 μg/mL after oral administration (see Table 7-1).267 Oral administration was based on a solution of 100 mg/mL. The elimination half-life was 8 hours in cats after oral administration, supporting a 12-hour dosing interval. The distribution volume in cats is supportive of a lipid-soluble drug. Adverse reactions were not noted in the six cats studied.
A major toxic concern with chloramphenicol for humans is both reversible dose-dependent and irreversible dose-independent (rare) bone marrow suppression. Reversible bone marrow suppression can also occur in animals. Dose-dependent bone marrow effects may reflect suppression of bone marrow precursor cells after mitochondrial damage. Irreversible bone marrow suppression may reflect reduction of the NO2 group to a toxic metabolite that causes stem cell damage. Irreversible suppression should be avoided with florfenicol, which lacks the NO2 group. Although cats appear more sensitive to chloramphenicol-induced reversible bone marrow suppression than do dogs, toxicosis appears rapidly reversible once the drug is discontinued. Toxicity to chloramphenicol occurs in cats after 7 days of therapy at 50 mg/kg, administered intramuscularly. The drug can, however, be used for 7 to 10 days safely in cats after oral administration of the crystalline form (capsules) at the rate of 50 mg/cat.265,266 The antianabolic effects of chloramphenicol may result in impaired protein synthesis in the patient; however, despite earlier concerns, impaired immune response to vaccines does not appear to occur.
Because they compete for the same ribosomal binding site, chloramphenicol should not be used in combination with macrolides. Because they target two different ribosomal sites, the combination of chloramphenicol with tetracyclines is appealing. Interestingly, the combined use of chloramphenicol with penicillins has been demonstrated to enhance penicillin (bacteriostatic) activity in Enterobacteraceae that are otherwise resistant to penicillins because of beta-lactamase production. Chloramphenicol inhibits product of beta-lactamases in these organisms.267a Chloramphenicol is a potent inhibitor of drug-metabolizing enzymes and inhibits the hepatic metabolism of other drugs, potentially causing toxicity should drug concentrations increase. Prolonged sleeping times have been documented after administration of pentobarbital to dogs and cats also receiving chloramphenicol;268 chloramphenicol has markedly prolonged phenytoin half-life269 and phenobarbital half-life (see Chapter 2) in dogs. Phenobarbital-induced sedation and ataxia have occurred in as few as 3 days of chloramphenicol therapy. Chloramphenicol decreases the rate of elimination of digoxin.270
The lincosamides, including lincomycin and its congener, clindamycin, are large glycosidic antimicrobials that contain an amino side chain (see Figure 7-5). They are often used in humans as penicillin substitutes to minimize the risk of penicillin hypersensitivity. The lincosamides inhibit the 50s subunit of the bacterial ribosomes but at a site distinct from that bound by the macrolides or chloramphenicol (see Figure 7-6). Peptidyl transferase is subsequently inhibited. Efficacy is reduced when the lincosamides are used concurrently with macrolides. The ribosomal action of the lincosamides results in a bacteriostatic action against susceptible organisms at recommended doses. Clindamycin is generally bacteriostatic but can be bactericidal at concentrations that can be achieved in some tissues. As with other bacteriostatic drugs, the lincosamides are classified as time dependent, implying that plasma or tissue drug concentrations should exceed the MIC of the infecting organism for the majority of the dosing interval; efficacy also may be related to the AUC/MIC.
The spectrum of the lincosamides varies with the drug. Clindamycin is more effective against susceptible bacteria compared with lincomycin and also has greater activity toward anaerobes. The spectrum of clindamycin includes aerobic gram-positive cocci, including Staphylococcus and Streptococcus spp. as well as Nocardia spp. and anaerobic organisms including B. fragilis, Fusobacterium spp., Clostridium perfringens, Peptostreptococcus, and Actinomyces spp. Clindamycin also is effective against cell wall–deficient organisms such as Mycoplasma spp. Plasmid-mediated resistance reflects changes in the ribosomes and appears to be increasing against Staphylococcus spp. and Bacteroides spp. Resistance to one lincosamide generally results in resistance to others. Occasionally, resistance to macrolides may confer resistance to clindamycin if the mechanism reflects methylation of the ribosome.80 Clindamycin is not a substrate for the macrolide efflux pump.
Because of its anaerobic and gram-positive spectrum, clindamycin often is chosen as one component of combination antimicrobial therapy. This combination also has been used to target P. aeruginosa; although generally ineffective as a sole agent, clindamycin may alter adherence of the microbe to epithelial cells, facilitating killing by the alternative drug.
Only oral preparations of clindamycin are approved in the dog and cat; an injectable preparation is approved for use in humans. Both clindamycin and lincomycin are bioavailable after oral administration, although clindamycin is more so. Food does not impair the absorption of clindamycin but does appear to impair absorption of lincomycin. Clindamycin is available as the hydrochloride, palmitate, or phosphate salts. The palmitate form is an oral prodrug, with the ester being rapidly hydrolyzed to yield free drug. The phosphate form is intended for parenteral administration, including subcutaneous, intramuscular (although it is painful), and intravenous routes. In the cat administration of 5.5 and 11 mg/kg orally generates serum concentrations above the MIC of most S. pseudintermedius organisms and previously S. aureus, but it is likely that resistance has resulted in less favorable PDI. Higher doses (11 to 20 mg/kg) will generate concentrations above the MIC of most susceptible anaerobes (see Table 7-1). In dogs oral administration of 11 mg/kg every 12 hours has been sufficient for treatment of most Staphylococcus spp. infections, but current MIC90 for clindamycin and susceptible Staphyloccocus spp. have not been reported. As a time-dependent drug, decreasing the interval to 8 to 6 hours may increase efficacy. Clindamycin is highly (>90%) protein bound. Distribution of the lincosamides includes most body tissues, with excellent concentrations being achieved in the skin and bones. However, it does not substantially penetrate the brain or CSF, although it can achieve concentrations effective for toxoplasmosis.80 Its Vd in both dogs and cats approximates 1.5 L/kg. Clindamycin has been cited for its efficacy in the treatment of chronic gingivitis or periodontal disease. Unlike many other drugs with a favorable spectrum, it is able to penetrate the biofilm that protects the causative organisms. Accumulation of clindamycin in white blood cells up to fortyfold or more may increase the probability of reaching bactericidal concentrations at some sites of infection. The lincosamides are eliminated primarily by biliary excretion.
After administration of 10 mg/kg intravenously, intramuscularly, and subcutaneously in dogs, in addition to Cmax and elimination half-life (see Table 7-1), the following were achieved: Tmax occurred at 73 ± 16 min (intramuscular) or 47 ± 20 min (subcutaneous) and CL (mL/min/kg) 6.1 ± 1.1. The elimination half-life may vary with the route (see Table 7-2), as does mean residence time at 143 ± 34, (intravenous), 700 ± 246 (intramuscular), or 364 ± 147 (subcutaneous) minutes. Bioavailability was 115% after intramuscular and 310% after subcutaneous administration. The long half-life coupled with the highest Cmax suggests that the subcutaneous route of administration is the preferred parenteral route for clindamycin.272 The reason for the very high bioavailability after subcutaneous administration is not clear, although enterohepatic circulation is anticipated to increase bioavailability regardless of route of administration.
Clindamycin disposition has been reported in cats after oral administration of either a capsule or aqueous solution (see Table 7-1). 271 Peak PDCs are equivalent for both preparations, but a longer half-life for the capsule may contribute to a (not statistically significant) greater AUC for the capsule (42.6 ± 12.2) compared with the solution (35 ± 9.2). The lack of statistical difference may reflect the marked variability in half-life mean residence time for both preparations, which was approximately 6.5 hours.
Pseudomembranous colitis is a reported side effect in humans caused by overgrowth of C. difficile. The negative impact on the intestinal microbiota may persist for more than 2 weeks.80 Because of similar mechanisms of action, this drug should not be combined with chloramphenicol or erythromycin. It has been combined with aminoglycoside treatment of polymicrobial infections involving gram-negative and anaerobic organisms. The use of clindamycin as combination antimicrobial therapy was addressed in the preceding section. Because of its ability to impair pili formation and thus adherance to tracheal epithelium, clindamycin has been associated with treatment of cystic fibrsosis associated with P. aeruginosa in humans, generally in association with antipseudomonadal antimicrobials.273 However, the macrolides are more generally accepted for this use. The use of clindamycin as part of combination chemotherapy targeting protozoal disease (toxoplasmosis) is addressed in Chapter 12).
The macrolides are named for their chemical structure, composed of a very large lactone (MW >750 to >1000) ring attached to a number of sugars.274 They include the azalides, which contain a nitrogen in the ring structure (see Figure 7-13). No macrolide derivative is approved for use in dogs or cats at the time of this publication. Human-medicine drugs include the14-member rings erythromycin and clarithromycin and the 15-member ring azithromycin (an azalide semisynthetic derivative of erythromycin), spiramycin, and dirithromycin (a prodrug converted to the active erythromycylamine). The methyl group that distinguishes clarithromycin from erythromycin and the additional methyl group on azithromycin increases acid stability and enhances tissue distribution. Telithromycin is a ketolide macrolide (discussed later). Tylosin, a drug approved for use in food animals, is used to treat intestinal disorders, largely in dogs. Of the human drugs, erythromycin (first-generation), azithromycin, and to a lesser extent, clarithromycin (second-generation) are used in dogs and cats.274 Tilmicosin is approved for use in selected food animals, but toxicity precludes its use in the injectable form in dogs and cats; information is not available regarding safety of other preparations. The second-generation macrolides differ from erythromycin only by the addition of a methyl group substitution. However, this simple change improves acid stability and tissue penetration. Further, because the methyl group enhances interaction with bacterial ribosomes, the spectrum also is improved.274
Macrolides inhibit bacterial ribosomal action by binding to the 50s subunit of susceptible organisms (see Figure 7-6), and impairing the translocation step of protein synthesis. The azalides macrolides bind the ribosome at two sites.275 Although macrolides are classified as bacteriostatic in vitro, they are bactericidal against very susceptible organisms. Further, selected drugs (e.g., azithromycin) accumulate in selected tissues at bactericidal concentrations. All macrolides generally accumulate in phagocytic white blood cells, which may facilitate distribution to the site of infection. Efficacy is enhanced in an alkaline pH, probably because of increased diffusion of the nonionized drug into organisms; as such, intracellular activity may be decreased in phagocytic cells. The antibacterial effects of the macrolides vary with the drug and are time dependent for erythromycin; antibacterial effects for azithromycin and clarithromycin are time dependent for some organisms and concentration dependent for others.
Like the lincosamides, the macrolides are often used in humans as penicillin substitutes to minimize the risk of penicillin hypersensitivity. Organisms are considered susceptible to the macrolides at an MIC below 2 μg/mL. For the first-generation drugs, gram-positive organisms accumulate erythromycin at concentrations that exceed that of gram-negative organisms by a hundredfold. As such, erythromycin is most effective against gram-positive organisms. Streptococcus spp. are susceptible at a range of 0.015 to 1 μg/mL, although resistance is increasing. Many Staphyloccocus organisms have remained susceptible to erythromycin, but MIC ranges of 0.12 to > 128 μg/mL for S. aureus indicate an increasing trend of resistance. Among the staphyloccoci, S. pseudintermedius remains the most susceptible. P. multocida, Bordetella pertussis, and Mycoplasma spp. are among the organisms susceptible to erythromycin. However, use should be based on C&S testing. Erythromycin generally is effective against anaerobic organisms, with the exception of Bacterioides spp. Macrolides are generally effective against Campylobacter spp.
The azolides were designed to overcome barriers presented to penetration of gram-negative organisms. Thus the spectrum of azithromycin and clarithromycin increases, particularly in terms of gram-negative bacteria, although efficacy toward selected gram-positive microbes may decrease, requiring higher MIC.275 The actions of the azolides are bactericidal for Streptococcus pyogenes and S. pneumoniae but bacteriostatic toward staphylococci and most aerobic gram-negative organisms. Clarithromycin is effective at lower concentrations than erythromycin against Streptococcus and Staphyloccus spp. but is similar to erythromycin in efficacy against other organisms. Azithromycin has less activity against gram-positive organisms compared with erythromycin and greater activity against selected gram-negative organisms and Mycoplasma spp.80 Although the spectrum of the macrolides generally includes Actinomyces spp., efficacy is generally less for Nocardia spp. Clarithromycin and azithromycin are effective against the Mycobacterium avium complex, Mycobacterium leprae, and Toxoplasma gondii. Compared to erythromycin, azithromycin and clarithromycin have enhanced activity against selected protozoa (e.g., T. gondii, Cryptosporidium spp.).
Controversy surrounds the classification of macrolides as either concentration or time dependent. The macrolides do exhibit a postantibiotic effect, with that of clarithromycin and azithromycin being longer than that of erythromycin. Azithromycin appears to be bacteriostatic against Staphylococcus or Streptococcus spp.; in vitro killing did not increase in a dose-dependent manner, suggesting that the drug is a time-dependent antimicrobial.276
Acquired mechanisms of resistance to macrolides include pump-driven drug efflux from the cell (particularly in staphylococci, group A streptococci, and S. pneumoniae) and altered ribosomal targets (methylase enyzme; MLSB phenotype) that also confer resistance to lincosamides, which bind at the same ribosomal site. Efflux pumps contribute to resistance in E. coli as well.179 Chromosomal mutations in Bacillus subtilis, Campylobacter spp., and gram-positive cocci alter the ribosomal binding site. Resistance of S. aureus to erythromycin generally is indicative of resistance to azithromycin and clarithromycin as well.The Enterobacteriaceae produce an esterase that hydrolyses the drug.
The macrolides and azolides are largely water insoluble and are unstable in the acidic gastric environment.274 However, each of the macrolides is available as an oral preparation. Erythromycin also is available as a topical and ophthalmic preparation. Erythromycin base preparations generally are coated to prevent gastric degradation. Oral absorption of enteric-coated or delayed-release products designed for humans may be unpredicatable in animals.2 Oral salts include the estolate and ethylsuccinate salts, which must be de-esterified after oral absorption, and the stearate (octadecanoate) and phosphate salts. The former (and possibly the latter) dissociate in the duodenum to be absorbed as the free base. The disposition of selected erythromycin salts has been described in dogs.277
After oral administration, the erythromycin base is incompletely but adequately absorbed. Food may increase acidity and thus delay absorption. Esters (stearate, estolate, ethylsuccinate) improve stability and absorption but do not appear to increase PDCs. Among the salts, estolate appears to be best absorbed orally and minimally affected by food. For the azolides, clarithromycin is characterized by greater acid stability compared with erythromycin. Clarithromycin is more rapidly absorbed (in humans), but food delays absorption and first-pass metabolism (to an active metabolite) further reduces oral bioavailability of the parent compound to 50%. Azithromycin also is absorbed rapidly, but, again, food decreases bioavailability to 43% (in humans). Erythromycin is approximately 75% protein bound; binding is as high as 96% (in humans) for the estolate salt. Protein binding for clarithromycin is concentration dependent and ranges from 40% to 70%. Despite their large moleculer size, macrolides are sufficiently lipid soluble that they diffuse through membranes, albeit slowly. With a Vd of 2 L/kg in dogs, erythromycin will reach effective concentrations in all tissues except the brain and CSF. In general, the macrolides act as weak bases and, as such, trapped in an acidic environment, including acidic intracellular organelles. Consequently, tissue concentrations will exceed plasma in many tissues. Although accumulation occurs in selected tissues (e.g., bile, bronchial secretions, phagocytic white blood cells), concentrations reach only 50% of plasma in the prostate and aqueous humor and less than 15% in the CSF. Concentrations in the middle ear will approximate 50% of those in plasma. Clarithromycin and its active metabolite are well distributed, achieving higher concentrations than erythromycin in both the middle ear and CNS. Among the macrolides, azithromycin distributes the most extensively, with a Vd that exceeds (in humans) 30 L/kg. Fibroblasts act as a reservoir, with transfer to phagocytic cells. Whereas erythromycin and azithyromycin are eliminated principally in the bile, clarithromycin is extensively metabolized to an active (14 hydroxy derivative) metabolite. Excretion is primarily by biliary secretion into the feces; enterohepatic circulation of active drug might be anticipated. Urine excretion is not significant (3% to 5%), with concentrations in urine being low (approximately 50% of plasma); an exception is clarithromycin, for which the active metabolite might achieve high concentrations in urine. The elimination half-life for azithromycin has been reported at 1 to 1.5 hours in dogs279,280 and cats.
The disposition of erythromycin as the estolate tablet and ethylsuccinate suspension and tablet has recently been described in dogs.277 Intravenous administration revealed a Vd of 4.8 L/kg (see Table 7-1) and a clearance of 2.64 ± 0.84 L/hr/kg. Oral absorption of all three products was poor: the ethylsuccinate tablets did not yield predictably detectable concentrations, whereas, based on mean AUC adjusted for differences in dose, the bioavailability of the estolate tablet was only 11% (Tmax 1.7 hr) and the ethylsuccinate suspension only 3% (Tmax, 0.7 hr). Absorption of the suspension, in particular, was described by the authors as erratic. Peak concentrations did not reach MIC90 for susceptible Staphylococcus spp. of 0.5 μg/mL (reported by the authors) for any of the oral preparations. The apparent efficacy of erythromycin, despite poor absorption, may reflect accumulation of drug in tissues such that higher concentrations are achieved at the site of infection.277 All dogs vomited after dosing, regardless of route of administration, with vomiting apparent 5 to 10 minutes after intravenous administration, approximately 45 minutes after oral succinate preparations, and 1 to 2 hours after the estolate tablet administration.
Limited information is available for the second-generation macrolides in animals. Azithromycin and clarithromycin absorption is influenced by uptake transporters in the intestinal epithelium. Whereas efflux transporters, such as P-glycoprotein, decrease absorption, others (organic anion-transporting proteins) facilitate uptake.278 Azithromycin has been studied in cats and dogs (see Table 7-1).279,280 Bioavailability in the dog is greater than 97%. Serum protein binding is less than 25%.
Clearance is 6.0 mL∗min/kg. In dogs 67% of the drug is eliminated in the bile and 33% in the urine.279 The majority of the drug (75%) is eliminated unchanged. The remaining portion is metabolized by cytochrome P450s into a number of metabolites, which, with one exception, are inactive. Tissue concentrations (based on homogenate) at 24 hours after 20 mg/kg orally were over 101, 20, and 39 μg/mL, respectively, for liver, kidney, and lungs. After 5 days of dosing, 23 μg/mL was achieved 24 hours after the last dose in the eye but only 1.2 μg/mL in the brain (at 30 mg/kg for 5 days). In cats the maximum drug concentration (Cmax) of 0.97 ± 0.65 μg/mL occurs at Tmax of 0.85 ± 0.72 hr. Plasma concentrations (μg/mL) range from approximately 8 at 1 hour to 0.1 at 12 hours after intravenous administration of 5 mg/kg and approximately 1 μg/mL to 0.1 μg/mL during the same times after oral administration of 5 mg/kg. Although the elimination half-life is long, concentrations in plasma are below 0.1 μg/mL after 12 hours. However, concentrations of azithromycin approximate 0.75 to 1 μg/mL in the femur, skin, and muscle versus 10 μg/mL in tissues characterized by reticuloendothelial cells (liver, spleen, and to a lesser degree lung) and the kidney with concentrations persisting for 72 hours or more. Because tissue concentrations were based on homogenate, it is not clear how much drug is available to interstitial fluid. Clearance is 0.64 ± 0.24 L∗hr/kg. Oral bioavailability is 52 ± 22%. The elimination half-life is 35 (range 29 to 51 hours).280 The Clinical Laboratory and Standards Institute susceptible breakpoint for azithromycin (human pathogens) is 4 μg/mL. Because concentrations decline to less than 0.1 μg/mL by 12 hours, daily dosing should be considered in both cats and dogs; because time to steady state will approximate 3 to 5 days, a 15 mg/kg loading dose should be considered followed by once-daily dosing at a minimum of 5 mg/kg. Although cats do metabolize azithromycin, the unchanged drug is the predominant form in tissues. Biliary excretion is a major route of clearance in the cat.280 Kinetics of clarithromycin become zero order (saturated) at higher doses. The large Vd of the macrolides contributes to their long elimination half-life. The half-life in cats exceeds 72 hours in some tissues.280 In contrast to azithromycin, urinary concentrations of clarithromycin can be signifcant: up to 40% of the parent drug or its metabolite are eliminated in the urine. The mean half-life in plasma is 35 hours but varies in tissues from a low of 13 hours (fat) to a high of 72 hours (cardiac muscle).
Side effects of the macrolides are limited. With injectable products, pain may occur with intramuscular injection and thrombophlebitis with intravenous injection. Reversible cholestatic hepatitis accompanied by jaundice has been reported in humans 10 to 20 days into erythromycin therapy, especially with the estolate preparation.
Gastrointestinal upset is the most common adverse effect of the macrolides. Up to 50% of animals treated with erythromycin may exhibit vomiting. Erythromicin is motilin-like in action and characterized by marked prokinetic effects on gastrointestinal motility. This effect is dose dependent in humans and may occur more commonly in younger animals. Abdominal cramping, epigastric pain, and increased gastric emptying resulting from increased gastric motility also may occur. However, because contraction is not coordinated, efficacy as a prokinetic is limited. Gastric emptying may decrease gastric maceration of ingested food, although the impact on digestion is not likely to be significant. Azithromycin and clarithromycin do not appear to have the same gastrointestinal side effects of erythromycin. In humans allergic reactions occur rarely and are manifested as fever or skin eruptions, which resolve once therapy is discontinued. Cholestatic hepatitis is an infrequent side effect in humans.
Antacids decrease the rate (and thus peak) but not extent of absorption of azithromycin, whereas food decreases the extent by close to 50%. The macrolides may inhibit cytochrome P450 enzymes, and CYP 3A4 in particular, impairing the metabolism of other drugs.280a Among the macrolides, erythromycin followed by clarithromycin is most likely to be involved in significant drug interactions, although all three drugs inhibit drug-metabolizing enzymes. The effects of drugs metabolized by the liver, including selected anticonvulsants, cardiac drugs, and theophylline, are likely to increase. Drugs affected in humans include glucocorticoids, digoxin, theophylline, and warfarin. The macrolide antimicrobials (clarithromycin, roxithromycin) also increase the risk of digoxin toxicity, although this effect may be more reflective of competitive interactions with P-glycoprotein transport proteins.281 Azithromycin is a substrate; others may be as well.282 Among the P-glycoprotein interactions with azithromycin in cats is cyclosporine; peak cyclosporine concentrations exceeded 4500 ng/mL in a cat receiving 5 mg/kg while being treated with azithromycin.
Because they are ribosomal inhibitors, care must be taken not to combine the macrolides with drugs whose efficacy requires rapid bacterial growth, unless scientific support exists, or “-cidal” concentrations of the macrolide are achieved at the target site for both drugs. For example, synergistic effects have been documented against B. fragilis when erythromycin is combined with cefamandole and against Nocardia asteroides when combined with ampicillin. The use of erythromycin in combination with other antimicrobials is limited in small animals. Erythromycin has been used in combination with rifampin to treat Rhodococcus equi in horses; a similar application has not been identified in dogs or cats. Synergistic antimicrobial actions also have been reported against P. aeruginosa for either azithromycin or clarithromycin when combined with sulfadiazine/trimethoprim or doxycycline. In humans azithromycin has been combined with antipseudomonadal drugs, particularly for treatment of cystic fibrosis–associated P. aeruginosa infections. This may reflect an apparent immunomodulatory effect of azithromycin or its ability to inhibit adherence of pseudomonad organisms to respiratory epithelium. Less commonly, synergism has been demonstrated for azithromycin when combined with ticarcillin/clavulanic acid, piperacillin/tazobactam, ceftazidime, meropenem, imipenem, ciprofloxacin, travofloxacin, chloramphenicol, or tobramycin.259
Although not included in their spectrum, the macrolides, like clindamycin, impair the ability of P. aeruginosa to adhere to tracheal epithelium, the first step in respiratory tract infection. The effect occurs at least at subinhibitory concentrations and reflects decreased ability to form pili.273 Decreased adherence to human mucins also has been demonstrated for azithromycin.283 Other proposed effects of azithromycin include decreased alginate formation and decreased biofilm. Azithromycin has been demonstrated to impede, but not prevent, biofilm formation by Pseudomonas spp.284 These attributes have led to its long-term use for treatment of cystic fibrosis in humans, generally in association with some level of antipseudomonadal antimicrobials. Antiinflammatory effects have also been attributed to azithromycin’s apparent long-term efficacy for treatment of cystic fibrosis.285,286
Tylosin is a classified as a macrolide, but it is structurally somewhat different from erythromycin, leading to differences in its mechanism and spectrum. Like erythromycin, it targets the 50s ribosomal subunit, but with different sequelae. It is stable in the gastric environment and does not require enteric coating for oral administration. Like erythromycin, tylosin is distributed well to most body tissues and is eliminated by hepatic metabolism and biliary excretion. Approved for use in the United States for treatment of swine dysentery and other large animal syndromes, tylosin also has been used in small animals to treat infections of the gastrointestinal tract (associated with chronic inflammatory bowel disease) and bacterial pyodermas. Its spectrum is not clear but includes selected gram-positive and gram-negative organisms.
Like the azolides, the ketolides are semisynthetic modifications of erythromycin designed to minimize barriers to penetration in gram-negative organisms.275 Telithromycin is the first ketolide approved for clinical use in humans; the drug was developed specifically for treatment of upper and lower respiratory tract infections caused by organisms resistant to the macrolides.287 Like the macrolides and azalides, the ketolides are well distributed into tissues, with concentrations being maintained in humans sufficiently long to allow a 24-dosing interval. Thus far, the ketolides have not been used or studied in veterinary medicine, perhaps because azithromycin currently meets the needs of infections that might otherwise be treated with ketolides.
Oxazolidinones are a new group of synthetic antimicrobials effective against gram-positive bacteria, including methicillin- and vancomycin-resistant staphylococci, vancomycin-resistant enterococci, penicillin-resistant pneumococci, and anaerobes.288 Linezolid is the first of this class of drugs to be approved for use in the United States (see Figure 7-4). Oxazolidinones inhibit the initiation of protein synthesis by binding at the P site of 50S ribosomal subunit; it also binds to the 70S subunit. Oxazolidinones compete with chloramphenicol and lincomycin for binding of the 50S subunit, which indicates that they have close binding sites, even though oxazolidinones do not inhibit peptidyl transferase as do chloramphenicol and lincomycin. Oxazolidinones may inhibit formation of the ribosomal initiation complex, similar to aminoglycosides. The mechanism is sufficiently different from other 50S binders that resistance to other protein synthesis inhibitors does not cross over to the oxazolidinones. Efficacy against Staphylococcus spp. is characterized by an MIC90 between 1 and 4 μg/mL in humans; methicillin resistance does not appear to affect susceptibility. Linezolid also is effective against enterococci; even intermediate isolates appear to be susceptible at 1 μg/mL.288 Streptococci also are susceptible. Anaerobic activity is comparable to clindamycin.289 Linezolid is effective toward atypical mycobacterium290 and both Actinomyces and Nocardia sp. Activity toward S. pneumoniae is generally bactericidal but bacteriostatic against staphylococci and enterococci. 291, 292 Antibacterial effects appear to be time dependent, with efficacy related to AUC/MIC. Resistance thus far is rare.
Disposition includes good oral absorption and good tissue penetration. Linezolid accumulates in bone, lung, vegetations, hematoma, and CSF. Concentrations in sanctuaries are lower than those in plasma.293 Linezolid has been approved by the FDA for treatment in humans of complicated skin infections or nosocomial pneumonia caused by MRSA, concurrent bacteremia associated with either vancomycin-resistant E. faecium or CA pneumonia caused by penicillin-resistant S. pneumoniae.288 It has become the drug of choice for treatment of resistant gram-positive infections. The oxazolidinones have been minimally used in dogs and cats, and their use is discouraged unless warranted on the basis of C&S testing and until kinetic studies are available in the target species (e.g., cats).
Linezolid PK has been described in the dog after oral and intravenous administration (see Table 7-1).293 Oral absorption is rapid and complete, allowing intravenous and oral dosing to be the same. The drug is minimally protein bound. Clearance is 2.0 ± 0.3 mL∗min/kg. The drug appears to undergo limited metabolism to inactive metabolites that are extensively enterohepatic recycled. Renal excretion occurs for parent compounds and metabolites. In humans renal disease causes accumulation of metabolites that may contribute to adverse effects.294
Linezolide appears to be well tolerated in humans. Myelosuppression has occurred in humans. Additionally, it is an inhibitor of monoamine oxidases, and care should be taken in patients also receiving serotonergic or adrenergic drugs or dietary supplements. Peripheral neuropathies have been associated with long-term use. Drug interactions involving cytochrome P450 do not appear to occur. Linezolide inhibits mitochondrial protein synthesis, causing hyperlactatemia in humans.294 Linezolid may decrease intracellular movement of aminoglycosides, affecting rapid killing. 94 Based on in vitro killing curve studies, linezolid efficacy against MRSA was enhanced most by rifampin, compared with vancomycin or gentamicin; indeed, efficacy of the latter was reduced by linezolid, with activity antagonistic toward gentamicin.
Daptomycin is a lipopeptide derived from Streptomyces that was discovered several decades ago but has been reconsidered for treatment of vancomycin-resistant gram-positive organisms. Its spectrum includes gram-positive and anaerobic microbes. However, its mechanism involves binding to the cell membrane, and although bactericidal, daptomycin is associated with an increased risk of toxicity. It acts in a concentration-dependent fashion.80 Vancomycin-resistant drugs require higher concentrations. Daptomycin is minimally orally bioavailable, requiring intravenous administration for systemic effects. It cannot be given intramuscularly because of direct toxicity. It is not involved in any clinically relevant drug interactions. Although largely renally excreted, it is approximately 92% bound to plasma proteins in humans. The result is a longer half-life that allows once-daily dosing in humans. Daptomycin causes skeletal muscle damage in dogs at doses that exceed 10 mg/kg and peripheral neuropoathies at higher doses.80 Dispostion has been described for Beagles after once-and twice-daily dosing.295 When given at 5, 25, or 75 mg/kg intravenously, peak serum concentrations were 58, 165, and 540 μg/mL, respectively (total drug); concentrations extrapolated from the terminal component of the curve approximated 30, 100, and 300 μg/mL, respectively. The elimination half-life appeared to be between 2 and 3 hours, which may indicate that the drug is not as highly protein bound in dogs compared with people. All doses caused skeletal muscle damage, as indicated by serum creatine phosphokinase; damage, however, was worse with 8-hour administration of 25 mg/kg than with once-daily administration of 75 mg/kg.295
Fusidic acid is a steroidlike antimicrobial that interferes with ribosomal translocation (peptidyl tRNA). Efficacy is limited to gram-positive bacteria. It is bactericidal at high concentrations against both coagulase-positive and coagulase-negative staphylococci. It is available as oral, intravenous, topical, and ocular preparations. In humans it is characterized by 90% oral bioavailability. Adverse reactions include granulocytopenia, rash, and hepatotoxicity; thrombophlebitis; and venospasm, which may accompany intravenous infusion. Resistance develops rapidly when used as a sole agent. Drugs with which it has been combined include the aminoglycosides, quinolones, rifampin, and vancomycin. However, combination therapy has not precluded development of MRSA.
The advantage of topical antimicrobials is achievement of very high concentrations at the site of infection and avoidance of side effects that otherwise might occur with systemic therapy.
Bacitracin is a complex polypeptide isolated from B. subtilis. It inhibits peptidoglycan synthesis in bacteria by interfering with the enzyme responsible for movement of cell components through the membrane. Its spectrum of activity includes gram-positive and very few gram-negative organisms. Systemic use causes nephrotoxicity, and use is limited to topical administration. The drug is not absorbed after oral administration and can be used to treat gastrointestinal infections caused by susceptible organisms.5,297
Polymyxins are a group of large acetylated decapeptides produced by Bacillus spp. At least six compounds have been identified, of which only two, polymyxin (polymyxin B) and colistin (polymyxin E), are used clinically. Polymyxins are cationic detergents that interact and interfere with the phospholipid of the bacterial cell membrane, resulting in increased permeability. The polymyxins are thus bactericidal. However, a number of compounds can interfere with their activity, including divalent cations, purulent exudate, fatty acids, and quaternary ammonium compounds. The spectrum of activity of the polymyxins includes most gram-negative organisms, including P. aeruginosa. Two exceptions include Proteus spp. and most Serratia spp. The drugs are weak bases (pKa 8 to 9) and are not orally bioavailable. As such, they have been used to “sterilize” the gastrointestinal tract.
Elimination is principally by way of the kidneys, which are also the primary sites of toxicity. Glomerular and tubular epithelial damage has limited their usefulness. Other side effects include respiratory paralysis (after rapid intravenous administration), CNS dysfunction, fever, and anorexia. Use of the polymyxins is primarily limited to topical administration. However, pemphigus vulgaris has been reported in association with topical use for otitis externa in the dog.296 Polymyxin protects against gram-negative endotoxemia by binding to the anionic lipid component of the lipopolysaccharide at concentrations much lower than those associated with toxicity. Relevance to treatment in dogs or cats is not established.
Novobiocin is derived from coumarin and is effective against both gram-positive and gram-negative organisms. The drug is particularly efficacious against Staphylococcus spp. Its mechanism of action is not certain but involves both cell membrane and cell wall synthesis. Novobiocin causes a number of toxic effects when used systemically, including bone marrow suppression, nausea, vomiting, and diarrhea. Its use is limited to topical application.5,297
Mupirocin (pseudomonic acid) is a naturally occurring fermentation product of Pseudomonas fluorescens. It is available as a cream or ointment, and its use has been largely limited to topical application. Although it acts to inhibit protein synthesis, its mechanism is novel in that it prevents incorporation of isoleucine into proteins by binding to isoleucyl transfer-RNA synthetase.297 Its unique mechanism precludes cross-resistance with other antibacterials. Resistance is unusual, low level, and generally overcome by higher concentrations. The spectrum of mupirocin includes aerobic gram-positive cocci (high efficacy toward S. aureus, S. epidermidis, and beta-hemolytic streptococci) and selected gram-negative cocci. An advantage to mucopirin is that it minimally affects normal flora. Its indications in human medicine include prophylaxis in ulcers, operative wounds, and burns and treatment of skin infections. In humans mupirocin has proved efficacious as an oral antibiotic. In addition, mupirocin has proven useful in the management of secondary pyodermas or superinfection of chronic dermatoses. Mupirocin is generally not associated with side effects; local burning, stinging, itching, or pain has been reported in about 1% of human patients.297
Silver sulfadiazine (see the discussion of sulfonamides) is approved for use in humans in a polypropylene glycol vehicle and in a water-soluble gel. It is approved for use in dogs combined with enrofloxacin as an otic preparation. The synergistic coupling of the silver with sulfadiazine results in efficacy against P. aeruginosa as well as a broad range of gram-positive and other gram-negative organisms. The silver component interferes with the cell wall. Silver sulfadiazine has been approved for use in the treatment of human burn patients, but other antimicrobials have proved more efficacious (e.g., iodophors; combinations of povidone iodine with neomycin, polymyxin, and bacitracin [Neosporin]; and silver sulfadiazine–cerium nitrate cream). However, the low toxicity, low hypersensitivity, and low level of resistance warrants its continued use in veterinary patients.297
The nitrofurans are synthetic compounds whose antimicrobial activity occurs through the 5-nitrofuran group (see Figure 7-12).5,297 Nitrofurantoin and furazolidone are examples. They are weak acids. These drugs block oxidative reactions necessary for formation of bacterial acetyl coenzyme A. They are bacteriostatic in action. The spectrum of activity of nitrofurantoin includes a number of gram-positive or gram-negative organisms, but its use should be based on C and S testing. The spectrum also includes selected protozoa. Nitrofurans are orally bioavailable but require an acidic environment to cross cell membranes. Use is limited to urinary tract infections, and ideally those associated with an acidic pH. Because 50% of nitrofurantoin is eliminated in urine in an active form, the drug is appropriate for treatment of urinary tract infections. Its use is, however, limited by gastrointestinal and systemic toxicity. Systemic toxicities in humans include peripheral neuropathy at therapeutic doses. The time to onset ranges from 3 weeks to over 12 months (median: 2 to 3 months). Although not common, peripheral neuropathy can be both severe and irreversible. Old age and renal disease increased the risk of toxicity.271 Albeit rare, pulmonary pneumonitis and fibrosis have been associated with long-term (6 months or more) use in humans and may be insidious in onset. The use of nitrofurantoin is limited to infections of the urinary tract that are not susceptible to other drugs. However, a current advantage to this drug is limited resistance among those organisms considered susceptible, including E. coli and selected other organisms.
Methenamine (hexamine; hexamethylenetetramine is the name for commercial uses) is a chemical that releases formaldehyde and ammonia on hydrolysis (see Figure 7-12). It is usually sold as the hippurate salt. The degree of hydrolysis, and thus antibacterial efficacy, is pH dependent, requiring an acidic pH. The drug is bactericidal in an acid environment and bacteriostatic in a more alkaline environment. Therefore it is less effective in the presence of urease-producing bacteria that alkalinize the urine. Its spectrum of activity includes both gram-positive and gram-negative organisms. Methenamine is orally bioavailable and reaches high concentrations in urine.220 The chemical is used primarily to treat urinary tract infections in dogs. Generally, it is used in combination with urinary acidifiers to enhance antibacterial actions. Its safety in cats could not be verified.
1. Albarellos G.A., Landoni M.F. Current concepts on the use of antimicrobials in cats. Vet J. 2009;180(3) (Jun):):304-316.
2. United States Pharmacopeia (USP) Antibiotic Monographs. J Vet Pharmacol Ther. 26(Suppl 2), 2003.
3. Mattoes H.M., Kim M-K Kuti J.L., et al. Clinical advantage of continuous infusion β–lactams. Serious Hospital Infections. 2001;13(3):1-8.
4. Rice L.B. Genetic and biochemical mechanisms of bacterial resistance to antimicrobial agents. In: Lorian V., editor. Antibiotics in laboratory medicine. Baltimore: Williams & Wilkins; 2005:441-508.
5. Neu H.C. Principles of antimicrobial use. In: Brody T.M., Larner J., Minneman K.P., et al, editors. Human pharmacology: molecular to clinical. St Louis: Mosby; 1994:616-701.
6. Kapusnik J., Miller R.T., Sande M.A. Antibacterial therapy in critical care. In: Schoemaker W.C., Ayers S., Grenvik A., et al, editors. Textbook of critical care. ed 2. Philadelphia: Saunders; 1989:780-801.
7. Bush L.M., Calmon J., Johnson C. Newer penicillins and lactamase inhibitors. Infect Dis Clin North Am. 1989;3:571-594.
8. Donowitz G.R. Third generation cephalosporins. Infect Dis Clin North Am. 1989;3:595-612.
9. Vaden S., Riviere J. Penicillins. In: Adams R., editor. Veterinary pharmacology and therapeutics. ed 8. Ames, Iowa: Iowa State University Press; 2001:818-827.
10. Williams J.D., Naber K.G., Bryskier A., et al. Classification of oral cephalosporins. A matter for debate. Inter J Antimicro Agents. 2001;17:443-450.
11. Caprile K.A. The cephalosporin antimicrobial agents: a comprehensive review. J Vet Pharmacol Ther. 1988;11:1-32.
12. Bramhill D. Bacterial cell division. Annu Rev Cell Dev Biol. 1997;13:395-424.
13. Anonymous. Antimicrobial treatment guidelines for acute bacterial rhinosinusitis. Arch Otolaryngol Head Neck Surg. 2004;130:S1-S45.
14. Chambers H. Penicillin-binding protein - mediated resistance in pneumococci and staphylococci. J Infect Dis. 1999;179(Suppl 2):S353-S359.
15. Bywater R.J., Palmer G.H., Buswell J.F., et al. Clavulanate-potentiated amoxycillin: activity in vitro and bioavailability in the dog. Vet Rec. 1985;116(2):33-36.
16. Brown S.A. Treatment of gram-negative infections. Vet Clin North Am. 1988;18:1141-1166.
17. Papich M. Treatment of gram-positive bacterial infections. Vet Clin North Am. 1988;18:1267-1286.
18. Sobel J.D. Imipenem and aztreonam. Infect Dis Clin North Am. 1989;3:613-624.
19. Yigit H., Queenan A.M., Anderson G.J., et al. Novel carbapenem-hydrolyzing β-lactamase, KPC-1, from a carbapenem-resistant strain of Klebsiella pneumoniae. Antimicrob Agents Chemother. 2001;45(4):1151-1161.
19a. Human Drug Product Not Equivalent to Veterinary Ceftiofur; CVM Update, July 1997. Accessed June 2010, at http://www.fda.gov/AnimalVeterinary/NewsEvents/CVMUpdates/ucm127855.
20. Stegemann M.R., Passmore C.A., Sherington J., et al. Antimicrobial activity and spectrum of cefovecin, a new extended-spectrum cephalosporin, against pathogens collected from dogs and cats in Europe and North American. Antimicrob Agents Chemother. 2006;50(7):2286-2292.
21. Lowy F.D. Antimicrobial resistance: the example of Staphylococcus aureus. J Clin Invest. 2003;111:1265-1273.
22. Helfand M.S., Bonomo R.A. β-Lactamases: a survey of protein diversity. Curr Drug Targets Infect Disord. 2003;3:9-23.
23. Li X.Z., Mehrotra M., Ghimire S., et al. Beta-lactam resistance and beta-lactamases in bacteria of animal origin. Vet Microbiol 15. 2007;121(3-4):197-214.
24. Rodriguez-Banõ J., Navarro M.D., Romero L., et al. Clinical and molecular epidemiology of extended-spectrum β-lactamase–producing Escherichia coli as a cause of nosocomial infection or colonization: implications for control. Clin Infect Dis. 2006;42:37-45.
25. Dancer S.J. The problem with cephalosporins. J Antimicro Chemother. 2001;48:463-478.
26. Vree T.B., Dammers E., van Duuren E. Variable absorption of clavulanic acid after an oral dose of 25 mg/kg of Clavubactin and Synulox in healthy dogs. J Vet Pharmacol Therap. 2003;26:165-171.
27. Malik S., Christensen H., Peng H., et al. Presence and diversity of the β-lactamase gene in cat and dog staphylococci. Vet Microbiol. 2007;123:162-168.
28. Chaudhary U., Aggarwal R. Extended spectrum -lactamases (ESBL): an emerging threat to clinical therapeutics. Indian J Med Microbiol. 2004;22(2):75-80.
29. Jacoby G.A., Medeiros A.A., O’Brien T.F., et al. Broad-spectrum, transmissible beta-lactamases. N Engl J Med. 1988;319:723-724.
30. Jacoby G.A., Medeiros A.A. More extended-spectrum beta-lactamases. Antimicrob Agents Chemother. 1991;35:1697-1704.
31. Jacoby G.A. Extended-spectrum beta-lactamases and other enzymes providing resistance to oxyimino-beta-lactams. Infect Dis Clin North Am. 1997;11:875-887.
32. Decre D., Gachot B., Lucet J.C., et al. Clinical and bacteriologic spectrum of extended-spectrum beta-lactamase-producing strains of Klebsiella pneumoniae in a medical intensive care unit. Clin Infect Dis. 1998;27:834-844.
33. Sturenburg E., Mack D. Extended-spectrum beta-lactamases: implications for the clinical microbiology laboratory, therapy, and infection control. J Infect Dis. 2003;47:273-295.
34. Lee N., Yuen K.Y., Kumana C.R. Clinical role of beta-lactam/beta-lactamase inhibitor combinations. Drugs. 2003;63(14):1511-1524.
35. Antonio O., Weigel L.M., Rasheed K., et al. Mechanisms of decreased susceptibility to cefpodoxime in. Escherichia coli, Antimicrob Agents Chemother. 2002;46(12):3829-3836.
36. Tenover F.C., Raney P.M., Williams P.P. Confirmation methods for Escherichia coli with isolates the flqA locus and genetic evidence that Topoisomerase IV is the primary target and DNA gyrase is the secondary target. The Fluoroquinolone Toxicity Research Foundation, Collected during Project ICARE. J Clin Microbiol. 2003;41:3142-3146.
37. Queenan A.M., Bush K. Carbapenemases: the versatile β-lactamases. Clin Microbiol Rev. 2007;20(3):440-458.
38. Devriese L.A., Hermans K., Baele M., et al. Staphylococcus pseudintermedius versus Staphylococcus intermedius. Vet Microbiol. 2009;133(1-2):206-207.
39. Leonard F.C., Markey B.K. Methicillin-resistant Staphylococcus aureus in animals: a review. Vet J. 2008;75:27-36.
40. Kania S.A., Williamson N.L., Frank L.A. Methicillin resistance of staphyloccci isolated from the skin of dogs with pyoderma. J Am Vet Med Assoc. 2004;65:1268-1285.
40a. Faires M.C., Tater K.C., Weese J.S. An investigation of methicillin-resistant Staphylococcus aureus colonization in people and pets in the same household with an infected person or infected pet. J Am Vet Med Assoc. 2009;235(5):540-543.
41. Gortel K., Campbell K.L., Kakoma I., et al. Methicillin resistance among staphylococci isolated from dogs. Am J Vet Res. 1999;60(12):1526-1530.
42. Weese J.S., van Duijkeren E. Methicillin-resistant Staphylococcus aureus and Staphylococcus pseudintermedius in veterinary medicine. Vet Microbiol Jan 27:. 2010;140(3-4):418-429.
43. Walther B., Wieler L.H., Friedrich A.W., et al. Methicillin-resistant Staphylococcus aureus (MRSA) isolated from small and exotic animals at a university hospital during routine microbiological examinations. Vet Microbiol Feb 5:. 2008;127(1-2):171-178.
44. Strommenger B., Kehrenberg C., Kettlitz C., et al. Molecular characterization of methicillin-resistant Staphylococcus aureus strains from pet animals and their relationship to human isolates. J Antimicrob Chemother. 2006;57:461-465.
45. Devriese L.A., Hermans K., Baele M., et al. Staphylococcus pseudintermedius versus Staphylococcus intermedius,. Vet Microbiol. 2009;133(1-2):206-207.
46. Ruscher C., Lubke-Becker A., Wleklinski C.-G., et al. Prevalence of methicillin-resistant Staphylococcus pseudintermedius isolated from clinical samples of companion animals and equidaes. Vet Microbiol Apr 14:. 2009;136(1-2):197-201.
47. El Zubeir I.E.M., Kanbar T., Alber J., et al. Phenotypic and genotypic characteristics of methicillin/oxacillin-resistant Staphylococcus intermedius isolated from clinical specimens during routine veterinary microbiological examinations. Vet Microbiol. 2007;121:170-176.
48. Pottumarthy S., Schapiro J.M., Prentice J.L., et al. Clinical isolates of Staphylococcus intermedius masquerading as methicillin-resistant Staphylococcus aureus. J Clin Microbiol. 2004;42(12):5881-5884.
49. Guardabassi L., Loeber M.E., Jacobson A. Transmission of multiple antimicrobial-resistant Staphylococcus intermedius between dogs affected by deep pyoderma and their owners. Vet Microbiol. 2004;98:23-27.
49a. Methicillin-resistant Staphylococcus Aureus. Accessed January 1, 2010, at http://www.avma.org/reference/backgrounders/mrsa_bgnd.asp
50. Braga P.C. Antibiotic penetrability into bronchial mucus: pharmacokinetic and clinical considerations. Curr Ther Res. 1989;49(2):300-327.
51. Bergogne-Bérézin E. Pharmacokinetics of antibiotics in respiratory secretions. In: Pennington J.E., editor. Respiratory infections: diagnosis and management. ed 2. New York: Raven Press; 1988:608-631.
52. Bergan T. Pharmacokientics of tissue penetration of antibiotics. Rev Infect Dis. 1981;3:45-66.
53. Bolton G.C. Allen GD, Filer CW: Absorption, metabolism and excretion studies on clavulanic acid in the rat and dog. Xenobiotica. 1984;14(6):483-490.
54. Brown SA: Pfizer Animal Health, personal communication.
55. Garg R.C., Keefe T.J., Vig M.M. Serum levels and pharmacokinetics of ticarcillin and clavulanic acid in dog following parenteral administration of Timentin. J Vet Pharmacol Ther. 1987;10(4):324-330.
56. Barker C.W., Zhang W., Sanchez S., et al. Pharmacokinetics of imipenem in dogs. Am J Vet Res. 2003;64:694-699.
57. Bidgood T.L., Papich M.G. Comparison of plasma and interstitial fluid concentrations of doxycycline and meropenem following constant rate intravenous infusion in dogs. Am J Vet Res. 2003;64:1040-1046.
58. Bidgood T., Papich M.G. Plasma pharmacokinetics and tissue fluid concentrations of meropenem after intravenous and subcutaneous administration in dogs. Am J Vet Res. 2002;63:1622-1628.
59. Krueger W.A., Neeser G., Schuster H., et al. Correlation of meropenem plasma levels with pharmacodynamic requirements in critically ill patients receiving continuous veno-venous hemofiltration. Chemotherapy. 2003;49(6):280-286.
59a. Papich M.G., Davis J.L., Floerchinger A.M., et al. Cefpodoxime and cephalexin plasma pharmacokinetics, protein binding, and tissue distribution after oral administration to dogs. Accepted. Am J Vet Res. 2010. in press
60. Carli S., Anfossi P., Villa R., et al. Absorption kinetics and bioavailability of cephalexin in the dog after oral and intramuscular administration. J Vet Pharmacol Ther. 1999;22:308-313.
61. Prados A.P., Ambros L., Montoya L., et al. Chronopharmacological study of cephalexin in dogs. Chronobiol Int. 2007;24(1):161-170.
62. Campbell B.G., Rosin E. Effect of food on absorption of cefadroxil and cephalothin in dogs. J Vet Pharmacol Ther. 1998;21:418-420.
63. Richardson D.C., Aucoin D.P., DeYoung D.J., et al. Pharmacokinetic disposition of cefazolin in serum and tissue during canine total hip replacement. Vet Surg. 1992;21(1):1-4.
64. Marcellin-Little D.J., Papich M.G., Richardson D.C., et al. Pharmacokinetic model for cefazolin distribution during total hip arthroplasty in dogs. Am J Vet Res. 1996;57(5):720-723.
64a. WHO Food Additives Series: 49 Toxicological evaluation of certain veterinary drug residues in food. Accessed June 2010, at http://www.inchem.org/documents/jecfa/jecmono/v49je04.htm).
65. Jung H.-H., Mischkowsky T. Measurements of cefuroxime concentration in bone in dogs and man. Arch Orthop Trauma Surg. 1979;94:25-28.
66. Spurling N.W., Harcourt R.A., Hyde J.J. An evaluation of the safety of cefuroxime axetil during six months oral administration to beagle dogs. J Toxicol Sci. 1986;11:237-277.
67. Sakamoto H., Hirose T., Mine Y. Pharmacokinetics of FK027 in rats and dogs. J Antibot. 1985;38(4):496-504.
68. Rebuelto M., Albarellos G., Ambros L. Pharmacokinetics of Ceftriaxone administered by the intravenous, intramuscular or subcutaneous routes to dogs. Vet Pharm Therap. 2002;25:73-76.
69. Moore K.W., Trepanier L.A., Lautzenhiser S.J., et al. Pharmaocinetics of ceftazidime in dogs following subcutanoeus administration and continuous infusion and the association with in vitro susceptibility of Pseudomonas aeruginosa. Am J Vet Res. 2000;61:1204-1208.
70. Albarellos G.A., Ambros L.A., Landoni M.F. Pharmacokinetics of ceftazidime after intravenous and intramuscular administration to domestic cats. Veterinary Journal. 2008;178:238-243.
71. Brown S.A., Arnold T.S., Hamlow P.J., et al. Plasma and urine disposition and dose proportionality of ceftiofur and metabolites in dogs after subcutaneous administration of ceftiofur sodium. J Vet Pharmacol Ther. 1995;18(5):363-369.
72. Stegemann M.R., Sherington J., Blanchflower S. Pharmacokinetics and pharmacodynamics of cefovecin in dogs. J Vet Pharmacol Therap. 2006;29:501-511.
73. Stegemann M.R., Sherington J., Coati N., et al. Pharmacokinetics of cefovecin in cats. J Vet Pharmacol Therap. 2006;29:513-524.
73a. Harmoinen J., Mentula S., Heikkilä M., et al. Orally administered targeted recombinant beta-lactamase prevents ampicillin-induced selective pressure on the gut microbiota: a novel approach to reducing antimicrobial resistance. Antimicrob Agents Chemother. 2004;48:75-79.
74. Trautmann M., Zick R., Rukavina T., et al. Antibiotic-induced release of endotoxin: in-vitro comparison of meropenem and other antibiotics. J Antimicrob Chemother. 1998;41(2):163-169.
75. Kumar G.A.V., Kothari V.M., Kirsihman A., et al. Benzathine penicillin, metronidazole and benzyl penicillin in the treatment of tetanus: a randomized, controlled trial. Ann Trop Med Parasitol. 2004;98:59-63.
76. Semel J.D., Allen N. Seizures in patients simultaneously receiving theophylline and imipenem or ciprofloxacin or metronidazole. South Med J. 1991;84:465-468.
77. Rees C.A., Boothe D.M. Evaluation of the effect of cephalexin and enrofloxacin on clinical laboratory measurements of urine glucose in dogs. J Am Vet Med Assoc. 2004;224(9):1455-1458.
78. Ingerman M.J., Santoro J. Vancomycin: a new old agent. Infect Dis Clin North Am. 1989;3:641-652.
79. Walsh T.R., Howe R.A. The prevalence and mechanisms of vancomycin resistance in Staphylococcus aureus. Annu Rev Microbiol. 2002;56:657-675.
80. Chambers HF: Protein synthesis inhibitors and miscellaneous antibacterial agents. In Brunton LL, Lazo JS, Parker KL, editors: Goodman & Gilman’s the pharmacological basis of therapeutics, ed 11. Accessed January 2009, at http://www.accessmedicine.com/content.aspx?aID=949328.
81. Fillgrove K.L., Pakhomova S., Schaab M.R., et al. Structure and mechanism of the genomically encoded fosfomycin resistance protein, FosX, from Listeria monocytogenes. Biochemistry. 2007;46:8110-8120.
82. Marchese A., Bozzolasco M., Gualco L., et al. Effect of fosfomycin alone and in combination with N-acetylcysteine on E. coli biofilms. Int J Antimicrob Agents. 2003;22(Suppl 2):95-100.
83. Marchese A., Gualco L., Debbia E.A., et al. In vitro activity of fosfomycin against gram-negative urinary pathogens and the biological cost of fosfomycin resistance. Int J Antimicrob Agents. 2003;22(Suppl 2):53-59.
84. Sauermann R., Marsik C., Steiner I., et al. Immunomodulatory effects of fosfomycin in experimental human endotoxemia. Antimicrob Agents Chemother. 2007;51(5):1879-1881.
85. Olay T., Vicente M.V., Rodríguez A. Efficacy of fosfomycin + vancomycin or gentamicin in experimental endocarditis due to methicillin-resistant Staphylococcus aureus (MRSA). J Chemother. 1989;1(4 Suppl):391-392.
86. Schito G.C. Why fosfomycin trometamol as first line therapy for uncomplicated UTI? Int J Antimicrob Agents. 2003;22(Suppl 2):79-83.
87. Sack K., Schulz E., Marre R., et al. Fosfomycin protects against tubulotoxicity induced by Cis-diaminedichloroplatin and cyclosporin A in the rat. J Mol Med. 1987;65(11):525-527.
88. Gutierrez O.L., Ocampo C.L., Aguilera J.R., et al. Pharmacokinetics of disodium–fosfomycin in mongrel dogs. Res Vet Sci. 2008;85:156-161.
89. Kawaguchi K., Hasunuma R., Kikuchi S., et al. Time- and dose-dependent effect of fosfomycin on suppression of infection-induced endotoxin shock in mice. Biol Pharm Bull. 2002;25(12):1658-1661.
90. Riviere J., Spoo W. Aminoglycosides. In: Adams R., editor. Veterinary pharmacology and therapeutics. ed 8. Ames, Iowa: Iowa State University Press; 2001:828-840.
91. Bamberger D.M., Foxworth J.W., Bridwell D.L., et al. Extravascular antimicrobial distribution and the respective blood and urine concentrations in humans. In: Lorian V., editor. Antibiotics in laboratory medicine. Baltimore: Williams & Wilkins; 2005:799-809.
92. Vakulenko S.B., Mobashery S. Versatility of aminoglycosides and prospects for their future. Clin Microbiol Rev. 2003;16:430-450.
93. Maller R., et al. Once- versus twice-daily amikacin regimen: efficacy and safety in systemic gram-negative infections. J Antimicrob Chemother. 1993;31:939-948.
94. Jacqueline C., Caillon J., Le Mabecque V., et al. In vitro activity of linezolid alone and in combination with gentamicin, vancomycin or rifampicin against methicillin-resistant Staphylococcus aureus by time-kill curve methods. J Antimicrob Chemother. 2003;51(4):857-864.
95. Powell S.H., Thompson W.L., Luthe M.A., et al. Once-daily vs. continuous aminoglycoside dosing: efficacy and toxicity in animal and clinical studies of gentamicin, netilmicin, and tobramycin. J Infect Dis. 1983;147:918-932.
96. Blaser J. Efficacy of once- and thrice-daily dosing of aminoglycosides in in vitro models of infection. J Antimicrob Chemother. 1991;27(Suppl C):21-28.
97. Reiner N.E., Bloxham D.D., Thompson W.L. Nephrotoxicity of gentamicin and tobramycin given once daily or continuously in dogs. J Antimicrob Chemother. 1978;4(Suppl A):85-101.
98. Vanhaeverbeek M., Siska G., Douchamps J., et al. Comparison of the efficacy and safety of amikacin once or twice a day in the treatment of severe gram-negative infections in the elderly. Int J Clin Pharmacol Ther. 1993;31:153-156.
99. Blaser J., Stone B.B., Zinner S.H. Efficacy of intermittent versus continuous administration of netilmicin in a two-compartment in vitro model. Antimicrob Agents Chemother. 1985;27:343-349.
100. Roudebush P., Fales W.H. Antibacterial susceptibility of gentamicin resistant organisms recovered from small companion animals. J Am Anim Hosp Assoc. 1982;18:649-652.
101. Asseray N., Caillon J., Roux N., et al. Different aminoglycoside-resistant phenotypes in a rabbit Staphylococcus aureus endocarditis infection model. Antimicrob Agents Chemother. 2002;46(5):1591-1593.
102. Forgan-Smith W.R., McSweeney R.J. Gentamicin and amikacin—an in vitro comparison using 1000 clinical isolates. Intern Med J. 2008;8:383-386.
103. Barclay M.L., Begg E.J. Aminoglycoside adaptive resistance: importance for effective dosage regimens. Drugs. 2001;61(6):713-721.
104. Mealey K.L., Boothe D.M. Nephrotoxicosis associated with topical administration of gentamicin in a cat. J Am Vet Med Asso. 1994;204:1919-1921.
104a. Kihlström E., Andåker L. Inability of gentamicin and fosfomycin to eliminate intracellular Enterobacteriaceae. J Antimicrob Chemother. 1985;15(6):723-728.
105. Ling G.V., Conzelman G.M.Jr., Franti C.E., et al. Urine concentrations of gentamicin, tobramycin, amikacin, and kanamycin after subcutaneous administration to healthy adult dogs. Am J Vet Res. 1981;42(10):1792-1794.
106. Peters A.M. The kinetic basis of glomerular filtration rate measurement and new concepts of indexation to body size. Eur J Nucl Med Mol Imaging. 2004;31(1):137-149.
107. Riviere J.E., Coppoc G.L. Pharmacokinetics of gentamicin in the juvenile dog. Am J Vet Res. 1981;42(9):1621-1623.
108. Wilson R.C., Duran S.H., Horton C.R.Jr. Bioavailability of gentamicin in dogs after intramuscular or subcutaneous injections. Am J Vet Res. 1989;50(10):1748-1750.
109. Baggot J.D., Ling G.V., Chatfield R.C. Clinical pharmacokinetics of amikacin in dogs. Am J Vet Res. 1985;46(8):1793-1796.
110. Wright L.C., Horton C.R.Jr., Jernigan A.D., et al. Pharmacokinetics of gentamicin after intravenous and subcutaneous injection in obese cats. J Vet Pharmacol Ther. 1991;14(1):96-100.
111. Jernigan A.D., Hatch R.C., Wilson R.C. Pharmacokinetics of tobramycin in cats. Am J Vet Res. 1988;49(5):608-612.
112. Jernigan A.D., Wilson R.C., Hatch R.C. Pharmacokinetics of amikacin in cats. Am J Vet Res. 1988;49(3):355-358.
113. Kukanich B., Coetzee J.F. Comparative pharmacokinetics of amikacin in Greyhound and Beagle dogs. J Vet Pharmacol Ther. 2007;31:102-107.
114. Jernigan A.D., Hatch R.C., Wilson R.C., et al. Pharmacokinetics of gentamicin in cats given Escherichia coli endotoxin. Am J Vet Res. 1988;49(5):603-607.
115. Jernigan A.D., Hatch R.C., Wilson R.C., et al. Pathologic changes and tissue gentamicin concentrations after intravenous gentamicin administration in clinically normal and endotoxemic cats. Am J Vet Res. 1988;49(5):613-617.
116. Kim H.-S., Sohn I.-J., Min D.I. Pharmacokinetics of amikacin and effect of ascites in Korean patients. Am J Health-Syst Pharm. 2002;59:1855-1857.
117. Kim M.-J., Bertino J.S.Jr., Erb T.A., et al. Application of bayes theorem to aminoglycoside-associated nephrotoxicity: comparison of extended-interval dosing, individualized pharmacokinetic monitoring, and multiple-daily dosing. J Clin Pharmacol. 2004;44:696-707.
118. Shy-Modjeska J.S., Riviere J.E., Rawlings J.O. Application of biplot methods to the multivariate analysis of toxicological and pharmacokinetic data. Toxicol Appl Pharmacol. 1984;72(1):91-101.
119. Riviere J.E., Hinsman E.J., Coppoc G.L., et al. Morphological and functional aspects of experimental gentamicin nephrotoxicity in young beagles and foals. Vet Res Commun. 1983;7(1):211-213.
120. Isoherranen N., Lavy E., Soback S. Pharmacokinetics of gentamicin C1, C1a, and C2 in beagles after a single intravenous dose. Antimicrob Agents Chemother. 2000;44(6):1443-1447.
121. Jernigan A.D., Hatch R.C., Brown J., et al. Pharmacokinetic and pathological evaluation of gentamicin in cats given a small intravenous dose repeatedly for five days. Can J Vet Res. 1988;52(2):177-180.
122. Jernigan A.D., Wilson R.C., Hatch R.C., et al. Pharmacokinetics of gentamicin after intravenous, intramuscular, and subcutaneous administration in cats. Am J Vet Res Jan:. 1988;49(1):32-35.
123. Grauer G.F., Greco D.S., Behrend E.N., et al. Estimation of quantitative enzymuria in dogs with gentamicin induced nephrotoxicosis using urine enzyme/creatinine ratios from spot urine samples. J Vet Intern Med. 1995;9(5):324-327.
124. Brown S.A., Garry F.B. Comparison of serum and renal gentamicin concentrations with fractional urinary excretion tests as indicators of nephrotoxicity. J Vet Pharmacol Ther. 1988;11:330-337.
125. Frazier D.L., Aucoin D.P., Riviere J.E. Gentamicin pharmacokinetics and nephrotoxicity in naturally acquired and experimentally induced disease in dogs. J Am Vet Med Assoc. 1988;192:57-63.
126. Boothe D.M. Adverse renal effects of selected drugs, Veterinary previews, Purina Publication for Veterinarians. Veterinary Learning Systems. 1995;2:10-13.
127. Hsu C.H., Kurtz T.W., Easterling R.E., et al. Potentiation of gentamicin nephrotoxicity by metabolic acidosis. Proc Soc Exp Biol Med. 1974;146:894-897.
128. Julien N., Karzazi M., Labrecque G., et al. Temporal modulation of nephrotoxicity, feeding, and drinking in gentamicin-treated rats. Physiol Behav. 2000;68(4):533-541.
129. Prins J.M., Weverling G.J., van Ketel R.J., et al. Circadian variations in serum levels and the renal toxicity of aminoglycosides in patients. Clin Pharmacol Ther. 1997;62:106-111.
130. Cowan R.H., Jukkola A.F., Arant B.S. Pathophysiologic evidence of gentamicin nephrotoxicity in neonatal puppies. Pediatr Res. 1980;14:1204-1211.
131. Brown S.A., Nelson R.W., Moncrieff C.S. Gentamicin pharmacokinetics in diabetic dogs. J Vet Pharmacol Ther. 1991;14:90-95.
132. Chambers H.F. Aminoglycosides. In Brunton L.L., Lazo J.S., Parker K.L., editors: Goodman & Gilman’s the pharmacological basis of therapeutics, ed 11, New York: McGraw Hill, 2006.
133. Kes P., Reiner Z. Symptomatic hypomagnesemia associated with gentamicin therapy. Magnes Trace Elem. 1990;9(1):54-60.
134. Ozbek E., Turkoz Y., Sahna E., et al. Melatonin administration prevents the nephrotoxicity induced by gentamicin. Br Med J (Intl Ed). 2000;85(6):742-746.
135. Kopple J.D., Ding H., Letoha A., et al. L-carnitine ameliorates gentamicin-induced renal injury in rats. Nephrol Dial Transplant. 2002;17:2122-2131.
136. Mazzon E., Britti D., de Sarro A., et al. Effect of N-acetylcysteine on gentamicin-mediated nephropathy in rats. Eur J Pharmacol. 2001;424(1):75-83.
137. Feldman L., Efrati S., Eviatar E., et al. Gentamicin-induced ototoxicity in hemodialysis patients is ameliorated by N-acetylcysteine. Kidney Int. 2007;72(3):359-363.
138. Konishi H., Goto M., Nakamoto Y., et al. Tobramycin inactivation by carbenicillin, ticarcillin and piperacillin. Antimicrob Agents Chemother. 1983;23:654-657.
139. Uber W.E., Brundage R.C., White R.L., et al. In vivo inactivation of tobramycin by piperacillin. Ann Pharmacother. 1991;25:357-359.
140. Ariano R.E., Kassume D.A., Meatherall R.C., et al. Lack of in vitro inactivation of tobramycin by imipenem/cilastin. Ann Pharmacother. 1992;26:1075-1077.
140a. Atilla A., Boothe H.W., Tollett M., Duran S., Campos Diaz H., Boothe D.M. In vitro elution of amikacin and vancomycin from impregnated plaster of Paris beads. Accepted for publication. Vet Surg 39: 715–721. 2010.
141. Riviere J.E., Carver M.P., Coppoc G.L., et al. Pharmacokinetics and comparative nephrotoxicity of fixed-dose versus fixed-interval reduction of gentamicin dosage in subtotal nephrectomized dogs. Toxicol Appl Pharmacol. 1984;75(3):496-509.
142. Barza M., Loannidis J.P.A., Cappelleri J.C., et al. Single or multiple daily doses of aminoglycosides: a meta-analysis. Br Med J. 1996;312:338-345.
143. Bailey T.C., Little J.R., Littenberg B., et al. A meta-analysis of extended-interval dosing versus multiple daily dosing of aminoglycosides. Clin Infect Dis. 1997;24(5):786-795.
144. Ali M.Z., Goetz M.B. A meta-analysis of the relative efficacy and toxicity of single daily dosing versus multiple daily dosing of aminoglycosides. Clin Infect Dis. 1997;24(5):796-809.
145. Gilbert D.N., Lee B.L., Dworkin R.J., et al. A randomized comparison of the safety and efficacy of once-daily gentamicin or thrice-daily gentamicin in combination with ticarcillin-clavulanate. Am J Med. 1998;105(3):182-191.
146. Wallace A.W., Jones M., Bertino J.S. Evaluation of four once-daily aminoglycoside dosing nomograms. Pharmacotherapy. 2002;22(9):1077-1083.
147. Woeltje K.F., Ritchie D.J. Antimicrobials. In: Carey C.F., Lee H.H., Woeltje K.F., editors. The Washington manual of medical therapeutics. ed 29. Philadelphia: Lippincott-Raven; 1998:250-251.
148. Rougier F., Claude D., Maurin M., et al. Aminoglycoside nephrotoxicity. Curr Drug Targets Infec Disord. 2004;4:153-162.
149. Kingsley P, Coulthard D G, Peckham SP, Conway et al: Therapeutic drug monitoring of once daily tobramycin in cystic fibrosis—caution with trough concentrations. Accessed April 2007, at http://www.cysticfibrosismedicine.com 6(2):125–130, 2001.
150. Neu H.C. The quinolones. Infect Dis Clin North Am. 1989;3:625-640.
151. Schmitz F.J., Higgins P.G., Mayer S. Activity of quinolones against gram-positive cocci: mechanisms of drug action and bacterial resistance. Eur J Clin Microbiol Infect Dis. 2002;21:647-659.
152. Petri W.A.Jr. Chemotherapy of tuberculosis, Mycobacterium avium complex disease, and leprosy. In Brunton L.L., Lazo J.S., Parker K.L., editors: Goodman & Gilman’s the pharmacological basis of therapeutics, ed 11, New York: McGraw-Hill, 2006.
153. Ng E.Y., Truckis M., Hooper D.C. Quinolone resistance mutations in topoisomerase IV: Relationship to target of fluoroquinolones in Staphylococcus aureus. Antimicrob Agents Chemother. 1996;40:1881-1888.
154. Ginsburg A.S., Grosset J.H., Bishai W.R. Fluoroquinolones, tuberculosis and resistance. The Lancet Infect Dis. 2003;3:432-442.
155. Power E.G.M., Bellido J.L.M., Phillips I. Detection of ciprofloxacin resistance in gram-negative bacteria due to alterations in gyrA. J Antimicrob Chemother. 1992;29:9-17.
156. Prescott J.F., Yielding K.M. In vitro susceptibility of selected veterinary bacterial pathogens to ciprofloxacin, enrofloxacin and norfloxacin. Can J Vet Res. 1990;54:195-197.
157. Witte W., Grimm H. Occurrence of quinolone resistance in Staphylococcus aureus from nosocomial infection. Epidemiol Infect. 1992;109:413-421.
158. Wetzstein HG, Schmeer N: Bactericidal activity of enrofloxacin against Escherichia coli growing under strictly anaerobic conditions, Proceedings of the 95th General Meeting, American Society of Microbiologists, Washington, DC, A-37, 1995.
159. van de Hoven R., Wagenaar J.A., Walker R.D. In vitro activity of difloxacin against canine bacterial isolates. J Vet Diagn Invest. 2000;12:218-233.
160. Wetzstein H.G. The in vitro postantibiotic effect of enrofloxacin, Proc 18th World Buiatrics Congress, Bologna. Italy. 1994;18:615-618.
161. Walker R.D., Stein G.E., Hauptman J.G., et al. Pharmacokinetic evaluation of enrofloxacin administered orally to healthy dogs. Am J Vet Res. 1992;53:2315-2319.
162. McKinnon P.S., Davis S.L. Pharmacokinetic and pharmacodynamic issues in the treatment of bacterial infectious diseases. Eur J Clin Microbiol Infect Dis. 2004;23:271-288.
163. Schentag J.J., Meagher A.K., Forrest A. Fluoroquinolone AUIC break points and the link to bacterial killing rates. Part 1: In vitro and animal models. Ann Pharmacother. 2003;37(9):1287-1298.
164. Boothe DM, Boeckh A, Slimpson RB, et al: Comparison of pharmacodynamic and pharmacokinetic indices of efficacy for 5 fluoroquinolones toward pathogens of dogs and cats, J Vet Intern Med 20(6):1297–13306, 2006.
165a. Shaheen B.S., Nayak R., Boothe D.M. Identification of novel mutations and high incidence of plasmid mediated quinolone resistance determinants among extended-spectrum cephalosporins in Escherichia coli isolates from companion animals in the United States. Toronto: Presented at the American Society of Microbiology Conference Antimicrobial Resistance in Zoonotic Bacteria and Foodborne Pathogens; June 8-11, 2010.
165. Hopkins K.L., Davies R.H., Threlfall E.J. Mechanisms of quinolone resistance in Escherichia coli and Salmonella: recent developments. Int J Antimicrob Agents. 2005;25:358-373.
166. Riddle C., Lemons C.L., Papich M.G., et al. Evaluation of ciprofloaxcin as a representative veterinary fluoroquinolone in susceptibility testing. J Clin Microbiol. 2000;38(4):1636-1637.
167. Pfaller M.A., Jones R.N. Trends in ciprofloxacin nonsusceptibility and levofloxacin resistance among Streptococcus pneumoniae isolates in North America. J Clin Microbiol. 2001;39(7):2748-2750.
168. Breitschwerdt E.B., Papich M.G., Hegarty B.C., et al. Efficacy of doxycycline, azithromycin, or trovafloxacin for treatment of experimental rocky mountain spotted fever in dogs. Antimicrob Agents Chemother. 1999;43(4):813-821.
169. Kang S.L., Rybak M.J., McGrath B.J., et al. Pharmacodynamics of levofloxacin, ofloxacin, and ciprofloxacin, alone and in combination with rifampin, against methicillin-susceptible and –resistant Staphylococcus aureus in an in vitro infection model. Antimicrob Agents Chemother. 1994;38(12):2702-2709.
170. Fritsche T.R., Sader H.S., Jones R.N. Epidemiology of antimicrobial resistance: species prevalence, susceptiblity profiles and resistance trends. In: Lorian V., editor. Antibiotics in laboratory medicine. Baltimore: Williams & Wilkins; 2005:815-850.
171. Hecht D.W., Wexler H.M. In vitro susceptibility of anaerobes to quinolones in the United States. Clin Infect Dis. 1996;23:S2-S8.
172. Drlica K., Zhao X., Blondeau J.M., et al. Low correlation between MIC and mutant prevention concentration. Antimicrob Agents Chemother. 2006;50(1):403-404.
173. Sánchez Navarro M.D., Sayalero Marinero M.L. Sánchez Navarro A: Pharmacokinetic/pharmacodynamic modelling of ciprofloxacin 250 mg/12 h versus 500 mg/24 h for urinary infections. J Antimicrob Chemother. 2002;50:67-72.
174. Marchbanks C.R., McKiel J.R., Gilbert D.H., et al. Dose ranging and fractionation of intravenous ciprofloxacin against Pseudomonas aeruginosa and Staphylococcus aureus in an in vitro model of infection. Antimicrob Agents Chemother. 1993;37:1756-1763.
175. Cole L.K., Kwochka K., Kowalski J., et al. Microbial flora and antimicrobial susceptibility patterns of isolated pathogens from the horizontal ear canal and middle ear in dogs with otitis media. J Amer Anim Hosp Assoc. 1998;212:534-538.
176. Boothe D.M., Smaha T., Carpenter M., et al. Emerging resistance in canine and feline Escherchia coli pathogens: a pilot surveillance study. Submitted to J Vet Int Med. March 2009.
177. Hooper D.C. Emerging mechanisms of fluoroquinolone resistance. Emerg Infect Dis. 7(2), 2001. Accessed November 12, 2009 at www.cdc.gov/ncidod/eid/vol7no2/hooper.htm
178. Debavalya N., Boothe D.M., Hathcock T. Multi-drug resistance in fecal Escherichia coli following routine enrofloxacin but not amoxicillin therapy in dogs. Am Coll Vet Int Med (Proceedings). 2008.
179. Shaheen B.S., Boothe D.M., Oyarzabal O., et al. The contribution of gyrA mutation and efflux pumps to fluoroquinolone and multi-drug resistance in canine and feline pathogenic Escherichia coli isolates from the US. In print. Am J Vet Res. 2010.
179a. Shaheen B.W., Boothe D.M., Oyarzabal O.A., et al. Antimicrobial resistance profiles and clonal relatedness of canine and feline Escherichia coli pathogens expressing multidrug resistance in the United States. J Vet Intern Med Mar-Apr:. 2010;24(2):323-330.
180. Wetzstein H.G. Comparative mutant prevention concentrations of pradofloxacin and other veterinary fluoroquinolones indicate differing potentials in preventing selection of resistance. Antimicrob Agents Chemother. 2005;49(10):4166-4173.
181. Pasquali F., Manfreda G. Mutant prevention concentration of ciprofloxacin and enrofloxacin against Echerichia coli, Salmonella Typhimurium and. Pseudomonas aeruginosa, Vet Microbiol. 2007;119:304-310.
182. Schwaber M., Cosgrove S.E., Gold H. Fluoroquinolones protective against cephalosporin resistance in gram negative nosocomial pathogens. Emerging Infectious Diseases. 2004;10(1):94-99.
183. Karlowsky J.A. Susceptibilities to levofloxacin in Streptococcus pneumoniae, Haemophilus influenzae, and Moraxella catarrhalis clinical isolates from children: results from 2000-2001 and 2001-2002 TRUST studies in the United States. Antimicrob Agents Chemother. 2003;47(6):1790-1797.
184. Seral C., Barcia-Macay M., Mingeot-Leclercq P., et al. Comparative activity of quinolones (ciprofloxacin, levofloxacin, moxifloxacin and garenoxacin) against extracellular and intracellular infection by Listeria monocytogenes and Staphylococcus aureus in J774 macrophages. J Antimicrob Chemother. 2005;55:511-517.
185. Seguin A., Papich M.G., Siegle K.J., et al. Pharmacokientics of enrofloxacin in neonatal kittens. Am J Vet Res. 2004;65:350-356.
186. Bidgood T.L., Papich M.G. Plasma and interstitial fluid pharmacokinetics of enrofloxacin, its metabolite ciprofloxacin, and marbofloxacin after oral administration and a constant rate intravenous infusion in dogs. J Vet Pharmacol Therap. 2005;28:329-341.
187. Isaccs A, Boothe DM, Axlund T: Comparative distribution of fluorquinolons in the brain and cerebral spinal fluid of dogs, 2010 (In preparation)
188. Boothe D.M., Boeckh A., Boothe H.W., et al. Tissue concentrations of enrofloxacin and ciprofloxacin in anesthetized dogs following single intravenous administration. Vet Ther. 2001;2:120-128.
189. Frazier D.L., Thompson L., Trettien A., et al. Comparison of fluoroquinolone pharmacokinetic parameters after treatment with marbofloxacin, enrofloxacin, and difloxacin in dogs. J Vet Pharmacol Therap. 2000;23:293-302.
190. Boothe D.M., Boothe H.W., Doornink M. Pharmacokinetic and pharmacodynamic indices of pradofloxacin efficacy in canine serum, and subcutaneous interstitial fluid. Berlin, Germany: Presented as a final report to Bayer Animal Health; 2006.
191. Regnier A., Concordet D., Schneider M., et al. Population pharmacokientics of marbofloxacin in aqueous humor after intravenous administration in dogs. Am J Vet Res. 2003;64:899-993.
192. Boothe D.M., Boeckh A., Boothe H.W., et al. Tissue concentrations of enrofloxacin and ciprofloxacin in anesthetized dogs following single intravenous administration. Vet Ther. 2001;2:120-128.
193. Boeckh A., Boothe D.M., Wilkie S., et al. Time course of enrofloxacin and its active metabolite in peripheral leukocytes of dogs. Vet Ther. 2001;2:334-344.
194. Hawkins E.C., Boothe D.M., Guinn A., et al. Concentration of enrofloxacin and its active metabolite in alveolar macrophages and pulmonary epithelial lining fluid of dogs. J Vet Pharmacol Ther: Feb:. 1998;21(1):18-23.
195. Boothe H.W., Jones S.A., Wilkie W.S., et al. Evaluation of the concentration of marbofloxacin in alveolar macrophages and pulmonary epithelial lining fluid after administration in dogs. Am J Vet Res. 2005;66:1770-1774.
196. Boothe D.M., Boeckh A., Boothe H.W. Evaluation of the distribution of enrofloxacin by circulating leukocytes to sites of inflammation in dogs. Am J Vet Res Jan:. 2009;70(1):16-22.
197. Carbon C. Clinical relevance of intracellular and extracellular concentrations of macrolides. Infection. 1995;23:S10-S14.
198. Lefebvre H.P., Schneider M., Dupouy V., et al. Effects of experimental renal impairment on disposition of marbofloxacin and its metabolites in the dog. J Vet Pharmcol Ther. 1998;21:453-461.
199. Boothe D.M., Boeckh A., Boothe H.W., et al. Plasma concentrations of enrofloxacin and its active metabolite ciprofloxacin in dogs following single oral administration of enrofloxacin at 7.5, 10, or 20 mg/kg. Vet Ther. 2002;3(4):409-419.
200. Heinen E. Comparative serum pharmacokinetics of the fluoroquinolones enrofloxacin, difloxacin, marbofloxacin, and orbifloxacin in dogs after single oral administration. J Vet Pharmacol Therap. 2002;25:1-5.
201. Abadıá A.R., Aramayona J.J., Munõz M.J., et al. Disposition of ciprofloxacin following intravenous administration in dogs. J Vet Pharmacol The. 1994;17:384-388.
202. Abadıá A.R., Aramayona J.J., Munõz M.J., et al. Ciprofloxacin pharmacokinetics in dogs following oral administration. J Vet Med. 1995;42:505-511.
203. Dalhoff A., Bergan T. Pharmacokinetics of fluoroquinolones in experimental animals. In: Kuhlman J., Dalhoff A., Zeiler H.J., editors. Quinolone antibacterials. Berlin: Springer Verlag; 1998:188-189.
204. Albarellos G., Montoya L., Ambros L., et al. Multiple once-daily dose pharmacokinetics and renal safety of gentamicin in dogs. J Vet Pharmacol Therap. 2004;27:21-25.
205. Albarellos G.A., Kreil V.E., Landoni M.F. Pharmacokinetics of ciprofloxacin after single intravenous and repeat oral administration to cats. J Vet Pharmacol Therap. 2004;27:155-162.
206. Miyamoto K. Use of plasma clearance of iohexol for estimating glomerular filtration rate in cats. Am J Vet Res. 2001;62(4):572-575.
207. Marchetti F., Viale P. Current and future perspectives for levofloxacin in severe Pseudomonas aeruginosa infections. J Chemother. 2003;15(4):315-322.
208. Fish D.N., Chow A.T. The clinical pharmacokinetics of levofloxacin. Clin Pharmacokinet. 1997;32(2):101-119.
209. Yoshida K., Yabe K., Nishida S., et al. Pharmacokinetic disposition and arthropathic potential of oral ofloxacin in dogs. J Vet Pharmacol Therap. 1998;21:128-132.
210. Albarellos G.A., Ambros L.A., Landoni M.F. Pharmacokinetics of levofloxacin after single intravenous and repeat oral administration to cats. J Vet Pharmacol Therap. 2005;28:363-369.
211. Intorre L., Mengozzi G., Maccheroni M., et al. Enrofloxacin-theophylline interaction: influence of enrofloxacin on theophylline steady state pharmacokinetics in the beagle dog. J Vet Pharmacol Ther. 1995;18:352-356.
212. Hirt R.A., Teinfalt M., Dederichs D. The effect of orally administered marbofloxacin on the pharmacokinetics of theophylline. J Vet Med Assoc. 2003;50:246-250.
213. Landor M., Iashinsky A., Waxman J. Quinolone allergy? Ann Allergy Asthma Immunol. 1996;77:273-276.
214. Davenport C.M., Boston R.C., Richards D.W. Enrofloxacin and magnesium deficiency on matrix metabolism in equie articular cartilage. Am M Vet Res. 2001;62:160-166.
215. Ergerbacher M., Edinger J., Tschulenk W. Effects of enrofloxcin and ciprofloxacin hydrochloride on canine and equine chondrocytes in culture. Am J Vet Res. 2001;63:704-708.
216. Stahlmann R., Forster C., Shakiboei M., et al. Magnesium deficiency induces joint cartilage lesions in juvenile rats which are identical to quinolone-induced arthopathy. Antimicrob Agents Chemother. 1995;39:2013-2018.
217. Ergerbacher M., Wolfesberger B., Gabler C. In vitro evidence for effects of magnesium supplementation on quinolone-treated horse and dog chondrocytes. Vet Pathol. 2001;38(2):143-148.
218. Huddleston P.M., Hanssen A.D., Rouse M.S., et al. Ciprofloxacin inhibition of experimental fracture-healing. The Fluoroquinolone Toxicity Research Foundation; 2003.
219. Shakibaei M., de Souza P., van Sickle D. Biochemical changes in achilles tendon from juvenile dogs after treatment with ciprofloxacin or feeding a magnesium-deficient diet. Arch Toxicol. 2001;75(6):369-374.
220. Halliwell R.F., Davey P.G., Lambert J.J. The effects of quinolones and NSAIDS upon GABA-evoked currents recorded from rat dorsal root ganglion neurons. J Antimicrob Chemother. 1991;27:209-218.
221. De Sarro A., Cecchetti V., Fravolini V., et al. Effects of novel 6-desfluoroquinolones and classic quinolones on Pentylenetetrazole-induced seizures in mice. Antimicrob Agents Chemother. 1999;43(7):1729-1736.
222. Lode H. Potential interactions of the extended-spectrum fluoroquinolones with the CNS. Drug Safety. 1999;21:123-135.
223. Cohn J.S. Peripheral neuropathy associated with fluoroquinolones. Ann Pharmacother. 34(1), 2001.
224. Gelatt K.N., Van der Woerdt A., Ketring K.L., et al. Enrofloxacin associated retinal degeneration in cats. Vet Ophthalmol. 2001;4:99-106.
225. Wiebe V., Hamilton P. Fluoroqinolone-induced retinal degeneration in cats. Am Vet Med Assoc. 2002;221:1508-1571.
226. Marutani K., Matsumoto M., Otabe Y., et al. Reduced phototoxicity of a fluoroquinolone antibacterial agen with a methoxy group at the 8 position in mice irradiated with long-wave length UV light. Antimicrob Agents Chemother. 1993;37:2217-2222.
227. Miller C.W., Prescott J.R., Mathews K.A., et al. Streptococcal toxic shock syndrome in dogs. J Am Vet Med Assoc. 1996;209:1421-1426.
228. Ingrey K.T., Ren J., Prescott J.F. A fluoroquinolone induces a novel mitogen-encoding bacteriophage in. Streptococcus canis, Infect Immun. 2003;71(6):3028-3033.
229. Baciewicz A.M., Chrisman C.R., Finch C.K., et al. Update on rifampin and rifabutin drug interactions. Am J Med Sci. 2008;335(2):126-136.
230. Ziglam H.M., Daniels I., Finch R.G. Immunomodulating activity of rifampicin. J Chemother. 2004;16(4):357-361.
231. Adachi J.A., DuPont H.L. Rifaximin: a novel nonabsorbed rifamycin for gastrointestinal disorders. Clin Infect Dis. 2006;42:541-547.
232. Phillips M.A., Stanley S.L.Jr. Chemotherapy of protozoal infections: amebiasis, giardiasis, trichomoniasis, trypanosomiasis, leishmaniasis, and other protozoal infections. In Brunton L.L., Lazo J.S., Parker K.L., editors: Goodman & Gilman’s the pharmacological basis of therapeutics, ed 11, New York: McGraw-Hill, 2006.
233. Muller M. Mode of action of metronidazole on anaerobic bacteria and protozoa. Surgery. 1983;93:165-171.
234. Levett P.N. Time-dependent killing effect of Clostridium difficile by metronidazole and vancomycin. J Antimicrob Chemother. 1991;27:55-62.
235. Karjagin J., Pähkla R., Karki T., et al. Distribution of metronidazole in muscle tissue of patients with septic shock and its efficacy against. Bacteroides fragilis in vitro, J Antimicrob Chemother. 2005;55:341-346.
236. Nix D.E., Tyrrell R., Muller M. Pharmacodynamics of metronidazole determined by a time-kill assay for Trichomonas vaginalis. Antimicrob Agents Chemother. 1995;39(8):1848-1852.
237. Neff-Davis C.A., Davis L.E., Gillette E.L. Metronidazole: a method for its determination in biological fluids and its disposition kinetics in the dog. J Vet Pharmacol Ther. 1981;4(2):121-127.
238. Sekis I., Ramstead K., Rishniw M., et al. Single-dose pharmacokinetics and genotoxicity of metronidazole in cats. J Feline Med Surg. 2009;11:60-68.
239. Fitch R., Moore M., Roen D. A warning to clinicians: metronidazole neurotoxicity in a dog. Probl Vet Neurol. 1991;2:307-309.
240. Heaney C.J., Campeau N.G., Lindell E.P. MR imaging and diffusion-weighted imaging changes in metronidazole (Flagyl)-induced cerebellar toxicity. Am J Neuroradiol. 2003;24:1615-1617.
241. Evans J., Levesque D., Knowles K. Diazepam as a treatment for metronidazole toxicosis: a retrospective study of 21 dogs. J Vet Intern Med. 2003;17:304-310.
242. Caylor K.B., Cassimatis M.K. Metronidazole neurotoxicosis in two cats. J Am Anim Hosp Assoc. 2001;37(3):258-262.
243. Olson E.J., Morales S.C., McVey A.S., et al. Putative metronidazole neurotoxicosis in a cat. Vet Pathol. 2005;42:665-669.
244. Davidson G. To benzoate or not to benzoate: cats are the question. Int J Pharm Compound. 2001;5:89-90.
245. Wu Y., Fassihi R. Stability of metronidazole, tetracycline HCl and famotidine alone and in combination. Int J Pharm. 2005;290:1-13.
246. Petri W.A.Jr. : Sulfonamides, trimethoprim-sulfamethoxazole, quinolones, and agents for urinary tract infections. In Brunton L.L., Lazo J.S., Parker K.L., editors: Goodman & Gilman’s the pharmacological basis of therapeutics, ed 11, New York: McGraw-Hill, 2006.
247. Hirsch D.C., Biberstein E.L., Jang S.S. Obligate anaerobes in clinical veterinary practice. J Clin Microbiol. 1979;10:188-191.
248. Hirsch D.C., Indiveri M.C., Jang S.S., et al. Changes in prevalence and susceptibility of obligate anaerobes in clinical veterinary practice. J Am Vet Med Assoc. 1985;186(10):1086-1089.
249. Indiveri M.C., Hirsch D.W. Susceptibility of obligate anaerobes to trimethoprim-sulfamethoxazole. J Am Vet Med Assoc. 1986;188:46-48.
249a. Kuo C.C., Wang S., Grayston J.T. Antimicrobial activity of several antibiotics and a sulfonamide against Chlamydia trachomatis organisms in cell culture. Antimicrob Agents Chemother. 1977;12(1):80-83.
250. Sharer W.C., Fair W.R. The pharmacokinetics of antibiotic diffusion in chronic bacterial prostatitis. Prostate. 1982;3(2):139-148.
251. Trepanier L.A., Ray K., Winand N.J., et al. Cytosolic arylamine N-acetyltransferase (NAT) deficiency in the dog and other canids due to an absence of NAT genes. Biochem Pharmacol. 1997;54(1):73-80.
252. Trepanier L.A. Idiosyncratic toxicity associated with potential sulfonamides in the dog. J Vet Pharmacol Therap. 2004;27:129-138.
253. Hall I.A., Campbell K., Chambers M.D., et al. Effect of trimethoprim-sulfamethoxazole on thyroid function in dogs with pyoderma. J Am Vet Med Assoc. 1993;202:1959-1962.
254. Trepanier L.A., Danhof R., Toll J., et al. Clinical findings in 40 dogs with hypersensitivity associated with administration of potentiated sulfonamides. J Vet Intern Med. 2003;17:647-652.
255. Chopra I. New developments in tetracycline antibiotics: glycylcyclines and tetracycline efflux pump inhibitors. Drug Resist Updat. 2002;5:119-125.
256. Westfall D.S., Twedt D.C., Steyn P.F., et al. Evaluation of esophageal transit of tablets and capsules in 30 cats. J Vet Intern Med. 2008;15(5):467-470.
257. Carlborg B., Densert O. Esophageal lesions caused by orally administered drugs. An experimental study in the cat. Eur Surg Res. 1980;12(4):270-282.
258. Apaydin B.B., Paksoy M., Art T., et al. Influence of pentoxifylline and interferon-alpha on prevention of stricture due to corrosive esophagitis. Eur Surg Res. 2001;33:225-231.
259. Saiman L., Liu Z., Chen Y. Synergistic activity of macrolide antibioitics paired with conventional antimicrobial agents against multiply antibiotic resistant P aeruginosa isolated from CF patients. Antimicrob Agents Chemother. 2002;46(4):1105-1107.
260. Park B.-K., Lim J.-H., Kim M.-S., et al. Pharmacokinetics of flor-fenicol and its metabolite, florfenicol amine, in dogs. Res Vet Sci. 2008;84:85-89.
261. Papich M.G., Riviere J. Chloramphenicol. In: Adams R., editor. Veterinary pharmacology and therapeutics. ed 8. Ames, Iowa: Iowa State University Press; 2001:868-897.
262. Watson A.D.J. Chloramphenicol in the dog: observations of plasma levels following oral administration. Res Vet Sci. 1974;16:147-151.
263. Glazko A.J., Wolf L.M., Dill W.A., et al. Biochemical studies on chloramphenicol (chloromycetin). J Pharm Exp Ther. 1949;96:445-449.
264. Watson A.D. Chloramphenicol 2. Clinical pharmacology in dogs and cats. Aust Vet J. 1991;68(1):2-5.
265. Watson A.D.J. Plasma chloramphenicol concentrations in cats after parenteral administration of chloramphenicol sodium succinate. J Vet Pharamcol Therap. 1979;2:123-127.
266. Watson A.D.J. Effect of ingesta on systemic availability of chloramphenicol from two oral preparations in cats. J Vet Pharmacol Therap. 1979;2:117-121.
267. Papich M.G. Florfenicol pharmacokinetics in dogs and cats (Abstract No. 235). Chicago, IL: 17th Annual Veterinary Medical Forum, American College of Veterinary Internal Medicine; June 10-13, 1999.
267a. Michel J., Bornstein H., Luboshitzky R., et al. Mechanisms of chlorampenicol-cephalordine synergism on Enterobacteiaeae. Antimicrob Agents Chemother. 1975;7(6):845-849.
268. Adams H.R., Dixit B.N. Prolongation of pentobarbital by chloramphenicol in dogs and cats. J Am Vet Med Assoc. 1970;156(7):902-905.
269. Sanders J.E., Yeary R.A., Fenner W.R., et al. Interaction of phenytoin with chloramphenicol or pentobarbital in the dog. J Am Vet Med Assoc. 1979;175(2):177-180.
270. Turco J.D. Pharmacokinetics and interactions of digoxin with phenobarbital in dogs. Am J Vet Res. 1987;48(8):1244-1249.
271. Boothe D.M., Brown S.A., Fate G.D., et al. Plasma disposition of clindamycin microbiologic activity in cats after single oral doses of clindamycin hydrochloride as either capsules or aqueous solution. J Vet Pharmacol Ther. 1996;19:491-494.
272. Lavy E., Ziv G., Shem-Tove M., et al. Pharmacokinetics of clindamycin HCl administered intravenously, intramuscularly, and subcutaneously to dogs. J Vet Pharmacol Therap. 1999;22:261-265.
273. Yamasaki T., Ichimiya T., Hirai K., et al. Effect of antimicrobial agents on the piliation of Pseudomonas aeruginosa and adherence to mouse tracheal epithelium. J Chemother. 1997;9:32-37.
274. Asaka T., Manaka A., Sugiyama H. Recent developments in macrolide antimicrobial research. Curr Top Med Chem. 2003;3:961-989.
275. Schlunzen F., Harms J.M. Structural basis for the antibiotic activity of ketolides and azalides. Structure. 2003;11:329-338.
276. Dorfman M.S., Wagner R.S., Jamison T., et al. The pharmacodynamic properties of azithromycin in a kinetics-of-kill model and implications for bacterial conjunctivitis treatment. Adv Ther. 2008;25(3):208-217.
277. Albarellos G.A., Kreil V.E., Ambros L.A., et al. Pharmacokinetics of erythromycin after the administration of intravenous and various oral dosage forms to dogs. J Vet Pharmacol Ther. 2008;31(6):496-500.
278. Garver E., Hugger E.D., Shearn S.P., et al. Involvement of intestinal uptake transporters in the absorption of azithromycin and clarithromycin in the rat. Drug Metab Dispos. 2008;36:2492-2498.
279. Shepard R.M., Falkner F.C. Pharmacokinetics of azithromycin in rats and dogs. J Antimicrob Chemother. 1990;25(Suppl A):49-60.
280. Hunter R.P., Lynch M.J., Ericson J.F. Pharmacokinetics, oral bioavailability and tissue distribution of azithromycin in cats. J Vet Pharmacol Ther. 1995;18(1):38-46.
280a. Haddad A., Davis M., Lagman R. The pharmacological importance of cytochrome CYP3A4 in the palliation of symptoms: review and recommendations for avoiding adverse drug interactions. Support Care Cancer. 2007;15(3):251-257.
281. Bizjak E.D., Mauro V.F. Digoxin-macrolide drug interaction. Ann Pharmacother. 1997;31(9):1077-1079.
282. Sugie M., Asakura E., Zhao Y.L., et al. Possible involvement of the drug transporters P glycoprotein and multidrug resistance-associated protein Mrp2 in disposition of azithromycin. Antimicrob Agents Chemother. 2004;48(3):809-814.
283. Carfartan G., Gerardin P., Turck D., et al. Effect of subinhibitory concentrations of azithromycin on adherence of Pseudomonas aeruginosa to bronchial mucins collected from cystic fibrosis patients. J Antimicrob Chemother. 2004;53:686-688.
284. Gillis R.J., Iglewski B.H. Azithromycin retards Pseudomonas aeruginosa biofilm formation. J Clin Microbiol. 2004;42(12):5842-5845.
285. Kourlas H. Anti-inflammatory properties of macrolide antibiotics. J Pharm Pract. 2006;19(5):326-329.
286. Amsden G.W. Anti-inflammatory effects of macrolides—an underappreciated benefit in the treatment of community-acquired respiratory tract infections and chronic inflammatory pulmonary conditions? J Antimicrob Chemother. 2005;55:10-21.
287. Lorenz J. Telithromycin: the first ketolide antibacterial for the treatment of community acquired respiratory infections. Int J Clin Pract. 2003;57:519-529.
288. Bozdogan B., Applebaum P.C. Oxazolidinones: activity, mode of action, and mechanism of resistance. Int J Antimicrob Agents. 2004;23:113-119.
289. Yagi B.H., Zurenko G.E. An in vitro time-kill assessment of linezolid and anaerobic bacteria. Anaerobe. 2003;9(1):1-3.
290. Ntziora F, Falagas ME: Linezolid for the treatment of patients with mycobacterial infections: a systematic review. Int J Tuberc Lung Dis. 2007;11(6):606-611.
291. Brown-Elliott B.A., Ward S.C., et al. In vitro activities of linezolid against multiple Nocardia species. Antimicrob Agents Chemother. 2001;45(4):1295-1297.
292. Smith A.J., Hall V., Thakker B., et al. Antimicrobial susceptibility testing of Actinomyces species with 12 antimicrobial agents. J Antimicrob Chemother. 2005;56(2):407-409.
293. Slatter J.G., Adams L.A., Bush E.C., et al. Pharmacokinetics, toxicokinetics, distribution, metabolism and excretion of linezolid in mouse, rat and dog. Xenobiotica. 2002;32(10):907-924.
294. Garrabou G., Soriano A., Lopez S., et al. Reversible inhibition of mitochondrial protein synthesis during linezolid-related hyperlactatemia. Antimicrob Agents Chemother. 2007;51(3):962-967.
295. Oleson F.B.Jr., Berman C.L., Kirkpatrick J.B., et al. Once-daily dosing in dogs optimizes daptomycin safety. Antimicrob Agents Chemother. 2000;44(11):2948-2953.
296. Rybníček J., Hill P.B. Suspected polymyxin B-induced pemphigus vulgaris in a dog. Vet Dermatol. 2007;18(3):165-170.
297. Spann C.T., Turtrone W.D., Weinberg J.M. Topical antibacterial agents for wound care: a primer. Dermatol Surg. 2003;29:620-626.