Drugs Affecting the Liver

Cholagogues and Choleretics

Substances that cause contraction of the gallbladder are called cholagogues. The resistance of the sphincter of Oddi decreases as bile flows freely into the duodenum. Dietary fat and concentrated magnesium sulfate introduced directly into the duodenum through a tube exert a cholagogue effect through release of CCK/pancreozymin from the upper small intestine. Vagus stimulation also promotes contraction of the gallbladder.

Substances that increase secretion of bile by the hepatocytes are known as choleretics. A drug that stimulates the liver to increase output of bile of low specific gravity is called a hydrocholeretic.

Bile acids are synthesized from cholesterol and secreted into bile. Their production is increased by stimulation of the vagus nerves and by the hormone secretin, which increases the water and bicarbonate content of bile. Physiologically, however, bile acids are mainly responsible for the “bile salt–dependent” component of bile secretion and flow. Bile salts secreted with bile are almost complete (95%) resorbed, undergoing enterohepatic circulation. Reabsorption occurs principally in the terminal ileum. The primary bile acids are cholic and chenodeoxycholic acids; secondary acids include deoxycholic and lithocholic acid; lithocholic acid and ursodeoxycholic acid (UDCA) represent less than 5% of the total bile salt components. The major salts of bile acids are glycine and taurine. Bile acids induce bile flow, inhibit (by feedback) cholesterol synthesis, and promote dispersion and absorption of lipids and fat-soluble vitamins.

A number of natural bile salts and several partially synthetic derivatives are used therapeutically as choleretics, including dehydrocholic acid, which is the most potent hydrocholeretic agent. Naturally occurring bile acid conjugates such as glycocholate and taurocholate enhance bile flow to a lesser extent. Bile salts used therapeutically have a dual action in directly promoting fat absorption and stimulating biliary secretion after they have been absorbed. Overdosage with these compounds tends to cause diarrhea. UDCA is a natural bile acid constituting a very small portion of the bile acid pool. It is a degradation production of chenodeoxycholic acid. Among the bile acids, UDCA has the lowest hydrophobic–hydrophilic balance, the lowest capacity to make micelles, and the least potential for cholestatic or cellular membrane toxicity. Because cholestatic liver disease may be associated with accumulation of toxic bile acids, treatment with UDCA is appealing. Its efficacy in a variety of chronic liver diseases has been established. Its mechanism of action is not well understood but is probably related to bile acid metabolism.171 The use of UDCA in dogs suffering from selected cholestatic liver diseases has been documented.172 The disposition of UDCA has been studied in healthy cats.173 Sporadic vomiting and diarrhea were reported, but otherwise the drug appears to be safe. Scientific studies establishing its safety and efficacy in diseased animals are indicated. Bile acids are effluxed from the canicular membrane into bial by a transport protein described as a “sister” to P-glycoprotein. However, selected bile acids, including UDCA, inhibit P-glycoprotein. 174 The therapeutic implications in patients with cholestasis or being treated with UDCA are not clear.

Liver Protectants and Hepatotropic Agents

A comprehensive review of the treatment and management of hepatic disease is beyond the purview of this chapter. However, a selection of the drugs for treatment of liver failure are listed and the rationale for their use noted. The major pharmacologic properties of many of these substances are discussed elsewhere in this volume. Hepatotropic agents are those having a special affinity for the liver or exerting a specific effect. Lipotropic agents hasten removal of fat or decrease its deposition in the liver. Use of lipotropic agents (choline, methionine, cysteine, betaine, lecithin, hydroxocobalamin) to increase mobilization of hepatic lipid is of proven value only in cases in which deficiencies of these substances exist. Deficiencies may be present in hepatic disease as a result of anorexia or insufficient dietary protein. Patients receiving and consuming a nutritious diet with adequate amounts of protein do not require supplementation with lipotropic agents, but their use has not been shown to be detrimental.

S-adenosyl-L-methionine

S-adenosyl-L-methionine (SAMe) is an endogenous coenzyme composed of ATP and the sulfur-containing amino acid methionine. The methyl group attached to the sulfur of methionine is chemically reactive such that it can be donated (i.e., transmethylation) to a number of acceptor substrates. Up to 80% of methionine in the body is used to form SAMe in the liver, which is the major, but not sole, site of methylation reactions. Regeneration of SAMe depends on vitamins B6 and B12 and folic acid.175 Transmethylation represents the primary function of SAMe, although sulfhydryl (transulfation) and aminopropyl (aminopropylation) groups also are donated. As such, SAMe is involved in more than 40 metabolic reactions in the body. Substrates are variable and include nucleic acids, proteins, and lipids. SAMe donates methyl groups to diverse compounds such as choline, creatine, carnitine, norepinephrine, DNA, and transfer RNA.

KEY POINT 19-38

Many hepatoprotectants either facilitate bile acid secretion or promote the activity of glutathione oxygen radical scavenging.

Aminopropylation yields compounds that affect cell and tissue repair. Transmethylation is important in detoxification, energy utilization, gene transcription and membrane functions that influence growth, and cellular signaling and adaptation. Transmethylation is necessary for formation of the phsopholipid bilayer of outer cell membranes and membrane fluidity. After donating its methyl group, SAMe is converted to S-adenosylhomocysteine, the precursor to transsulfuration reactions. Transsulfuration also is important, resulting in the formation of sulfur and thiolated compounds, glutathione (GSH; one of the principle antioxidants of the liver), cysteine (the rate-limiting substrate for GSH formation), and sulfates. Each is particularly important in detoxification and conjugation reactions, including bile conjugation and bile flow. Glutathione has many additional effects on cellular biology, including gene transcription, triggering proinflammatory cell signaling and apoptosis, and also is important as an intracellular scavenger of oxygen radical.

The importance of SAMe in GSH synthesis suggested a possible role in liver disease. Studies in humans have demonstrated that SAMe is associated with an increase in hepatic GSH in chronic liver disease. Bile flow increases, presumably in response to increased sulfate bioavailability; sulfated bile acids are more soluble than others. Lieber175 reviewed the role of SAMe in the body and its role in the treatment of liver disease in humans.176 In an original research report, Center and coworkers177 also reviewed the role of SAMe, with an emphasis in the liver. Multiple hepatic functions are dependent on GSH. Howver, SAMe formation from methionine decreases with severe liver disease as a result of downregulation of SAME synthetase. Sequelae include impairment of multiple hepatic functions as well as retention of methionine, which may contribute to hepatic insufficiency.

Because SAMe is unstable and is destroyed in the GI tract, it must be administered as an enteric-coated product. After absorption, oral bioavailability of SAMe is low on account of first-pass metabolism. Gender differences in oral bioavailability exist for SAMe in humans, with peak concentrations threefold to sixfold higher in women compared with men. SAMe is minimally bound to serum proteins (in humans) and is able to penetrate into and accumulate in cerebrospinal fluid.174

A case report178 described the successful treatment of acetaminophen overdose (1 g/kg) in a dog with SAMe and supportive care; treatment was not begun until 48 hours after ingestion. Center and coworkers177 examined the safety and effects of SAMe on the redox potential in red blood cells and the liver in normal, healthy cats (n=15) receiving 48 mg/kg of SAMe (Denosyl) orally once daily for 113 days. Plasma SAMe concentrations increased in concert with each dose. Peak concentrations approximating 1 to 1.5 μg/mL occurred between 2 to 4 hours after each dose; the disappearance half-life after oral dosing approximated 3 hours. Concentrations did not accumulate during the 113-day dosing period. Unmetabolized SAMe appears to be eliminated almost equally in urine and feces. In humans and normal cats, SAMe was not associated with adverse events. In clinically normal cats, the redox status of both red blood cells and hepatocytes was increased after 113 days of SAMe administration. Further, red blood cells became more resistant to osmotic lysis. Hepatic cysteine, GSH, and protein increased, the latter attributed to an anabolic effect of SAME. Bile flow also increased, and histologic improvement was noted in the seemingly normal cats that had asymptomatic nonsuppurative portal inflammation.

SAMe is among the supplements for which comparison of labeled and actual contents may not match. The hygroscopic nature of SAMe contributes to quality assurance issues in that improper formulation and storage can result in the loss of active ingredient. The lack of premarket approval suggests that consumers should take a proactive approach to ensuring the quality of purchased products. Manufacturers appear to have improved the predictability of high-quality product in that eight of eight manufactured products tested by Consumer Laboratories and reported on its website in 2007 passed evaluation (as of January 2010). This compares to 2003, in which one product was found with only 30% of its listed amount, and in 2000, when close to 50% of tested products contained less than the labeled content (www.consumerlab.com). Enteric-coated products are available to protect SAMe from GI acidity; among the tests implemented by Consumer Laboratories is assurance of proper dissolution of enteric-coated products.

KEY POINT 19-39

The hygroscopic nature of SAMe may result in poor-quality product, and efforts should be taken to prescribe a product of known quality.

SAMe is available in several salt forms, each of which weighs a different amount and thus contributes variably to the total weight in mg. Care should be taken to dose on the active ingredient (i.e., SAMe). Salts include tosylate, disulfate tosylate, disulfate ditosylate, and 1,4-butanedisulfonate (Denosyl). For example, 200 mg of S-adenosyl-methionine disulfate tosylate contains only 100 mg of SAMe. The salt name should be included on the label as part of the chemical name, and the label should indicate the amount of active ingredient. SAMe also can be reviewed in the Herb and Plant Supplement at consumerlab.com179

SAMe may be involved in a number of drug–diet interactions. Because it inhibits uptake of serotonin, any other compound that does likewise should be avoided or used cautiously. This includes antidepressant behavior-modifying drugs such as monoamine oxidase inhibitors, selective serotonin reuptake inhibitors, tricyclics, tramadol, and selected herbs (such as St. John’s wort).

Animal studies and clinical human studies indicate that SAMe improves biochemical parameters of liver function.180

Endogenous concentrations are reduced in patients with cirrhotic liver disease. Production of sulfated compounds and phosphatidylcholine subsequently is reduced. In animal studies SAMe improved bile secretion impaired by a variety of toxins and by pregnancy. Drug-induced hepatotoxicity and chronic liver disease were also reduced, without occurrence of serious side effects.176 SAMe may act synergistically with UDCA for treatment of chronic progressive liver disease.181 Although absorbed well after oral administration, SAMe undergoes extensive first-pass metabolism.180 The compound is so hygroscopic that it is unstable unless protected; it might be most.

prudent to use products in bubble packets. Tablets cannot be broken without risking loss of efficacy. Caution is recommended when purchasing SAMe; an independent investigation that compared the content of SAMe with the labeled amounts found 6 of 13 products to be mislabeled. SAMe is marketed with silymarin (discussed in the next section) by Nutramax Laboratories for use in animals.

Milk Thistle

Silymarin has been reviewed by the Herb and Plant Supplement Encyclopedia at consumerlab.com. A member of the daisy family (Asteraceae), silymarin is one of the most important medical constituents of ripe seeds of the blessed milk thistle plant (Silybum marianum). The name reflects the legend that attributes the white leaf veins to a drop of the Virgin Mary’s milk and the location of the hidden infant Jesus during the family’s flight from Egypt. Other synonyms for milk thistle include Marian thistle, Mary thistle, St. Mary’s thistle, Lady’s thistle, Holy thistle, sow thistle, thistle of the blessed virgin, Christ’s crown, Venus thistle, heal thistle, variegated thistle, and wild artichoke.

Silymarin is a combination of seven flavonolignans (a flavanoid). The most prevalent is silybinin (silybin), which itself is two different compounds, with others including silychristin (or silicristin), and silydianin (also silidianin). Although all parts of the plant have been used medicinally,182 the concentration of silymarin is highest in the seeds and leaves. Generally, the flavonoids are extracted. Silybin is often complexed with phosphatidylcholine, which improves its bioavailability. Dosing should be based on silybinin equivalents. As with flavonoids, the active ingredient of silymarin are potentially potent antioxidants, scavenging hydroxyl radicals, superoxide anions, and lipid oxygen radicals owing to lipid peroxidation.

Animal models (including tetrachloromethane hepatotoxicity in dogs and a variety of other hepatotoxicants) support a hepatoprotective effect. Its mechanism of hepatoprotection is not known. Suggested mechanisms include increased hepatic regeneration (by stimulating RNA polymerase, ribosomal RNA, and potentially DNA synthesis), scavenging of oxygen radicals, “stabilization of ” hepatocyte cell membranes, and competitive inhibition of toxins that might otherwise bind to hepatic and damage cell membrane receptors (e.g., Amanita phalloides). In rat and mice models of acetaminophen toxicosis, silymarin was associated with higher concentrations of hepatic glutathione and superoxide dismutase in experimental compared with control animals. Silymarin can also bind to steroid receptors, although the clinical relevance of this is not clear. Antifibrotic effects have been demonstrated in animal models. Finally, antinflammatory effects may reflect inhibition of lipoxygenase and subsequent leukotriene synthesis.182

KEY POINT 19-40

SAMe and milk thistle are among the few dietary supplements for which evidence of efficacy for treatment or prevention of liver disease exists.

Silymarin is poorly water soluble, and concentrated products are considered necessary for effective absorption. According to Consumer Laboratories, most studies involving silymarin are based on extract products that contain 70% to 80% silymarin on a weight basis. However, some products composed of seed powder contain only 1.5% silymarin. Quality assurance is likely to be an issue with silymarin: of eight products tested by Consumer Laboratories, only two contained the labeled amount. The remaining seven products ranged from 19% to 85% (median 64%). When properly labeled, the source of milk thistle (generally the seed) should be noted on the product. Care must be taken when dosing on the basis of label information; doses range from 15 to 1200 mg, reflecting in part the source and type of preparation. For example, pills made from seed powder contain about 9 to 15 mg of silymarin compared with 112 to 240 mg for pills made from dry extracts. Differences in doses may also reflect different salt preparations. In humans the phosphatidylcholine preparation may be more orally bioavailable, leading to a dose that is lower (1.5 to 3 mg/kg twice daily) compared with a 70% extract (3 mg/kg twice to thrice daily).

As a flavonoid, silymarin is an inhibitor of P-glycoprotein and should be used cautiously with drugs known to serve as substrates for the transport protein. Silymarin may also inhibit selected CYP450 (specifically P450 2C9) enzymes.

Milk thistle is used to prevent or treat a number of medical conditions, including hepatitis (acute or chronic), and to protect the liver from toxicants, including medications. Milk thistle is the most common alternative medicine used in humans for treatment of liver diseases, with more than 50% of users claiming efficacy. Other proposed health benefits of silymarin include improved diabetic control. An intravenous preparation of silybinin is available in Europe for treatment of mushroom poisoning caused by A. phalloides. Note that as of January 2010, of the 10 milk thistle products tested by Consumer Laboratories, nine did not pass, the primary reason being that the actual amount was less than the label claimed amount.

Miscellaneous Vitamins, Minerals, and Nutrients

Choline

Choline is an indispensable metabolite of the body. It forms part of a number of endogenous compounds, particularly phospholipids. Phosphatidylcholine, lysophospholipids, plasmalogens, and sphingomyelins are phospholipids that contain choline. The mode of action of choline as a lipotropic agent is unknown. It may promote conversion of liver fat into choline-containing phospholipids, which are more rapidly transferred from the liver into blood. Choline is also essential for synthesis of phospholipids that are used in intracellular membranes concerned with lipoprotein synthesis. It is thought that the lipotropic agents methionine, betaine, and lecithin are beneficial because they contain choline or promote choline synthesis. The requirement for choline is well recognized in all conditions predisposing to fatty infiltration of the liver, including diabetes mellitus, malnutrition, and cirrhosis. Greater than normal quantities of choline seem to be necessary for prevention of a fatty liver when the liver is already damaged. Choline deficiency is not the only cause of fatty liver in these conditions, nor will choline supplementation alone restore the liver to full functional competence. Choline is, however, extremely valuable in the multitherapeutic approach to prevention and cure of fatty liver.

Selenium and Vitamin E

Selenium is now known to be essential for tissue respiration and is protective against dietary hepatic necrosis. It is extremely active and is only required in minute amounts. Vitamin E enhances the action of selenium, but both are required.

Selenium is an essential component of glutathione peroxidase, which catalyzes oxidation of reduced glutathione:


image


This glutathione peroxidase catalyzes removal of hydrogen peroxide and fatty acid hydroperoxides and thus exerts a protective effect on all cells but especially on muscle, liver, and erythrocytes. The essential substances required for removal of peroxides are reduced glutathione and glutathione peroxidase. Vitamin E maintains glutathione in the reduced form by preventing formation of hydroperoxides; it is an antioxidant and thus reduces the amount of glutathione peroxidase required. Cysteine (N-acetylcysteine) is required for the reduced sulfhydryl radical of glutathione and is generally present in adequate quantities. Selenium and vitamin E enhance each other’s action and together protect cells, especially hepatocytes, against harmful buildup of peroxides.

Vitamins

In the presence of liver disease, the fat-soluble vitamin K should be supplemented because hepatic stores may be quite rapidly depleted. The water-soluble vitamins of the B-complex group are frequently employed in therapeutic regimens for hepatic insufficiency. Few controlled studies have been carried out in this regard, but the rationale behind their clinical use is based on ensuring an adequate supply of metabolic cofactors.

Hydroxocobalamin (previously called vitamin B12) is stored in the liver, mainly in mitochondria, but there is also a microsomal fraction. This microsomal vitamin may be of importance in hepatic protein metabolism. Microsomal cell fractions from the livers of hydroxocobalamin-deficient animals are defective in the incorporation of methionine and alanine into protein. General liver protein synthesis is depressed in hydroxocobalamin deficiency.

Hydroxocobalamin has a lipotropic effect. It is involved in metabolism of labile methyl groups and in formation of choline. Hydroxocobalamin is also necessary for overall utilization of fat. When intake is low, however, the demand for this vitamin in hematopoiesis exceeds that for any other clinically recognizable physiologic function.

Glucose and Fructose

The liver resists many forms of injury when its stores of carbohydrate and protein are adequate; its efficiency is impaired when hepatocytes are laden with fat. Administration of a hypertonic solution of glucose and fructose produces favorable responses in a variety of hepatic abnormalities. A high glycogen content appears to protect liver cells from damage, and inhibition of gluconeogenesis (which occurs with administration of both insulin and glucose) may play an important role. Under the influence of insulin, hepatocytes undergo glycogen storage, hypertrophy, and hyperplasia. Insulin has a major anabolic effect on the liver.

Treatment of Specific (Non-Infective) Disorders of the Gastrointestinal Tract

Treatment of gastrointestinal disorders frequently involves drugs that target the disease rather than the gastrointestinal tract. Enhanced discussion of such conditions or the drugs used to treat them might be found in their respective chapters (i.e., Treatment of Bacterial Infections [Chapter 8], Glucocorticoids [Chapter 30] or Immunomodulators [Chapter 31]). Treatment of specific infectious diseases also will be found in chapters that address treatment of the causative organisms.

Diseases of the Oral Cavity

The primary treatment for the feline eosinophilic granuloma complex is glucocorticoids,183 given orally (prednisone, 1 to 2 mg/kg twice daily), subcutaneously (methylprednisolone acetate, 20 mg every 2 weeks), or intralesionally (triamcinolone, 3 mg weekly). Progestational compounds such as methylprogesterone may prove beneficial, but side effects associated with long-term use (including hyperadrenocorticism and diabetes mellitus) should limit their use to cases that have not responded to any other (properly administered) drug therapy. Up to 50% of treated cats may relapse. Because of the potential impact of leukotrienes on eosinophil trafficking (see the section on inflammatory bowel disease), leukotriene receptor antagonists might be considered. Stomatitis may be a reflection of an autoimmune skin diseases, renal disease, microbiologic infection (viral, bacterial, or fungal) or may be idiopathic. Antimicrobial therapy should be considered in cases of idiopathic stomatitis; therapy may be necessary on a chronic, intermittent basis. Drugs should target anaerobic organisms (e.g., metronidazole, a penicillin derivative, or clindamycin). Glucocorticoid therapy should be used cautiously in stomatitis unless an autoimmune disorder has been diagnosed or other causes (including infectious ones) have been ruled out.

Diseases of the Esophagus

Megaesophagus

Myasthenia gravis is the most common cause of secondary megaesophagus in dogs. It is diagnosed on the basis of response to edrophonium chloride, a short-acting anticholinesterase.184 Effects of the drug on skeletal muscle occur within 1 minute of intravenous administration and last up to 10 minutes or longer in some myasthenic patients. Drug therapy of megaesophagus associated with myasthenia gravis targets improvement in muscular activity with anticholinesterase therapy (pyridostigmine bromide, 1 to 3 mg/kg orally every 12 hours) and suppression of the immune response with glucocorticoid or other immunosuppressive therapy (e.g., azathioprine). Pyridostigmine improves appendicular muscle strength but may not improve pharyngeal or esophageal function. Prednisone tends to be the preferred glucocorticoid but may contribute to muscle weakness. Azathioprine does not have many of the side effects of glucocorticoids, but remission takes longer to achieve (up to several weeks), and neutropenia may limit treatment. Mycophenolate mofetil is a lymphocyte-inhibiting immunomodulator used orally to prevent graft-versus-host rejection in human renal transplant patients. The drug inhibits purine synthesis but only in lymphocytes (both B and T lymphocytes), and side effects are limited to GI upset. The drug can be given orally, causing response within 4 hours of administration. The drug has proved efficacious for treatment of myasthenia gravis in dogs (5 to 10 mg/kg every 12 hours orally).185 Thyroid hormone replacement (thyroxine) may also prove helpful. Treatment for megaesophagus for which an underlying cause cannot be found is difficult. A number of drugs that stimulate GI smooth muscle have been recommended, including metoclopramide and cisapride, with varying reports of success. Neither of these two prokinetic drugs are likely to be effective in the striated muscle of the esophagus; further, enhanced lower esophageal sphincter tone might impede esophageal emptying. Bethanechol may stimulate propagating contractions in selected dogs. Drugs that relax the lower esophageal sphincter (anticholinergics and calcium channel blockers) have not proved effective. A major focus for treatment of myasthenia gravis is prevention of aspiration pneumonia and treatment or prevention of esophagitis.

Esophagitis

Esophagitis should be treated by correction of the underlying etiology. It is commonly associated with ingestion of a corrosive or hot (thermally) material. The feline esophagus appears to be particularly susceptible to drug-induced esophagitis (discussed in the following section). Antibiotic therapy in such cases should be reserved for esophageal perforation. The mucosa can be protected by administration of sucralfate administered as a slurry (1 g in 10 mL warm water), 5 to 10 mL every 6 to 8 hours. Lidocaine solution (Xylocaine viscous solution) may be administered orally (2 mg/kg every 4 to 6 hours) to minimize pain. Esophagitis caused by gastroesophageal reflux should respond well to medical management. In addition to protecting the damaged mucosa with sucralfate, drug therapy targets increasing gastric pH and tightening the lower esophageal sphincter. Antisecretory drugs (e.g., famotidine, omeprazole) help minimize damage induced by gastric acid and pepsin. Among the antihistaminergic drugs, ranitidine and nizatidine have prokinetic activity in humans comparable to that of cisapride, an effect evident within 1 hour after administration.128 Interestingly, a peppermint–caraway oil preparation induced relief from dyspepsia equal to that produced by cisapride in human patients.186 Antacids may also prove beneficial; products containing alginic acid may provide additional protection of the esophagus by providing a barrier of foam. Prokinetic drugs should be administered to tighten the lower esophageal sphincter. Indeed, metaclopramide or cisapride is probably as effective as antisecretory drugs in preventing further esophageal damage associated with gastric reflux. Glucocorticoids can be used to minimize esophageal stricture formation resulting from damage to the esophagus that extends into the muscular layers. Therapy should include tapered doses of glucocorticoids followed by reevaluation at 2-week intervals. Pentoxifylline may be an alternative or adjuvant therapy, particularly for drug-induced esophagitis.187

Gastroesophogeal Reflux

Up to 60% of humans are considered to be at risk for developing perioperative gastroesophogeal reflux (GER), whereas GER was demonstrated in 17% to 55% of healthy dogs. The difference in incidence of GER between these studies might be attributable to differences in the anesthetic agents used.

Metaclopramide prevents GER in unanesthetized dogs, as was demonstrated experimentally.188 Wilson and coworkers189 also prospectively studied the effect of metoclopramide on GER (defined when esophageal pH to <4 or an increase to >7.5 that lasted more than 30 seconds) in anesthetized dogs. Animals were undergoing orthopedic surgical procedures; anesthetic protocols (morphine and acepromazine as premeds and thiopental for induction) and surgical positioning were the same in treatment and placebo (saline) groups. Blinded and randomization methods were not clear. The authors determined, after reviewing the medical records of dogs undergoing the same surgical procedures and anesthetic protocols, that 55% of dogs experienced GER. A high dose (1 mg/kg followed by constant-rate infusion [CRI] 1 mg/kg/hr) but not a low dose (0.4 mg/kg followed by 0.3 mg/kg/hr CRI) of metoclopramide reduced the relative risk of GER by 54%. The timing of metoclopramide administration in relation to anesthesia was not provided. Vomiting occurred preoperatively in 55% of animals but was not a predictive factor for the risk of GER. No adverse effects attributed to treatment were identified. Preoperative vomiting in response to preanesthetic agents was not a risk factor for GER in dogs, whereas the duration of surgery (but not anesthesia) tended to be a risk factor.190 Wilson and coworkers190 compared the effects of pre-anesthetic morphine to mepiridine with or without acepromazine in healthy dogs (n = 30) using a randomized design. When compared with morphine, treatment with meperidine alone or with acepromazine before anesthesia was associated with a 55% and 27% risk reduction in risk of developing GER. In this same study, GER was detected in 51 of 90 dogs within 36 minutes of induction isoflurane (n = 14), halothane (n = 19) or sevoflurane (n = 18).

Esophogeal Motility and Oral Medications

The association of doxycycline administration with esophageal lesions in cats191,191a has led to several studies focusing on passage of medications through the feline esophagus. In one study, approximately 50% of capsules were retained in the midcervical esophagus of cats for longer than 240 seconds. Trapped capsules passed into the stomach if a small amount of food was administered.192 A prospective study used fluoroscopy to document the esophageal transit of barium tablets (20 mg) or capsules (size 4) in cats.193 The percentage of swallows needed for successful passage (movement of the medication into the stomach) of each medication was determined when administered with or without a follow-up bolus of water (6 mL). Success was only 36.7% 5 minutes after administration, compared with 90% at 30 seconds and 100% at 90 seconds if the medication was followed with a water bolus. The study was performed in a nonrandomized fashion, with the “wet” study always following 5 minutes after a “dry” study. Retention of dry medicaments was generally in the cervical esophageal region.193 Treatment should consist of an antisecretory drug, sucralfate, and metoclopramide. An anti-inflammatory may be helpful; glucocorticoids have been used, but pentoxyfylline might also be considered.

Diseases of the Stomach

Acute Gastritis

Acute gastritis is best treated by resolution of the underlying cause, whether it is diet, infectious agents or chemicals (including drugs or toxins), or metabolic diseases (e.g., renal or liver disease). Chemicals, including hydrochloric acid and bile acids, can induce vomiting as a result of direct damage or hypertonicity. Inflammation, if allowed to progress, can result in erosion and ulceration. Although most patients with acute gastritis improve in 1 to 5 days, some patients require supportive therapy.194 Depending on the patient, supportive therapy (in addition to nothing given orally) may include fluid therapy, antiemetics, and protectants or adsorbents. Fluid therapy with balanced crystalloids may require the addition of potassium. Bicarbonate is rarely indicated; glucose supplementation may be indicated in some patients. Any of the antiemetics previously discussed can be used, although phenothiazine derivatives should be withheld until volume replacement has begun. Metaclopramide is useful when given either peripherally or centrally; maropitant is likely to be very effective. Among the protectants, bismuth subsalicylate has proved most useful in decreasing vomiting associated with acute gastritis. However, gastric distention from the drug may cause the animal to vomit; therefore prudence is indicated in its use.

Gastric Ulceration and Erosion

There is no sensitive indicator of damage to the GI mucosa; damage may be quite extensive before hematemesis or melena is noted. Damage to the GI mucosa (erosion or ulceration) probably occurs more frequently than anticipated. For example, up to 25% of human patients admitted to intensive care units have gastric erosions; by the third day of hospitalization, this number increases to 90%. The risk of translocation of enteric pathogens is increased in these patients, suggesting an important role for prophylactic anti-ulcer therapy. Sucralfate has been recommended as the preferred method of prophylaxis in patients in whom enteral nutrition is not possible.195 Diseases in which mucosal damage should be anticipated and treatment implemented include but are not limited to mast cell disease, renal disease, liver disease, and IBD. The underlying cause of ulceration must be resolved; additionally, antisecretory drugs and antacids are indicated for treatment of mucosal damage.

Nonsteroidal antiinflammatory drugs

The most commonly recognized cause of GI ulceration in dogs is probably use of NSAIDs. The primary mechanism of ulceration by NSAIDs is inhibition of both the constitutive and inducible forms of cyclooxygenase, the enzyme responsible for formation of the cytoprotective prostaglandin E (see Chapter 29). The latter (inducible) enzyme is particularly important for healing in the damaged gastrointestinal mucosa. Alteration of ion (probably hydrogen) transport across the mucosa also has a variable role in ulcer formation, depending on the NSAID used (e.g., aspirin).196 Ulcerogenic drugs (e.g., glucocorticoids, which also inhibit the inducible cycclooxygenase) but potentiate ulceration caused by NSAIDs. Treatment of NSAID-induced GI ulceration includes discontinuation of the drug; replacement of missing prostaglandins by administration of cytoprotectants such as misoprostol; providing cytoprotection through sucralfate; and inhibiting acid secretion by administration of an antisecretory drug (e.g., ranitidine or omeprazole). Sucralfate, misoprostol, H2-receptor antagonists and proton-pump blockers have been studied either as sole agents or in various combinations for their ability to prevent GI ulceration in patients requiring high doses or long-term NSAID therapy. Among them, misoprostol probably provides the most consistent protection, followed by sucralfate and then antisecretory drugs. Although famotidine was found to be equal in efficacy to misoprostol for treatment of NSAID-induced ulceration in humans,197 the H2-receptor blockers are described as having only limited efficacy, particularly for ulcers in the stomach, unless it is used at higher than (generally twice) recommended doses198 Proton pump inhibitors have proved superior to H2-receptor antagonists for treatment of NSAID-induced ulceration.198

KEY POINT 19-41

Sucralfate has been associated with effective prevention of ulcers associated with nonsteroidal antiinflammatory drugs and stress.

The effect of misoprostol (3 mg/kg orally every 8, 12, or 24 hours) on gastric injury induced by aspirin (25 mg/kg orally every 8 hours) was studied prospectively in normal dogs (n = 24; 6 dogs per group) using a parallel, placebo-controlled design. Animals were dosed with aspirin and either placebo or misoprostol, the latter at 8- or 12-hour intervals, for 28 days with gastroscopy performed on -9, 5, 14, and 28 days. Visible lesions were scored on a scale of 1 (mucosal hemorrhage) to 11 (perforating ulcer). Median total scores between the placebo or misoprostol at 24 hours were significantly greater than scores in the group receiving misoprostol at 8-or 12-hour intervals. In contrast, no differences were measured between placebo and misoprostol at 24-hour intervals. Differences were not detected among groups for vomiting, diarrhea, or anorexia.199

In cases of NSAID overdosing (including accidental ingestion), prophylactic administration of both sucralfate and misoprostol is recommended along with an antisecretory drug. The duration of antiulcer therapy depends in part on the elimination half-life of the drug and the amount of NSAID ingested. The elimination half-life of some of the drugs is several days, suggesting that toxic concentrations may remain in the bloodstream for some time (1 to 2 weeks). Omeprazole may prolong the half-life of the NSAID, suggesting that treatment duration should err on the side of too long rather than too short. Prevention of NSAID-induced ulceration in humans is best accomplished with proton pump inhibitors; omeprazole was superior to either ranitidine or misoprostol after 6 months of NSAID therapy for reducing the risk of either gastric or duodenal ulcer.200 Omeprazole also may be the preferred choice in dogs; at least twice daily dosing also is recommended for famotidine. 87 Because of its potential to impair drug metabolism, omeprazole might be less desirable than the antihistaminergic antisecretory drugs when preventing ulcers in order to avoid prolonging NSAID half-life. Administration of cytoprotective agents for treatment of ulcers depends on the drug and the amount ingested. In general, treatment should extend at least 1-2 weeks after the inciting NSAID has been eliminated (i.e., at least 5 half-lives of the inciting NSAID or longer in the case of accidental ingestion that saturates drug metabolizing enzymes). Administration of cytoprotective antacids such as magnesium–aluminum hydroxide combinations with a meal may provide further protection.

Stress ulcers

A European meta-analysis (Mantel–Haenszel test) found that sucralfate was superior to H2 antisecretory drugs and equal to antacids in the prevention of macroscopic stress bleeding in long-term ventilated patients in an intensive care unit. Further, the incidence of pneumonia (caused by nosocomial organisms) was less in those ventilated patients receiving sucralfate compared with antacids or antisecretory drugs.201

The efficacy of omeprazole (20 mg orally once daily) in prevention of exercise-induced gastric ulceration in Racing Alaskan Sled Dogs (three teams of 16) was tested using a randomized placebo design.202 Response was based on endoscopic scoring of the gastric mucosa after completion of the race (0=least, 3=numerous bleeding ulcers). Mean score was significantly less (0.65) for treatment compared with placebo (1.09), although diarrhea was worse in the treatment group (54%) compared with placebo (21%). The percentage of animals that developed ulcers between the two groups was not reported. Williamson and coworkers203 subsequently reported on the efficacy of famotidine in this study.

Helicobacter species

H. pylori infects over 50% of the global human population. Infection will persist for life, unless specifically treated. All infected persons develop gastritis, with 15% of infected persons developing peptic ulceration. Infection also is associated with an increased risk of gastric adenocarcionoma and mucosa-associated lymphoid tissue lymphoma; indeed, it is classified as a class 1 carcinogen by the World Health Organization.209a The role of Helicobacter species as causal agents in GI diseases in dogs and cats is being elucidated. It is likely that a causal relationship exists between the organism and the disease.

A causal relationship also has been suggested in ferrets, cheetahs, and cats. Clinical signs attributed to Helicobacter organisms in dogs and cats include chronic vomiting and diarrhea, inappetence, pica, and fever.210 More than 70% of human patients with GI ulceration associated with Helicobacter organisms are cured of their ulcerative disease if Helicobacter is successfully eradicated. The pathophysiology of Helicobacter in part reflects urease-mediated conversion of urea to ammonia and bicarbonate. Ammonia causes local tissue damage, whereas bicarbonate appears to facilitate deeper colonization of the organism into the mucosa.210 Cytokine production by the organism appears to be associated with inflammation and ulcerogenesis; biochemical changes in the mucosa also contribute to disease. Treatment includes a colloidal bismuth (i.e., bismuth subsalicylate), an antibacterial that targets Helicobacter (metronidazole, amoxicillin, or clarithromycin), and an antacid (an H2-receptor antagonist or omeprazole). Bismuth accumulation causes cell wall damage and subsequent cell lysis. Among the antibacterials selected, amoxicillin is associated with the greatest and clarithromycin with the least amount of microbial resistance in humans. Resistance to metronidazole is also increasing. The duration of therapy in humans is several weeks. Reports of similar therapy in dogs and cats support but do not conclusively prove this approach may be beneficial in dogs and cats suffering from selected GI diseases. One study reported marked improvement in 90% of dogs and cats treated with a combination of metronidazole, amoxicillin, and famotidine for 3 weeks. Sucralfate may also be of benefit.

Gastric Dilatation–Volvulus

Therapy for gastric–dilatation volvulus (GDV) focuses on management of this acute and potentially life-threatening disease and long-term prevention. Medical management of the patient with acute disease focuses on resolution of the dilatation or volvulus and treatment of the sequelae of the syndrome. The sequelae of GDV that are most life-threatening are decreased cardiac preload (compression of the posterior vena cava and hepatic portal systems by the enlarged stomach), ischemia of the gastric wall (with loss of the mucosal barrier and increased risk of perforation), and congestion of abdominal viscera with subsequent endotoxemia and disseminated intravascular coagulation. Although they are potentially later in onset, cardiac arrhythmias also may become life-threatening. Shock should be treated with a balanced crystalloid electrolyte solution or hypertonic saline. Shock doses of glucocorticoids (methylprednisolone) may ameliorate some of the signs or clinical sequelae of endotoxemia although evidence for such use is lacking. Free radical scavengers (deferoxamine or allopurinol) may help reduce damage caused by reperfusion. Methylprednisolone may also be helpful in minimizing the effects of oxygen radicals The impact of prophylactic lidocaine on reperfusion injury was retrospectively studied in dogs (n = 51; 47 nontreated dogs served as control). Animals receiving lidocaine for cardiac arrhythmias were excluded from study. Dogs received either a loading dose (2 mg/kg intravenously) followed by CRI or a CRI alone (0.05 mg/kg/min) for at least 3 hours. No difference was detected in survival or complications between the two groups, although hospitalization was longer in the treatment group.204

Decompression of the dilated stomach can be facilitated by chemical restraint or sedation. Oxymorphone may be the drug of choice. Cardiac arrhythmias are most commonly ventricular in origin but may include atrial arrhythmias such as fibrillation. Intravenous lidocaine is the preferred drug for ventricular arrhythmias, administered initially as an intravenous bolus followed by a CRI (75 μg/kg/min). Procainamide can be used (intravenously, including CRI if lidocaine is ineffective; mexiletine) may be preferred. Amiodarone may also be used to acutely treat atrial fibrillation. Use of anti-secretory drugs is recommended to minimize the effects of hydrochloric acid on the already damaged mucosa. Although antimicrobials may be indicated, their use ideally is limited to situations in which contamination is known (i.e., surgical) or translocation is likely. Corrective surgery, including gastropexy, may be indicated. Medical management of chronic GDV has not been well established. Motility modifiers (metaclopramide, cisapride) and H2-receptor antagonists may be indicated, particularly after an acute episode, to minimize the accumulation of gastric secretions. However, their efficacy has not been established.

Gastric Motility Disorders

Dietary management of delayed gastric emptying should precede pharmacologic management. Underlying causes, including electrolyte abnormalities, should be corrected, and use of concurrent drugs (e.g., anticholinergics, alpha-adrenergics, opioid antagonists) that might alter motility or response to prokinetic agents should be discontinued. Prokinetic agents can be added when dietary management fails; cisapride is preferred.

Vomiting

For many causes of vomiting, studies regarding the etiology, prevention or treatment in animals are lacking. Accordingly, much of the information is drawn from the human literature. The discussions are offered as points of consideration, recognizing that relevance to dogs or cats remains to be demonstrated.

Chemotherapy-induced Emesis

Ellebaek and Herrstedt205 reviewed the prevention and treatment of nausea associated with chemotherapy in humans. The relative emetogenic potential of antineoplastic agents was described. In humans level 1 agents (lowest potential; <10% of patients) include chlorambucil, hydroxyurea, interferon α, tamoxifen, and vincristine; level 2 (10% to 30%): asparaginase, fluorouracil, gemcitabine, melphalan, paclitaxel, and thiopeta; level 3 (30% to 60%): cyclophosphamide (at <600 mg/m2), doxorubicin, methotrexate (<1000 mg/m2), and mitomycin; level 4 (60% to 90%): busulfan, carboplatin, cyclophosphamide (≥600 mg/m2), doxorubicin (>60 mg/m2), methotrexate (≥1000 mg/m2), and mitoxantrone; and level 5, the highest (>90% incidence): cisplatin (≥60 mg/m2), cyclophosphamide (1000 mg/m2), pentostatin, and others.205a

Chemotherapy-induced vomiting appears to reflect the release of serotonin from enterochromaffin cells of the GI mucosa with subsequent stimulation of type 3 vagal afferent 5-HT3 serotonin receptors in the mucosa, the nucleus tractus solitarius of the medulla oblongata, and the CRZ. At the CRZ, D2, opioid, and 5-HT3 receptors stimulate vomiting. Neurokinin receptors in humans are responsible for the delayed phase of vomiting associated with cisplatin anticancer chemotherapy. Maropitant appears to be effective for control of chemotherapy-induced emesis in dogs.205a,b Rau and coworkers demonstrated the efficacy of maropitant (2 mg/kg PO once daily) for control of vomiting associated with doxorubicin administration in dogs (n = 25) with cancer. Dogs were studied using a randomized, placebo-controlled study (n = 24) for 5 days posttreatment. Both vomiting and diarrhea were significantly less in the treatment group.205a D2 antagonists, such as phenothiazines and butyrophenones, have limited efficacy in humans in preventing acute vomiting associated with chemotherapy, with their use limited primarily to combination therapy for breakthrough nausea and vomiting. In a comparative study of humans undergoing cisplatin chemotherapy, antiemetic efficacy of metoclopramide was evident at 0.54 mg/L with efficacy as an antiemetic being dose dependent.206 Glucocorticoids, and specifically dexamethasone, improve the antiemetic efficacy of metoclopramide in humans; trials in dogs or cats are lacking. However, acute nausea induced by chemotherapeutic agents appears to be best prevented by the combination of dexamethasone (0.22 mg/kg intravenously) with serotonin antagonists. Among the antiserotinergic drugs, no differences in clinical efficacy appear to exist between ondansetron and dolasetron; however, granisetron, although not necessarily more effective, may last twice as long, perhaps because of its tighter affinity for serotonin receptors.

Postoperative Nausea and Vomiting

The use of antiemetics to prevent or treat postoperative nausea was reviewed by Ku and Ong.207 The incidence of postoperative vomiting in humans receiving volatile anesthesia ranges from 20% to 30%. Risk or predictive factors include age, gender (greater in females), history of vomiting or motion sickness, and duration and type of surgery and anesthesia. Opioid preanesthetics increase and alpha-2 agonists decrease the risk. Intraoperative drugs that increase the risk in humans include nitrous oxide (especially in patients already vomiting), whereas the more potent volatile anesthetics are associated with less vomiting compared with the less potent anesthetics. Propofol as an inducing agent causes less vomiting than do thiopental, etomidate, and ketamine; propofol also may reduce vomiting associated with general anesthesia.20 The impact of anticholinergics (atropine) or neuromuscular blockade antagonists (neostigmine) on postoperative emesis is less clear, although the incidence is reduced with the combination of the two agents. As with preoperative opioids, postoperative opioids increase the risk of nausea and emesis (due to direct stimulation of the CTZ and decreased GI motility) even if pain is effectively controlled. The incidence of vomiting associated with opioid-induced analgesia can be reduced by using those opioids associated with less emesis, and balanced analgesia.

Treatment of postoperative emesis is approached by either preventing or rescuing the patient. Drugs used as preventives include antagonists of H1 receptors (dimenhydrinate), muscarinic receptors (scopolamine patch), and 5-HT3 receptors (ondansetron or other carbazalone derivatives). Glucocorticoids (dexamethasone [0.11 to 0.14 mg/kg] and methylprednisolone), whose antiemetic mechanisms are unknown, also are recommended (administered intravenously) in patients with a high risk. Their use is more effective when combined with other antiemetics, particularly 5-HT3 antagonists. The NK1 receptor antagonists have recently undergone investigation, with early information in humans suggesting that NK1 antagonists may be more effective than 5HT3 receptor antagonists; accordingly, maropitant is reasonable choice in dogs or cats. Combinations are recommended over single drugs when treating breakthrough vomiting or nausea. Examples include 5-HT3 receptor antagonists with antihistaminergic drugs (e.g., ondansetron with cyclizine or promethazine) or combinations of droperidol, metoclopramide, or dimenhydrinate.20 Combinations with an NK1 receptor antagonist should be considered. Ideally, the combination would not include a drug that has already failed to control vomiting.

Postoperative Ileus

In humans, the time for gastric motility to recovery post-operatively ranges from 24 to 48 hours, the colon 48 to 72 hour. Thus, even with uncomplicated ileus, the time to recovery after surgery is 72 hours or less. The efficacy of metoclopramide in treating or preventing postoperative ileus has been studied in humans with conflicting results.208 In an observational, prospective study design, Seta and Kale-Pradhan208 found no difference in time to first bowel movement in intensive care unit patients that received metaclopramide postoperatively compared with those that did not. The drawbacks of this study include its design (nonrandomized, nonblinded, and non–placebo controlled) and very small sample size (n = 32; 16 per group). The authors reviewed the impact of metoclopramide on the effects of postoperative ileus in the report. Differences in dose, poor end points, and variability in study subjects (including surgical procedures) preclude consensus among the studies. Further, differences in opioid use among treatment groups generally has not been addressed. Time to reduction in oral feeding was a more frequent positive indicator of response to metoclopramide than was time to first bowel movement. However, the authors caution that the time to first bowel movement more likely reflects initiation of an oral diet, and studies using both outcome measures must address cause and effect. The review of the studies regarding the effect of metaclopramide on postoperative ileus are inconclusive.

The effect of lidocaine as a prokinetic for treatment of postoperative ileus after abdominal surgery has been the subject of a Cochrane Review of randomized clinical trials in humans.209 A review of 39 trials found that the data were insufficient (generally owing to inconsistent outcomes, small sample size, or poor data-collection methods) to recommend the use of CCK-like drugs, cisapride, dopamine antagonists, propranolol, or vasopressin. The authors noted that cisapride has been withdrawn from the marked, and further consideration was not given. Fifteen trials had reviewed alvimopan, a peripheral mu receptor antagonist that currently is an investigational drug. The authors noted that alvimopan may prove effective but further information was needed to define the criteria for use. Erythromycin was found to be consistently ineffective. Of the drugs studied, only intravenous lidocaine or neostigmine were considered potentially effective, but further assessment based on clinically relevant outcomes was indicated. No major adverse events to any of the drugs was noted in the review.

For postoperative ileus induced by opioids, treatment options include stimulant laxatives, oral naloxone, prokinetic agents (metoclopramide should follow an opioid antagonist), and potentially alvimopan. Misoprostol (high dose; approximately 6 μg/kg in humans) also might be considered; because this dose might be associated with nausea (in humans), a lower dose, given more frequently, may be necessary. Therapy should begin early.

Small Intestinal Diseases

Diarrhea

Acute diarrhea

Like vomiting, diarrhea should be managed by providing supportive therapy, treating symptoms, and resolving the underlying cause with specific therapy.210 Generally, intestinal causes of acute diarrhea include diet, toxins or drugs, and infections (including viral, microbial, and parasitic). A number of extraintestinal diseases include diarrhea as a manifestation. For many diarrheas rehydration and maintenance of hydration and electrolyte balance are the cornerstones of therapy Antiemetics should be used to control vomiting accompanied by acute diarrhea. Those antiemetics that cause hypotension (e.g., phenothiazine derivatives) should be withheld until fluid replacement has begun. Protectants and adsorbents are indicated for diarrheas associated with toxins (including “garbage enteritis”) and may be used for nonspecific (undiagnosed) causes of acute diarrhea. Kaolin may be useful for its adsorbent properties. Bismuth subsalicylate provides both adsorbent and antiinflammatory effects and is the preferred antidiarrheal agent for toxin-associated diarrheas.

Motility modifiers must also be used with discretion for treatment of diarrhea. Hypomotility rather than hypermotility is the more likely abnormality, and most motility modifiers cause hypomotility. Further, such drugs are often associated with side effects. However, among the motility modifiers, opioid drugs such as loperamide increase resistance to outflow as well as provide antisecretory effects. As such, of the motility modifiers, the opioid derivatives are preferred for short-term use as long as toxins, drugs, or obstructive disease have been ruled out as causes. Anticholinergic motility modifiers are reserved for psychogenic causes of acute diarrhea.

Viral Enteritis

There is no specific treatment for diarrheas of viral origin. Fluid therapy, electrolyte replacement, and antiemetics are indicated, depending on the severity of clinical signs. Among the viral causes of diarrhea, canine parvovirus stands out for its severity and life-threatening nature. Intensive, aggressive care such as that offered at tertiary hospitals can increase survival rates to 96% compared with 67% at local practitioners. The rate of survival with no therapy may range from 64% to 79%.211 Supportive therapy centers around the intravenous administration of balanced electrolytes (e.g., lactated Ringer’s solution) with potassium replacement. Damage to the mucosal barrier and risk of bacterial translocation should be addressed with parenteral antibiotics. The use of the viral neuromidase inhibitor olstamavir may be helpful in this regard. Antibiotics should target both aerobes and anaerobes. E. coli was identified as an organism associated with septicemia in canine parvovirus212,213; however, C. perfringens may also play a role.214 Because of the life-threatening nature of sepsis, combination therapy with a beta-lactam antibiotic (amoxicillin may be preferred to cephalexin because of a better anaerobic spectrum) and an aminoglycoside (gentamicin, amikacin) is recommended. Vomiting and the life-threatening nature of the illness preclude oral administration of antibiotics. Ceftiofur has been used by some clinicians because of its efficacy toward E.coli; however, its limited spectrum (toward anaerobes) might limit efficacy for treatment of parvovirus-associated bacteremia; adverse events also may be an issue. Fluorinated quinolones should be avoided if possible because of the risk of cartilage defects in young, growing animals. Because fluid therapy is likely to be intensive in these patients, and because most patients are pediatric, an increased volume of distribution should be anticipated and higher higher doses of antimicrobials, particularly water soluble, may be indicated. Treatment of septic shock may include drugs that target eicosanoids and other mediators of endotoxic shock. Prevention of endotoxemia or its effects, is reasonable, although reaching consensus regarding effect through randomized clinical trials is limited by study designs.211 Glucose may be added when indicated by clinical signs consistent with septicemia. Glucocorticoids and flunixin meglumine have historically been advocated to ameliorate some of the negative sequelae resulting from endotoxemia, with benefits more likely if treatment occurs within 4 hours of the onset of endotoxemia. Shock doses of glucocorticoids should be used. Controversy regarding the use of flunixin meglumine centers primarily on the risk of GI damage. However, damage is generally so severe at the time of clinical presentation that it is reasonable to assume that the use of a single dose of flunixin meglumine is not likely to contribute to further damage. An additional benefit of flunixin meglumine is its potent visceral analgesic effect, which may be important is alleviating the marked pain and its negative pathophysiologic sequelae. Because cyclooxygenase 2 is the predominant eicosanoid mediating the sequelae of shock, any of the newer injectable NSAIDs that target cyclooxygenas 2 (e.g., carprofen, meloxicam, firocoxib) presumably should be equally effective in treatment or prevention of endotoxic shock. Opioids should be avoided because of their inhibitory effect on expelling luminal contents.

KEY POINT 19-43

Although gram-negative coliforms clearly should be targeted in patients with viral enteritis for which translocation is a concern, anaerobes also should be targeted.

Although currently experimental, compounds targeting endotoxin may prove to be an important adjuvant to patients suffering from sepsis. Examples include endotoxin serum or a bacterial toxoid (Salmonella typhimurium; 10 mL/kg). The latter is commercially available. Transfusions with fresh whole blood or plasma can be beneficial in some dogs, particularly those that are hypoproteinemic or anemic. The impact of a recombinant amino terminal fragment of bactericidal permeability-increasing protein (rBPI21; an antimicrobial and endotoxin-neutralizing agent) was studied in dogs (n = 40) with viral enteris. Treatment (3 mg/kg intravenously over 30 minutes, followed by 3 mg/kg intravenously over 5.5 hours) was implemented using a randomized placebo (n = 9 dogs treated with canine plasma protein) controlled design. Outcome measures included plasma endotoxin concentration, severity of clinical signs, and survival of parvovirus. No treatment effect could be identified ; however, contributing factors were insufficient sample size and biased patient selection toward animals that are less ill than the average infected animal.211

Treatment of diarrhea associated with parvovirus is generally not indicated. Until the mucosa has had time to heal, drugs intended to prevent or resolve diarrhea are not likely to be effective.

Bacterial Enteritis

The role of antimicrobial therapy in the treatment of diarrhea should be closely examined. Bacterial infection is not a major cause of diarrhea, nor does infection appear to perpetuate small intestinal diseases. More important, use of antimicrobial agents does not appear to improve the course of most acute diarrheas. Antimicrobials (neomycin, ampicillin) may, in fact, worsen diarrhea, perhaps because of suppression of normal microflora. Use of antimicrobials for treatment of diarrhea should be based on a diagnosis of intestinal bacterial infection (overgrowth detection, based in part, on fecal gram staining) or in cases of mucosal damage sufficiently severe to allow bacterial translocation. In the latter case, clinical signs generally include hemorrhagic diarrhea, fever, and abnormal white blood cell counts. Systemic antimicrobial therapy is indicated for bacterial translocation.

Despite the low incidence (less than 4% of cases of acute diarrhea), bacterial infections have been associated with both acute and chronic enterotoxigenic diarrhea of both small and large intestines. Diagnosis and antibacterial treatment are best based on culture and susceptibility data when possible. The most likely therapy for each of the infecting organisms is enrofloxacin, trimethoprim–sulfonamide combinations, and chloramphenicol for Salmonella; erythromycin, enrofloxacin, furazolidone, doxycycline, neomycin, clindamycin, or chloramphenicol for C. jejuni; a prolonged course of trimethoprim–sulfonamide combinations, tetracycline, or chloramphenicol for Yersinia enterocolitica (prognosis is guarded); metronidazole for C. difficile; and amoxicillin, ampicillin, metronidazole, tylosin, or clindamycin for C. perfringens. For E. coli the role will be difficult to establish. Clostridium piliformis (formerly Bacillus piliformis: Tyzzer’s disease) is a less common, although rapidly fatal, cause of acute hemorrhagic enterocolitis. The organism appears to be nonresponsive to antimicrobials, wih therapy being supportive in nature. Salmon poisoning (Neorickettsia helminthoeca) is an endemic, fatal cause of diarrhea in dogs in the Pacific Northwest. As with other rickettsial organisms, tetracycline (oxytetracycline, doxycycline) is the treatment of choice. Oral therapy (in the absence of vomiting) includes tetracycline, chloramphenicol, sulfonamides, and penicillins. Therapy should continue for 2 to 3 weeks; the trematode vector can be treated with fenbendazole for 10 to 14 days (50 mg/kg, once daily). Because bacterial infections as a cause of diarrhea are often associated with some type of toxin production, motility modifiers should be avoided. Bismuth subsalicylate may be beneficial for both its adsorbent and antiinflamamtory effects.

Hemorrhagic Gastroenteritis

This syndrome of uncertain etiology is characterized by a packed cell volume (PCV) that may be as high as 80%. Hemoconcentration rather than dehydration is the cause. Treatment requires prompt and rapid but appropriate volume replacement with a balanced electrolyte solution until the PCV falls below 50%. Fluid therapy should continue for an additional 24 hours to maintain the PCV at 50% or lower. Disseminated intravascular coagulopathy may develop if fluid therapy is not instituted rapidly.

Chronic Diarrhea

Treatment of chronic diarrhea should be based on removing the underlying causes. This is perhaps more important than in acute diarrhea because drugs used to symptomatically treat acute diarrhea should not be continued on a long-term basis. Chronic IBD is discussed later as a separate entity.

Bacterial overgrowth is increasingly being recognized as a cause of chronic intermittent small bowel diarrhea in dogs. Because no sensitive, specific, and widely available diagnostic test is available, diagnosis is difficult unless an underlying cause (e.g., partial intussusceptions, tumors, foreign body) can be identified. Oral treatment should include broad-spectrum antibiotics, such as tylosin (10 to 20 mg/kg every 12 hours) or metronidazole, a drug effective against anaerobes (10 to 20 mg/kg every 12 hours). The use of probiotics should be considered as previously discussed.

Intestinal fungal disease may also manifest as chronic diarrhea. Prognosis is generally poor to fair. Intestinal histoplasmosis should be treated with an orally administered azole drug. Itraconazole is the drug of choice (5 to 10 mg/kg every 12 hours) followed by ketoconazole (10 to 15 mg/kg every 12 hours); both drugs should be given 3 to 4 months after clinical signs of remission. Voriconazole also might be considered. Amphotericin B can be given in addition to an azole drug, particularly for severe cases. Currently, no antifungal drug has been effective for treatment of pythiosis (previously phycomycosis). Surgical resection followed by imidazole therapy (itraconazole or ketoconazole) is indicated in animals in which the disease is diagnosed before tissue damage and infiltration.

Protozoal diseases of the small intestine include coccidioidomycosis, cryptosporidiosis, and giardiasis. Pentatrichomonas hominis also has been associated with diarrhea, particularly in puppies and kittens. Diagnosis of each infection is based on identification of the organism or of cysts in feces. Treatment of coccidioidomycosis includes sulfadimethoxine (50 to 65 mg/kg once daily orally for 10 days); trimethoprim–sulfonamide combinations (30 mg/kg orally once daily for 10 days), quinacrine (10 mg/kg orally once daily for 5 days), or amprolium (100 mg [small dogs] to 200 mg [large dogs] of 20% powder once daily in gelatin capsules, or 1 to 2 teaspoons of 9.6% amprolium per gallon of free-choice water for 1 to 2 weeks). There is no effective treatment of cryptosporidiosis in dogs. This infection is generally self-limiting in immunocompetent animals. A number of therapies can be used to treat giardiasis. Metronidazole (25 to 30 mg/kg orally twice daily for 5 to 10 days) is the treatment of choice, although up to a third of animals may not respond. Metronidazole benzoate has demonstrated efficacy in treatment of feline giardiasis based on a study in 26 chronically infected cats, 10 of which also were infected with Cryptosporidium parvum.215 Cats were treated with 25 mg/kg orally twice daily for 7 days; the drug was prepared as a solution. It is not clear if the dose was based on active ingredient (i.e., metronidazole base) or total drug. All cats were negative for three consecutive fecal examinations (based on indirect immunofluorescence assay) for the 15 day after treatment study period. Albendazole (25 mg/kg orally every 12 hours for 2 days) can be used in dogs, but safety and efficacy have not been reported in cats. Fenbendazole (50 mg/kg orally once a day for 3 days) may also be effective. Furazolidone (4 mg/kg orally every 12 hours for 5 to 10 days) can be used in cats, although toxicity may limit its use. For nonresponsive cases in dogs, quinacrine (6.6 mg/kg orally every 12 hours for 5 days) can be used, although side effects (anorexia, lethargy, vomiting, and fever) are common. Ipronidazole (126 mg/L drinking water) is a poultry drug that can be used for treating groups of animals. Tinidazole (currently not available in the United States; 44 mg/kg once orally) may also be useful. Pentatrichomoniasis should respond to a 5-day course of metronidazole therapy; tinidazole for 3 days may also be useful. Both drugs can be used according to previously described dosing regimens.

Short Bowel Syndrome

Short bowel syndrome occurs after surgical removal of a large portion of the small intestine. Resultant malabsorption results in malnutrition and diarrhea. The impact of the resection on bowel function and the ability of the remaining bowel to adapt to the loss depend on the extent and site of resection. Dogs have functioned with an absence of clinical signs following resection of up to 85% of the small intestine. Preservation of the ileum is important because of its role in slowing transit and absorption of vitamin B12 and bile acids. Medical management focuses primarily on correction of secondary ill effects. Exocrine pancreatic insufficiency should be treated with enzyme supplementation. Gastric hypersecretion should be treated with H2-receptor antagonists. Bacterial overgrowth should be treated with appropriate antimicrobial therapy (e.g., metronidazole, tylosin). Cholestyramine can be used to bind excessive bile acids that result in diarrhea. Occasionally, motility modifiers may be indicated to slow transit time. The opioids are preferred because of their effects on segmentation and thus retention of luminal contents.

Bacterial translocation

Bacterial translocation refers to the movement of gastrointestinal origin microbes or their products across the intact gastrointestinal tract into normally sterile tissues and subsequent direct infection or inflammation causing tissue injury, organ failure, and death. The treatment of subsequent sepsis is addressed in Chapter 8. Bacterial translocation ideally is prevented.151a-c Among the choices to prevent bacterial translocation are antimicrobials (selective gastrointestinal or digestive decontamination; SDD) and probiotics. The concept of SDD has been promoted and studied in the human critical care patient. Steinberg has reviewed the impact of bacterial translocation (in the surgical patient)215a and Schultz and co workers have reviewed the conclusions of several metaanalysis that focus on SDD in the critical care patient as part of their report of a clinical trial involving SDD with a favorable outcome. 215b In general, scientific clinical evidence supporting SDD is lacking, and as such, SDD has not become a standard of care for the critical care patient. Nonetheless, Schultz has demonstrated a benefit of SDD in a population of critical care patients receiving ventilator support. In their review, the authors indicate that the risk of emergent resistance as a sequelae of SDD—a major antagonist argument against its routine implementation—did not occur in one clinical trial: indeed, SDD was associated with decreased resistance in this trial. This might be a reasonable expectation particularly if principles of judicious use are applied (see Chapter 6). Care must be taken when extrapolating the results of meta-analysis regarding SDD use in humans to the veterinary patient in that the applicability of the sample human populations may not represent the diseases with which veterinary criticalists are faced. Drug choices for SDD include oral drugs that target gram-negative coliforms but are not orally bioavailable, coupled with systemic antimicrobials targeting the same. However, Schulz and coworkers have also demonstrated that oral absorption of drugs normally characterized by no oral bioavailability may indeed occur in the critical care patient.215c As such, further caution is recommended when using SDD. There is a need for well-designed clinical trials that address SDD with antimicrobial therapy are lacking in the veterinary critical care patient.

The use of probiotics to prevent colonic bacterial translocation in human patients undergoing abdominal surgery was reviewed by Lenoir-Wijnkoop and coworkers.151 The rationale targets the increased risk of bacterial translocation associated with surgical trauma, portal hypertension, decreased hepatic function and immunosuppression. Several controlled clinical trials have been implemented using synbiotics. One study in human liver transplant recipients found that the incidence of infection was much lower (3%) in patients receiving antibiotics plus a highly concentrated combination product of four fibers (2.5 g each of beta-glucan, resistant starch, inulin, and pectin) and four probiotics (1010 Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus mesenteroides, and Pediococcus pentosaceu) compared with those patients that received antimicrobials only (48%). A recent controlled clinical trial in humans undergoing pancreatic surgery found that the combination of probiotics and selective bacterial decontamination had no effect on bacterial translocation and other selective outcome measures.215d

Diseases of the Large Intestine

Diarrhea

Diarrhea associated with the large intestine should be approached in the same manner as diarrhea of the small intestine. Chronic IBD is discussed as a separate entity.

Irritable bowel syndrome

IBS, spastic colon, or nervous colitis is a poorly described functional disorder afflicting dogs and is diagnosed by ruling out other causes of large bowel diarrhea. Effective treatment is complicated by the intermittent nature of the syndrome.216 Dietary management should be stressed for long-term management. Some large bowel diarrheas may respond to dietary fiber (psyllium). Intermittent bouts of diarrhea attributed to IBS can be managed with administration of short-term opioid antidiarrheals (1 week or less). Anticholinergics can be used to reduce intestinal spasms, particularly those associated with pain and tenesmus. Combination anticholinergics and sedatives (e.g., chlordiazepoxide and clidinium) also may prove useful.

Treatment of IBS in humans was reviewed by Spanier and coworkers.217 Whereas the prevalence in humans is as high as 24%, the role of IBS as a cause of diarrhea in dogs or cats is not clear. In humans no specific therapy emerges as clearly effective, causing afflicted patients to seek out alternative therapies. A review of clinical trials reveals therapies of variable efficacy to include probiotics (L. acidophilus, Candida and others); herbal products such as aloe or peppermint oil; and nonmedicinal therapies such as colonic irrigation, acupuncture, psychotherapy, and meditation. None was well supported, but positive scientific evidence was greatest for Chinese herbal therapy and psychological therapy.217

Clostridium spp

C. difficile has reached epidemic proportions in human medicine, with antimicrobial-induced suppression of normal flora as a major risk factor. Those drugs most commonly associated with its emergence included clindamycin, penicillins, and cephalosporins; fluoroquinolones have also been identified in humans (see Chapter 8). Environmental contamination and fecal-to-oral transmission are important, with hand carriage by health care personnel occurring in human patients in much the same way as with methicillin-resistant Staphylococcus aureus. In humans a new strain has emerged with increased virulence. Outbreaks have increased in the hospital setting, with community-acquired infections increasing in North America and Europe.218 Clinical signs range from mild diarrhea to pseudomembranous colitis to toxic (and potentially fatal) megacolon. Clostridial toxins increase with deletion of the gene that downregulates production. Pathophysiology reflects binding of toxin by intestinal cells, disruption of epithelial tight junctions, and inflammation. Watery diarrhea is the clinical hallmark of infection. A seasonal pattern has been described for selected human hospitals.220 Enterotoxicosis associated with C. perfringens is also emerging as a cause of large bowel diarrhea primarily in dogs (see Chapter 8). Diagnosis is based on a reverse latex agglutination test available in many human laboratories. Acute treatment includes metronidazole, ampicillin, or amoxicillin. Tylosin may be effective for cases requiring long-term treatment. High-fiber diets (or psyllium) may also be helpful. Other bacterial diseases of the large intestine were discussed as causes of diarrhea in the small intestine.

Clostridial resistance may have already emerged toward newer 8-methoxy fluoroquinolones used to treat the organism (e.g., gatifloxacin and moxifloxacin).221

Antibiotic-responsive diarrheas

Several chronic enteropathies afflicting dogs are reported to respond to a number of antibiotics, leading to the term antibiotic-responsive diarrhea (ARD). The distinction of antibiotic-responsive diarrhea from small bowel diarrheas associated with (idiopathic) small intestinal bacterial overgrowth is not clear.222 Small bowel diarrhea of German Shepherd Dogs typifies the syndrome. Antibiotics to which animals have responded include tetracycline, metronidazole, ampicillin, tylosin, and enrofloxacin.

Tylosin-responsive chronic diarrhea was described in dogs (n = 14).222 Middle-aged, large-breed dogs are more commonly affected with clinical signs referable to both the small and large bowels. However, the study is complicated by study design (e.g., all dogs were treated with tylosin 1 month before starting the study, and animals that responded to sequential therapies were dropped from the study). Nonetheless, the authors reported that all animals responded to tylosin (6 to 16 mg/kg orally once daily) within 3 days (most commonly within 24 hours), with clinical signs recurring within 30 days of discontinuing therapy in 86% of dogs. Other failed therapies attempted with recurrence included prednisone treatment (for 3 days; partial response) or Lactobacillus rhamnosus probiotics (no responders). On the basis of response to tylosin, potential pathogens have been proposed, including C. perfringens, campylobacters, and Lawsonia intracellularis. A proposed rationale for the syndrome is the existence of an as of yet to be identified specific enteropathogenic common to the canine GI tract that is susceptible to tylosin.

Several antimicrobials have beneficial immunomulatory effects in the GI mucosa (as reviewed by Westermarck and coworkers222). These include metronidazole and fluoroquinolones (ciprofloxacin and enrofloxacin). The use of enrofloxacin to treat inflammatory bowel disease is discussed later. Probiotics have been shown to be effective for pediatric antimicrobial diarrhea, but not that associated with C. difficile. Trichomoniasis is caused by Entamoeba histolytica and Balantidium coli, protozoal organisms associated with diarrhea of the large intestine in dogs and cats. Treatment of trichomoniasis was delineated in the section on diseases of the small intestine.

Treatment of B. coli infection has not been delineated in animals but, based on the response in humans, might include the use of tetracyclines or metronidazole.

Megacolon

Initial medical management of megacolon associated with mild constipation should include bulk laxatives and more active laxatives such as bisacodyl or docusate sodium suppositories. As constipation progresses to obstipation, enemas and evacuation under general anesthesia are implemented. In severe cases broad-spectrum antimicrobials may be indicated to decrease the potential for bacterial translocation across the damaged mucosa. Long-term medical management should be accompanied by dietary management. Laxatives and periodic enemas are indicated. Prokinetics such as cisapride have had variable success but should be tried. Earlier use is more likely to be prevent progression from constipation to obstipation in cats. Erythromycin demonstrated a prokinetic effect on colonic motility but did not provide a clinically evident benefit in human patients with postoperative colonic ileus.183 Antisecretory drugs with anticholinesterase activity (e.g., ranitidine, nitazidine) might also be considered.

Inflammatory Bowel Disease

Pathophysiology

IBD is characterized by infiltration of inflammatory cells in the gastric or intestinal mucosa (or both).194,223 Its emergence in predisposed animals is facilitated by the large immune system and its multivariate responses presented by the GI tract, coupled with the vast number of antigens from ingested microbes, parasites, food, toxins, endogenous microbes or their products, and other materials. Increased intestinal permeability to antigens probably plays a role, but it is not clear if this is an initiating event or a sequela. Disruption of the intricate balance between microbe and host leads to breakdown of mucosal tolerance. This key event leads to initiation, progression, and reemergence of disease.223 Loss of mucosal tolerance generally requires a loss of the normal barrier, as might occur with loss of cadherins. Immunologic dysfunction largely reflects altered T-cell [CD-4] activity. High concentrations of IL-10 and 18, and TGF-β promote T cell differentiation to the Th-1 phenotypes, resulting in high concentrations of IL-2, INF-γ, and TNF-alpha.224 A third factor in the loss of mucosal tolerance is the presence of endogenous microflora.

KEY POINT 19-44

Effective treatnment of of IBD is complicated by the complex interaction between microbes, the gastrointestinal tract, and the local immune response.

German and coworkers223 reviewed IBD in dogs, and Allenspach and coworkers225 described risk factors for therapeutic failure. Canine IBD is a group of diseases that are highly variable in cause and presentation. Manifestations and treatment depend on cell type (with lymphocytic–plasmacytic most common, followed by eosinophilic), region affected (any location but most commonly small intestine) and breed (e.g., histiocytic [large macrophages] ulcerative colitis of Boxers, protein-losing enteropathies of Soft Coated Wheaten Terriers, gluten sensitivity of Irish Setters, and immunoproliferative enteropathy of Basenjis).223 The type of predominating cell (lymphocytic, plasmocytic, eosinophilic, or histocytic) that causes inflammation can serve as a basis of the classification and, to some degree, treatment of IBD.226 Overgrowth of small intestinal bacteria in response to either a primary (e.g., idiopathic) or secondary (e.g., acquired) disorder has been studied as a potential cause, particularly in German Shepherd Dogs; IgA deficiency has been suggested.223 Although commonalities exist between human and canine IBD, the most common forms in human include ulcerative colitis and Crohn’s disease. Ulcerative colitis is diffuse and superficial, involving predominantly neutrophils, with some lymphocytes and plasma cells, particularly in the ileum. Crohn’s disease is focal and segmental, characterized by chronic pyogranulomatous inflammation. When extrapolating therapeutic options between human and canine or feline IBD, considerations must also include differences in the pathophysiology of the diseases among species. Even within species, preferred therapies and extent of response is too variable among canine and feline populations to be predicted without supportive diagnostic (histopathologic) data. Accordingly, treatment should be approached individually (Table 19-5). A scoring system has been proposed for dogs to facilitate treatment.226

Table 19-5 Products for Treatment of Inflammatory Bowel Disease

Category Product Example Drug or Active Compound
  Undigested carbohydrates  
  Undigested proteins  
Supplement Iron  
  Protein  
  Water- and fat-soluble vitamins  
  Cyanocobalamine  
  Trace elements  
  Electrolytes  
Antiinflammatories Aminosalicylates  
  Mesalazine (5 aminosalicylic acid, 5 ASA) Asacol (Mesalamine) Increased stability in acid medium; absorption slowed)
    Salofalk (coated with ethylcellulose)
  Sulfasalazine Sulfapyridine diazotized to 5 ASA
  Pentoxyfylline  
  Leukotriene receptor antagonists  
Immunomodulators Cyclosporine  
  Glucocorticoids  
  Systemic  
    Prednisolone (preferred to prednisone in cats and possibly some dogs)
    Prednisone
  Topical (oral) Budesonide
    Fluticasone
    Tixocortol pivolate
  Topical (foams; rectal) Hydrocortisone acetate
    Prednisolone metasulphobenzoate
    Hydrocortisone sodium phosphate
    Prednisolone sodium phosphate
  Enema Methylprednisolone
    Betamethasone valerate
    Beclometasone diproprionate
    Budesonide
Directed polypeptides Role not yet elucidated  
Probiotics Lactobacilus acidophilus, bulgaricus With fructose oligosaccharide
  Bifidobacterium?  
  Others  
Antibiotics Metronidazole  
  Fluoroquinolone Enrofloxacin
  Others (e.g., Tylosin)  
Protection Sucralfate  
  Antisecretory drugs Famotidines, proton pump inhibitors
  Polysulfated glycosaminoglycans Glucosamine, chondroitin sulfates

Foams adhere to mucosa and are for colonic disease. Administered at night.

Histocytic ulcerative colitis.

The “4-R’s” approach to treatment of Crohn’s disease in humans includes removal (underlying causes such as inappropriate diet), replacement (missing nutrients, such as vitamin B12), re-inoculation with “friendly” bacteria (L. acidophilus and L. bulgaricus along with fructose oligosaccharides), and repair. Dietary considerations in the role of (human) IBD have been reviewed by Shah.227 The role of gut flora, GI immunity, and IBD based on models and spontaneous disease has been reviewed in humans.228,229 A series of studies using interleukin 10–deficient mouse colitis models demonstrated the influence of different bacterial substrates at different sites of inflammation. Inflammation best responded to a combination of vancomycin–imipenem or neomycin–metronidazole compared with ciprofloxacin and metronidazole. Response to the latter was effective for acute but not chronic colonic inflammation. Narrow-spectrum antimicrobials such as ciprofloxacin were more effective in preventing but not treating experimentally induced colitis230 (see also the discussion of antibiotic-responsive diarrhea). Studies have demonstrated that inflammatory disease will not evolve experimentally in microbe-free environments but can be experimentally induced by transfer of T-cells reactive to bacterial antigens. In human patients with IBD, lesions are worse in those areas with highest microbial counts. Although the role of potential pathogens has been intensively studied, emerging data suggest that it is the commensal, rather than pathogenic, microbes that are contributing to the disease. As such, perpetuation of the disease may reflect abnormal signaling between host immune system and microbes and a loss of host tolerance for microbes. Accordingly, treatment for IBD targets not only suppression of the inflammatory response but also manipulation of the contributing microbiota with either antimicrobials or probiotics.

Unfortunately, IBD is a diagnosis of exclusion and is made after other causes of inflammatory disease of the GI tract have been ruled out. Food allergies or intolerance; infections by fungal, bacterial, or parasitic organisms; and neoplasms must be ruled out or their role in the pathophysiology identified and treated accordingly. Confirmation of the diagnosis is based on biopsy, which also is necessary to identify the predominant inflammatory cell associated with the disease and the most appropriate therapy. For example, eosinophilic infiltrates may respond to dietary management alone. Monitoring response to therapy should include, in addition to clinical signs, serum folate and cyanocobalamin concentrations; serum albumin levels can be used as a monitoring tool in animals with protein-losing enteropathy. Allenspach and coworkers225 retrospectively determined that hypocobalaminemia (<200 ng/mL), along with hypoalbuminemia, is a risk factor for therapeutic failure.

Initial medical management may vary with the severity and length of disease. In dogs, particularly, elimination of an irritating diet and antibiotic-responsive causes might be considered before biopsy. This is best accomplished by a well-designed clinical trial in the patient. The role of diet in human IBD has been well reviewed.227 Feeding the animal an elimination diet containing novel or highly digestible protein foods or (particularly for colonic disease) a high-fiber diet (e.g., yams, sweet potatoes, pumpkin) might be considered first. The role of diets with altered n:3 to n:6 omega-fatty acid ratios or hydrolyzed diets (protein molecules are small and presumably nonantigenic) is not yet clear but may be promising.223

Antiinflammatories remain the cornerstone of therapy for IBD in dogs and cats. Use of glucocorticoids should be reserved for animals in which biopsy has confirmed a diagnosis of IBD. Indiscriminate use of glucocorticoids can be dangerous, particularly in areas in which fungal causes of GI disease are not uncommon. In addition, use of glucocorticoids in patients with GI lymphoma may render the neoplasia resistant to further glucocorticoid therapy used as part of a combination antineoplastic regimen. Glucocorticoids are indicated in dogs and cats with lymphocytic–plasmacytic IBD. Prednisolone (2.2 mg/kg/day orally) should result in clinical response within 1 to 2 weeks. Therapy should continue at the same rate for another 2 weeks (beyond clinical response) and then slowly be tapered. More severe cases of IBD or cases that do not initially respond to prednisolone may respond to dexamethasone (0.22 mg/kg/day orally).Because glucocorticoids impair healing in the gastrointestinal mucosa (by virtue of cyclooxygenase 2 inhibition), antisecretory and cytoprotectant drugs are indicated. Indeed, their use is indicated in general to facilitate healing development of multidrug resistance has been implicated as a cause of therapeutic failure with glucocorticoids in some human patients with IBD; expression of mucosal multidrug resistance may ultimately be used to determine response of IBD patients to therapy.231 Several mechanisms of glucocorticoid resistance have been described in humans and may be relevant to dogs or cats.232 These include heterogenicity of the disease process itself, overexpression of the MDR1 gene causing increased P-glycoprotein–mediated efflux of glucocorticoid from target cells; impaired glucocorticoid–receptor signaling; and activiation of epithelial proinflammatory mediators such as nuclear factor kappa B.

KEY POINT 19-45

Although glucocorticoids are the cornerstone of inflammatory bowel disease therapy, glucocorticoids also impair gastrointestinal healing, and gastroprotective therapies should simultaneously be implemented.

Humans generally do not tolerate systemic glucocorticoids as well as dogs or cats, leading to effective alternative therapies for IBD.233 For example, 80% of human ulcerative colitis cases are successfully controlled with a variety of 5-aminosalicylic acid preparations. Rectal glucocorticoid foams have also proven useful for diseases involving the colon. “Topical” budesonide is as effective as systemic glucocorticoids and is better tolerated than systemic glucocorticoids. Glucocorticoids are the cornerstone of therapy for Crohn’s disease; more severe disease responds to higher doses of steroids. The extrapolation of these treatments to dogs and cats is limited in part because colonic disease is not as common (limiting applicability of therapies targeting the colon). Further, the extent of first-pass metabolism of budesonide is not known. A number of therapies should be considered in dogs or cats that do not respond to initial therapy or for which glucocorticoids are not tolerated or contraindicated. Various immunomodulators should be considered. In humans, these have included cyclosporine and mycophenolate mofetil. Allenspach and coworkers234 reported on the use of cyclosporine for treatment of IBD in dogs (Chapter 19). Overall, cyclosporine was considered effective in 78% of the animals. In humans, long-term cyclosporine may facilitate long-term control when used in combination (but not alone) with other immunomodulating drugs. Leukotrienes appear to be involved in chronic allergic diseases such as IBD, atopy, and asthma. A role has been described in signaling and trafficking between eosinophils and lymphocytes in affected tissues, and in the IL-5 eotaxin induced differentiation, proliferation, and release of eosinophils at the level of the bone marrow. Accordingly, leukotriene receptor antagonists (e.g., zafirlukast or montelukast; see Chapter 29) might be considered as the sole agent in mild disease in those animals in which glucocorticoids are contradicted, in combination for nonresponders, or as dose-sparing agents. The role of TNF-alpha in Crohn’s disease of humans has led to some scientific support for pentoxifylline (also referred to as oxpentifylline).235 It has proved useful in mouse models of colitis236 and in humans as a dose-sparing agent when combined with glucocorticoids or directed polypeptide therapy.237

Sulfasalazine (20 mg/kg every 12 to 24 hours orally in cats; 50 mg/kg/day divided every 8 to 12 hours in dogs) may also be beneficial in cats and dogs with IBD. Response may take 1 to 2 weeks. As a sulfonamide, sulfasalazine may cause immune-mediated diseases ascribed to other sulfonamide antibiotics; use of the drug should be based on a histologic diagnosis whenever possible. Newer 5-aminosalicylate (sulfasalazine-like drugs) such as mesalazine and olsalazine (10 to 20 mg/kg every 12 hours) might be considered; both may decrease tear production in dogs. Omega fatty acid (fish oil) products also may be helpful for their antiinflammatory effects; response may take several weeks.

Animals that continue to be unresponsive to medical management of IBD may respond to azathioprine (0.3 mg/kg every other day, orally in cats; 2.2 mg/kg/day in dogs). Response may take up to 5 weeks. Side effects of azathioprine are sufficiently severe that a diagnosis of severe IBD should be confirmed (based on biopsy) before its use. White blood cell counts should be monitored weekly and the drug temporarily discontinued if neutrophil counts drop below 2000/uL.

The role of directed polypeptides in the management of refractory human IBD is emerging.238,239 Emerging therapies are targeting co-stimulatory molecules which are responsible for initial interactions between T cell receptors and macrophage MHC antigen complexes.224 Endotoxin and cytokines interact directly or indirectly with a variety of co-stimulatory molecules, including CD40-ligands (with T-cells) and B-7, an immunoglobulin that serves as a ligand for C28 (also a T cell co-stimulatory molecule). Because many of these therapies represent proteins foreign to dogs and cats and because animals are already affected by a dysfunctional immune system, caution is recommended with their use. Side effects can be profound in humans and may preclude adaptation to dogs or cats. Use in dogs and cats should be implemented only after collecting intensive scientific support.

Antibiotic therapy is intended to resolve bacterial overgrowth that might be contributing to the inflammatory process and either mimicking or contributing to IBD. However, response may just as likely reflect immunomodulation rather than antimicrobial therapy. Therapy for overgrowth in the large intestine should target clostridial organisms (metronidazole or ampicillin), whereas broader-spectrum drugs (tylosin, ampicillin) should be used for small intestinal disease. C. perfringens overgrowth in the small intestine may be difficult to detect; drug therapy that targets this organism includes tylosin and ampicillin. Metronidazole therapy in conjunction with glucocorticoids is indicated not only for its antibacterial effects but also because it appears to have immunomodulatory capabilities; indeed, this may explain why it may be effective as the sole therapy in some cases of IBD.

Nonhypoproteinemic dogs with lymphocytic–plasmacytic enteritis responded to a combination of oral antiinflammatory agents (prednisone, 1 mg/kg twice daily slowly decreased to 0.5 mg/kg every 48 hours) and antimicrobial agents (metronidazole 10 mg/kg twice daily for 21 days); most dogs also received oral cimetidine (0.5 mg/kg bid) and metaclopramide (0.5 mg/kg bid) for 90 days.240 Treatment dogs (n = 16) received a prescription diet. A group of normal animals (n = 9) were studied as untreated controls. Dogs were studied for 120 days, with outcome measures including clinical signs, endoscopic lesions and histopathy of endoscopic biopies. After treatment, the mean activity index diminished from 7.3 at baseline to 1.7, 0.8, 0.5, and 0.19 at baseline, on days 30, 60, 90, and 120, respectively. Further, gastric and duodenal endoscopic lesions decreased in 75% of animals, although no significant reduction was detected histologically.

Hostutler and coworkers241 have retrospectively described a series of cases (n = 9) of canine histoycytic ulcerative colitis responsive to antibiotics. The common drug among all nine dogs was enrofloxacin, with or without combinations of amoxicillin or metronidazole. Four of the dogs had failed to respond to antiinflammatory therapy that included combinations of prednisolone, azathioprine, or sulfasalazine, with or without other antibiotics, for a duration of 1 to 20 weeks. The remaining five dogs responded to antibiotics alone. Diarrhea resolved within 3 to 12 days of treatment with enrofloxacin at standard recommended doses. Although three dogs remained asymptomatic for 7 to 14 months, some dogs have required therapy for 2 to 21 months or longer.

Among the more promising approaches of therapy that do not directly suppress inflammation is the use of probiotics (see the earlier discussion of biotherapeutics). Probiotics are intended to replace the pathogens with healthy flora that have developed host tolerance. Their use in patients with IBD should be strongly considered; however, notably lacking is scientific evidence regarding their use. Necssary information ranges from characterization of the normal state of microbiota in the canine and feline gastrointestinal tract to its state in patients with IBD; the optimal replacement microbiota; and clinical trial evidence of response when used as either sole or combined therapy. Further, deficiencies in product quality may limit effective response. None the less, attention must be given to this approach to therapy. The role of probiotics in the treatment of IBD in humans has been reviewed.228,229 Randomized controlled clinical trials in humans have demonstrated resolution or improvement of IBD compared with controls (placebo or 5-aminosalicylic acid) after treatment with probiotics containing bifidobacteria, lactobacilli, and streptococci as core microbes.228,229 However, because the pathophysiology of IBD and endogenous microbiota (and presumed response to biotherapeutic) varies with site, age, diet, species, and other factors, scientific evidence of efficacy of biotherapeutics in IBD may be slow to emerge. Human and mice model data are not necessarily relevant to either dogs or cats; studies in target species are needed to support efficacy. Further, the microbiota of individuals with IBD is different from that in normal animals and (has been described as unstable). Accordingly, although biotherapeutics are largely safe in normal animals, the unknown impact of colonization of microbes in the diseased intestine mandates that discretion (and knowledge) accompany biotherapeutic use. Marteau and coworkers228 reviewed the role of biotherapeutics in the treatment of IBD. Several studies suggest that E. coli and Bacteroides vulgatus, both normal flora, in particular, may be reasonable targets of therapy. Members of bifidobacteria and lactobacilli generally tend to be the most likely organisms to provide protection against IBD, but species differences in normal flora are likely to mandate clinical trials in target species as a basis of proof.

KEY POINT 19-46

The mechanism of efficacy of selected antimicrobials for treatment of inflammatory bowel disease may reflect immunomodulation more that antibacterial effects.

Associated clinical signs of IBD that may require medical management include vomiting, small or large bowel diarrhea, flatulence, and occasionally GER. Hematochezia may be present with colitis. Diarrhea is the primary presentation of IBD in dogs. Vomiting occurs less commonly with gastric and enteric IBD; hematochezia occurs consistently in colitis. Anorexia and weight loss occur to variable degrees in IBD. Use of antisecretory drugs is indicated, in part to provide GI protection; sucralfate likewise is indicated, particularly in the presence of erosive or ulcerative disease and glucocorticoid therapy.

Supplementation of cobalamin should be considered at least in cats with chronic inflammatory disase of the small intestine associated with hypocabalaminemia. Ruaux and coworkers242 studied 19 cats severly deficient in cobalamine (based on serum concentrations) during and after treatment (250 units subcutaneously once weekly) for 4 weeks.

Treatment for Helicobacter spp. might also be considered. Leib and coworkers216 demonstrated marked improvement in dogs with IBD when they were treated for Helicobacter spp. Chronically vomiting dogs with spirochetes and either normal or inflamed stomach or duodenum (based on biopsy samples) were studied. Dogs were assigned to receive twice daily for two weeks “triple” therapy (amoxicillin 15 mg/kg, metronidazole 10 mg/kg and bismuth subsalicylate at 13 to 26 mg/kg [0.25 to 2 262-mg tablets, depending on body size), either with or without famotidine (0.5 mg/kg). Potential therapeutic benefits of bismuth subsalicylate include antibacterial effects, altered microbial adhesion, protection agains ulcerative effects, and decreased resistance to metronidazole (as reviewed by Leib and coworkers216). Placebos apparently were not given and blinding was not ascribed; assignment to either group was by coin toss, and although other therapies were discouraged, some dogs did receive other antibiotics as well as antiinflammatory therapies. Dogs were reevaluated at 4 weeks and 6 months. No significant treatment effect emerged in the famotidine group, with the frequency of vomiting reduced by 86% and organisms eradicated in approximately 75% of dogs in both groups. On the basis of this study, the authors concluded that famotidine did not enhance response to therapy; however, caution is recommended in basing therapy on this conclusion, in part becausethe ability of the study to detect a famotidine effect was not identified. In humans eradication of Helicobacter might be expected in more than 90% of human patients receiving “quadruple” (i.e., with antisecretory drugs) therapy. As with other investigators, recrudescence or re-infection of dogs with Helicobacter after presumably successful eradiction (based on the presence of the organism rather than molecular techniques) is not unusual. In Leib’s study216 close to 50% of dogs negative for Helicobacter at 4 weeks were positive by 6 months, suggesting improved therapy is still needed.

Liver Diseases

With few exceptions, treatment of liver disease is nonspecific, being primarily supportive and symptomatic.243 Feline hepatic lipidosis is largely a nutritionally managed disease and as such is not discussed in this chapter; however, future considerations should be made in the role of adipose hormones in the initiation or perpetuation of the syndrome. Recommended supplements include L-carnitine (250 to 500 mg/cat), taurine (250 mg), B vitamins at twice the standard recommended dose, vitamin C (30 mg/kg), vitamin E (100 to 400 mg/cat), and elemental zinc (7 to 8 mg/cat). Supplemenation with vitamin K (e.g., 0.5 to 1.5 mg/kg) also may be indicated.

Acute Hepatic Failure

Supportive therapy includes intensive fluid therapy with a balanced electrolyte solution to which potassium chloride, B vitamins, and (particularly in the presence of hypoglycemia or septicemia) glucose has been added. Coagulopathies are likely to reflect disseminated intravascular coagulopathy (stimulated by massive endothelial damage in the liver), impaired coagulation protein synthesis, or both. Clinical coagulopathies should be treated with heparin and replacement therapy (fresh whole blood or plasma or fresh frozen plasma). Rapid destruction of hepatic storage sites of vitamin K may also contribute to bleeding disorders, and replacement therapy may be indicated. Gastric ulceration should be anticipated and GI bleeding minimized by the use of antisecretory drugs. However, cimetidine is not recommended because of its negative effects on hepatic enzyme activity; omeprazole likewise might be used only cautiously. Antibiotics are indicated because of increased risk of bacteremia. Bacteria are likely to be gram-negative coliforms or anaerobes from the GI tract or Staphylococcus spp. Combination antimicrobial therapy is indicated for full antibacterial coverage. No documented studies have established the usefulness of drugs intended to support the liver as it heals or overcomes acute hepatic necrosis. Intrahepatic glutathione is an important scavenger of oxygen radicals, and its depletion probably contributes to inflammatory damage. Replacement in the form of acetylcysteine (e.g., Mucomyst) is certainly indicated for acetaminophen overdose but also might be considered in any case of acute hepatic necrosis. Cimetidine, a potent inhibitor of hepatic microsomal enzymes, might be considered in cases of acute hepatic failure associated with the formation of toxic drug metabolites, such as acetaminophen. However, its routine use in other cases of acute disease is discouraged because of its inhibitory effects.

KEY POINT 19-47

Acute drug hepatopathies might be treated with intravenous N-acetylcysteine.

Treatment for hepatic encephalopathy focuses on decreased absorption of encephalotoxins generated by microbes from protein and fat degradation. Medical management should be implemented in conjunction with dietary management. Lactulose is a semisynthetic disaccharide that is metabolized by colonic bacteria to lactic acid. In addition to the osmotic laxative effect, which causes evacuation of the luminal contents, acidification of the contents results in ionization of ammonia, precluding its absorption across the rectal mucosa. It can be administered either orally or, in severe cases of encephalopathy, as a retention enema (three parts lactulose to seven parts saline, administered at 20 mL/kg every 4 to 6 hours). The enema should be retained for 15 to 20 minutes. Lactitol is an alternative to lactulose that is less sweet and perhaps better tolerated. It is administered as a powder (500 mg/kg daily orally). Oral doses for long-term management with either lactulose or lactitol should generate two to three soft stools a day. Povidone–iodine (10%) given as an enema also acidifies luminal contents and provides some antibacterial activity. Selective microbial decontamination may reduce formation of encepahlotoxins. Neomycin (22 mg/kg orally twice daily or as an enema in water) also decreases bacteria responsible for formation of encephalotoxins. Other antimicrobials used for long-term management of hepatic encephalopathy include metronidazole (7.5 mg/kg orally every 8 to 12 hours) and ampicillin (22 mg/kg orally every 8 hours). With severe encephalopathy glucose-containing fluids may help prevent accumulation of ammonia in neurons.

Benzodiazepine receptors increase in patients with hepatic encephalopathy; use of benzodiazepine receptor antagonists such as flumazenil can be effective in human patients, but its efficacy is less well established in animals. If the drug is used, animals should be monitored for seizures. Intracranial pressures may increase in some patients; treatment should include mannitol (1 mg/kg of a 20% solution intravenously over 30 minutes, at 4-hour intervals) and furosemide. Glucocorticoids appear to offer no advantage to patients suffering from hepatic encephalopathy and may be contraindicated for treatment of increased intracranial pressure associated with hepatic encephalopathy.

Vomiting in patients with acute or chronic liver disease should be treated with antiemetics active at the CTZ or emetic center. Metoclopramide has been the first drug of choice, followed by a phenothiazine derivative; maropitant might reasonably replace either. Impaired hepatic function may increase the duration of action of the drug, whereas dehydration may increase plasma drug concentrations. Dosing regimens should take these changes into account. Volume replacement should take place before treatment with phenothiazine antiemetics in the dehydrated patient.

Chronic Hepatic Diseases

Halting hepatic inflammation

As with acute hepatic disease, treatment of chronic disease focuses on removal or correction of the inciting cause and supportive and symptomatic therapy. Long-term management should be accompanied by discontinuation of any drugs that are contributing to the chronic damage to the liver and dietary regimen. Drugs intended to remove the inciting cause are used in diseases for which the diagnosis is clear. For example, cecoppering agents are indicated in dogs predisposed to copper storage disease. However, Poldervaart and coworkers244 retrospectively reviewed hepatitis in dogs and concluded that the role of copper as a cause or contributor to acute or chronic hepatitis may be underestimated. Drugs used to treat copper-related hepatic disease include D-penicillamine (10 to 15 mg/kg orally 30 minutes before a meal, every 12 hours; start with a lower dose and increase after the first week) and, for animals that cannot tolerate D-penicillamine, trientine (2,2,2-tetramine, 10 to 15 mg/kg orally twice daily). In Bedlington Terriers with copper hepatotoxicosis, 2,3,2-tetramine (7.5 mg/kg orally every 12 hours) may be used instead of trientine (and may result in greater copper elimination), but the drug must be reformulated. A more controversial treatment for copper storage disease focuses on decreased absorption of copper in the diet by treatment with zinc acetate (5 to 10 mg/kg or 100 mg for the first 3 months and 50 mg thereafter orally every 12 hours, 1 hour before each meal). This treatment should be started at a young age, before hepatic accumulation of copper has occurred. Monitoring plasma zinc concentrations every 2 to 3 months has been recommended to ensure therapeutic concentrations and to prevent toxic concentrations of zinc that might lead to hemolytic anemia (therapeutic range of zinc is 200 to 500 μg/dL; higher than 1000 μg/dL is considered toxic).

Suppression of hepatic inflammation in chronic liver disease is problematic but critical if the progression of chronic to cirrhotic disease is to be halted. Underlying causes should be identified and removed. Consideration should be given to the role that insulin resistance, adioponectin, and adiponectin have in perpetuating the inflammatory response.4 Hepatic damage by drugs often is reversible if the drug is discontinued before fibrosis has occurred. Drug-induced hepatic disease is often dose and duration dependent, meaning that the risk of toxicity increases with higher doses (plasma drug concentrations) and long-term therapy. Consequently, single doses or short-term therapy with a hepatotoxic drug is not likely to lead to chronic hepatic disease. Examples of drugs associated with chronic liver disease in dogs are discussed in Chapter 4. Anticonvulsants (primidone, phenobarbital, and phenytoin) and heartworm preventive (oxibendazole–diethylcarbamazine) are among the most commonly used drugs associated with hepatic disease. However, any drug metabolized by the liver and to exposure is considerable (ie, large initial doses, or long duration of exposure) might be considered as a possible cause of liver disease.

Identifying the role of infection as a continued cause of liver disease may be difficult. However, with the exception of ascending chronic cholangiohepatitis, bacterial infection as a cause of chronic liver disease is uncommon. Because the liver is well perfused, any antimicrobial with a good gram-negative spectrum should be effective. However, as disease progresses and fibrosis deposition occurs, drugs that are more lipid-soluble should be considered Because of the loose but increasingly characterized association of Helicobacter spp. and cholangiohepatitis in cats,245 treatment for helicobacter might be considered.

Idiopathic chronic hepatitis (chronic active hepatitis or chronic active liver disease) is generally detected by increases in serum alanine transferase activity (greater than 10 times normal) and alkaline phosphatase activity (greater than 5 times normal). Biopsy should provide a confirmation as well as a histologic description on which therapy and response to therapy can be based. Inflammation usually is controlled with immunosuppressant drugs; evidence of piecemeal necrosis, bridging necrosis, and fibrosis indicates their need. Prednisolone (1 to 2 mg/kg orally a day) should be administered until clinical remission occurs (generally 7 to 10 days) and the dose then gradually tapered (decreased every 10 days) until a minimum effective dose has been established. Clinical signs, clinical pathologic changes (at 1- to 2-week intervals), and ultimately a repeat hepatic biopsy (at 2 to 3 months) should be monitored for response to therapy. Note that glucocorticoids can increase serum bile acids, and the failure of these to decrease is not necessarily indicative of continued damage. More aggressive immunosuppressive therapy is implemented if glucocorticoids cannot be tolerated or if the progression of hepatic disease cannot be halted with glucocorticoids. Azathioprine therapy is initiated (2 mg/kg/day or 50 mg/m2 orally given every day for 7 days, then every other day), with prednisolone therapy continued for the first 7 days and then alternated with azathioprine thereafter. Weekly white blood cell counts should be performed to detect bone-marrow suppression by azathioprine; therapy should be suspended for 5 to 7 days if the neutrophil count drops below 2000 cells/μL or the platelet count below 50,000/μL. Lymphocytic or sclerosing cholangitis/cholangiohepatitis in cats may also respond to glucocorticoid therapy (2.2 mg/kg orally a day). In order to avoid the adverse events associated with azaothioprine or other anti-inflammatory drugs, alternative therapies might be considered. Pentoxifylline is among these alternative drugs for additional control of inflammatory disease.246 The role of mycophenolate or cyclosporine in treating immune-mediated liver disease has not been addressed.

Ursodeoxycholic acid has proved beneficial in both dogs and cats with chronic liver disease, particularly if it is associated with a significant cholestatic component. The dose in patients with chronic hepatitis (8 to 10 mg/kg) is less than that in patients with primary biliary cirrhosis or sclerosing cholangitis (10 to 5 mg/kg/day). Note that more studies are needed to describe the clinical efficacy of ursodeoxycholic acid in dogs and cats. The drug appears to be safe; cats showed no evidence of adversity when dosed with 10 mg/kg orally for 8 weeks. Dehydrocholic acid (10 to 15 mg/kg orally every 12 hours) has also been recommended in cats with cholangiohepatitis and “ludged bile.” Note, however, that less evidence is available to support the efficacy of this bile acid and that it is among the lipid-soluble and thus potentially hepatotoxic bile acids. A deletion in an efflux transport protein has been identified as the underlying cause of biliarly mucoceole in dogs; its role in feline diseases has yet to be identified. Ascorbic acid (25 mg/kg/day orally) has been suggested as supportive therapy in dogs with chronic hepatic disease because the liver is less able to produce this vitamin. Zinc therapy has also been suggested to reduce copper deposition in the damaged liver.

When the progression of disease cannot be halted and, specifically, fibrotic tissue deposition continues, antifibrotic drugs can be considered. Prednisolone provides some prevention of collagen deposition. Colchicine appears to improve (histologically) the progression of cirrhosis in human patients, but evidence is lacking in dogs because of lack of controlled trials. Adverse reactions have not been reported in dogs receiving colchicine (0.03 mg/kg/day orally) for 6 to 30 months.

Sequelae of Chronic Liver Disease

Management of GI ulceration was previously discussed. As disease progresses, the likelihood of ulceration increases not only because of impairment of the mucosal barrier but also because of increased risk of bleeding resulting from coagulopathies. Bleeding into the GI tract increases the risk of hepatic encephalopathy. Treatment of hepatic encephalopathy was discussed under acute hepatic failure.

Control of ascites can be difficult with chronic disease. Fluid accumulation is more likely to reflect increased sodium and water retention (stimulated by portal hypertension) rather than decreased albumin, although hypoalbuminemia may contribute to ascitic fluid formation. Dietary restriction of sodium should be the targeted method by which ascitic fluid formation is controlled. If this is insufficient, diuretic therapies should be instituted. Because ascites may be associated with high aldosterone concentrations, spironolactone (1 to 2 mg/kg orally every 12 hours) might be the first diuretic used. The dose may be doubled in 1 week if there has been little response. Note that its cardioprotective effects (see Chapter 15) may also potential contribute to control of inflammation in the liver. Because spironolactone is a potassium-sparing diuretic, potassium supplementation may not be necessary and may be dangerous. If the patient continues not to respond to spironolactone, furosemide therapy can be instituted (1 to2 mg/kg orally every 8 to 12 hours initially and then titrated to a minimum effective dose daily, every other day, or every third day). Care must be taken not to dehydrate the patient. Total eradication of ascitic fluid need not be the goal of diuretic therapy.

Animals with chronic (including cirrhotic) liver disease are increasingly susceptible to bacterial infections and specifically to septicemia. However, routine use of antimicrobials is not recommended in order to avoid advent of resistance; fluoroquinolones in particular should be avoided routinely. Previous exposure to antibials should be considered as drugs are empirically selected to treat septicemia. Both gram-negative coliforms and anaerobes should be targeted with antimicrobial therapy.

As hepatic disease progresses to end-stage disease, note that patients are more susceptible to disseminated intravascular coagulation. This syndrome should be anticipated in patients and managed accordingly.

A review of of 41 relevant articles regarding the use of SAMe in human patients with liver disease, including a focus on cholestasis.179 Most studies enrolled only a small number of patients, and the quality of the studies were markedly variable. The review concluded that SAMe was more effective than placebo in reducing hyperbilirubinemia and pruritis associated with cholestasis, in studies comparing SAMe with traditional therapy (ursodeoxcycholic acid) for liver disease, although two clinical trials indicated that ursodeoxycholic acid was preferred to SAMe for cholestatic pruritis associated with pregnancy. The remaining studies were too diverse with regard to diagnosis to allow conclusions to be drawn.

Sixteen placebo-controlled clinical trials were reviewed in human patients with a variety of chronic liver diseases who were receiving milk thistle. As with the SAMe studies, poor study methods or reporting limited effective evaluation, causing reviewers to have difficulty with interpretation of results. In general, meta-analyses indicated small treatment effects, with some statistical significance favoring milk thistle for treatment of selected liver disorders based on improvements in aminotransferases and liver function tests. Milk thistle was associated with few adverse events, and these generally were considered minor.247 Cholangitis and cholangiohepatitis in the cat are frequently associated with IBD or pancreatitis. In the cat, the common association may reflect the proximity of the bile duct to the pancreatic duct. Treatment is similar to that for chronic liver disease, including SAMe and silymarin, choleretics, and antimicrobials (to decrease bacterial and toxin load), which target gastrointestinal microflora. Immune suppression is indicated in nonsupprative disease, particularly if associated with IBD. In the dog, the presence of fibrosis may indicate the need for antifibrotics (e.g., colchicine or elemental zinc).

Portosystemic Shunting

In his original report of cerebrospinal fluid concentrations of potential mediators of hepatic encephalopathy, Holt and coworkers248 reviewed the pathophysiology of neurologic abnormalities associated with portosystemic shunting.

Among the proposed altered mediators, which might serve as targets of drug therapy, are monoamine and amino acid neurotransmitter systems, endogenous benzodiazepines and their receptors, and ammonia. Upregulation of genes encoding peripheral GABA-like receptors has been proposed as a cause of altered neurotransmission. Accumulation of potential neurotoxins, including GABA, has also been proposed. Others include include short-chain fatty acids, mercaptans, false neurotransmitters such as tyramine, octopamine, beta-phenylethanolamines; manganese, and ammonia. Ammonia is associated with increased cerebrospinal fluid tryptophan, possibly because of direct stimulation by way of the neutral amino acid transport proteins at the blood–brain barrier. In the absence of a urea cycle, CNS ammonia is removed by transamination of glutamate into glutamine, which occurs in astrocytes by way of a specific astroglial enzyme in rats. Glutamine, in turn, however, may competively inhibit blood–brain barrier transport proteins responsible for efflux of the large, neutral amino acids. Supporting this mechanism, clinical signs of hepatic encephalopathy resolved in rats with portosystemic shunting treated with methionine sulfoximine, an inhibitor of glutamine synthetase.

A Cochrane review of clinical trials studying the use of probiotics for treatment of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis found insufficient evidence to support or refute treatment. However, the use of probiotics and synbiotics to support liver function or treat liver also disease has been reviewed by Lenoir-Wijnkoop and coworkers.151 The rationale reflects the impact that these agents might have on the microbiota of the gut–liver axis, a term used in human medicine to refer to the impact that gut-derived endotoxins and active metabolites have on the liver. Upregulation of proinflammatory cytokines may contribute to inflammation progressing to fibrosis and lipid peroxidation. Theoretically, modulation of the gut microflora might reduce these detrimental microbiota effects. Again, in a mouse model, a probiotic containing three species of Bifidobacterium, four species of Lactobacillus, and Streptococcus thermophilus decreased the extent of (alcohol-induced) liver disease. In a human clinical trial, treatment of a synbiotic containing fructooligosaccharides, Bifidobacterium, and seven species of Lactobacillus (L. acidophilus,L. rhamnosus, L. plantarum, L. salivarius, L. bulgaricus, L. lactis, L. breve plus) decreased liver enzymes and (in the alcoholic group) increased hepatic function in patients with nonalcoholic fatty liver disease and alcoholic cirrhosis. More intriguing, a prospective, randomized study in human patients with liver cirrhosis associated with minimal hepatic encephalopathy positively responded to a symbiotic containing four probiotic non–urease-producing strains (L. plantarum, L. paracasei, Leuconostoc mesenteroides, Pediococcus pentosaceus) and four fibers (beta-glucan, resistant starch, inulin, pectin). The fecal microbiota was recolonized with non–urease-producing Lactobacillus spp., urinary pH decreased along with serum ammonia and endocoxin. Hepatic encephalopathy was reversed in 50% of patients, and hepatic function improved in 50% of patients receiving the symbiotic.

Diseases of the Pancreas and Acute Pancreatitis

The combination of feline trypsinogen-like imunoreactivity and abdominal ultrasound findings appear to be able to diagnose feline pancreatitis with high sensitivity and specificity.249 Steiner et al provides evidence that exocrine pancreatic insufficiency does occur in cats and can be diagnosed on the basis of feline trypsin-like immunoactivity.250 Cobalamin but not folic acid absorption is impaired in exocrine pancreatic insufficiency. Decreased folic acid indicates concurrent intestinal disease.

Medical management of acute pancreatitis is supportive and symptomatic, allowing the pancreas to “rest” and heal. Drugs that may contribute to pancreatitis251,252 should be discontinued. Suspected drugs include thiazide diuretics, furosemide, azathioprine, L-asparaginase, sulfonamides, and tetracyclines. Glucocorticoids, bromide, phenobarbital, and H2-receptor antagonists have also been implicated. Glucocorticoids may impair macrophage clearance of alpha-macroglobulin complexes (protease inhibitors complexed with proteolytic enzymes), thus predisposing the pancreas to stimulation by CCK.

Animals should be fasted to prevent pancreatic stimulation. Fluid therapy consisting of balanced electrolytes should be administered for at least 3 to 4 days, depending on the severity of the case. Electrolytes and acid–base therapy should be monitored; hypokalemia should be anticipated and treated accordingly. Because of the risk of subclinical hypocalcemia, sodium bicarbonate should be used cautiously because alkalosis can precipitate a hypocalcemic episode in these patients. Antiemetics should be used in animals that continue to vomit. Ideally, a drug that acts both centrally and peripherally, such as metoclopramide, should be chosen.

Analgesic therapy is indicated in patients with moderate to severe pain. Opioid analgesics such as butorphanol or buprenorphine should be considered. Meperidine has been recommended as well, although its short duration of action may preclude effective use. Fresh whole blood or plasma may replace alpha macroglobulins responsible for clearing the pancreas of proteolytic enzymes and may increase plasma albumin. This may be important, particularly in the case of severe pancreatitis or that associated with disseminated intravascular coagulation. The advent of disseminated intravascular coagulationshould be treated accordingly.

Protease inhibitors such as aprotinin (250 mg or 1,500,000 kallikrein inhibitory units intraperitoneally every 6 to 8 hours) may be more effective in dogs than in humans because of differences in potency.253 However, the drug may be prohibitively expensive. Alternatively, 5000 kallikrein inhibitory units/kg intravenously every 6 hours has been recommended but is not as preferred as intraperitoneal injection. Selenium (0.1 mg/kg every 24 hours by intravenous infusion administered as selenious acid [40 μg/mL]) may be helpful.

Because oxidative stress plays a major role in the early stage of acute pancreatitis, antioxidant therapy might be considered. Among the antioxidants studied is N-acetylcysteine, which intracellularly is converted to a reduced GSH provider, which directly scavenges reactive oxygen species. N-acetyl-cysteine (1000 mg/kg every 3 hours intraperitoneal [IP]) was effective in reducing outcome measures (cytokines, conjugated dienes, lung injury, survival) associated with acute pancreatic in a mice ceruline or diet model when administered prophylactically (1 hour before induction or with the diet).253 Glucocorticoid therapy is controversial because of the potential for these drugs to contribute to pancreatitis. Even in patients suffering from shock, the role of glucocorticoids is not clear. However, it is unlikely that a very short-term administration of glucocorticoid therapy (i.e., one to two doses) will be harmful in patients with fulminating pancreatitis. Methylprednisolone succinate is probably preferred because of the oxygen-scavenging ability of this glucocorticoid compared to others. Inhibition of gastric secretions with H2-receptor antagonists, proton pump inhibitors, antacids, or drugs targeting the pancreas and its secretion (e.g., atropine, calcitonin, and somatostatin) has not yet proved effective for the treatment of acute pancreatitis. With time, natural or synthetic enzyme inhibitors directed toward pancreatic secretions may become useful (and available). In very acute cases or repetitive cases, insulin therapy may be indicated in the presence of persistent hyperglycemia indicative of diabetes mellitus.

Manipulation of microflora may be a target of treatment for pancreatitis. The role of parenteral antibiotics is not clear in the treatment of acute pancreatitis, and caution is recommended because of the risk of emergent multidrug-resistant microorganisms. If the decision is made to use parenteral antimicrobials, a number of drugs will penetrate the pancreas effectively. As with any infection involving the abdomen, gram-negative coliforms should be the primary target, but anaerobes should not be overlooked. Trimethoprim–sulfonamide combinations have been suggested, although sulfonamides are one of the groups of drugs implicated in the cause of pancreatitis. Bacterial depopulation of the GI tract by antimicrobials decreases the risk of bacterial translocation, thus potentially reducing the incidence of systemic organ failure. However, the advent of multidrug-resistant bacteria is negatively affecting widespread implementation of antibacterial prophylaxis. The use of probiotics for treatment of pancreatitis was reviewed in humans.151 Use targets that small proportion of patients for which acute pancreatitis shifts from mild to severe and life threatening. As reviewed by Lenoir-Wijnkoop and coworkers,151 a clinical trial in humans found the incidence of pancreatic necrosis to be less (1 of 22, or 5%) in a group receiving a probiotic compared with a group that received a heat-inactivated probiotic (7 of 23, or 30%). A multicenter clinicial trial in Europe is further investigating the use of B. bifidum W23, B. infantis W52, L. acidophilus W70, L. casei W56, Lactobacillus salivarius W24, and Lactococcus lactis W58; these specific organisms were selected on the basis of their ability to survive in the GI environment associated with pancreatitis. In a rat model, probiotic use increased survival in acute pancreatitis. Although the use of probiotics in prevention of acute pancreatitis appears promising, the potential risk for (pathogenic) colonization of the necrotic pancreas with the probiotic microorganisms has not been effectively addressed. In human medicine the advent of pancreatitis actually has been associated with IBD. Chronic pancreatitis may reflect generation of autoantibodies toward pancreatic acinar cells, which may reflect hapten formation and drug allergies.255

A number of drugs used (in humans) to treat IBD have been associated with the advent of acute pancreatitis in those patients. These include mercaptopurine, azathioprine, coricosteroids, sulfasalazine, and 5-aminosalicylic acid products.255 For the former, treatment of chronic pancreatitis should focus primarily on treatment of IBD, whereas the latter includes discontinuation of the inciting drug.

Exocrine Pancreatic Insufficiency

Clinical signs associated with exocrine pancreatic insufficiency (e.g., diarrhea, weight loss, polyphagia) reflect decreased intraduodenal concentrations of pancreatic enzymes and, to a lesser degree, bicarbonate or other materials. These deficiencies result in malassimilation of fats, carbohydrates, proteins, fat-soluble vitamins, and cobalamin. Additionally, the number and composition of the small intestinal bacterial flora may change, contributing to the clinical signs. Accordingly, supplementation of B vitamins and cobalamin and treatment with probiotics should be considered.

Medical management of exocrine pancreatic deficiency should be supported by dietary management. Enzyme replacement using commercially available products should be sufficient in most animals. Use of the powder in two daily feedings (two teaspoons of the nonenteric product per 20 kg) with each meal should resolve diarrhea within 3 to 4 days and promote weight gain. Because commercial dried pancreatic extracts are relatively expensive, chopped pig or cow pancreas (certified as healthy) can be used (3 to 4 ounces per 20 kg) in lieu of the commercial preparation. Fresh pancreatic tissue can be frozen for 3 to 4 months without apparent loss of pancreatic enzyme activity.

Commercial powders are not particularly efficient; much of the enzyme activity is rapidly lost due to inactivation by gastric acidity. For animals that do not respond to therapy initially, attempts can be made to improve the action of the enzymes. Of the methods suggested to improve efficiency or reduce gastric loss (including preincubation with food and addition of bile acids), inhibition of gastric acid secretions appears most useful. An H2-receptor antagonist can be given with food or, to further improve efficacy, 30 to 60 minutes before feeding. Enteric coating not only does not appear to improve efficiency but may further decrease availability of the enzymes.

Supplementation of vitamin B12 (250 μg intramuscularly or subcutaneously once weekly for 1 month) and vitamin A (tocopherol; 400 to 500 IU once daily with food for 30 days) may be necessary for some patients and might be considered in animals in whom diarrhea persists despite enzyme replacement. Bacterial overgrowth may become a problem in some patients because of the presence of undigested nutrients that serve as a nutrient source for bacteria. Long-term antibiotic therapy is discouraged because of the risk of altered microflora and damage to the GI mucosa. Short-term therapy with oral metronidazole, tylosin, or oxytetracycline should prove beneficial in cases where bacterial overgrowth is causing malabsorption and diarrhea. Occasionally, animals may also have IBD, which contributes to clinical signs. Treatment with glucocorticoids may be indicated for 7 to 14 days.

References

1. Sugrue M.F. Neuropharmacology of drugs affecting food intake. Pharmacol Ther. 1987;32:145-182.

2. Havel P.J., Section I.V. lipid modulators of islet function, update on adipocyte hormones, regulation of energy balance and carbohydrate/lipid metabolism. Diabetes. 2004;53(Suppl 1):S143-S151.

3. Margetic S., Gazzola C., Pegg G.G., et al. Leptin: a review of its peripheral actions and interactions. Int J Obes. 2002;26:1407-1433.

4. Tsochatzis E.T., Manolakopoulos S., Papatheodoridis G.V., et al. Insulin resistance and metabolic syndrome in chronic liver diseases: old entities with new implications. Scand J Gastroenterol. 2009;44(1):6-14.

5. Spaulding M. Recent studies of anorexia and appetite stimulation in the cancer patient. Oncology. 1989;3(Suppl 8):17-23.

6. Bernstein J.G. Psychotropic drug induced weight gain: mechanisms and management. Clin Neuropharmacol. 1988;11:S194-S206.

7. Timmer C.J., Ad Sitsen J.M., Delbressine L.P. Clinical pharmacokinetics of mirtazapine. Clin Pharmacokinet. 2000;38(6):461-474.

8. Macy D.W., Gasper P.W. Diazepam-induced eating in anorexic cats. J Am Anim Hosp Assoc. 1985;21:17-20.

9. Center S.A., Elston T.H., Rowland P.H., et al. Fulminant hepatic failure associated with oral administration of diazepam in 11 cats. J Am Vet Med Assoc. 1996;209(3):618-625.

10. Hughes D., Moreau R.E., Overall K.K., et al. Acute hepatic necrosis and liver failure associated with benzodiazepam therapy in six cats, 1986-1995. J Vet Am Crit Care. 1997;6:13-20.

11. Norris C.R., Boothe D.M., Esparza T., et al. Disposition of cyproheptidine in cats after intravenous or oral administration of a single dose. Am J Vet Res. 1998;59:79-82.

12. Femia R.A., Goyette R.E. The science of megestrol acetate delivery. Biodrugs. 2005;19(3):179-187.

13. Loprinzi C.L., Kugler J.W., Sloan J.A., et al. Randomized comparison of megestrol acetate versus dexamethasone versus fluoxymesterone for the treatment of cancer anorexia/cachexia. J Clin Oncol. 1999;17(10):3299-3306.

14. Miller K.J. Serotonin 5-ht2c receptor agonists: potential for the treatment of obesity. Mol Interv. 2005;5(5):282-291.

15. Long J.P., Greco S.C. The effect of propofol administered intravenously on appetite stimulation in dogs. Contemp Top Lab Anim Sci. 2000;39(6):43-46.

16. Johnson S.E. Clinical pharmacology of antiemetics and antidiarrheals. Proc Kal Kan Waltham Symp Treat Small Anim Dis. 1984;8:7-15.

17. Johnson S.E. Clinical pharmacology of antiemetics and antidiarrheals. Gastroenterology. 1985;80:1008.

18. Andrews P.L.R., Rapeport W.G., Sanger G.J. Neuropharmacology of emesis induced by anti-cancer therapy. Trends Pharmacol Sci. 1988;9:334-341.

19. Merrifield K.R., Chaffee B.J. Recent advances in the management of nausea and vomiting caused by antineoplastic agents. Clin Pharmacol. 1989;8:187-199.

20. Saito R., Takano Y., Kamiya H.O. Roles of substance P and NK(1) receptor in the brainstem in the development of emesis. J Pharmacol Sci. 2003;91(2):87-94.

21. Diemunsch P., Grelot L. Potential of substance P antagonists as antiemetics. Drugs. 2000;60:533-546.

22. Hickman M.A., Cox S.R., Mahabir S., et al. Safety, pharmacokinetics and use of the novel NK-1 receptor antagonist maropitant (CereniaTM) for the prevention of emesis and motion sickness in cats. J Vet Pharmacol Therap. 2008;31:220-229.

23. Kohler D.R., Goldspiel B.R. Ondansetron: a serotonin receptor (5-HT3) antagonist for antineoplastic chemotherapy-induced nausea and vomiting. Ann Pharmacother. 1991;25:367-380.

24. Pasricha P.J. Treatment of disorders of bowel motility and water flux, antiemetics, agents used in biliary and pancreatic disease. 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.

25. Hikasa Y., Ogasawara S., Takase K. Alpha adrenoceptor subtypes involved in the emetic action in dogs. J Pharmacol Exp Ther. 1992;261:746-754.

26. Hikasa Y., Takase K., Ogasawara S. Evidence for the involvement of alpha-adrenoceptors in the emetic action of xylazine in cats. Am J Vet Res. 1989;50:1348-1350.

27. Peroutka S.J., Snyder S.H. Antiemetics: neurotransmitter receptor binding predicts therapeutic actions. Lancet. 1982;1:658-659.

28. Costall B., Naylor R.J. Neuropharmacology of emesis in relation to clinical response. Br J Cancer Suppl. 1992;19:S2-S8.

29. Hesketh P.J., Younger J., Sanz-Altamira P., et al. Aprepitant as salvage antiemetic therapy in breast cancer patients receiving doxorubicin and cyclophosphamide. Support Care Cancer. Dec 6, 2008.

30. Conder G.A., Sedlacek H.S., Boucher J.F., et al. Efficacy and safety of maropitant, a selective neurokinin 1 receptor antagonist, in two randomized clinical trials for prevention of vomiting due to motion sickness in dogs. J Vet Pharmacol Ther. 2008;31(6):528-532.

31. de la Puente-Redondo V.A., Siedek E.M., Benchaoui H.A., et al. The anti-emetic efficacy of maropitant (Cerenia™) in the treatment of ongoing emesis caused by a wide range of underlying clinical aetiologies in canine patients in Europe. J Small Anim Pract. 2007;48:93-98.

32. Sedlacek H.S., Ramsey D.S., Boucher J.F., et al. Comparative efficacy of maropitant and selected drugs in preventing emesis induced by centrally or peripherally acting emetogens in dogs. J Vet Pharmacol Ther. 2008;31(6):533-537.

33. Tyers M.B. Pharmacology and preclinical antiemetic properties of ondansetron. Semin Oncol. 1992;19(Suppl 10):1-8.

34. Reynolds J.C. Prokinetic agents: a key in the future of gastroenterology. Gastroenterol Clin North Am. 1989;18:437-457.

35. Burrows C.F. Metoclopramide. J Am Vet Med Assoc. 1983;183:1341-1343.

36. Urbie M., Ballesteros A., Strauss R., et al. Successful administration of metoclopramide for the treatment of nausea in patients with advanced liver disease. Gastroenterology. 1985;88:757-762.

37. Albibi R., McCallum R.W. Metoclopramide: pharmacology and clinical application. Arch Intern Med. 1983;98:86-95.

38. Shinkai T., Saijo N., Eguchi K., et al. Control of cisplatin-induced delayed emesis with metoclopramide and dexamethasone: a randomized controlled trial. Jpn J Clin Oncol. 1989;19:40-44.

39. Gralla R.J., Itri L.M., Pisko S.E., et al. Antiemetic efficacy of high-dose metoclopramide: randomized trials with placebo and prochlorperazine in patients with chemotherapy-induced nausea and vomiting. N Engl J Med. 1981;305:905-909.

40. Howard J.M., Chremos A.N., Collen M.J., et al. Famotidine, a new, potent, long-acting histamine H2-receptor antagonist: comparison with cimetidine and ranitidine in the treatment of Zollinger–Ellison syndrome. Gastroenterology. 1985;88:1026-1033.

41. Aapro M., Johnson J. Chemotherapy-induced emesis in elderly cancer patients: the role of 5-HT3-receptor antagonists in the first 24 hours. Gerontology. 2005;51(5):287-296.

42. Burrows C.F. Ondansetron: a new drug to control nausea and emesis in cancer patients. Vet Clin Briefs. 1990;8:3.

43. Dow J., Francesco G.F., Berg C. Comparison of the pharmacokinetics of dolasetron and its major active metabolite, reduced dolasetron, in dog. J Pharm Sci. 1996;85(7):685-689.

44. Ogilvie G.K. Dolasetron: a new option for nausea and vomiting. J Am Anim Hosp Assoc. 2000;36:481-483.

45. Robert A., Kauffman G.L.Jr. Stress ulcers, erosions and gastric mucosal injury. In: Sleisenger M.H., Fordtran J.S., editors. Gastrointestinal diseases: pathophysiology, diagnosis, management. ed 4. Philadelphia: Saunders; 1989:773-855.

46. Moreland K.J. Ulcer disease of the upper gastrointestinal tract in small animals: pathophysiology, diagnosis and management. Compend Contin Educ Pract Vet. 1988;10:1265-1279.

47. Kleiman R.L., Adair C.G., Ephgrave K.S. Stress ulcers: current understanding of pathogenesis and prophylaxis. DICP Ann Pharmacother. 1988;22:452-460.

47a. Abelö A., Holstein B., Eriksson U.G., et al. Gastric acid secretion in the dog: a mechanism-based pharmacodynamic model for histamine stimulation and irreversible inhibition by omeprazole. J Pharmacokinet Pharmacodyn. 2002;29(4):365-382.

48. Hoogerwerf W.A., Pasricha P.J. Pharmacotherapy of gastric acidity, peptic ulcers, and gastroesophageal reflux disease. 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. 2006

49. Wolfe M.M., Soll A.H. The physiology of gastric acid secretion. N Engl J Med. 1988;319:1707-1715.

50. Akimoto M., Nagahata N., Furuya A., et al. Gastric pH profiles of beagle dogs and their use as an alternative to human testing. Eur J Pharm Biopharm. 2000;49(2):99-102.

51. Abelö A., Holstein B., Eriksson U.G., et al. Gastric acid secretion in the dog: a mechanism-based pharmacodynamic model for histamine stimulation and irreversible inhibition by omeprazole. J Pharmacokinet Pharmacodyn. 2002;29(4):365-382.

52. Baker K.J. Binding of sulfobromophthalein sodium and indocyanine green by plasma alpha1 lipoproteins. Proc Soc Exp Biol Med. 1966;122:957-963.

53. Toutain P.L., Alvinerie M., Ruckebusch Y. Pharmacokinetics of dexamethasone and its effect on adrenal gland function in the dog. Am J Vet Res. 1983;44:212-217.

54. Shorrock C.J., Rees W.D.W. Overview of gastroduodenal mucosal protection. Am J Med. 1988;84(Suppl 2A):25-34.

55. Miller T.A. Protective effects of prostaglandins against gastric mucosal damage: current knowledge and proposed mechanisms. Am J Physiol. 1983;245:G601-G623.

56. Charlet N., Gallo-Torres H.E., Bounameaux Y., et al. Prostaglandins and the protection of the gastroduodenal mucosa in humans: a critical review. J Clin Pharmacol. 1985;25:564-582.

57. Szelenyi I., Brune K. Possible role of sulfhydryls in mucosal protection induced by aluminum hydroxide. Dig Dis Sci. 1986;31:1207-1210.

58. Whittle B.J.R., Garner A. New targets for anti-ulcer drugs. Trends Pharmacol Sci. 1988;9:187-189.

59. Muir W.W. Small volume resuscitation using hypertonic saline. Cornell Vet. 1990;80:7-12.

60. Krishna D.R., Ulrich K. Newer H2-receptor antagonists clinical pharmacokinetics and drug interaction potential. Clin Pharmacokinet. 1988;15:205-215.

61. Hirschowitz B.I., Gibson R.G. Effect of cimetidine on stimulated gastric secretion and serum gastrin in the dog. Am J Gastroenterol. 1987;70:437-447.

62. Bemis K., Bendele A., Clemens J., et al. General pharmacology of nizatidine in animals. Drug Res. 1989;39:240-250.

63. Coruzzi G., Bertaccini G., Noci M.T., et al. Inhibitory effect of famotidine on cat gastric secretion. Agents Actions. 1986;19(3-4):188-193.

64. Price A.H., Brogden R.N. Nizatidine: a preliminary review of its pharmacodynamic and pharmacokinetic properties, and its therapeutic use in peptic ulcer disease. Drugs. 1988;36:521-539.

65. Brogden R.N., Carmine A.A., Heel R.C., et al. Ranitidine: a review of its pharmacology and therapeutic use in peptic ulcer disease and other allied diseases. Drugs. 1982;24:267-303.

66. Boom S.P., Hoet S., Russel F.G. Saturable urinary excretion kinetics of famotidine in the dog. J Pharm Pharmacol. 1997;49(3):288-292.

67. Ames T.R., Patterson E.B. Pharmacokinetics of a long-acting oxytetracycline injectable in healthy and diseased calves. Proc 13th World Cong Dis Cattle. 1984;2:931-935.

68. Sedman A.J. Cimetidine-drug interactions. Am J Med. 1984;76:109-114.

69. Gibaldi M. Drug interactions: part I. Ann Pharmacother. 1992;26:709-713.

70. Jackson J.E. Cimetidine protects against acetaminophen toxicity. Life Sci. 1982;31:31-35.

71. Jackson J.E. Reduction of liver blood flow by cimetidine. N Engl J Med. 1981;307:99-101.

72. Guharoy S.R. Streptokinase versus recombinant tissue-type plasminogen activator. DICP Pharmacother. 1991;25:1271-1272.

73. el-Omar E., Banerjee S., Wirz A., et al. Marked rebound acid hypersecretion after treatment with ranitidine. Am J Gastroenterol. 1996;91:355-359.

74. Fullarton G.M., MacDonald A.M.I., McColl K.E.L. Rebound hypersecretion after H2-antagonist withdrawal: a comparative study with nizatidine, ranitidine and famotidine. Aliment Pharmacol Ther. 1991;5:391-398.

75. Marks I.N., Johnston D.A., Young G.O. Acid secretory changes and early relapse following duodenal ulcer healing with ranitidine or sucralfate. Am J Med. 91(Suppl 2A), 1991. 2A-95S–2A-100S

76. Yamaji Y., Abe T., Omata T., et al. Effects of successive doses of nizatidine, cimetidine and ranitidine on serum gastrin level and gastric acid secretion. Drug Res. 1991;41:954-957.

77. Larsen K.R., Dajani E.Z., Ives M.M. Antiulcer drugs and gastric mucosal integrity effects of misoprostol, 16, 16-dimethyl PGE2, and cimetidine on hemodynamics and metabolic rate in canine gastric mucosa. Dig Dis Sci. 1992;37:1029-1037.

78. Boudville N. The predictable effect that renal failure has on H2 receptor antagonists—increasing the half-life along with increasing prescribing errors. Nephrol Dial Transplant. 2005;20:2315-2317.

79. Lampkin T.A., Ouellet D., Hak L.J., et al. Omeprazole: a novel antisecretory agent for the treatment of acid-peptic disorders. DICP Ann Pharmacother. 1990;24:393-402.

80. Tutuian R., Katz P.O., Ahmed F., et al. Over-the-counter H(2)-receptor antagonists do not compromise intragastric pH control with proton pump inhibitors. Aliment Pharmacol Ther. 2002;16(3):473-477.

81. Fändriks L., Lönroth H., Pettersson A., et al. Can famotidine and omeprazole be combined on a once-daily basis? Scand J Gastroenterol. 2007;42(6):689-694.

82. Larsson H., Mattsson H., Carlsson E. Gastric acid antisecretory effect of two different dosage forms of omeprazole during prolonged oral treatment in the gastric fistula dog. Scand J Gastroenterol. 1988;23:1013-1019.

83. Säfholm C., Havu N., Forssell H., et al. Effect of 7 years’ daily oral administration of omeprazole to beagle dogs. Digestion. 1994;55(3):139-147.

84. Andersson T. Omeprazole drug interaction studies. Clin Pharmacokinet. 1991;21:195-212.

85. Pauli-Magnus C., Rekersbrink S., Klotz U., et al. Interaction of omeprazole, lansoprazole and pantoprazole with P-glycoprotein. Naunyn Schmiedebergs Arch Pharmacol. 2001;364(6):551-557.

85a. Li X.Q., Andersson T.B., Ahlstron M., et al. Comparison of inhibitory effects of the proton pump-inhibiting drugs omeprazole, esomeprazole, lansoprazole, pantoprazole, and rebeprazole on human cytochrome P450 activities. Drug Metabolism and Disposition. 2004;32(8):821-827.

86. Waldon H.L., Arnestad J.S., Brenna E., et al. Marked increase in gastric acid secretory capacity after omeprazole treatment. Gut. 1996;39:649-653.

87. Bersenas A.M., Mathews K.A., Allen D.G., et al. Effects of ranitidine, famotidine, pantoprazole, and omeprazole on intragastric pH in dogs. Am J Vet Res. 2005;66(3):425-431.

88. Misoprostol Monograph: Misoprostol: Preclinical and Clinical Review. Glenview, Ill: Physicians and Scientists Publishing Co; 1990.

89. Jones J.B., Bailey R.T.Jr. Misoprostol: a prostaglandin E1 analog with antisecretory and cytoprotective properties. DICP Ann Pharmacother. 1989;23:276-282.

90. Babakhin A.A., Nolte H., DuBuske L.M. Effect of misoprostol on the secretion of histamine from basophils of whole blood. Ann Allergy Asthma Immunol. 2000;84:361-365.

91. Morrissey J.F., Barreras R.F. Drug therapy, antacid therapy. N Engl J Med. 1974;290:550-554.

92. Vergin H., Kori-Lindner C. Putative mechanisms of cytoprotective effect of certain antacids and sucralfate. Dig Dis Sci. 1990;35:1320-1327.

93. Siepler J.K., Mahakian K., Trudeau W.T. Current concepts in clinical therapeutics: peptic ulcer disease. Clin Pharmacol. 1986;5:128-142.

94. Steinberg W.M., Lewis J.H., Katz D.M. Antacids inhibit absorption of cimetidine. N Engl J Med. 1982;307:400-404.

95. Untersmayr E., Schöll I., Swoboda I., et al. Antacid medication inhibits digestion of dietary proteins and causes food allergy: a fish allergy model in Balb/c mice. J Allergy Clin Immunol. 2003;112:616-623.

96. McCarthy D.M. Sucralfate. Drug Ther. 1991;325:1017-1023.

97. Tarnawski A., Hollander D., Gergely H. The mechanism of protective, therapeutic and prophylactic actions of sucralfate. Scand J Gastroenterol. 1987;22(Suppl 140):7-13.

98. Hickey A.R., Wenger T.L., Carpenter V.P., et al. Digoxin immune fab therapy in the management of digitalis intoxication: safety and efficacy results of an observational surveillance study. J Am Coll Cardiol. 1991;17:590-598.

99. Hollander D., Tarnawski A. The protective and therapeutic mechanisms of sucralfate. Scand J Gastroenterol. 1990;25(Suppl 173):1-5.

100. Konturek S.J., Brzozowski T., Drozdowicz D., et al. Role of acid milieu in the gastroprotective and ulcer-healing activity of sucralfate. Am J Med. 91, 1991. 2A-20S–2A-29S

101. Jensen S.L., Jensen P.F. Role of sucralfate in peptic disease. Dig Dis. 1992;10:153-161.

102. Furukawa O., Matsui H., Suzuki N. Effects of sucralfate and its components on acid- and pepsin-induced damage to rat gastric epithelial cells. Jpn J Pharmacol. 1997;75(1):21-25.

103. Slomiany B.L., Piotrowski J., Tamura S., et al. Enhancement of the protective qualities of gastric mucus by sucralfate: role of phosphoinositides. Am J Med. 91(Suppl 2A), 1991. 2A-30S–2A-36S

104. Wada K., Kamisaki Y., Kitano M., et al. Effects of sucralfate on acute gastric mucosal injury and gastric ulcer induced by ischemia-reperfusion in rats. Pharmacology. 1997;54(2):57-63.

105. Slomiany B.L., Piotrowski J., Slomiany A. Cell cycle progression during gastric ulcer healing by ebrotidine and sucralfate. Gen Pharmacol. 1997;29(3):367-370.

106. Konturek S.J., Brzozowski T., Majka J., et al. Role of nitric oxide and prostaglandins in sucralfate-induced gastroprotection. Eur J Pharmacol. 1992;211:277-279.

107. Szabo S., Vattay P., Scarbough E., et al. Role of vascular factors, including angiogenesis, in the mechanisms of action of sucralfate. Am J Med. 19(Suppl 2A), 1991. 2A-158S–2A-160S

108. Katz P.O., Geisinger K.R., Hassan M., et al. Acid-induced esophagitis in cats is prevented by sucralfate but not synthetic prostaglandin E. Dig Dis Sci. 1988;33:217-224.

109. Holzer P. Opioids and opioid receptors in the enteric nervous system: from a problem in opioid analgesia to a possible new prokinetic therapy in humans. Neurosci Lett. 2004;6;361(1-3):192-195.

110. Crema A., De Ponti F. Recent advances in the physiology of intestinal motility. Pharmacol Res. 1989;21:67-73.

111. Grider J.R. Reciprocal activity of longitudinal and circular muscle during intestinal peristaltic reflex. Am J Physiol Gastrointest Liver Physiol. 2003;284(5):G768-75.

112. Wilson D.V., Evans A.T., Mauer W.A. Influence of metoclopramide on gastroesophageal reflux in anesthetized dogs. Am J Vet Res. 2006;67(1):26-31.

113. Demol P., Ruoff H.-J., Weihrauch T.R. Rational pharmacotherapy of gastrointestinal motility disorders. Eur J Pediatr. 1989;148:489-495.

114. Hedge S.S., Eglen R.M. Peripheral 5-HT-4 receptors. FASEB J. 1996;10:1398-1407.

115. Tinker J., Cox A.G. Effect of metoclopramide on transport in the small intestine of the dog. Gut. 1969;10(2):986-989.

116. Hunt J.M., Gerring E.L. A preliminary study of the effects of metoclopramide on equine gut activity. J Vet Pharmacol Ther. 1986;9:109-112.

117. Wingate D., Pearce E., Hutton M., et al. Effect of metoclopramide on interdigestive myoelectric activity in the conscious dog. Dig Dis Sci. 1980;25:15-21.

118. Sojka J.E., Adams S.B., Lamar C.H., et al. Effect of butorphanol, pentazocine, meperidine, or metoclopramide on intestinal motility in female ponies. Am J Vet Res. 1988;49:527-529.

119. Clark E.S., Becht J.L. Clinical pharmacology of the gastrointestinal tract. Vet Clin North Am Equine Pract. 1987;3:101-123.

120. Cox S.R., Harrington E.L., Capponi V.J. Bioavailability studies with ciglitazone in beagles. II. Effect of propantheline bromide and metoclopramide HCL on bioavailability of a tablet. Biopharm Drug Dispos. 1985;6(1):81-90.

121. Prados A.P., Kreil V., Albarellos G., et al. Metoclopramide modifies oral cephalexin pharmacokinetics in dogs. J Vet Pharmacol Therap. 2007;30:127-131.

122. Takahashi T., Kurosawa S., Wiley J.W., et al. Mechanism for the gastrokinetic actions of domperidone. Gastroenterology. 1991;101:703-710.

123. McCallum R.W., Prakash C., Campoli-Richards D.M., et al. Cisapride: a preliminary review of its pharmacodynamics and pharmacokinetic properties, and therapeutic use as a prokinetic agent in gastrointestinal motility disorders. Drugs. 1988;36:652-681.

124. LeGrange S., Boothe D.M., Herndon S., et al. Pharmacokinetics and suggested oral dosing regimen of cisapride: a study in healthy cats. J Am Anim Hosp Assoc. 1997;33:517-523.

125. Piquett R.K. Torsade de pointes induced by cisapride/clarithromycin interaction. Drug Sat. 1997;17:265-275.

126. Ueki S., Seiki M., Yoneta T., Aita H., et al. Gastroprokinetic activity of nizatidine, a new H2-receptor antagonist, and its possible mechanism of action in dogs and rats. J Pharmacol Exp Ther. 1993;264(1):152-157.

127. Pouderoux P., Kahrilas P.J. A comparative study of cisapride and ranitidine at controlling oesophageal acid exposure in erosive oesophagitis. Aliment Pharmacol Ther. 1995;9(6):661-666.

128. Zarling E.J. Prokinetic activity of nizatidine: implications for the management of patients with gastroesophageal reflux disease. Clin Ther. 1999;21:2038-2046.

129. Briejer M.R., Prins N.H., Schuurke J.A.J. Effects of the enterokinetic prucalopride (R093877) on colonic motility in fasted dogs. Neurogastroenterol Motil. 2001;13(5):465-472.

129a Fargeas M.J., Fioramonti J., Bueno L. Time-related effects of benzodiazepines on intestinal motility in conscious dogs. J Pharm Pharmacol 1984. 1984;36(2):130-132.

129b. Birnbaum D., Karmeli F., Tefera M. The effect of diazepam on human gastric secretion. Gut. 1971;12(8):616-618.

130. Roebel L.E., Cavanagh R.L., Buyniski J.P. Comparative gastrointestinal and biliary tract effects of morphine and butorphanol (Stadol). J Med. 1979;10(4):225-238.

131. Scrivani P.V., Bednarski R.M., Myer C.W. Effects of acepromazine and butorphanol on positive-contrast upper gastrointestinal tract examination in dogs. Am J Vet Res. 1998;59(10):1227-1233.

132. Smith A.A., Posner L.P., Goldstein R.E., et al. Evaluation of the effects of premedication on gastroduodenoscopy in cats. J Am Vet Med Assoc. 2004;225:540-544.

133. Hughes S. Acute secretory diarrhoeas: current concepts in pathogenesis and treatment. Drugs. 1983;26:80-90.

134. Greenwood B., Davison J.S. The relationship between gastrointestinal motility and secretion. Am J Physiol. 1987;252:G1-G7.

135. Kromer W. Endogenous and exogenous opioids in the control of gastrointestinal motility and secretion. Pharmacol Rev. 1988;40:121-162.

136. Allescher H.D., Ahmad S., Kostka P., et al. Distribution of opioid receptors in canine small intestine: implications for function. Am J Physiol. 1989;256:G966-G974.

137. Shook J.E., Lemcke P.K., Gehrig C.A., et al. Antidiarrheal properties of supraspinal mu and delta and peripheral mu, delta and kappa opioid receptors: inhibition of diarrhea without constipation. J Pharmacol Exp Ther. 1989;249:83-90.

138. Schiller L.R., Santa Ana C.A., Morawski S.G., et al. Mechanism of the antidiarrheal effect of loperamide. Gastroenterology. 1984;86:1475-1480.

139. Ishaq S., Green J.R.B. Tolerability of aminosalicylates in inflammatory bowel disease. Biodrugs. 2001;15(5):339-349.

140. Sellin J.H., Pasricha P.J. Pharmacotherapy of inflammatory bowel disease. 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.

141. Wilcke J.R., Turner J.C. The use of absorbents to treat gastrointestinal problems in small animals. Semin Vet Med Surg. 1987;2:266-273.

142. DuPont H.L. Bismuth subsalicylate in the treatment and prevention on diarrheal disease. DICP Ann Pharmacother. 1987;21:687-693.

143. Papich M.G., Davis C.A., Davis L.E. Absorption of salicylate from an antidiarrheal preparation in cats and dogs. J Am Anim Hosp Assoc. 1987;23:221-228.

144. Watson W.A. Factors influencing the clinical efficacy of activated charcoal. DICP Ann Pharmacother. 1987;21:160-166.

145. Dimski D.S. Constipation: pathophysiology, diagnostic approach, and treatment. Semin Vet Med Surg. 1989;4:247-254.

146. Horn D. The impact of fecal impactions. Cornell Fel Health Cent Info Bull. 1987;2:1-8.

147. Burrows C.F. Gastrointestinal pharmacology. AAHA Ann Meet. 1984;51:197-200.

148. Daugherty M.A., Leib M.S., Rossmeisl J.H., et al. Safety and efficacy of oral low-volume sodium phosphate bowel preparation for colonoscopy in dogs. J Vet Intern Med. 2008;22:31-36.

149. Atkins C.E., Tyler R., Greenlee P. Clinical, biochemical, acid-base, and electrolyte abnormalities in cats after hypertonic sodium phosphate enema administration. Am J Vet Res. 1985;46:980-988.

150. Schrezenmeir J., de Vrese M. Probiotics, prebiotics, and synbiotics: approaching a definition. Am J Clin Nutr. 2001;73:361S-364S.

151. Lenoir-Wijnkoop I., Sanders M.E., Cabana M.D., et al. Probiotic and prebiotic influence beyond the intestinal tract. Nutr Rev. 2007;65(11):469-489.

151a. Steinberg S.M. Bacterial translocation: what it is and what it is not. Am J Surg. 2003;186(3):301-305.

151b. Schultz M.J., de Jonge E., Kesecioglu J. Selective decontamination of the digestive tract reduces mortality in critically ill patients. Crit Care. 2003;7(2):107-110.

151c. ol M., van Kan H.J., Schultz M.J., de Jonge E. Systemic tobramycin concentrations during selective decontamination of the digestive tract in intensive care unit patients on continuous venovenous hemofiltration. Intensive Care Med. 2008;34(5):903-906.

152. Blaut M., Clavel T. Metabolic diversity of the intestinal microbiota: implications for health and disease. J Nutr. 2007;137:751S-755S.

153. Marchesi J., Shanahan F. The normal intestinal microbiota. Curr Opin Infect Dis. 2007;20:508-513.

154. Rastall R.A. Bacteria in the gut: friends and foes and how to alter the balance. J Nutr. 2004;134:2022S-2026S.

155. Mentula S., Harmoinen J., Heikkilä M., et al. Comparison between cultured small-intestinal and fecal microbiotas in beagle dogs. Appl Environ Microbiol. 2005;71(8):4169-4175.

156. Davis C.P., Cleven D., Balish E., Yale C.E. Bacterial association in the gastrointestinal tract of beagle dogs. Appl Environ Microbiol. 1977;34:194-206.

157. Winkler P., Ghadimi D., Schrezenmeir J., et al. Molecular and cellular basis of microflora–host interactions. J Nutr. 2007;137:756S-772S.

158. Corthesy B., Gaskins H.R., Mercenier A. Cross-talk between probiotic bacteria and the host immune system. J Nutr. 2007;137:781S-790S.

159. Dethlefsen L., McFall-Ngai M., Relman D.A. An ecological and evolutionary perspective on human–microbe mutualism and disease. Nature. 2007;449(7164):811-818. 18

160. de Vrese M., Schrezenmeir J. Probiotics, prebiotics, and synbiotics. Adv Biochem Eng Biotechnol. 2008;111:1-66.

161. Drisko J.A. Probiotics in health maintenance and disease prevention—probiotics. Altern Med Rev. 2003;8(2):143-155.

162. Johnston B.C., Supina A.L., Ospina M., et al. Probiotics for the prevention of pediatric antibiotic-associated diarrhea. Cochrane Database of Sys Rev. 2007;2:CD004827. doi: 10.1002/14651858.CD004827.pub2

163. Kwon J.H., Farrell R.J. Probiotics and inflammatory bowel disease. Biodrugs. 2003;17(3):179-186.

163a. Abad C.L., Safdar N. The role of lactobacillus probiotics in the treatment or prevention of urogenital infections−a systematic review,. J Chemother. 2009;21(3):243-252.

164. Duncan S.H., Richardson A.J., Kaul P., et al. Oxalobacter formigenes and its potential role in human health. Appl Environ Microbiol. 2002;68(8):3841-3847.

165. Lieske J.C., Goldfarb D.S., De Simone C., et al. Use of a probiotic to decrease hyperoxaluria. Kidney Int. 2005;68:1244-1249.

166. Weese J.S., Weese H.E., Yuricek L., et al. Oxalate degradation by intestinal lactic acid bacteria in dogs and cats. Vet Microbiol. 2004;101(3):161-166. Jul 14

167. Isakow W., Morrow L.E., Kollef M.H. Probiotics for preventing and treating nosocomial infections, review of current evidence and recommendations. Chest. 2007;132:286-294.

168. Quigley E.M., Flourie B. Probiotics and irritable bowel syndrome: a rationale for their use and an assessment of the evidence to date. Neurogastroenterol Motil. 2007;19(3):166-172.

169. Baillon M.L.A., Marshall-Jones Z.V., Butterwick R.F. Effects of probiotic Lactobacillus acidophilus strain DSM13241 in healthy adult dogs. Am J Vet Res. 2004;65:338-343.

170. Rinkinen M., Jalava K., Westermarck E., et al. Interaction between probiotic lactic acid bacteria and canine enteric pathogens: a risk factor for intestinal Enterococcus faecium colonization? Vet Microbiol. 2003;92(1-2):111-119.

171. Heller F.R., Martinet J.P., Henrion J., et al. The rationale for using ursodeoxycholic acid in chronic liver disease. Proc Natl Acad Sci USA. 1991;88:9543-9547.

172. Meyer D.J., Thompson M.B. Bile acids: beyond their value as a liver function test. Biochem J. 1992;285:929-932.

173. Nicholson B.T., Center S.A., Rowland P.J., et al. Evaluation of the safety of ursodeoxycholic acid in healthy cats. Mech Ageing Dev. 1989;48:145-155.

174. Mazzanti R., Fantappié O., Kamimoto Y., et al. Bile acid inhibition of P-glycoprotein-mediated transport in multidrug-resistant cells and rat liver canalicular membrane vesicles. Hepatology. 1994;20(1 Pt 1):170-176.

175. Lieber C.S. S-adenosyl-L-methionine: its role in the treatment of liver disorders. Am J Clin Nutr. 2002;76(5):1183S-1187S.

176. Lieber C.S., Packer L. S-adenosylmethionine: molecular, biological, and clinical aspects:an introduction. Am J Clin Nutr. 2002;76(5):1148S-1150S.

177. Center S.A., Randolph J.F., Warner K.L., et al. The effects of S-adenosylmethionine on clinical pathology and redox potential in the red blood cell, liver, and bile of clinically normal cats. J Vet Intern Med. 2005;19:303-314.

178. Wallace K.P., Center S.A., Hickford F.H., et al. S-Adenosyl-L-Methionine (SAMe) for the treatment of acetaminophen toxicity in a dog. J Am Anim Hosp Assoc. 2002;38:246-254.

179. Herb and Plant Supplement, Consumerlab.com, reviewed October 2007 by EBSCO CAM Review Board. Accessed January 5, 2010, at www.consumerlab.com/tnp.asp?chunkiid=21460.

180. Osman E., Owen J.S., Burroughs A.K. Review article: S-adenosyl-L-methionine: a new therapeutic agent in liver disease. Aliment Pharmacol Ther. 1993;7:21-28.

181. Concari M, et al: S-adenosyl-L-methionine enhances hepatoprotective effects of ursodeoxycholic acid in HEPG2 cell line. AASLD abstract: Hepatology 29(4): pt 2, No. 196, 1996.

182. Lawrence V, Jacobs B, Dennehy C et al: Report on milk thistle: effects on liver disease and cirrhosis and clinical adverse effects, AHRQ Publication No. 01–E025 October 2000.

183. Smith A.J., Nissan A., Lanouette N.M., et al. Prokinetic effect of erythromycin after colorectal surgery: randomized, placebo-controlled, double-blind study. Dis Colon Rectum. 2000;43:333-337.

184. Jergens A.E. Diseases of the esophagus. In Ettinger S.J., Feldman E.C., editors: Textbook of veterinary internal medicine, ed 6, St Louis: Saunders, 2005.

185. Dewey C., Boothe D.M., Rinn K.L., et al. Treatment of a myasthenic dog with mycophenolate mofetil. J Vet Emerg Crit Care. 2000;10(3):177-187.

186. Madisch A., Heydenreich C.J., Wieland V., et al. Treatment of functional dyspepsia with a fixed peppermint oil and caraway oil combination preparation as compared to cisapride. A multicenter, reference-controlled double-blind equivalence study. Arzneimittelforschung. 1999;49:925-932.

187. 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.

188. Stanciu C., Bennett J.R. Metoclopramide in gastrooesophageal reflux. Gut. 1973;14(4):275-279.

189. Wilson D.V., Evans A.T., Mauer W.A. Influence of metoclopramide on gastroesophageal reflux in anesthetized dogs. Am J Vet Res. 2006;67:26-31.

190. Wilson D.V., Tom Evans A., Mauer W.A. Pre-anesthetic meperidine: associated vomiting and gastroesophageal reflux during the subsequent anesthetic in dogs. Vet Anaesth Analg. 2007;34(1):15-22.

191. Melendez L.D., Twedt D.C., Wright M. Suspected doxycycline-induced esophagitis with esophageal stricture formation in three cats. Feline Pract. 2000;28:10-12.

191a. German A.J., Cannon M.J., Dye C. Oesophageal strictures in cats associated with doxycycline therapy. J Fel Med Surg. 2005;7(1):33-41.

192. Graham J.P., Lipman A.H., Newell S.M., et al. Esophageal transit of capsules in clinically normal cats. Am J Vet Res. 2000;61:655-657.

193. 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. 2001;15:467-470.

194. Simpson K.W. Diseases of the stomach. In Ettinger S.J., Feldman E.C., editors: Textbook of veterinary internal medicine, ed 6, St Louis: Saunders, 2005.

195. Marino P. The ICU book, ed 2. Philadelphia: Lippincott Williams & Wilkins; 1997.

196. La Corte R., Caselli M., Castellino G., et al. Prophylaxis and treatment of NSAID-induced gastroduodenal disorders. Drug Saf. 1999;20:527-543.

197. Wu C.S., Wang S.H., Chen P.C., et al. Does famotidine have similar efficacy to misoprostol in the treatment of non-steroidal anti-inflammatory drug-induced gastropathy? Int J Clin Pract. 1998;52:472-474.

198. Brown G.J., Yeomans N.D. Prevention of the gastrointestinal adverse effects of nonsteroidal anti-inflammatory drugs: the role of proton pump inhibitors. Drug Saf. 1999;21:503-512.

199. Ward D.M., Leib M.S., Johnston S.A., et al. The effect of dosing interval on the efficacy of misoprostol in the prevention of aspirin-induced gastric injury. J Vet Intern Med. 2003;17:282-290.

200. Lazzaroni M., Bianchi Porro G. Non-steroidal anti-inflammatory drug gastropathy: clinical results with H2 antagonists and proton pump inhibitors. Ital J Gastroenterol Hepatol. 1999;31(Suppl1):S73-S78.

201. Tryba M. Sucralfate versus antacids or H2-antagonists for stress ulcer prophylaxis: a meta-analysis on efficacy and pneumonia rate. Crit Care Med. 1991;19(7):942-949.

202. Davis M.S., Willard M.D., Nelson S.L., et al. Efficacy of omeprazole for the prevention of exercise-induced gastritis in racing Alaskan sled dogs. J Vet Intern Med. 2003;17(2):163-166.

203. Williamson K.K., Willard M.D., McKenzie E.C., et al. Efficacy of famotidine for the prevention of exercise-induced gastritis in racing Alaskan sled dogs. J Vet Intern Med. 2007;21(5):924-927.

204. Buber T., Saragusty J., Ranen E., et al. Evaluation of lidocaine treatment and risk factors for death associated with gastric dilatation and volvulus in dogs: 112 cases (1997-2005). J Am Vet Med Assoc. 2007;230(9):1334-1339.

205. Ellebaek E., Herrstedt J. Optimizing antiemetic therapy in multiple day and multiple cycles of chemotherapy. Curr Opin Support Palliat Care. 2008;2(1):28-34.

205a. Vail D.M., Rodabaugh H.S., Conder G.A., et al. Efficacy of injectable maropitant (Cerenia) in a randomized clinical trial for prevention and treatment of cisplatin-induced emesis in dogs presented as veterinary patients,. Vet Compar Oncol. 2007;5:38-46.

205b. Rau S.E., Barber L.G., Burgess K.E. Efficacy of maropitant in the prevention of delayed vomiting associated with administration of doxorubicin to dogs. J Vet Intern Med. 2010;24(6):1452-1457.

206. Brechot J.M., Dupeyron J.P., Delattre C., et al. Continuous infusion of high-dose metoclopramide: comparison of pharmacokinetically adjusted and standard doses for the control of cisplatin-induced acute emesis. Eur J Clin Pharmacol. 1991;40(3):283-286.

207. Ku C.M., Ong B.C. Postoperative nausea and vomiting: a review of current literature. Singapore Med J. 2003;44(7):366-374.

208. Seta M.L., Kale-Pradhan P.B. Efficacy of metoclopramide in postoperative ileus after exploratory laparotomy. Pharmacotherapy. 2001;21(10):1181-1186.

209. Traut U., Brügger L., Kunz R., et al. Systemic prokinetic pharmacologic treatment for postoperative adynamic ileus following abdominal surgery in adults. Cochrane Database of Systematic Reviews. 2008. Issue 1, Art. No: CD004930, doi: 10.1002/14651858. CD004930.pub3

209a. Correa P., Houghton J. Carcinogenesis of Helicobacter pylori. Astroenterology. 2007;133(2):659-672.

210. Hall E.J., German A.J. Diseases of the small intestine. In Ettinger S.J., Feldman E.C., editors: Textbook of veterinary internal medicine, ed 6, St Louis: Saunders, 2005.

211. Otto C.M., Jackson C.B., Rogell E.J., et al. Recombinant bactericidal/permeability-increasing protein (rBPI21) for treatment of parvovirus enteritis: a randomized, double-blinded, placebo-controlled trial. J Vet Intern Med. 2001;15:355-360.

212. Isogai E., Isogai H., Onuma M. Escherichia coli associated endotoxemia in dogs with parvovirus infection. Nippon Juigaku Zasshi. 1989;51:597-606.

213. Turk J., Miller M., Brown T., et al. Coliform septicemia and pulmonary disease associated with canine parvoviral enteritis: 88 cases (1987-1988). J Am Vet Med Assoc. 1990;196:771-773.

214. Turk J., Faks W., Miller M., et al. Enteric Clostridium perfringens infection associated with parvoviral enteritis in dogs: 74 cases (1987-1990). J Am Vet Med Assoc. 1992;200:991-994.

215. Scorza A.V., Lappin M.R. Metronidazole for the treatment of feline giardiasis. J Feline Med Surg. 2004;6:157-160.

215a. Steinberg S.M. Bacterial translocation: what it is and what it is not. Am J Surg. 2003;186(3):301-305.

215b. Schultz M.J., de Jonge E., Kesecioglu J. Selective decontamination of the digestive tract reduces mortality in critically ill patients. Crit Care. 2003;7(2):107-110.

215c. ol M., van Kan H.J., Schultz M.J., de Jonge E. Systemic tobramycin concentrations during selective decontamination of the digestive tract in intensive care unit patients on continuous venovenous hemofiltration. Intensive Care Med. 2008;34(5):903-906.

215d. Diepenhorst G.M., van Ruler O., Besselink M.G., et al. Influence of prophylactic probiotics and selective decontamination on bacterial translocation in patients undergoing pancreatic surgery: A randomized controlled clinical trial. Shock. June 22, 2010. [Epub ahead of print] PubMed PMID: 20577144

216 Leib M.S., Duncan R.B., Ward D.L. Triple antimicrobial therapy and acid suppression in dogs with chronic vomiting and gastric Helicobacter spp. J Vet Intern Med. 2007;21:1185-1192.

217. Spanier J.A., Howden C.W., Jones M.P. A systematic review of alternative therapies in the irritable bowel syndrome. Arch Intern Med. 2003;163:265-274.

218. Kuijper E.J., Coignard B., Tull P. Emergence of Clostridium difficile–associated disease in North America and Europe. Clin Microbiol Infect. 2006;12(Suppl 6):2-18.

219. Loo V.G., Poirier L., Miller M.A., et al. A predominantly clonal multi-institutional outbreak of Clostridium difficile–associated diarrhea with high morbidity and mortality. N Engl J Med. 2005;353:2442-2449.

220. Miller M.A., Hyland M., Ofner-Agostini M., et al. Canadian Hospital Epidemiology Committee. Canadian Nosocomial Infection Surveillance Program. Morbidity, mortality, and healthcare burden of nosocomial Clostridium difficile–associated diarrhea in Canadian hospitals. Infect Control Hosp Epidemiol. 2002;23(3):137-140.

221. Drudy D., Quinn T., O’Mahony R., et al. High-level resistance to moxifloxacin and gatifloxacin associated with a novel mutation in gyrB in toxin-A-negative, toxin-B-positive. Clostridium difficile, J Antimicrob Chemother. 2006;58(6):1264-1267.

222. Westermarck E., Skrzypczak T., Harmoinen J., et al. Tylosin-responsive chronic diarrhea in dogs. J Vet Intern Med. 2005;19:177-186.

223. German A.J., Hall E.J., Day M.J. Chronic intestinal inflammation and intestinal disease in dogs. J Vet Intern Med. 2003;17:8-20.

224. Maerten P., Liu Z., Ceuppens J.L. Targeting of costimulatory molecules as a therapeutic approach in inflammatory bowel disease. Biodrugs. 2003;17(6):395-411.

225. Allenspach K., Wieland B., Gröne A., et al. Chronic enteropathies in dogs: evaluation of risk factors for negative outcome. J Vet Intern Med. 2007;21:700-708.

226. Jergens A.E., Schreiner C.A., Frank D.E., et al. A scoring index for disease activity in canine inflammatory bowel disease. J Vet Intern Med. 2003;17:291-297.

227. Shah S. Dietary factors in the modulation of inflammatory bowel disease activity. Med Gen Med. 2007;9(1):60.

228. Marteau P., Lepage P., Mangin I., et al. Review article: gut flora and inflammatory bowel disease. Aliment Pharmacol Ther. 2004;20(Suppl 4):18-23.

229. Sheil B., Shanahan F., O’Mahony L. Probiotic effects on inflammatory bowel disease. J Nutr. 2007;137:819S-824S.

230. Hoentjen F., Harmsen H.J.M., Braat H., et al. Antibiotics with a selective aerobic or anaerobic spectrum have different therapeutic activities in various regions of the colon in interleukin 10 gene deficient mice. Gut. 2003;52:1721-1727.

231. Farrell R.J., Murphy A., Long A., et al. High multidrug resistance(P-glycoprotein 170) expression in inflammatory bowel disease patients who fail medical therapy. Gastroenterology. 2000;118:279-288.

232. Farrell R.J., Kelleher D. Mechanisms of steroid action and resistance in inflammation. J Endocrinol. 2003;178:339-346.

233. Schölmerich J. Review article: systemic and topical steroids in inflammatory bowel disease. Aliment Pharmacol Ther. 2004;20(Suppl 4):66-74.

234. Allenspach K., Rüfenacht S., Sauter S., et al. Pharmacokinetics and clinical efficacy of cyclosporine treatment of dogs with steroid-refractory inflammatory bowel disease. J Vet Intern Med. 2006;20:239-244.

235. Tilg H., Kaser A. Antitumour necrosis factor therapy in Crohn’s disease. Expert Opin Biol Ther. 2002;2(7):715-721.

236. Murthy S., Cooper H.S., Yoshitak H., et al. Combination therapy of pentoxifylline and TNF-α monoclonal antibody in dextran sulphate–induced mouse colitis. Aliment Pharmacol Ther. 1999;13:251-260.

237. Sands B.E., Kaplan G.G. The role of TNF-alpha in ulcerative colitis. J Clin Pharmacol. 2007;47(8):930-941.

238. Baumgart D.C., Dignass A.U. Current biological therapies for inflammatory bowel disease. Curr Pharm Des. 2004;10:4127-4147.

239. Kaser A., Tilg H. Novel therapeutic targets in the treatment of IBD. Expert Opin Ther Targets. 2008;12(5):553-563.

240. Garcia-Sancho M., Rodriguez-Franco F., Sainz A., et al. Evaluation of clinical, macroscopic, and histopathologic response to treatment in nonhypoproteinemic dogs with lymphocytic-plasmacytic enteritis. J Vet Intern Med. 2007;21:11-17.

241. Hostutler R.A., Luria B.J., Johnson S.E., et al. Antiobiotic-responsive histiocytic ulcerative colitis in 9 dogs. J Vet Intern Med. 2004;18:499-504.

242. Ruaux C.G., Steiner J.M., Williams D.A. Early biochemical and clinical responses to cobalamin supplementation in cats with signs of gastrointestinal disease and severe hypocobalaminemia. J Vet Intern Med. 2005;19:155-160.

243. Johnson S.E. Diseases of the liver. In: Ettinger S.J., Feldman E.C., editors. Textbook of veterinary internal medicine. ed 4. Philadelphia: Saunders; 1995:1313-1357.

244. Poldervaart J.H., Favier R.P., Penning L.C., et al. Primary hepatitis in dogs: a retrospective review (2002-2006). J Vet Intern Med. 2009;23(1):72-80.

245. Greiter-Wilke A., Scanziani E., Soldati S., et al. Association of Helicobacter with cholangiohepatitis in cats. J Vet Intern Med. 2006;20:822-827.

246. Tuncer I., Uygan I., Dülger H., et al. The comparative effects of pentoxifylline and ursodeoxycholic acid on IL-1β, IL-6, IL-8 and TNF-α levels in nonalcoholic fatty liver. East J Med. 2003;8(2):27-32.

247. Milk Thistle: Effects on Liver Disease and Cirrhosis and Clinical Adverse Effects. Summary, Evidence Report/Technology Assessment: Number 21, September 2000. Agency for Healthcare Research and Quality, Rockville, MD. Accessed January 5, 2010, at www.ahrq.gov/clinic/epcsums/milktsum.htm.

248. Holt D.E., Washabau R.J., Djali S., et al. Cerebrospinal fluid glutamine, tryptophan, and tryptophan metabolite concentrations in dogs with portosystemic shunts. Am J Vet Res. 2002;63(8):1167-1171.

249. Forman M.A., Marks S.L., De Cock H.E., et al. Evaluation of serum feline pancreatic lipase immunoreactivity and helical computed tomography versus conventional testing for the diagnosis of feline pancreatitis. J Vet Intern Med. 2004;18(6):807-815.

250. Steiner J.M., Williams D.A. Serum feline trypsin-like immunoreactivity in cats with exocrine pancreatic insufficiency. J Vet Intern Med. 2000;14:627-629.

251. Scarpelli D.C. Toxicology of the pancreas. Toxicol Appl Pharmacol. 1989;101:543-554.

252. Simpson K.W. Diagnosis and treatment of acute pancreatitis in the dog. In: Ettinger S.J., Feldman E.C., editors. Textbook of veterinary internal medicine. ed 4. Philadelphia: Saunders; 1995:117-124.

253. Steiner J.M. Canine pancreatitis. In Bonagura J., editor: Kirk’s current veterinary therapy, ed 14, St Louis: Saunders, 2009.

254. Demols A., Van Laethem J.L., Quertinmont E., et al. N-acetylcysteine decreases severity of acute pancreatitis in mice. Pancreas. 2000;20(2):161-169.

255. Inamura H., Kashiwase Y., Morioka J., Kurosawa M. Acute pancreatitis possibly caused by allergy to bananas. J Invest Allergol Clin Immunol. 2005;15(3):222-224.