Which of the following routes of administration typically produces the quickest onset of effects?

Onset of action is the duration of time it takes for a drug's effects to come to prominence upon administration. With oral administration, it typically ranges anywhere from 20 minutes to over an hour, depending on the drug in question. Other methods of ingestion such as smoking or injection can take as little as seconds to minutes to take effect. The determination of the onset of action, however, is not completely dependent upon route of administration. There are several other factors that determine the onset of action for a specific drug, including drug formulation, dosage, and the patient receiving the drug.

Effect of Administration Route on the Onset of Action[edit]

A drug's pharmacological effects can only occur once it has been fully solubilized and has entered the blood stream. For most drugs administered orally, the drug must be ingested, pass through the stomach, and into the small intestine, where the drug molecules enter the blood stream through the villi and microvilli.[1] A few drugs such as alcohol are absorbed by the lining of the stomach, and therefore tend to take effect much more quickly than the vast majority of oral medications which are absorbed in the small intestine. Gastric emptying time can vary from 0 to 3 hours,[2] and therefore plays a major role in onset of action for orally administered drugs. For intravenous administration, the pathway is much shorter because the drug is administered (usually already in solution) directly to the bloodstream.

References[edit]

  1. ^ Pang, K. Sandy (2003). "MODELING OF INTESTINAL DRUG ABSORPTION: ROLES OF TRANSPORTERS AND METABOLIC ENZYMES (FOR THE GILLETTE REVIEW SERIES)". Drug Metabolism and Disposition. 31 (12): 1507–1519. doi:10.1124/dmd.31.12.1507. PMID 14625347.
  2. ^ Bummer, Paul (2012). Gastro Intestinal Tract and Oral Drug Absorption. University of Kentucky.

  • Journal List
  • J Am Assoc Lab Anim Sci
  • v.50(5); 2011 Sep
  • PMC3189662

J Am Assoc Lab Anim Sci. 2011 Sep; 50(5): 600–613.

Published online 2011 Sep.

Abstract

Administration of substances to laboratory animals requires careful consideration and planning to optimize delivery of the agent to the animal while minimizing potential adverse experiences from the procedure. For all species, many different routes are available for administration of substances. The research team and IACUC members should be aware of reasons for selecting specific routes and of training and competency necessary for personnel to use these routes effectively. Once a route is selected, issues such as volume of administration, site of delivery, pH of the substance, and other factors must be considered to refine the technique. Inadequate training or inattention to detail during this aspect of a study may result in unintentional adverse effects on experimental animals and confounded results.

Administration of substances to laboratory animals is often a critical component of experimental design. Administered substances may include: infectious disease agents; various therapeutics, such as vaccinations, antimicrobials, pharmacologic agents, anesthetics, and analgesics; chemical test agents; radiocontrast agents; electrolytes and other fluids; and nutritive support. Because substances may be administered repeatedly to the same animal or to multiple animals on the same study, the dosing methodology is an important consideration when planning an experiment and during protocol review by animal care and use committees and represents an essential opportunity for refining treatment of research subjects. Specific considerations for delivery of substances to animals are numerous and include factors such as absorption, distribution, metabolism and excretion of therapeutic or chemical agents; route, volume, and frequency of administration; duration of treatment; pH, stability, homogeneity, and osmolality of the substance to be administered; selection of vehicle or solvent for delivering substances that cannot be administered in a solid or particulate state; solution preparation, including considerations for sterility if the substance is being administered parenterally; and dosing apparatus and animal restraint necessary for specific routes of delivery. In addition, research teams should be aware of potential adverse effects related to substance administration to avoid confounding effects with other aspects of study design and to permit accurate interpretation of research findings.

Although understanding the basic pharmacology of any administered therapeutic or chemical agent is important for experimental planning, it is beyond the scope of this article to review principles of pharmacokinetics and pharmacodynamics, and readers are referred to several excellent texts dealing with these subjects.22,102,106 This article is the first of a 2-part review of substance delivery to laboratory animals and summarizes recommended practices for various routes of administration to a range of species and factors to consider during experimental planning. The second part of this review examines dosing equipment and apparatus needed for substance delivery, considerations for selecting vehicles, and solute preparation and handling.134

Routes of Administration

Selection of a route.

Substances are administered to laboratory animals by a wide variety of routes. A key factor determining the route selected is whether the agent is being administered for a local or systemic (either enteral [through the digestive tract] or parenteral [outside the digestive tract]) effect. Parenteral administration methods typically produce the highest bioavailability of substances because these methods avoid the first-pass effect of hepatic metabolism, which occurs commonly with orally administered chemicals and therapeutics. Parenteral routes also circumvent some of the unpredictability associated with enteral absorptive processes. Furthermore, regulatory requirements may influence the selection of a particular route, depending on the purpose of the study (for example, nonclinical safety testing, in which the route of delivery to animals should closely resemble the projected route of administration to humans).37,38

A substance may be given into the mouth (orally) or delivered directly into the stomach (gastric gavage); delivered into a blood vessel (intravenous); delivered onto, into, under, or across the skin or into a muscle (epicutaneous, intradermal, subcutaneous, transdermal, and intramuscular administration, respectively); instilled onto or into the eye (transcorneal or intraocular, respectively); into the brain (intracerebral) or the space surrounding the dura mater or that surrounding the distal spinal cord (epidural and intrathecal, respectively); administered into the peritoneal cavity (intraperitoneal), directly into the marrow cavity (intraosseous); sprayed into the nose for absorption across the nasal mucous membranes or into the lungs (intranasal) or delivered into the lungs by direct tracheal instillation (intratracheal) or inhalation; or administered by a range of less common routes using other body orifices, surgical exposures, and species-specific anatomic features (for examples, see references 16, 41, 60, 64, 73, 91, and 127).

In laboratory species, many of the commonly used methods of delivery require restraint, sedation, or general anesthesia. The use of such manipulations should be considered when selecting the administration route to refine procedures so that they are less invasive or aversive to the animals. In addition, each route has advantages and disadvantages that should be considered depending on the final effect to be achieved, and ultimately the route selected will markedly affect the pharmacokinetics of the substance. This pharmacokinetic effect of route of administration is exemplified by naloxone, a potent opioid antagonist. Given intravenously, naloxone rapidly reverses opioid-induced central nervous system depression,28 but when given enterally, the drug can be used to treat opioid-induced bowel stasis without antagonism of the analgesic effects of systemically administered opioids.52 Another consideration regarding once-daily administration of substances to animals is their chronobiology or circadian rhythm. Depending on the aims and objectives of the experiment, the timing of substance administration may need to be considered carefully, for example, to administer a therapeutic when an animal's system is most or least metabolically active to induce or minimize toxicity.119

Enteral administration.

Administration of substances directly into the mouth, admixed in diet or other foodstuffs, or by orogastric or nasogastric gavage is common in laboratory animal medicine and research. Per rectum administration of substances by enema or suppository is less common in animals than in humans. The oral route is economical, convenient, relatively safe, and some animals can be trained to cooperate voluntarily, depending on the compound being administered (Figure 1 A through C). Although voluntary consumption of the material being administered is ideal, this dosing technique may not be reliable in all animals or dose groups or for long-term studies, because of individual preferences for flavors, palatability issues, and changes in behavior over time. For substances being tested for safety, oral dosing mimics the most commonly used mode of administration of substances to humans. When placing substances directly into the mouth, it is important to ensure that tablets or gelatin capsules containing test material are placed far back in the mouth and that the animal swallows, to ensure receipt of the full dose. The number and size of capsules or tablets administered should be proportional to the size of the animal being dosed, to minimize regurgitation. Gavage (esophageal or gastric) is often used in research settings, instead of mixing substances in water or food, to ensure precise and accurate dosing of animals (Figure 1 D).

Which of the following routes of administration typically produces the quickest onset of effects?

(A) Rat voluntarily consuming nutritional supplement from a syringe. Photo courtesy of Colette Wheler. (B) Macaque voluntarily drinking medication from a syringe. Photo courtesy of Andrew Winterborn. (C) Pig voluntarily accepting medication when administered in a marshmallow. (D) Oral gavage of fish. Photo courtesy of Gerald Johnson.

Selection of appropriate tubing size for orogastric or nasogastric gavage is important to minimize discomfort while optimizing delivery of substances. Nasogastric tubes are used commonly in rabbits for enteral nutrition and in nonhuman primates for dose administration and typically comprise 3- to 8-French soft rubber pediatric feeding tubes.18,104 Tubing is measured from the external nares to the last rib and marked. To minimize discomfort, a small amount of xylocaine jelly can be placed on the end of the tubing or a drop of 0.5% proparicaine hydrochloride ophthalmic solution is placed directly in the nares prior to introducing the tubing into the ventromedial meatus (Figure 2).

Which of the following routes of administration typically produces the quickest onset of effects?

Chronic nasogastric catheter placement in a rabbit for enteral nutrition. Photo courtesy of Colette Wheler.

Except when given in the diet or admixed with food, oral administration of substances typically requires some form of restraint. In many species, including rodents and nonhuman primates, restraint can be the greatest adverse effect of a procedure.25,78,135 Habituation or positive reinforcement training to restraint may reduce the stress associated with the procedure.1,107,120 In addition, the administration of large volumes of substances by orogastric or nasogastric gavage may cause stress due to gastric distension in species that are unable to vomit, such as rodents.21 Therefore, using the smallest volume possible is recommended for the oral route of administration, optimally 5 mL/kg for all species (Table 1). When rats underwent gavage at this volume, no difference was noted between the stress induced by gavage compared with that induced by restraint alone.135 When large volumes must be administered by gavage, a slower delivery rate may be better tolerated by animals.

Table 1.

Recommended volumes and sites of Administration of substances to laboratory animals

Route Species Optimal volume (range) Site(s) References
Gavage
All 5 mL/kg (to 20 mL/kg) Mammals: intragastric 21, 82, 134
Fish: esophageala 127
Fish 2 g/kg (gel capsules) 16
Intravenous
All Up to 5 mL/kg (bolus) Rodents: tail or saphenous vein Rabbits: ear or cephalic vein Larger species: jugular, cephalic, femoral, or saphenous vein 82
Fish: caudal vein or arteryab 16
2 mL/kg hourly (to 4 mL/kg/h) continuous infusion)c 82, 89
Subcutaneous
Mammals Maximum of 5 mL/kg per site Intrascapular, neck, shoulder, flank 82
Fish 1 mL/kg Midline and just anterior to dorsal fin 53
Intradermal
All 0.05-0.1 mL per site Skin 82
Intramuscular
All Maximum of 0.05 mL/kg per site (rodents, rabbits, small nonhuman primates, fish) Mammals: triceps, quadriceps, dorsal lumbar, semimembranosus, semitendinosus muscles 82
Fish: base of dorsal fin or between dorsal fin and lateral line 16
Epidural
Mammals 0.15–0.2 mL/kgd (6 mL total volume in patients up to 35 kg) 47, 73, 138
Intraperitoneal
All Maximum of 10 mL/kg See text 82
Intranasal
Rodents Minimum of 35µL per animale (50 µL) 82, 121
Dog, cats, nonhuman primates, rabbits 200 to 500 µL per animal 82

Limitations of oral dosage may include a slower onset of action compared with parenteral delivery, a potentially significant first-pass effect by the liver for those substances metabolized through this route with reduced efficacy, lack of absorption of substances due to chemical polarity or interference with absorption by ingesta, poor compliance with voluntary consumption because of poor palatability or local irritation, lack of systemic absorption from the digestive tract, degradation of substances by digestive enzymes and acid, and inability to use this route in animals that are unconscious or have clinically significant diarrhea or emesis.11 Oral gavage requires moderate technical skill and confidence. Research personnel should have training and practice prior to study initiation to minimize adverse events associated with the technique and to ensure that it is performed accurately, rapidly, and humanely in experimental animals.

Intravenous administration.

The intravenous route of delivery is the most efficient means of delivering substances to animals because it bypasses the need for solute absorption. With this method, substances are administered as a bolus or infusion directly into blood vessels on either an acute or chronic basis (Figure 3). Precision electronic infusion pumps equipped with alarms to indicate flow interruptions and microdrop infusion sets are used to ensure accurate chronic intravenous delivery of many substances; however, less expensive precision and spring-operated disposable pumps have become available for this purpose in recent years and may represent a more economical alternative for experimental intravenous substance delivery, depending on the nature of the material to be administered and the duration of treatment.2,32,117

Which of the following routes of administration typically produces the quickest onset of effects?

Different routes of skin administration of substances. Depicted are intramuscular (IM), intravenous (IV), subcutaneous (SC), and intradermal (ID) routes. Illustration courtesy of Gianni Chiappetta.

Although fluids and parenteral nutrition typically are infused on a continuous basis over several hours or days, the decision to administer other substances by the intravenous route often depends on the pharmacokinetics of the substance, as well as the maximum tolerated dose, the time interval over which delivery is required (referred to as dosing intensity), and the need to minimize variations in peak and trough blood levels in the substance being administered. The actual technique involves aseptic preparation of skin for percutaneous venous injection or surgical exposure of blood vessels for substance administration. Intentional intraarterial administration of substances should be avoided routinely and used only for specific experimental conditions, because of the potential for severe complications with this route, including blindness, cerebrovascular stroke, permanent motor deficits, and limb gangrene.75,114,116,142 Suggested sites and volumes for intravenous injection and infusion of substances are given in Table 1.

Researchers designing experiments requiring single or repeated intravenous treatments should consider technique refinements that may enhance animal comfort, including the use of the smallest needle or catheter size possible to minimize injection trauma, butterfly needles for single injections to minimize perivascular trauma, indwelling catheters and vascular access ports for animal comfort and locomotor freedom, topical anesthetic creams and ointments prior to needle placement to minimize injection pain, and external pump packs to minimize the restriction of animal movement associated with tethering. Excellent recent reviews of techniques, equipment, and refinements for using catheters and vascular access ports in animals have been published.16,53,89,124,125,128 A more detailed discussion of dosing equipment for intravenous delivery can be found in the companion article to the current work.134

Intraosseous administration of substances, particularly crystalloid fluids, is used in human pediatric medicine and emergency avian and rabbit medicine as an alternative for the intravenous route in hypovolemic patients with inaccessible or collapsed veins.31,80,129 The medullary cavity contains noncollapsing venous sinuses that directly enter into the central venous circulation and substances administered intraosseously are generally detectable immediately after administration. The technique is difficult to perform without advanced training and is potentially invasive, with considerable risk for postprocedural osteomyelitis, fat embolization, iatrogenic fracture and growth plate injury, and pain. Intraosseous administration typically is conducted in fully anesthetized animals.

Substances administered intravenously or intraosseously must be delivered aseptically and should be sterile; free of particulates that may induce foreign body emboli; and minimally irritating to vascular endothelia, to prevent vasculitis and thrombosis, and to erythrocytes, to minimize lysis. Certain oily substances, such as cremaphor, and various alcohols, surfactants, and other vehicles and excipients may induce hemolysis when introduced intravenously; these substances should be avoided, whenever possible, or first evaluated in vitro for safety.4,79,90 The intravenous route of substance delivery, although efficient, can be risky in animals, and persons conducting this technique require training and practice to ensure competency. Careful control of hemostasis must be instituted when the catheter or needle is removed, to minimize blood loss and painful hematoma formation. When fluids or infusions are administered chronically, animals should be monitored closely for signs of fluid overload and pulmonary edema, such as dyspnea and cyanosis.77 Chronically implanted catheters and vascular access ports require regular cleaning and maintenance to ensure patency and prevent infection.

Administration to skin and muscle.

Some substances can be administered directly to the skin surface (epicutaneous administration) for a topical affect. The extent of absorption of materials through the skin and into the systemic circulation (that is, percutaneous or transdermal delivery) depends on: the surface area over which the substance is applied; the concentration of the substance administered; the lipid solubility of the material or vehicle; whether the skin surface is intact; the skin thickness at the site of application; the length of time that the material is in contact with the skin surface; and the degree of skin hydration and surface occlusion, in that covered and well-hydrated skin absorbs substances faster than does uncovered or dry skin.87 For fish, specialized chambers can be constructed to expose the skin or gills specifically to test substances.16,53 When administering substances topically to the skin of mammals, overlying hair is clipped to minimize matting and maximize contact with the material to be applied, and the skin surface is cleaned prior to application. Absorption of substances across the epidermis occurs through paracellular and transcellular mechanisms into the stratum corneum, to the stratum spinosum, and then to the basal layers of the skin and later, the dermis, as well as into the subcutaneous space through hair follicles and accessory glands.42,93

Caution must be exercised to avoid applying caustic or irritating material directly onto the skin, and some substances may induce local sensitization reactions. Consideration should be given to the potential for systemic toxicity when administering substances topically, particularly if the site is readily accessible for grooming.46 Application of thin layers of cream or ointment to the skin at more frequent intervals may be more efficacious with less potential for systemic toxicity than is less frequent application of thicker layers.

Transdermal or percutaneous delivery represents a similar route of administration except that materials are applied to the skin surface deliberately, usually by means of a patch, for absorption across the epithelial barrier into the systemic circulation. Typically, this method produces very constant blood levels of the substance being administered. Percutaneous delivery is an attractive alternative to other parenteral routes, avoiding the need for repeated animal restraint, painful injections, and sharps hazards. In addition, materials can readily be removed from the skin surface if dosing needs to be interrupted or if adverse effects are noted. Transdermal delivery of substances may be acute or chronic, and current techniques for delivering substances by this route have been reviewed recently.7,45,100 The skin is prepared as for topical delivery. When a transdermal delivery system will be used, the agent and delivery system (for example, patch) must be applied in advance of when the desired effect needs to occur, based on the pharmacokinetics of substance absorption. The product should be applied in such a way to protect it from ingestion and contamination, and the signs of toxicity after inadvertent ingestion by the animal should be known. Commercially available human transdermal products can be difficult to use in animals because of the much larger doses of substances impregnated into products intended for adult human use. Cutting transdermal patches to scale-down the dose being administered is not recommended; however, covering a portion of the patch to limit substance administration may be used. Animals should be observed closely for toxicity, and as for topical delivery methods, skin sensitization may occur over time with transdermal product use.84 Animals must be prevented from removing and ingesting patches.

Nonirritating substances may be given subcutaneously, which represents a rapid, inexpensive, and simple method of parenteral substance administration (Figure 3). Substances administered subcutaneously often are absorbed at a slower rate compared with other parenteral routes, providing a sustained effect. The exact mechanism of absorption is unknown but is thought to be due to uptake of macromolecules within the subcutis by small capillaries underling the skin, with minimal lymphatic absorption.56 Substances delivered subcutaneously can be aqueous or oily fluids, depots of oily materials for slow absorption, solid pellets, or injected into suitably sized osmotic minipumps or other implantable pumps, which subsequently are surgically inserted into a subcutaneous pocket. Because the subcutaneous space is largely a virtual space, it can be an excellent site for large volume fluid delivery in small or dehydrated animals, avoiding technical difficulties and problems sometimes seen with direct intravenous administration, such as fluid overload and pulmonary edema, because excess subcutaneous fluid is excreted rapidly by the kidneys. Compared with intravenous delivery, the subcutaneous route is a simple one to master; however, training and competency of personnel should be monitored to ensure that substances are delivered accurately and that inadvertent intravenous injection is avoided. Careful consideration should be given to using an appropriately sized needle, and humane and aseptic periinjection techniques. The skin overlying the site selected for injection should be loose to minimize discomfort, and the needle should be inserted at a shallow angle to minimize damage to underlying tissues. Passing a small-gauge needle through a thick rubber stopper to fill an attached syringe prior to injection may dull the needle point, enhancing injection discomfort. Contaminated substances injected subcutaneously typically will result in abscess formation. Recommended volumes and locations for subcutaneous injections are presented in Table 1. Inadvertent subcutaneous administration is a common complication of intradermal injections, and small, sharp needles are required for success with intradermal delivery.82

Intramuscular administration of substances is a common parenteral route in large animals and humans but often is avoided in smaller species because of the reduced muscle mass. Generally, intramuscular injections result in uniform and rapid absorption of substances, because of the rich vascular supply (Figure 3). Smaller volumes are administered intramuscularly than for subcutaneous delivery (Table 1). The intramuscular technique requires more skill than does subcutaneous injection and should be conducted only by well-trained personnel. Intramuscular injection of irritating substances or inadvertent injection of nerves may result in paresis, paralysis, muscle necrosis, and localized muscle sloughing.103 Repeated injections may result in muscle inflammation and necrosis.30 Other considerations and cautions for using the intramuscular route for substance delivery are similar to the subcutaneous route.

Epidural and intrathecal administration.

For rapid effects of substances on cerebrospinal tissues or meninges, substances can be administered into the epidural or subarachnoid (intrathecal) space of the spinal cord (Figure 4, Table 1). This technique avoids absorptive problems otherwise presented by the blood–brain barrier. The route is used commonly to induce spinal anesthesia or to introduce contrast media for visualizing vertebral bodies or the spinal cord of large animal species. The technique requires animals to be sedated heavily and given a local anesthetic block over the spinal needle insertion site; alternatively animals can undergo general anesthesia prior to implementation.23,138 Aseptic preparation of the skin overlying the injection site and use of sterile technique for needle insertion are critical for success and animal recovery. The exact location of needle insertion and volume of injectate will vary between species and for intrathecal compared with epidural administration, and several factors contribute to procedural success (see reference 138 for review). Epidural fat, lipophilicity of the substance being administered, leakage of injectate through intervertebral spaces, and pronounced meningovertebral ligaments all will limit or alter the spread of material being introduced by epidural or intrathecal routes.58 This limitation may be problematic, in that increased quantities of substances may need to be administered for effect, with the possibility of spill-over into systemic circulation, resulting in adverse effects, such as profound respiratory depression requiring prolonged ventilation. Visualization of cerebrospinal fluid after spinal needle insertion confirms intrathecal placement of the needle. If this fluid is noted when attempting an epidural injection, the needle should be withdrawn and repositioned, or the dose of the substance administered should be reduced, because the kinetics of substance absorption from epidural compared with intrathecal delivery can be markedly different.138

Which of the following routes of administration typically produces the quickest onset of effects?

Epidural (ED) compared with intrathecal (IT) injections in the distal lumbar spine. Illustration courtesy of Gianni Chiappetta.

Intrathecal or epidural administration of substances requires considerable technical skill and in-depth knowledge of anesthesia, analgesia, and spinal cord and vertebral column anatomy. These techniques should be performed only by well-trained personnel. Adverse events associated with epidural administration of substances to small animals include prolonged time for hair regrowth over the injection site, pruritus, urinary retention, nausea, vomiting, and prolonged and severe respiratory depression.132

Intraperitoneal administration.

Injection of substances into the peritoneal cavity is a common technique in laboratory rodents but rarely is used in larger mammals and humans. Intraperitoneal injection is used for small species for which intravenous access is challenging and it can be used to administer large volumes of fluid safely (Table 1) or as a repository site for surgical implantation of a preloaded osmotic minipump. Absorption of material delivered intraperitoneally is typically much slower than for intravenous injection. Although intraperitoneal delivery is considered a parenteral route of administration, the pharmacokinetics of substances administered intraperitoneally are more similar to those seen after oral administration, because the primary route of absorption is into the mesenteric vessels, which drain into the portal vein and pass through the liver.74 Therefore substances administered intraperitoneally may undergo hepatic metabolism before reaching the systemic circulation. In addition, a small amount of intraperitoneal injectate may pass directly across the diaphragm through small lacunae and into the thoracic lymph.3

In mammals, intraperitoneal administration typically is conducted in conscious animals by using firm manual restraint, with the head and body tipped downward to move viscera away from the surface of the ventral abdomen. Injections in rodents are made in the lower right abdominal quadrant away from the midline to avoid inadvertent injection into the urinary bladder or cecum.26 The syringe plunger may be withdrawn prior to injection, specifically looking for urine, blood, or digesta in the needle hub; if these fluids are seen, the needle should be withdrawn, replaced, and repositioned prior to injection. The most common mistake is to puncture the skin at too acute an angle, resulting in subcutaneous rather than intraperitoneal administration. For intraperitoneal injections in fish, the animals are restrained on their side on a flat surface, and the needle should enter along the midline, just anterior to the pelvic fins. Larger fish may require sedation or light anesthesia for appropriate restraint.16

Materials injected intraperitoneally should be sterile, isotonic, and nonirritating. Irritating substances injected intraperitoneally may induce painful ileus and peritonitis in rodents, with subsequent adhesions.43 This drawback is typified by the effects of undiluted chloral hydrate when administered intraperitoneally in rats.36 Injections of identical doses of chloral hydrate in less concentrated solutions may avoid peritoneal irritation,137 and this technique may be used for other potentially irritating substances. Although technically a simple procedure to perform, training and competency of personnel should be monitored to ensure that substances are delivered accurately and that inadvertent intracecal or intracystic injections are avoided.

Intranasal, intratracheal, and inhalational administration.

In research settings, animals generally are sedated or anesthetized47 for the intranasal and intratracheal routes of delivery, to minimize struggling and sneezing. Volumes administered intranasally are small compared with those of other routes (Table 1), to minimize the potential for suffocation and death. The technique may not be useful in animals with signs of rhinitis or conjunctivitis. Intranasal delivery is readily taught and simple to perform in an anesthetized animal. Substances administered by this route should be nonirritating to minimize sneezing, posttreatment rhinitis, and epistaxis.

Intranasal techniques may be used for either local (for example, vaccinations or decongestant sprays) or systemic delivery of substances. The nasal mucosa lines the nasal cavity and is richly supplied with blood vessels, potentially resulting in rapid substance absorption and subsequent systemic effects, avoiding the hepatic first-pass effect seen with oral delivery. Blood drug levels of substances administered intranasally may approach those seen after intravenous administration, and small, lipophilic molecules are absorbed more rapidly by this route than are large molecular weight or highly polar substances.54

The lung has a large surface area, which is supplied by a dense capillary network, making absorption from this site rapid. Intrapulmonary delivery is the most common route by which substances are administered to fish. With this method, substances are dissolved in a static or flow-through aquatic system into which fish are placed. Material is absorbed rapidly across the gills, which are richly supplied with capillaries, resulting in systemic uptake. Because the entire fish is submerged in the tank, the dissolved substance should not be corrosive or irritating to minimize skin and ocular damage (Figure 5 A and B).

Which of the following routes of administration typically produces the quickest onset of effects?

(A) Deep anesthesia of a fish with tricaine methane sulfonate (MS222), achieved by immersion in aqueous solution with the drug. Drug is taken up across the gills during respiration and, to a lesser extent, across the skin. Photo courtesy of Gerry Johnson. (B) Amphibians may be similarly dosed in aqueous chambers as in the Xenopus laevis depicted; however, substance uptake is solely through trancutaneous absortion.

Intrapulmonary delivery to other species is accomplished by either intratracheal instillation or inhalation. Intratracheal instillation is an easier delivery method requiring less specialized equipment and knowledge; however, this route typically is not as effective as are inhalational techniques in ensuring even pulmonary exposure to a substance. Intratracheal instillation involves injecting small volumes of solutions directly into the trachea of anesthetized animals and results in rapid but localized and uneven distribution of material over a relatively small volume of the lung. Volumes administered by the intratracheal route must be small to avoid suffocation. Those performing the intratracheal technique should be competent at intubating the species being treated, or a surgical cutdown can be used to expose the trachea for direct injection.

Inhalational delivery typically uses vapors (for example, volatile anesthetic gases) or aerosols of nebulized particles in solution. Animals are conscious with this delivery method and are restrained with or without a specialized nose mask to optimize delivery. Substances administered by aerosol are deposited by gravitational sedimentation, inertial impaction, or diffusion. As a rule of thumb, larger particles are deposited in the airways by gravitational sedimentation and inertial impaction, whereas smaller particles make their way into distal alveolar spaces by diffusion. Particles less than 3 µm in diameter penetrate the alveoli, and those that are 3 to 5 µm in diameter distribute uniformly throughout the lung. Materials deposited in the oropharynx, proximal trachea, or airways will be transported up the trachea by the mucociliary apparatus, into the mouth, and swallowed with subsequent first pass-effect after absorption.95 In addition, solvent and propellant effects must be taken into account, because evaporation may cause particles to change in size.

Inhalational administration is a highly complex technique requiring specialized equipment and knowledge, and it is beyond the scope of this article to discuss this methodology in further detail (for more information, see references 99, 113, and 140). Substances administered by this route should be nonirritating to minimize pharyngeal edema, bronchial spasm, anaphylaxis, peracute death, and chronic pulmonary fibrosis. Animals should be conditioned to restraint devices and nose masks prior to experimental initiation.

Factors to Consider for Substance Administration

There are a number of factors to consider to optimize substance delivery to animals and to minimize complications associated with delivery. Complications may arise from the method of delivery as well as those associated with volume of substance administered, rate of administration, temperature of substance, fasting state, and subject age. Checklists may be developed for use in experimental planning to ensure that all factors have been considered adequately; these factors should also be considered during ethical review of study protocols.82

Adverse effects associated with dosing route.

Any method of substance administration has inherent potential side effects. For enteral administration, complications depend on the delivery method: force feeding, pilling, delivery in food, or gavage. Oral gavage can result in passive reflux if the stomach is overfilled, aspiration pneumonia, pharyngeal, esophageal, and gastric irritation or injury with stricture formation, esophageal and gastric rupture (Figure 6), and stress.17,21,40,85 Even when small volumes are used, microaspiration has been suggested to occur in as many as one third of gavaged mice, resulting in detection of radiolabeled particles outside the gastrointestinal tract.27 Highly viscous substances can affect both the risk of aspiration and the systemic stress response to the gavage procedure, and oily vehicles increase the likelihood of both.21 Highly viscous substances are difficult to deliver through a small-diameter dosing needle or catheter and should be diluted, whenever possible, for ease of administration.

Which of the following routes of administration typically produces the quickest onset of effects?

Inadvertent esophageal rupture (arrows) with food contamination and local cellulitis after oral gavage in a mouse. Photo courtesy of David Hobson.

Habituation to restraint and gavage may reduce struggling and the risk of associated injuries. Without habituation, rats and mice have increased blood pressure and heart rate for as long as 1 h after gavage, as well as higher serum corticosterone levels.5,17,51 Repeated periods of brief restraint in the week before experimental gavage reduce physiologic responses to the gavage procedure in rats.135 Generally, rodents adjust quickly to repeated gavage, and corticosterone levels return to baseline after the second day of gavage in mice.51 Heart rate and blood pressure return to normal by the third day of oral gavage in rats.92 In addition, elevations in corticosterone levels in mice can be mitigated by dipping the gavage needle in a sucrose solution prior to gavage.51 Adverse effects may also be reduced by using soft gavage tubing. In rats, changes in heart rate and blood pressure were reduced if soft gavage tubes (Teflon) were used in place of stainless steel dosing needles.92,141 A potential drawback of soft tubing for oral gavage is the possibility that an animal may bite through the tube. Sedation prior to gavage is not necessarily a refinement and may interfere with pharmacokinetic measurement. Prolonged gastric retention occurred in rats that were anesthetized briefly with halothane for oral gavage.85 Although not clinically significant in the cited study,27 incomplete gastric retention of substances can result in variable rates of absorption and immune stimulation, thereby affecting study outcome.

Other methods of enteral administration include sprinkling or mixing the substance with food, food treats, or water, and introducing substances directly into the mouth by using a syringe or pill. When substances are mixed with food or water, dehydration, weight loss, and inefficient drug administration can occur if palatability is poor or if taste aversion develops.5,82 In addition, gingivitis, tooth decay, and tooth overgrowth can result from diets high in sugar or after the addition of diet softeners, such as polyethylene glycol.19,62,86,105,122,131 Finally, marked species-, breed-, strain-, and stock-associated variations in food and water consumption exist, so caution must be applied when using standard estimates of consumption of test substance in food or water, to avoid over- or under-dosing animals.6,35,68,69,70,94 Obtaining accurate body weights prior to dosing and throughout the study is critical.

Syringe feeding or feeding by dropper requires training and timing with relation to natural feeding. This technique can be time-consuming, particularly with studies of more than 20 animals.5 Not all animals readily acclimate to this method of delivery, and it may not be possible to use this method in studies in which strict accuracy of animal dosing is required. Palatability and taste aversion both can affect this type of delivery, similar to mixing substances into food or water. Absorption directly across the oral mucosa can affect substance pharmacokinetics, an effect that should be considered when oral routes of delivery are employed.5

With common parenteral routes of administration complications associated with substance administration include local irritation, pain, infection, and damage to the surrounding tissue depending on the species, method of restraint, route, volume, and substance administered. Generally, administering smaller volumes over multiple injection sites will minimize adverse reactions and can be used for subcutaneous, intramuscular, or intradermal delivery.82,143 Some substances cause species-specific complications. For example, complete Freund adjuvant can result in pulmonary granulomata in rodents, irrespective of the site of administration.97 The mechanism underlying this reaction is unknown.

Different routes of parenteral administration may be associated with specific inherent complications. Intramuscular injections can cause muscle necrosis or inflammation of the nerves, resulting in lameness and self-mutilation of the affected area. Pain, necrosis, and self-mutilation of the feet have been reported in response to intramuscular injection in rabbits, rodents, and other species.14,39,63,118,123,136 With intranasal injections, aspiration pneumonia and suffocation can occur, depending on the volume and formulation of the compound administered.48 Dosing can be inaccurate, because animals often sneeze in response to intranasal administration. Deep sedation or light anesthesia can be useful adjuncts to this procedure to ensure dosing accuracy.

Intraperitoneal delivery represents a theoretically easy method of introducing material into rodents, but the associated accuracy can be questionable. In one study in rats, 19.6% of intraperitoneal injections conducted by competent staff resulted in the material being injected in the gastrointestinal tract, subcutaneously, retroperitoneally, or into the urinary bladder.67 In addition, the true prevalence of associated complications likely is underestimated, given that many animals are not necropsied after injection. Potential complications include infection, pain, local irritation and chemical peritonitis, formation of fibrous tissue and adhesions within the abdominal cavity, perforation of an abdominal organ, hemorrhage, and respiratory distress or discomfort from administration of too large a volume. Repeated administration can result in a cumulative irritant effect and needle-induced damage.82

Complications associated with intravenous delivery methods are more readily apparent than after intraperiotenal delivery. Asepsis is critical, as intravenous administration of contaminated material can result in bacteremia and septicemia (Figures 7 and 8). Extravascular delivery of compounds that are irritating may result in local soft tissue damage, infection, pain, and tissue sloughing. In all species, injection of compounds that contain particulate material or are of low pH that precipitate when mixed with blood can result in vascular occlusion, emboli, and thrombosis of local and distant capillary beds such as those found in the ears, tail, toes, or lungs.8,50,61,81,98,126 Substances also may induce hemolysis, coagulation, or anaphylaxis when administered intravenously, and these complications may vary depending on the species and the nature of the material being administered. For example, the vehicle Tween 80 causes anaphylaxis when administered intravenously to dogs but not rodents.44,130 For studies involving multiple venipunctures and injections, those evaluating histologic sections should remember that pulmonary microthrombi or foreign-body granulomas related to shedding of catheter materials or hair fragments are not uncommon histologic findings in chronic infusion studies (Figure 9 A and B).24,34,81,98

Which of the following routes of administration typically produces the quickest onset of effects?

Photomicrograph of atrial thrombosis with secondary bacterial infection and myocarditis in a rat with a chronic indwelling jugular vein catheter. A large septic thrombus (T; bacterial colonies indicated by arrows) is firmly adherent to the endocardium and there is significant infiltration of the myocardium (C) with neutrophils. The thrombus has not entirely occluded the atrium, as a small lumen (L) is present. Hematoxylin and eosin stain; magnification, ×40.

Which of the following routes of administration typically produces the quickest onset of effects?

Photomicrograph demonstrating multifocal suppurative encephalitis with perivascular neutrophilic cuffing (arrows) after inadvertent contamination of an indwelling jugular vein catheter in a rat. Hematoxylin and eosin stain; magnification, ×40. Photo courtesy of Leah Schutt.

Which of the following routes of administration typically produces the quickest onset of effects?

Photomicrographs demonstrating incidental findings. (A) Raft of epithelial cells (arrows) forming microthrombus within a pulmonary vessel of a chronically infused rat. Hematoxylin and eosin stain; magnification, ×200. Photo courtesy of Igor Mikaelian. (B) Hair shaft embedded centrally within pulmonary microthrombus of rat receiving a bolus IV injection. Hematoxylin and eosin stain; magnification, ×400. Photo courtesy of Heather Workman.

For continuous infusion studies, the nature of the catheter material may affect irritation at the site of catheterization, and this consideration is important when catheters will be in place long term (see Table 2 of reference 134).134 In long-term continuous infusion studies, the local concentration of the substance in the cannulated vessel can be higher close to the catheter insertion site for a longer period of time when low flow rates are used. In combination with the mild local inflammation that is typically associated with the implanted catheter, this higher concentration may result in phlebitis and vascular thrombosis.49 Even substances that do not induce phlebitis when given as a rapid intravenous bolus may cause irritation when given by continuous infusion, because of the background inflammation in catheterized vessels.49

Considerations for administration volumes.

The volume of solution that can be given varies with species, strain, route, frequency of administration, speed of administration, and composition of the solution. For example, gavage administration of large volumes (20 mL/kg or more) of oil-based formulations is associated with greater toxicity than are aqueous-based formulations.21 Large volumes (10 mL/kg or more) administered by oral gavage can result in absorption changes associated with rapid shunting of the compounds to the duodenum88,133,144 or aspiration pneumonia associated with passive reflux of the material into the esophagus.21 Large volumes given subcutaneously, intramuscularly, and intradermally can result in pain, necrosis, and changes in absorption as well as leakage from the site of injection. Volume of administration also influences the absorption of substances given intraperitoneally,9,20,33 and larger volumes can result in pain and respiratory distress.82

The volume of substances given intravenously should be calculated carefully, because large volumes can result in immediate distress, pulmonary and cardiac abnormalities, and death. The maximum volume of substances that can be administered depends on dosing rate, in which smaller total volumes should be given for bolus administration (over 1 min or less) than for slow infusion (5 to 10 min) or chronic (continuous) infusion. In rats, large volumes (40 mL/kg or greater) of fluid given as a slow infusion (1.0 mL/min) induce clinical signs of distress, including tachypnea and porphyrin pigment staining, as well histologic evidence of pulmonary changes.81 Large volumes of substances given by bolus administration caused increased central venous pressure, hemodilution, acid–base disturbances, and diuresis. 30, 6,82

Generally, best practices for intravenous substance delivery suggest that the blood volume should not acutely be increased more than 4%.82 Special practices may require larger volumes, such as in hydrodynamic gene delivery, in which volumes ranging from 25% to more than 100% of the circulating blood volume are administered to deliver genes to the liver.111 In rodents, volumes of 80 to 100 mL/kg body weight typically are injected into the tail vein over 5 s, resulting in expression levels of reporter genes in approximately 40% of the hepatocytes without the use of viral vectors or other carriers (for review, see reference 111). Similar methods attempted in other species have been less successful.111 Hydrodynamic gene delivery results in swelling of the liver and outflow obstruction, which is believed to be critical to the gene delivery. Although rodents generally survive, this delivery method has considerable side effects. Blood pressure and heart rate drop dramatically (in mice, from 500 to 200 beats per minute), and cardiac electrical abnormalities are observed.110,145 Other approaches have been used to minimize some of these generalized effects and include inferior vena cava delivery and regional methods, such as delivery directly to particular lobes of the liver or isolated muscle groups.57,111,112

With slow or chronic infusion, the composition of the compound, the type of excipient 134 and the age, size, and sex of the recipient can induce potential complications.134 For example, a chronic infusion rate of 2 to 3 mL/kg/h usually is well tolerated in rats; however, increased fetal toxicity has been reported in pregnant rats at infusion rates exceeding 1 mL/kg/h.49 In general, chronic infusion results in decreased water consumption and weight loss. Higher rates of slow or chronic infusion (greater than 1 to 1.5 times the total circulating blood volume per 24 h) can result in hemodilution and diuresis.49

Considerations for substance temperature during administration.

Substances given at or near body temperature will have fewer side effects in animals. The administration of large volumes of cold substances (below body temperature) intraperitoneally or intravenously can induce distress and hypothermia.96,115 Local absorption rate can be influenced by the temperature of substances administered intraperitoneally.13,55

The effect of feeding and fasting prior to substance delivery.

The timing of dosing related to the diurnal rhythm of various species may affect absorption and toxicity in animals and introduce unwanted variability, regardless of the route of administration.12,66 For example, the activity of several hypnotic drugs including ketamine, pentobarbital, propofol, midazolam, and ethanol was tested at different times of the photoperiod, and longer periods of sleeping and anesthesia were observed when drugs were administered in the early active phase (early in the dark phase) as compared with during the early inactive period (early light phase).109 Aminoglycosides have greater toxicity when administered during the resting period than during the active period.10

In addition, fasting and water deprivation affect the absorption of many substances.29,65,108,139 Duration of fasting will vary with the species involved, in that gastric emptying times vary considerably across species, with mice and rats having significantly shorter gastric emptying times than those of larger animals.139

Considerations for subject age when administering substances.

Neonatal animals that receive experimental manipulations may undergo maternal rejection. Important techniques to minimize rejection and cannibalism include wearing gloves to mask hand odors, handling all young in a litter, and ensuring that pups are rewarmed before returning them to their dams.71 Neonatal stress associated with maternal separation for experimental purposes can profoundly affect behavioral indices later in life.72 However, when care is given to use volumes appropriate for the size, species, and route, neonatal animals can be dosed at early time points. Because they are undergoing rapid growth, establishing accurate body weights prior to administering substances is critical. In rodents, oral gavage can be administered as early as postnatal day 1, although waiting until postnatal day 4 is more common.83 In neonates, esophageal tissues are very thin, and care must be taken to use appropriately sized equipment and correct technique. Typically for oral gavage of rodent pups, a 30-gauge needle attached to size 10 polyethylene tubing is used. The end is lubricated, and a small amount (up to 10%) of food coloring may be added to the material to be gavaged (typically 0.05 mL for mouse pups) to permit immediate visualization of the substance within the stomach (through the body wall) after administration.101 Intravenous injections can be started at postnatal day 3 for rodents, and the external jugular and superficial temporal veins are readily accessible sites.59 Intraperitoneal and subcutaneous injections can be given early, although careful attention to volume is required and intraperitoneal injections are more difficult due to limited space in the abdomen. Administration of irritating substances or large volumes that result in discomfort may influence the outcome of the study significantly, in that either of these properties could affect the nursing behavior of the pups, causing them to go off feed.15 In many species, dams lick and consume the feces and urine of their litters, and consideration should be given to the effects on the dam of any test substance or its metabolites that are excreted by the pups.

Conclusion

The administration of substances to animals is a key component of many scientific projects. There are many factors that must be considered by the research team, veterinarian, institutional animal caregivers, and animal ethics committee members to ensure that studies involving experimental administration of substances to animals are planned and conducted appropriately. Careful attention to detail and consideration of the route of administration will contribute to experimental refinement and minimize adverse effects on animals.

References

1. Abbott CR, Small CJ, Sajedi A, Smith KL, Parkinson JR, Broadhead LL, Ghatei MA, Bloom SR. 2006. The importance of acclimatisation and habituation to experimental conditions when investigating the anorectic effects of gastrointestinal hormones in the rat. Int J Obes (Lond) 30:288–292 [PubMed] [Google Scholar]

2. Abe C, Tashiro T, Tanaka K, Ogihara R, Morita HJ. 2009. A novel type of implantable and programmable infusion pump for small laboratory animals. J Pharmacol Toxicol Methods 59:7–12 [PubMed] [Google Scholar]

3. Abu-Hijleh MF, Habbal OA, Moqattash ST. 1995. The role of the diaphragm in lymphatic absorption from the peritoneal cavity. J Anat 186:453–467 [PMC free article] [PubMed] [Google Scholar]

4. Amin K, Dannenfelser R-M. 2006. In vitro hemolysis: guidance for the pharmaceutical scientist. J Pharm Sci 95:1173–1176 [PubMed] [Google Scholar]

5. Atcha Z, Rourke C, Neo AH, Goh CW, Lim JS, Aw CC, Browne ER, Pemberton DJ. 2010. Alternative method of oral dosing for rats. J Am Assoc Lab Anim Sci 49:335–343 [PMC free article] [PubMed] [Google Scholar]

6. Bachmanov AA, Reed DR, Beauchamp GK, Tordoff MG. 2002. Food intake, water intake, and drinking spout side preference of 28 mouse strains. Behav Genet 32:435–443 [PMC free article] [PubMed] [Google Scholar]

7. Ball AM, Smith KM. 2008. Optimizing transdermal drug delivery. Am J Health Syst Pharm 65:1337–1346 [PubMed] [Google Scholar]

8. Ball PA. 2003. Intravenous in-line filters: filtering the evidence. Curr Opin Clin Nutr Metab Care 6:319–325 [PubMed] [Google Scholar]

9. Barrett JS, Wagner JG, Fisher SJ, Wahl RL. 1991. Effect of intraperitoneal injection volume and antibody protein dose on the pharmacokinetics of intraperitoneally administered IgG2a κ murine monoclonal antibody in the rat. Cancer Res 51:3434–3444 [PubMed] [Google Scholar]

10. Beauchamp D, Labrecque G. 2007. Chronobiology and chronotoxicology of antibiotics and aminoglycosides. Adv Drug Deliv Rev 59:896–903 [PubMed] [Google Scholar]

11. Becker DE. 2006. Drug therapy in dental practice: general principles. Part 1—pharmacokinetic considerations. Anesth Prog 53:140–146 [PMC free article] [PubMed] [Google Scholar]

12. Bélanger PM. 1993. Chronopharmacology in drug research and therapy. Adv Drug Res 24:1–80 [Google Scholar]

13. Bendavid Y, Leblond FA, Dubé P. 2005. A study of the effect of temperature on the pharmacokinetic profile of raltitrexed administered by intraperitoneal route in the rat. Med Sci Monit 11:BR1–BR5 [PubMed] [Google Scholar]

14. Beyers TM, Richardson JA, Prince MD. 1991. Axonal degeneration and self-mutilation as a complication of the intramuscular use of ketamine and xylazine in rabbits. Lab Anim Sci 41:519–520 [PubMed] [Google Scholar]

15. Biesemeier JA, Beck MJ, Silberberg H, Myers NR, Ariano JM, Bodle ES, Sved DW, Jacobi S, Stump DG, Hardy M, Stedeford T. 2010. Effects of dose, administration route, and/or vehicle on decabromodiphenyl ether concentrations in plasma of maternal, fetal, and neonatal rats and in milk of maternal rats. Drug Metab Dispos 38:1648–1654 [PubMed] [Google Scholar]

16. Black MC. 2000. Routes of administration for chemical agents. : Ostrander GK. The laboratory fish. London (UK): Academic Press [Google Scholar]

17. Bonnichsen M, Dragsted N, Hansen AK. 2005. The welfare impact of gavaging laboratory rats. Anim Welf 14:223–227 [Google Scholar]

18. Brady AG. 2000. Research techniques for the squirrel monkey (Saimiri spp.). ILAR J 41:10–18 [PubMed] [Google Scholar]

19. Branch-Mays GL, Dawson DR, Gunsolley JC, Reynolds MA, Ebersole JL, Novak KF, Mattison JA, Ingram DK, Novak MJ. 2008. The effects of a calorie-reduced diet on periodontal inflammation and disease in a nonhuman primate model. J Periodontol 79:1184–1191 [PMC free article] [PubMed] [Google Scholar]

20. Bredberg E, Lennernäs H, Paalzow L. 1994. Pharmacokinetics of levodopa and carbidopa in rats following different routes of administration. Pharm Res 11:549–555 [PubMed] [Google Scholar]

21. Brown AP, Dinger N, Levine BS. 2000. Stress produced by gavage administration in the rat. Contemp Top Lab Anim Sci 39:17–21 [PubMed] [Google Scholar]

22. Brunton L, Blumenthal D, Buxton I, Parker K. 2007. Goodman and Gilman's manual of pharmacology and therapeutics, 11th ed New York (NY): McGraw-Hill [Google Scholar]

23. Caron JP, LeBlanc PH. 1989. Caudal epidural analgesia in cattle using xylazine. Can J Vet Res 53:486–489 [PMC free article] [PubMed] [Google Scholar]

24. Chamanza R, Marxfield HA, Blanco AI, Naylor SW, Bradley AE. 2010. Incidences and range of spontaneous findings in control cynomolgus monkeys (Macaca fascicularis) used in toxicity studies. Toxicol Pathol 38:642–657 [PubMed] [Google Scholar]

25. Cinelli P, Rettich A, Seifert B, Bürki K, Arras M. 2007. Comparative analysis and physiological impact of different tissue biopsy methodologies used for the genotyping of laboratory mice. Lab Anim 41:174–184 [PubMed] [Google Scholar]

26. Coria-Avila GA, Gavrila AM, Ménard S, Ismail N, Pfaus JG. 2007. Cecum location in rats and the implications for intraperitoneal injections. Lab Anim (NY) 36:25–30 [PubMed] [Google Scholar]

27. Craig MA, Elliott JF. 1999. Mice fed radiolabeled protein by gavage show sporadic passage of large quantities of intact material into the blood, an artifact not associated with voluntary feeding. Contemp Top Lab Anim Sci 38:18–23 [PubMed] [Google Scholar]

28. Dahan A, Aarts L, Smith TW. 2010. Incidence, reversal, and prevention of opioid-induced respiratory depression. Anesthesiology 112:226–238 [PubMed] [Google Scholar]

29. De Leo L, Di Toro N, Decorti G, Malusà N, Ventura A, Not T. 2010. Fasting increases tobramycin oral absorption in mice. Antimicrob Agents Chemother 54:1644–1646 [PMC free article] [PubMed] [Google Scholar]

30. Diehl KH, Hull R, Morton D, Pfister R, Rabemampianina Y, Smith D, Vidal JM, van de Vorstenbosch C; European Federation of Pharmaceutical Industries Association and European Centre for the Validation of Alternative Methods 2001. A good practice guide to the administration of substances and removal of blood, including routes and volumes. J Appl Toxicol 21:15–23 [PubMed] [Google Scholar]

31. Dubé C, Dubois I, Struthers J. 2011. Intravenous and intraosseous fluid therapy in critically ill birds of prey. J Exot Pet Med 20:21–26 [Google Scholar]

32. Eagle CC, Capes DF. 1993. Use of a new syringe pump (Springfusor) for muscle relaxant infusion. Anaesth Intensive Care 21:444–446 [PubMed] [Google Scholar]

33. Esquis P, Consolo D, Magnin G, Pointaire P, Moretto P, Ynsa MD, Beltramo JL, Drogoul C, Simonet M, Benoit L, Rat P, Chauffert B. 2006. High intraabdominal pressure enhances the penetration and antitumor effect of intraperitoneal cisplatin on experimental peritoneal carcinomatosis. Ann Surg 244:106–112 [PMC free article] [PubMed] [Google Scholar]

34. Evans JG, Kerry PJ. 2000. Common pathological findings in continuous infusion studies. : Healing G, Smith D. Handbook of preclinical intravenous infusion. New York (NY): Taylor and Francis [Google Scholar]

35. Finke MD. 1991. Evaluation of the energy requirements of adult kennel dogs. J Nutr 121 Suppl:S22–S28 [PubMed] [Google Scholar]

36. Fleischman RW, McCracken D, Forbes W. 1977. Adynamic ileus in the rat induced by chloral hydrate. Lab Anim Sci 27:238–243 [PubMed] [Google Scholar]

37. Food and Drug Administration. [Internet]. Guideline for industry. toxicokinetics: the assessment of systemic exposure in toxicity studies. Center for Drug Evaluation Research (CDER). ICH S3A—March 1995. [Cited May 2010]. Available at: http://www.fda.gov/cder/guidance/index.htm.

38. Food and Drug Administration. [Internet]. 2009. Draft guidance for industry. Animal models: essential elements to address efficacy under the animal rule. Center for Drug Evaluation Research (CDER). [Cited May 2010]. Available at: http://www.fda.gov/cder/guidance/index.htm.

39. Gaertner DJ, Boschert R, Schoeb TR. 1987. Muscle necrosis in Syrian hamsters resulting from intramuscular injections of ketamine and xylazine. Lab Anim Sci 37:80–83 [PubMed] [Google Scholar]

40. Germann PG, Ockert D. 1994. Granulomatous inflammation of the oropharyngeal cavity as a possible cause for unexpected high mortality in a Fischer 344 rat carcinogenicity study. Lab Anim Sci 44:338–343 [PubMed] [Google Scholar]

41. Gerwin N, Hops C, Lucke A. 2006. Intraarticular drug delivery in osteoarthritis. Adv Drug Deliv Rev 58:226–242 [PubMed] [Google Scholar]

42. Gonzalez-Mariscal L, Nava P, Hernandez S. 2005. Critical role of tight junctions in drug delivery across epithelial and endothelial cell layers. J Membr Biol 207:55–68 [PubMed] [Google Scholar]

43. Gotloib L, Wajsbrot V, Shostak A. 2005. A short review of experimental peritoneal sclerosis: from mice to men. Int J Artif Organs 28:97–104 [PubMed] [Google Scholar]

44. Gough WB, Zeiler RH, Barreca P, El-Sherif N. 1982. Hypotensive action of commercial intravenous amiodarone and polysorbate 80 in dogs. J Cardiovasc Pharmacol 4:375–380 [PubMed] [Google Scholar]

45. Guy RH. 2010. Transdermal drug delivery. Handb Exp Pharmacol 197:399–410 [PubMed] [Google Scholar]

46. Hahn IH, Hoffman RS, Nelson LS. 2004. EMLA-induced methemoglobinemia and systemic topical anesthetic toxicity. J Emerg Med 26:85–88 [PubMed] [Google Scholar]

47. Hall LW, Clarke KW, Trim CM. 2001. Veterinary anaesthesia, 10th ed Philadelphia (PA): Saunders [Google Scholar]

48. Hayward AM, Lemke LB, Bridgeford EC, Theve EJ, Jackson CN, Cunliffe-Beamer TL, Marini RP. 2007. Biomethodology and surgical techniques. : Fox JG, Barthold SW, Davisson MT, Newcomer CE, Quimby FW, Smith AL. The mouse in biomedical research. Oxford (UK): Elsevier [Google Scholar]

49. Hickling K, Smith D. 2000. The contribution of vehicles, rates of administration and volumes to infusion studies. : Healing G, Smith D. Handbook of preclinical intravenous infusion. New York (NY): Taylor and Francis [Google Scholar]

50. Hill SE, Heldman LS, Goo ED, Whippo PE, Perkinson JC. 1996. Fatal microvascular pulmonary emboli from precipitation of a total nutrient admixture solution. JPEN J Parenter Enteral Nutr 20:81–87 [PubMed] [Google Scholar]

51. Hoggatt AF, Hoggatt J, Honerlaw M, Pelus LM. 2010. A spoonful of sugar helps the medicine go down: a novel technique to improve oral gavage in mice. J Am Assoc Lab Anim Sci 49:329–334 [PMC free article] [PubMed] [Google Scholar]

52. Holzer P. 2010. Opioid antagonists for prevention and treatment of opioid-induced gastrointestinal effects. Curr Opin Anaesthesiol 23:616–632 [PubMed] [Google Scholar]

53. Horsberg TE. 1994. Experimental methods for pharmacokinetic studies in salmonids. Annual Rev Fish Dis 4:345–358 [Google Scholar]

54. Illum L. 2002. Nasal drug delivery: new developments and strategies. Drug Discov Today 7:1184–1189 [PubMed] [Google Scholar]

55. Jacquet P, Averbach A, Stuart OA, Chang D, Sugarbaker PH. 1998. Hyperthermic intraperitoneal doxorubicin: pharmacokinetics, metabolism, and tissue distribution in a rat model. Cancer Chemother Pharmacol 41:147–154 [PubMed] [Google Scholar]

56. Kagan L, Gershkovich P, Mendelman A, Amsili S, Ezov N, Hoffman A. 2007. The role of the lymphatic system in subcutaneous absorption of macromolecules in the rat model. Eur J Pharm Biopharm 67:759–765 [PubMed] [Google Scholar]

57. Kamimura K, Zhang G, Liu D. 2010. Image-guided, intravascular hydrodynamic gene delivery to skeletal muscle in pigs. Mol Ther 18:93–100 [PMC free article] [PubMed] [Google Scholar]

58. Kapural L, Szabova A, Mehkail MA. 2003. Intraspinal drug delivery routes for treatment of chronic pain and spasticity. Seminars in Pain Medicine 1:254–259 [Google Scholar]

59. Kienstra KA, Freysdottir D, Gonzales NM, Hirsch KK. 2007. Murine neonatal intravascular injections: modeling newborn disease. J Am Assoc Lab Anim Sci 46:50–54 [PubMed] [Google Scholar]

60. Kliem MA, Wichmann T. 2004. A method to record changes in local neuronal discharge in response to infusion of small drug quantities in awake monkeys. J Neurosci Methods 138:45–49 [PubMed] [Google Scholar]

61. Knowles JB, Cusson G, Smith M, Sitrin MD. 1989. Pulmonary deposition of calcium phosphate crystals as a complication of home total parenteral nutrition. JPEN J Parent Enter Nutr 13:209–213 [PubMed] [Google Scholar]

62. König KG. 1962. Effects of particle size of corn and sugar diets and of mastication on caries incidence in Osborne–Mendel rats. J Dent Res 41:966–985 [PubMed] [Google Scholar]

63. Latt RH, Echobion DJ. 1984. Self-mutilation in guinea pigs following the intramuscular injection of ketamine and xylazine. Lab Anim Sci 34:516 [Google Scholar]

64. Leary Swan EE, Mescher MJ, Sewell WF, Tao SL, Borenstein JT. 2008. Inner ear drug delivery for auditory applications. Adv Drug Deliv Rev 60:1583–1599 [PMC free article] [PubMed] [Google Scholar]

65. Lee JH, Oh JM, Lee MG. 2008. Effects of water deprivation on drug pharmacokinetics: correlation between drug metabolism and hepatic CYP isozymes. Arch Pharm Res 31:951–964 [PubMed] [Google Scholar]

66. Lévi F, Focan C, Karaboué A, de la Valette V, Focan-Henrard D, Baron B, Kreutz F, Giacchetti S. 2007. Implications of circadian clocks for the rhythmic delivery of cancer therapeutics. Adv Drug Deliv Rev 59:1036–1053 [Google Scholar]

67. Lewis RE, Kunz AL, Bell RE. 1966. Error of intraperitoneal injections in rats. Lab Anim Care 16:505–509 [PubMed] [Google Scholar]

68. Lewis RM, Emmans GC. 2010. Feed intake of sheep as affected by body weight, breed, sex, and feed composition. J Anim Sci 88:467–480 [PubMed] [Google Scholar]

69. Lewis SR, Ahmed S, Dym C, Khaimova E, Kest B, Bodnar RJ. 2005. Inbred mouse strain survey of sucrose intake. Physiol Behav 85:546–556 [PubMed] [Google Scholar]

70. Lewis SR, Dym C, Chai C, Singh A, Kest B, Bodnar RJ. 2007. Genetic variance contributes to ingestive processes: a survey of eleven inbred mouse strains for fat (Intralipid) intake. Physiol Behav 90:82–94 [PubMed] [Google Scholar]

71. Lipman NS, Newcomer CE, Fox JG. 1987. Rederivation of MHV and MEV antibody positive mice by cross-fostering and use of the microisolator caging system. Lab Anim Sci 37:195–199 [PubMed] [Google Scholar]

72. Lu L, Mamiya T, Lu P, Niwa M, Mouri A, Zou LB, Nagai T, Hiramatsu M, Nabeshima T. 2009. The long-lasting effects of cross-fostering on the emotional behavior in ICR mice. Behav Brain Res 198:172–178 [PubMed] [Google Scholar]

73. Ludvig N, Baptiste SL, Tang HM, Medveczky G, von Gizycki H, Charchaflieh J, Devinsky O, Kuzniecky RI. 2009. Localized transmeningeal muscimol prevents neocortical seizures in rats and nonhuman primates: therapeutic implications. Epilepsia 50:678–693 [PubMed] [Google Scholar]

74. Lukas G, Brindle SD, Greengard P. 1971. The route of absorption of intraperitoneally administered compounds. J Pharmacol Exp Ther 178:562–566 [PubMed] [Google Scholar]

75. Lynch JC, Shehabi Y. 1995. Stroke caused by inadvertent intraarterial parenteral nutrition. Anaesth Intensive Care 23:358–360 [PubMed] [Google Scholar]

76. Mann WA, Kinter LB. 1993. Characterization of maximal intravenous dose volumes in the dog (Canis familiaris). Gen Pharmacol 24:357–366 [PubMed] [Google Scholar]

77. Mazzaferro EM. 2008. Complications of fluid therapy. Vet Clin North Am Small Anim Pract 38:607–619 [PubMed] [Google Scholar]

78. Meijer MK, Spruijt BM, van Zutphen LF, Baumans V. 2006. Effect of restraint and injection methods on heart rate and body temperature in mice. Lab Anim 40:382–391 [PubMed] [Google Scholar]

79. Montaguti P, Melloni E, Cavalletti E. 1994. Acute intravenous toxicity of dimethyl sulfoxide, polyethylene glycol 400, dimethylformamide, absolute ethanol, and benzyl alcohol in inbred mouse strains. Arzneimittelforschung 44:566–570 [PubMed] [Google Scholar]

80. Morris RE, Schonfeld N, Haftel AJ. 1987. Treatment of hemorrhagic shock with intraosseous administration of crystalloid fluid in the rabbit model. Ann Emerg Med 16:1321–1324 [PubMed] [Google Scholar]

81. Morton D, Safron JA, Glosson J, Rice DW, Wilson DM, White RD. 1997. Histologic lesions associated with intravenous infusions of large volumes of isotonic saline solution in rats for 30 days. Toxicol Pathol 25:390–394 [PubMed] [Google Scholar]

82. Morton DB, Jennings M, Buckwell A, Ewbank R, Godfrey C, Holgate B, Inglis I, James R, Page C, Sharman I, Verschoyle R, Westall L, Wilson AB; Joint Working Group on Refinement 2001. Refining procedures for the administration of substances. Report of the BVAAWF/FRAME/RSPCA/UFAW Joint Working Group on Refinement. British Veterinary Association Animal Welfare Foundation/Fund for the Replacement of Animals in Medical Experiments/Royal Society for the Prevention of Cruelty to Animals/Universities Federation for Animal Welfare. Lab Anim 35:1–41 [PubMed] [Google Scholar]

83. Moser VC, Walls I, Zoetis T. 2005. Direct dosing of preweaning rodents in toxicity testing and research: deliberations of an ILSI RSI expert working group. Int J Toxicol 24:87–94 [PubMed] [Google Scholar]

84. Murphy M, Carmichael AJ. 2000. Transdermal drug delivery systems and skin reactions. Incidence and management. Am J Clin Dermatol 1:361–368 [PubMed] [Google Scholar]

85. Murphy SJ, Smith P, Shaivitz AB, Rossberg MI, Hurn PD. 2001. The effect of brief halothane anesthesia during daily gavage on complications and body weight in rats. Contemp Top Lab Anim Sci 40:9–12 [PubMed] [Google Scholar]

86. Nakfoor EC, Hunt HR, Hoppert CA. 1952. Fracturing of the molar teeth in caries-susceptible and caries-resistant albino rats (Rattus norvegicus). J Dent Res 31:143–150 [PubMed] [Google Scholar]

87. Ngo MA, Maibach HI. 2010. Dermatotoxicology: historical perspective and advances. Toxicol Appl Pharmacol 243:225–238 [PubMed] [Google Scholar]

88. Nickerson DF, Weaver ML, Tse FL. 1994. The effect of oral dose volume on the absorption of a highly and a poorly water-soluble drug in the rat. Biopharm Drug Dispos 15:419–429 [PubMed] [Google Scholar]

89. Nolan TE, Klein HJ. 2002. Methods in vascular infusion biotechnology in research with rodents. ILAR J 43:175–182 [PubMed] [Google Scholar]

90. Nornoo AO, Osborne DW, Chow DSL. 2008. Cremophor-free intravenous microemulsions for paclitaxel: I: formulation, cytotoxicity, and hemolysis. Int J Pharm 349:108–116 [PubMed] [Google Scholar]

91. Ojewole E, Mackraj I, Naidoo P, Govender T. 2008. Exploring the use of novel drug delivery systems for antiretroviral drugs. Eur J Pharm Biopharm 70:697–710 [PubMed] [Google Scholar]

92. Õkva K, Tamoševiciute E, Cižiute A, Pokk P, Rukšenas O, Nevalainen T. 2006. Refinements for intragastric gavage in rats. Scand J Lab Anim Sci 33:243–252 [Google Scholar]

93. Otberg N, Patzelt A, Rasulev U, Hagemeister T, Linscheid M, Sinkgraven R, Sterry W, Lademann J. 2008. The role of hair follicles in the percutaneous absorption of caffeine. Br J Clin Pharmacol 65:488–492 [PMC free article] [PubMed] [Google Scholar]

94. Pasley JN, Powell EW, Halberg F. 1987. Strain differences in circadian drinking behaviors of ethanol and water in rats. Prog Clin Biol Res 227B:467–471 [PubMed] [Google Scholar]

95. Pauwels R, Newman S, Borgström L. 1997. Airway deposition and airway effects of antiasthma drugs delivered from metered-dose inhalers. Eur Respir J 10:2127–2138 [PubMed] [Google Scholar]

96. Pekow C, Baumans V. 2003. Common nonsurgical techniques and procedures, p 351–391 : Hau J, Schapiro SJ, Van Hoosier GL. Handbook of laboratory animal science, 2nd ed Vol 2: animal models. Boca Raton (FL): CRC Press [Google Scholar]

97. Percy DH, Barthold SW. 2007. Pathology of laboratory rodents and rabbits, 3rd ed Ames (IA): Blackwell Publishing [Google Scholar]

98. Perkin CJ, Stejskal R. 1994. Intravenous infusion in dogs and primates. Int J Toxicol 13:40–47 [Google Scholar]

99. Phalen RF, Mendez LB. 2009. Dosimetry considerations for animal aerosol inhalation studies. Biomarkers 14 Suppl 1:63–66 [PMC free article] [PubMed] [Google Scholar]

100. Prausnitz MR, Mitragotri S, Langer R. 2004. Current status and future potential of transdermal drug delivery. Nat Rev Drug Discov 3:115–125 [PubMed] [Google Scholar]

101. Ramig RF. 1988. The effects of host age, virus dose, and virus strain on heterologous rotavirus infection of suckling mice. Microb Pathog 4:189–202 [PubMed] [Google Scholar]

102. Rang HP, Dale MM, Ritter JM, Flower R. 2007. Rang and Dale's pharmacology. Philadelphia (PA): Churchill Livingstone [Google Scholar]

103. Rasmussen F. 1978. Tissue damage at the injection site after intramuscular injection of drugs. Vet Res Commun 2:173–182 [Google Scholar]

104. Rosen LB. 2011. Nasogastric tube placement in rabbits. J Exot Pet Med 20:27–31 [Google Scholar]

105. Rosen S, Hoppert CA, Hunt HR. 1960. A comparison of the Harvard 700 diet (modified) with the HWC diet (modified) for inducing occlusal caries in resistant and susceptible rats. J Dent Res 39:106–109 [PubMed] [Google Scholar]

106. Rowland M, Tozer TN. 2010. Clinical pharmacokinetics and pharmacodynamics: concepts and applications, 4th ed Philadelphia (PA): Lippincott, Williams & Wilkins [Google Scholar]

107. Ruys JD, Mendoza SP, Capitanio JP, Mason WA. 2004. Behavioral and physiological adaptation to repeated chair restraint in rhesus macaques. Physiol Behav 82:205–213 [PubMed] [Google Scholar]

108. Sagawa K, Li F, Liese R, Sutton SC. 2009. Fed and fasted gastric pH and gastric residence time in conscious beagle dogs. J Pharm Sci 98:2494–2500 [PubMed] [Google Scholar]

109. Sato Y, Seo N, Kobahashi E. 2005. The dosing-time dependent effects of intravenous hypnotics in mice. Anesth Analg 101:1706–1708 [PubMed] [Google Scholar]

110. Sawyer GJ, Dong X, Whitehorne M, Grehan A, Seddon M, Shah AM, Zhang X, Fabre JW. 2007. Cardiovascular function following acute volume overload for hydrodynamic gene delivery to the liver. Gene Ther 14:1208–1217 [PubMed] [Google Scholar]

111. Sawyer GJ, Rela M, Davenport M, Whitehorne M, Zhang X, Fabre JW. 2009. Hydrodynamic gene delivery to the liver: theoretical and practical issues for clinical application. Curr Gene Ther 9:128–135 [PubMed] [Google Scholar]

112. Sawyer GJ, Zhang X, Fabre JW. 2010. Technical requirements for effective regional hydrodynamic gene delivery to the left lateral lobe of the rat liver. Gene Ther 17:560–564 [PubMed] [Google Scholar]

113. Schmid O, Möller W, Semmler-Behnke M, Ferron GA, Karg E, Lipka J, Schulz H, Kreyling WG, Stoeger T. 2009. Dosimetry and toxicology of inhaled ultrafine particles. Biomarkers 14 Suppl 1:67–73 [PubMed] [Google Scholar]

114. Sen S, Chini EN, Brown MJ. 2005. Complications after unintentional intra-arterial injection of drugs: risks, outcomes, and management strategies. Mayo Clin Proc 80:783–795 [PubMed] [Google Scholar]

115. Sharp PE, La Regina MC. 1997. The laboratory rat. Boca Raton (FL): CRC Press LLC [Google Scholar]

116. Sintenie JB, Tuinebreijer WE, Kreis RW, Breederveld RS. 1992. Digital gangrene after accidental intra-arterial injection of phenytoin (epanutin). Eur J Surg 158:315–316 [PubMed] [Google Scholar]

117. Skryabina EA, Dunn TS. 2006. Disposable infusion pumps. Am J Health Syst Pharm 63:1260–1268 [PubMed] [Google Scholar]

118. Smiler KL, Stein S, Hrapkiewicz KL, Hiben JR. 1990. Tissue response to intramuscular and intraperitoneal injections of ketamine and xylazine in rats. Lab Anim Sci 40:60–64 [PubMed] [Google Scholar]

119. Smolensky MH, Peppas NA. 2007. Chronobiology, drug delivery, and chronotherapeutics. Adv Drug Deliv Rev 59:828–851 [PubMed] [Google Scholar]

120. Smythe JW, McCormick CM, Rochford J, Meaney MJ. 1994. The interaction between prenatal stress and neonatal handling on nociceptive response latencies in male and female rats. Physiol Behav 55:971–974 [PubMed] [Google Scholar]

121. Southam DS, Dolovich M, O'Byrne PM, Inman MD. 2002. Distribution of intranasal instillations in mice: effects of volume, time, body position, and anesthesia. Am J Physiol Lung Cell Mol Physiol 282:L833–L839 [PubMed] [Google Scholar]

122. Stookey GK, Warrick JM, Miller LL, Greene AL. 1995. Animal caries models for evaluating fluoride dentifrices. Adv Dent Res 9:198–207 [PubMed] [Google Scholar]

123. Svendsen O, Andersen CB, Mørkhøj CB, Lauritzen B. 2006. Spinal nociception induced by intramuscular injection of oxytetracycline preparations in rats and pigs. Basic Clin Pharmacol Toxicol 99:58–61 [PubMed] [Google Scholar]

124. Swindle MM, Nolan T, Jacobson A, Wolf P, Dalton MJ, Smith AC. 2005. Vascular access port (VAP) usage in large animal species. Contemp Top Lab Anim Sci 44:7–17 [PubMed] [Google Scholar]

125. Tan RH, Dart AJ, Dowling BA. 2003. Catheters: a review of the selection, utilization, and complications of catheters for peripheral venous access. Aust Vet J 81:136–139 [PubMed] [Google Scholar]

126. Taniguchi T, Yamamoto K, Kobayashi T. 1998. Precipitate formed by thiopentone and vecuronium causes pulmonary embolism. Can J Anaesth 45:347–351 [PubMed] [Google Scholar]

127. Tashjian D, Hung SSO. [Internet]. 2005. Noninvasive surgery techniques in fish research: a review on esophageal intubation, dorsal aorta cannulation, and urinary catheterization in sturgeon. In: Sakai Y, McVey JP, Jang D, McVey E, Caesar M. Aquaculture and pathobiology of crustacean and other species. Proceedings of the 32nd US–Japan Meeting on Aquaculture. [Cited 5 November 2010]. Available at: http://www.lib.noaa.gov/japan/aquaculture/proceedings/report32/hung_corrected.pdf

128. Thomsen M, Caine SB. 2007. Intravenous drug self-administration in mice: practical considerations. Behav Genet 37:101–118 [PubMed] [Google Scholar]

129. Tobias JD, Kinder Ross A. 2010. Intraosseous infusions: a review for the anesthesiologist with a focus on pediatric use. Anesth Analg 110:391–401 [PubMed] [Google Scholar]

130. Torres-Arraut E, Singh S, Pickoff AS. 1984. Electrophysiologic effects of Tween 80 in the myocardium and specialized conduction system of the canine heart. J Electrocardiol 17:145–151 [PubMed] [Google Scholar]

131. Touger-Decker R, van Loveren C. 2003. Sugars and dental caries. Am J Clin Nutr 78:881S–892S [PubMed] [Google Scholar]

132. Troncy E, Junot S, Keroack S, Sammut V, Pibarot P, Genevois JP, Cuvelliez S. 2002. Results of preemptive epidural administration of morphine with or without bupivacaine in dogs and cats undergoing surgery: 265 cases (1997–1999). J Am Vet Med Assoc 221:666–672 [PubMed] [Google Scholar]

133. Tsuzuki O, Matsumoto M, Kouno K. 1981. Effect of fluid volume on gastric emptying and gastrointestinal absorption of thiopental-Na and aminopyrine in mouse. Yakugaku Zasshi 101:548–555 [PubMed] [Google Scholar]

134. Turner PV, Pekow C, Brabb T, Vasbinder MA. 2011. Administration of substances to laboratory animals: equipment considerations, vehicle selection, and solute preparation. J Am Assoc Lab Anim Sci 50:614–627 [PMC free article] [PubMed] [Google Scholar]

135. Turner PV, Vaughn E, Sunohara J, Junkin A, Ovari J, Leri F. Oral gavage in rats: an animal welfare issue? J Am Assoc Lab Anim Sci. 2011 In press. [Google Scholar]

136. Vachon P. 1999. Self-mutilation in rabbits following intramuscular ketamine–xylazine–acepromazine injections. Can Vet J 40:581–582 [PMC free article] [PubMed] [Google Scholar]

137. Vachon P, Faubert S, Blais D, Comtois A, Bienvenu JG. 2000. A pathophysiological study of abdominal organs following intraperitoneal injections of chloral hydrate in rats: comparison between two anaesthesia protocols. Lab Anim 34:84–90 [PubMed] [Google Scholar]

138. Valverde A. 2008. Epidural analgesia and anesthesia in dogs and cats. Vet Clin North Am Sm Anim Pract 38:1205–1230 [PubMed] [Google Scholar]

139. Vermeulen JK, De Vries A, Schlingmann F, Remie R. 1997. Food deprivation: common sense or nonsense. Anim Technol 48:45–54 [Google Scholar]

140. Warheit DB. 1989. Interspecies comparisons of lung responses to inhaled particles and gases. Crit Rev Toxicol 20:1–29 [PubMed] [Google Scholar]

141. Wheatley JL. 2002. A gavage dosing apparatus with flexible catheter provides a less stressful gavage technique in the rat. Lab Anim (NY) 31:53–56 [PubMed] [Google Scholar]

142. Wilkinson WS, Morgan CM, Baruh E, Gitter KA. 1989. Retinal and choroidal vascular occlusion secondary to corticosteroid embolisation. Br J Ophthalmol 73:32–34 [PMC free article] [PubMed] [Google Scholar]

143. Workman P, Aboagye EO, Balkwill F, Balmain A, Bruder G, Chaplin DJ, Double JA, Everitt J, Farningham DA, Glennie MJ, Kelland LR, Robinson V, Stratford IJ, Tozer GM, Watson S, Wedge SR, Eccles SA; Committee of the National Cancer Research Institute 2010. Guidelines for the welfare and use of animals in cancer research. Br J Cancer 102:1555–1577 [PMC free article] [PubMed] [Google Scholar]

144. Yuasa H, Numata W, Ozeki S, Watanabe J. 1995. Effect of dosing volume on gastrointestinal absorption in rats: analysis of the gastrointestinal disposition of L-glucose and estimation of in vivo intestinal membrane permeability. J Pharm Sci 84:476–481 [PubMed] [Google Scholar]

145. Zhang G, Gao X, Song YK, Vollmer R, Stolz DB, Gasiorowski JZ, Dean DA, Liu D. 2004. Hydroporation as the mechanism of hydrodynamic delivery. Gene Ther 11:675–682 [PMC free article] [PubMed] [Google Scholar]


Articles from Journal of the American Association for Laboratory Animal Science : JAALAS are provided here courtesy of American Association for Laboratory Animal Science


How do psychoactive drugs reach the brain tissue?

How do psychoactive drugs reach the brain? All psychoactive drugs reach the brain tissue by way of the blood stream. Most psychoactive drugs are taken by one of four routes: by mouth, insufflation, injection, or inhalation.

Which of the following would be a reason for a drug company not to pursue expensive clinical trails with a new drug?

Which of the following would be a reason for a drug company not to pursue expensive clinical trials with a new drug? The therapeutic index is less than 1. prevents many drugs from entering the brain.

What is a stimulant in psychology?

Stimulants are a class of drugs that speed up messages travelling between the brain and body. They can make a person feel more awake, alert, confident or energetic.

When the Pure Food and Drugs Act was first passed quizlet?

The 1906 Pure Food and Drugs Act required drug manufacturers to prove that their drugs were an effective treatment for some diseases. In the early 1800s, _____________ was the medical doctor's most reliable and effective medicine that was used for a variety of conditions, but mainly for pain relief.