Lecture-Notes-1-26
Lecture-Notes-1-26
Lesson 01
Inflammation and
Allergy
Acute inflammation comprises interrelated vascular and cellular changes in affected tissues,
which are aimed at removing or neutralizing the offending stimulus, and repairing injured
tissues. Acute inflammation is necessary for the survival of the affected individual. Although
it serves to protect the individual, the inflammatory process may at times be brought
inappropriately against otherwise harmless substances, such as pollen or against certain body
tissues. The response under such circumstance may then produce damage, which may
constitute a disease process (e.g. anaphylaxis, autoimmune hemolytic anemia and lupus
erythematous). Such cases require drug therapy.
Before taking up drug used for therapy of inflammation and allergy, let us first review the
features of acute inflammatory reaction and immune response. A good knowledge of these
processes is essential to understanding the action of anti-inflammatory and
immunosuppressant drugs.
Page |2
[Remember P A R I S ]
These signs result from the vascular and cellular changes occurring in the affected tissue.
How these signs are generated during inflammation will be discussed shortly.
These components are discussed in sequence. In following the discussion, it will be helpful to
keep in mind a picture of a staphylococcal skin infection in a puppy, or perhaps an acute
inflammation you had say a splinter or infected ingrown nail.
Following an initial vasoconstriction, small blood vessels adjacent to the area of tissue
damage become dilated with increased blood flow, then the flow through these vessels slows
down and become static.
Partially due to anoxia and lack of nutrient supply, the vascular endothelial cells swell and
retract so that they no longer form a complete intact internal lining.
The vessels become leaky, permitting the passage of water, salts and small molecular-sized
proteins to leak out is the small soluble molecular sized proteins from plasma into the
damaged area (exudation).
One of the main proteins to leak out is the small soluble molecule fibrinogen, which is
eventually transformed into insoluble fibrin.
Circulating neutrophils initially adhere to the swollen endothelial cells (margination), and
then actively migrate through the basement membrane vascular epithelium (emigration),
passing into the area of tissue damage. Later, small numbers of blood monocytes (or
macrophages) and lymphocytes migrate in a similar way.
The objectives of these initial reactions to an inflammatory stimulus are three fold, and are
summarized below.
Functions of Acute Inflammation
Removal of inflammatory tissue debris The damaged tissue can be broken down and partly
(cleans up the battlefield) liquefied, and the debris removed from the site of
damage.
As noted above the vascular and cellular events overlap. In fact, the division of the
inflammatory process into vascular and cellular events in mainly for ease of discussion.
These events however constitute a continuum of inflammatory events of the cells involved in
the inflammation some (e.g. vascular endothelial cells, mast platelets, and tissue
macrophages) are normally present in tissues while others (e.g. platelets, and leukocytes) gain
access into the tissue from the blood.
The main cellular events in acute inflammation, all of which are caused by chemical
mediators, overlap with the vascular events as we have seen above. Let us now look into
some features of these cells relevant to inflammation.
During inflammation the endothelial cells of the small arterioles are “activated” to
secrete:
a. Nitric oxide (NO) and prostacyclin, both of which induce vascular relaxation and
inhibit
platelet aggregation
b. Endothelin, plasminogen activator, platelet-activating factor (PAF), thromboxane A2,
angiotensin II, and several cytokines, all of which cause vasoconstriction
c. These cells also express or manufacture several intercellular adhesion molecules
(ICAM), and a variety of receptors including those for histamine, acetylcholine, and
interleukin-1.
Endothelial cell function also involved in angiogenesis (formation of new blood vessels),
which occurs in wound repair, chronic inflammation and cancer. Neutrophils and other
migrating blood cells adhere to the vascular endothelium only when the endothelial cells are
“activated”.
The PMNs are characterized by having nuclei with varying shapes (banded, lobed or
segmented). All of them are found in circulating blood, but may migrate to sites of
inflammation.
Page |5
The neutrophils are the first of the leukocytes to enter the area of inflammation – the “first
line” of defense. Normally inactive, neutrophils have to be activated to enhance their capacity
for phagocytosis, bacterial killing, and generation of some inflammatory mediators. They
have to develop the ability to more actively in a directional fashion (chemotaxis) from the
vessels towards the area of tissue damage.
The eosinophils have capacities similar to those of the neutrophils and in addition, release a
number of potent granule constituents that can damage multicellular parasites such as worms
and mites. These constituents include eosinophil cationic protein, a peroxidase, the eosinophil
major basic protein, and a neurotoxin.
The basophils are very similar in many aspects to mast cells (see below.)
Mast cells
The mast cell morphologically similar to basophil, but are predominantly found in tissues
rather than in the circulating blood. Mast cell’s membrane contains surface receptors for both
immunoglobulin E (IgE) and for complement components C3a and C5a (see under Activated
Complement Components).
Mast cells can be activated to secrete histamine (and other mediators) through stimulation of
these receptors and by direct physical damage.
Monocytes/Macrophages
The monocytes enter the area of inflammation at a later stage, several hours after the
polymorphs. Like neutrophils, they also adhere to endothelium, and migrate into the tissue in
response to specific chemotaxic substances (chemokines). In the tissue, monocytes are
transformed into macrophages (literally “big eaters”, the neutrophils used to be called
macrophages, the “little eaters”). The macrophages bind with bacterial lipopolysaccharides
(LPS) by means of cell surface receptors, and subsequently generate and release cytokines
that act on vascular endothelial cells to further increase vascular permeability, attract other
leukocytes, and cause fever. In areas of inflammation macrophages engulf debris, dead cells
and microorganisms.
Platelets
Page |6
Involved primarily in blood coagulation, platelets also play a role in inflammation. Also
called thrombocytes, they generate thromboxane A₂ (TXA₂) and platelet-activating factor
radicals, and pro-inflammatory cationic proteins. Platelet-derived growth factor
contributes to the repair processes following tissue damage.
Neurons
Some neurons (type C and Aծ fibers) in inflamed areas release inflammatory neuropeptides
when appropriately stimulated. Type C and Aծ fibers transmit pain impulses to the central
nervous system. During injury and inflammation, they generate chemical mediators such as
kinins, serotonin (5-hydroxytryptamine, 5-II), which act on receptors and stimulate the
release of neuropeptides (e.g. neurokinin A and substance P). These substances mediate
pain sensation.
Natural killers
Natural killer (NK) cells are specialized lymphocytes that are active in non-immunological
reactions. They kill target cells, such as virus-infected cells and cancer cells that lack the
major histocompatibility complex (MHC) molecules. MHC molecules normally present in
normal cells inhibit NK cells by acting on the inhibitory receptors on the NK cells
themselves.
How inflammatory mediators bring about the cellular, as well as the vascular events will be
taken up a bit later, but let us, for the moment, talk about inflammatory exudates, after which
will discuss the specific immunological response to inflammation.
The inflammatory exudate is composed of fluids, electrolytes, proteins, cellular elements and
some inflammatory mediators. All of these components are derived from the blood as a result
of vascular and cellular changes of the innate non-immunological response that occur in the
blood vessels in the surviving tissue around the area of damage.
The fluid in the acute inflammatory exudates carries nutrients, mediators and
immunoglobulins.
Fluids and salts may dilute or buffer any locally produced toxins
Glucose and oxygen can diffuse into the area of inflammation to support the macrophages.
The fluid is constantly circulating to the local lymph nodes and assists in later development
of a specific immune response.
The host’s immunological defense mechanism is more specific and therefore more efficient
than the innate response against invading pathogens. The major cells involved here are the
lymphocytes, which are of three main groups.
Natural killer Specialized non-T and non-B lymphoid cells that are active in
cells non-immune innate response (see above)
This phase involves the production of B cells and T cells from uncommitted lymphocytes.
The antigenic molecules are presented to the lymphocytes by the antigen-presenting cells or
APCs. The APCs engulf and process the antigen, and present it to uncommitted CD4+ T
helper lymphocytes (also called T helper precursor) in association with histocompatibility
complex (MHC) molecules
These CD4 cells develop interleukins-2 (II-2), a cytokine which act on the same cells
(CD4+) causing their proliferation, producing clones of activated T cells (called Th0 cells)
Th0 cells give rise to two subsets of T helper cells Th1 and Th2 cells.
The action of specific interleukins determines whether Th1 cells or Th2 cells develop II-12
(produced by APC’s) stimulates development down the Th1 pathway, II-4 (produced by
some Th0) determines development down the Th2 pathway.
Page |8
Some Th1 secrete macrophage-activating cytokines and other cytokines. Some secrete
interferon-gamma (IFN-y), which activates CD8+T cells to become cytotoxic cells (Tc) that
kill virally infected host cells. II-4 derived from Th2 cells inhibits Th1 function.
The Th2 pathway controls antibody (humoral)-mediated responses: IFN-у derived from Th1
cells inhibits Th2 function.
This phase involves how the cells, which are produced in the induction phase, act to eliminate
antigens. This action may be antibody-mediated or cell-mediated.
Antibody-mediated response
When antigens bind with the Fb fractions of the antibodies, antigen-antibody complexes are
formed. The complement proteins bind with the exposed (Fc) portions of the antibody, which
results in the activation of the complement sequence producing, among other things, C3a (an
anaphylotoxin), C3b (a chemotactic factor), and C3b (an opsonin)
Phagocytic cells (neutrophils and macrophages) also have receptors to the Fe portion of the
antibody. The antibody, by acting as an opsonin, greatly facilitates phagocytosis. Antibody
can form a link between a parasite (e.g. worm) and eosinophils, which are then able to
damage or kill the parasite. IgE antibody can attach to mast cells and basophils to stimulate
these cells to release histamine and other inflammatory mediators (see under “Histamine”
below). Histamine is implicated in certain allergic reaction.
In virally infected cells, the virus-derived peptides, in association with MHC, are presented to
the cell surface. CD8+ T cells recognize this MHC-peptide complex and destroy the virus
infected cells. The cytokine-producing T cells also activate macrophages also produce and
release many other cytokines, toxic oxygen and neutral proteases (to kill extracellular
microorganisms), complement components, eicosanoids, fibroblast stimulating factor,
pyrogens (fever-inducing substances), and factor that activate coagulation cascades. The
production of memory cells during first exposure to antigen greatly accelerates and makes
more effective the immunological response to subsequent exposure.
Page |9
Acute inflammation starts when there is damage to tissue. The tissue reacts through vascular
and cellular changes. These changes are mediated by substances, which are collectively
called autocoids. Autocoids (from Greek autos=self and akos=medicinal agent or remedy)
are substances present in the body and act near the sites of their synthesis. They are
sometimes called local hormones, and are presented below.
I. Mediators derived from plasma
Specific Mediators
Origin
Bradykinin and Kallidin
Kinin system
Activated Hageman Factor (XII)
Clotting system
Plasmin
Thrombolic System
C3a, C3b, and C5a, C5b
Complement pathway
II. Mediators derived from cells
HISTAMINE
Histamine is derived from the amino acid histidine. It is distributed throughout the animal
and plant kingdom, present in venom, bacteria and plants. The blood of the goat and rabbit is
relatively histamine, while blood of horse, dog, cat and rat is relatively low in histamine.
Histamine is always present in damaged tissues, decomposing tissue extract, and putrefying
(decaying) ingesta rich in proteins.
Histamine is stored mainly by mast cells, which are abundant in the gastrointestinal mucosa,
bronchial mucosa, and skin. It is also stored in basophils circulating in the blood. It is
implicated in allergic reactions. Non-mast-cell histamine is secreted by histaminocytes in the
stomach and by histaminergic neurons in the brain.
In allergy, histamine is released as a result of interaction of the antigen and the antibody IgE
on the surface of mast cells (or basophils). The presence of an antigen stimulates the
production of IgE, which then binds to the surface of receptors on mast cells. [Note that C3a
and C5a can also induce receptor-mediated histamine secretion; see under “Mast Cells” on
page 5.] The antigen that binds to the IgE that is attached to the mast cells.
The antigen-antibody complex activates phospholipase C in the mast cell membrane, which
catalyzes the breakdown of membrane phospholipids (specifically phosphatidyl inositol)
resulting in the release of diacyglycerol (DAG) and inositol-1, 4, 5-triphosphate (ITP). DAG
P a g e | 10
and ITP by some mechanism cause the release of calcium ions (Ca²⁺) from the intracellular
stores (such as the endoplasmic reticulum), or allow calcium entry into the cytosol.
The elevated free cytosolic Ca²⁺ induces the fushion of the mast cell secretory granules with
the cell membrane resulting in the release of the histamine and other proinflammatory
substances associated with it.
Other than that mediated by the antigen-antibody complex, histamine may also be released
following administration of many compounds including drugs such as amides, amidine,
quaternary ammonium compounds, pyridinium compounds, piperidines, alkaloids, and
antibiotic bases. These drug-stimulated histamine release are not allergic in the nature since
there is no involvement of an antibody.
Intradermally injected histamine produces the so-called triple response of Lewis (first
described by Sir Thomas Lewis). Not all signs of inflammation, however, can be produced by
histamine so administered. Histamine therefore is not the only mediator of inflammation. At
best it is involved only in the initial stage of the inflammatory process
Aside from vascular reactions described above, histamine also causes pruritus (itch) but not
pain. Because of the vascular reactions associated with it, histamine is thought to have a role
in the development of inflammation. Histamine’s effects, however, occur early, is short-lived
and incomplete, and therefore, it may be not be essential for the development of the most
characteristic changes which produce the lasting tissue alteration during inflammation.
Three distinct histamine receptors have been recognized, more may be described in the
future.
Selective
Receptor Function Agonist
Antagonist
Mediates most inflammatory
H1 and are antagonized by the Histamine Mepyramine
classical anti-histamines
Has a role in gastric acid
H2 Histamine
secretion; an action blocked Cimetidine
Dimaprit
by specific H2 antagonists
Histamine
H3 Description is still
(R)-methyl Thloperamide
incomplete
histamine
There are three types of drugs that may be used to prevent or reverse the action of histamine
in clinical cases. These are summarized in the table below.
Dosages
Epinephrine
Dog/Cat: 0.5 to 1.5 ml of 1:10,000 solution IV, repeat in 30 min (for
anaphylaxis)
Horse/Ruminant: 1.0 ml of 1:1,000 solution per 45 kg b.w, IM, IV, SC.
Theophylline
P a g e | 12
Aminophylline:
Dog: 10mg/kg tid, PO, IM, IV
Cat: 5mg/kg bid-tid PO or 4 mg/kg bid PO, IM
CAUTION: monitor for drug toxicity
Cromolyn sodium
Horse: 80 mg nebulized once daily, or
200 to 300 mg nebulized per horse
NOTES:
Cromolyn sodium (Cromoglycate sodium) and its analog Nedicromyl
Cromolyn sodium
Cromolyn sodium is poorly absorbed from the gut, and is usually given inhalation as
nebulized solution or powder. Only 10% of inhale drug is absorb 50% of absorbed portion is
excreted I bile ad 50% in urine. Nedicromyl is better absorbed than cromolyn sodium.
Mode of Action
They inhibit phosphodiesterase (PDE) enzyme that catalysis the breakdown of cyclic
AMP. This action results in an increase (accumulation) of cyclic AMP, which
attenuates the action of inflammation.
Also, they cause relaxation on smooth muscle, which could be also be related to their
effect on cyclic guanine monophosphate (cyclic PDE)
Drugs that inhibit microsomal cytochrome P450 enzymes which may increase the serum
level and the likelihood of adverse effects:
Erythromycin
Ciprofloxacin
Ca2+¿ ¿ channel blockers
Allopurinol
Fluconazole and
Cimetidine,
Because of the very narrow margin of safety, extreme precaution should be taken in
administering theophylline and aminophylline.
Epinephrine (Adrenaline)
The classical or H1 antihistamines are not as widely used in animals as in people because of
two reasons: (a) antihistamines have varying effectiveness in animals, and (b) glucocorticoids
(see Lesson 2) are available and more effective. However, in some cases, antihistamines may
be used in treating dogs and cats with chronic pruritus of undetermined causes, or those with
short-term pruritus nut do not tolerate glucocorticoids. The antihistamines tend to be effective
when used for treatment of type I anaphylaxis, including urticarial and atrophy.
Antihistamines may have varying degrees of anticholinergic, sedative (CNS depression) and
sometimes local anesthetic effects. When histamines are used for purely peripheral action, all
CNS effects such as dizziness are unwanted. Excessive dose may cause convulsion especially
in young animals. The peripheral ant muscarinic effects, such as dryness of the mouth, are
always unwanted.
action]
THE EICOSANOIDS
(Prostaglandins, Prostacyclin, Thromboxane’s and Leukotrienes)
Arachidonic acid (20 carbons), the most abundant precursor of eicosanoids (eicosa – 20, may
be derived by elongation of dietary linoleic acid (18 carbons), and is usually esterified to
second carbon (C2) of cell membrane phospholipids. Arachidonic acid is liberated from this
site during cell damage by the action of activated phospholipase A2. Alternatively,
phospholipase C is first activated, followed by an increase in cytosolicC a2, which in turn,
stimulates phospholipase A2 that eventually release arachidonic acid.
The prostaglandins (PG) are also called prostanoids because they share the chemical features
of a hypothetical substance 20-carbon prostanoic acid with a five-member cyclopentane ring
and two hydrocarbon side-chains.
Prostaglandins are not stored in tissues, but are rapidly formed and released when tissue is
damaged. Thus, the synthesis of prostaglandins is intimately related to their release and
action. Phospholipase A2 is involved in the regulation of biosynthesis of prostaglandins by
causing the release of the precursor arachidonic acid. Two major enzyme systems – cyclo-
oxygenage and lipoxygenase – use arachidonic acid as substance
1. Cyclooxygenase -1 (COX-1)
A constitutive enzyme, which sub serve a number of physiologic role.
Inhibition of COX-1 is believed to underlie the most significant “side effects”
of non-steroidal anti-inflammatory drugs (NSAIs: aspirin-like drugs)
2. Cyclo-oxygenase-2 (COX-2)
COX-2, on the other hand, is induced during inflammation, and inhibition of
COX-2 accounts for the therapeutic action of NSAIDs.
From the endoperoxides, the synthetic process may follow three different paths.
1. 15-HPETE
HPETE’s are unstable and may be converted to their corresponding hydroxyl fatty acid,
known for short HETE.
LTC₄ and LTD₄ are the slow-acting substance of anaphylaxis (SRS-A). They are 1000 times
as potent as histamine in increasing vascular permeability.
Prostaglandins are pathologically important in inflammation but not as essential. They have
an array of metabolic effects. Even in the same organ-system, a PG can oppose the action of
another PG. As mediators of inflammation, PGs cause edema but less than the produced by
histamine and bradykinin. Leukocytes produce PGs a function enhanced during phagocytosis.
PGE is chemotactic for leukocytes. PGs are release during fever and may produce fever
P a g e | 17
themselves. They do not cause pain but sensitize pain receptors to mechanical and chemical
stimulation so that pain seems to be magnified.
Action of Prostaoids
Prostanoids act on specific prostanoid receptors – IP, TP, EP (EP1, EP2, EP3), FP,
and DP
Action of leukotrienes
LTB₄ acts on specific LTB₄-receptors and is a powerful chemotactic agent for bit neutrophils
and macrophages. It also increase the formation of membrane adhesion molecules and
production of toxic oxygen products and release of granules enzyme. LTD₄ and LTC₄ appear
to have specific receptors too. They cause increased respiratory mucus secretion, reduced
blood pressure, coronary vessel constriction, wheals and flare, and increased nasal vascular
P a g e | 18
Prostanoids. PGE and PGF are rapidly metabolized (bio transformed) in the lungs by
prostaglandin dehydrogenase. The depression of capacity of the lungs to metabolized PGs
may result in increased levels of PE₂ and PGF₂ₐ.
4. For estrus synchronization in mares, cows, awes and sows with the use of analogues:
fuprostenol, cloprostenol, dinoprost, tromethamine, prostalene and tiaprost
5. For cardiovascular therapy to ensure the patency of ductus arteriosus in some instance
of congenital heart disease before corrective surgery (use PGI₂), in peripheral vascular
disease of the legs (use PGE₁), in cardiopulmonary by-pass operation. Prostacyclin
(PGI₂), a platelet de-aggregator, is used to prevent blood clotting during the operation
6. gastrointestinal therapy to promote the healing of gastric and duodenal ulcers (use
PGEs) by to mechanisms – inhibition of gastric acid secretion and cytoprotection of
gastric lining and to correct paralytic ileus (use PGF₂ₐ), and (f) as bronchodilators
(use aerosol form of PGE₁ or PGE₂)
P a g e | 19
The NSAID’s are a heterogeneous group of compound which are often chemically unrelated.
The prototype of these agents is aspirin, hence they are also called aspirin –like drugs.
Central analgesia probably has limited contribution to overall efficacy of NSAIDs but
peripheral analgesia is more important. Careful selection of individual NSAIDs may be
necessary to reduce the likelihood of adverse gastrointestinal, renal or hepatic effects.
NSAIDs are shown to stimulate T-suppressor cells in their -action against T-helper
cells and autoantibody-producing B cell
NSAIDs are used as an adjunct in the non-special treatment of pics affected with
porcine respiratory and reproductive syndrome (PRRS), a viral infection
Methylsalicylic acid
Used externally as a counter-irritant.
(Oil of Wintergreen)
Mefenamic acid
Tolfenamic acid
The platelet-activating factor (PAF) is a polar lipid released from leukocytes and causes
platelets to aggregate. The term, however, is quite misleading since PAF has action on variety
of different target cells. Like the eicosanoids, it is not stored but synthesized in response to
stimulation. It is generated and released from most inflammatory cells when these are
stimulated. Its inflammatory effects, which incidentally are also inhibited by NSAIDs, are
mediated through action on specific receptors.
CYTOKINES
Role of Cytokines:
1. Induction of cell adhesion molecules on endothelium
P a g e | 25
Nitric Oxide
Nitric oxide is a small molecule that is locally synthesized by endothelium and macrophage
through the activity of the enzyme, nitric oxide synthesis. It is powerful cause of vasodilation
and increase vascular permeability. As an important re-active oxygen intermediary, it can
also mediate cell and bacterial killing.
Activation of kinins
Kinins are activated by protease enzyme kallikrein. Kallikreins are produced by activation of
the inactive precursor kallikrein, which is present in plasma. The clotting factor XII
(Hageman factor), initially activated on exposure to collagen, cleave pre-kallikreins into
active kallikreins. The kallikreins then cleave the substrate kininogen to form active kinins.
The types of kininogens are high-molecular weight kininogen (HMW-K) which is activated
to bradykinin, and lower molecular weight kininogen which activated to kallidin.
Both bradykinin and kallidin have very short half-life (about 15 seconds) in the plasma. They
are readily metabolized by kininase II, which is identical to angiotensin converting enzyme
(ACE), and by the slower- acting kininase.
Complement is a collective term referring to heat labile factor in the extra cellular fluids. If
antibodies, which are directed against the surface of the cells, and are able to fix complement,
a cytotoxic reaction may occur.
Complement Activation
Complement can be activated either by:
1. The Classical Complement Activation Pathway or
2. The Alternative Complement Activation Pathway
Activation of C4
C1s activates C4 by cleaving a fragment and forms, C1,4,
which causes immune-coagulation and virus neutralization
Activated C4 has several functions; binds to cell membrane
P a g e | 27
C3b exist in trace amounts in It combines with a serum factor called factor b,
normal serum forming a complex C3bB
P a g e | 28
When platelets or mast cells are the target or the complement reaction, mediators of
inflammation may be released and activated
Blood clotting is initiated by activation of Hageman factor (Factor XII). Factor XII
interacts with the kinin-kallikrein system resulting in the generation of vasoactive kinins.
Factors XII by itself increase vascular permeability. In disseminated intravascular
coagulation (DIC) as that occurring in toxemia, the involvement of the inflammatory process
in such abnormality of blood coagulation may be a critical outcome or the disease.
The best “anti-inflammatory” agent in such clinical setting is heparin (an anticoagulant)
THROMBOLYTIC SYSTEM
Lesson 02
P a g e | 30
Glucocorticoids
and
Immunosuppressan
t Agents
G lucocorticoids are steroidal agent produced and released by the adrenal cortex.
Cells of the adrenal cortex secrete endogenous glucocorticoids. The term corticosteroid
applies to any steroidal hormone secreted by the adrenal cortex but is often used
synonymously with glucocorticoids. Adrenal steroids are divided into three main groups
based on their physiological activity.
2. Mineralocorticoids
Aldosterone, predominantly affect electrolyte and water metabolism
The ruminant secretes sizable amount of both. Both of these glucocorticoids have some
degree of mineralocorticoids activity. The structure of cortisol is shown below. These are
four rings in the steroid structure and are designated here as rings A, B, C, and D.
substitutions in each ring affect its glucocorticoids activity.
The glucocorticoid base refers to the non-esterified steroid component and determines its
glucocorticoid, an anti-inflammatory potency, and extents its duration of action.
COMPARISON OF GLUCOCORTICOIDS
SHORT ACTING
Hydrocortisone 1 ++ <12
Cortisone 0.8 ++ <12
INTERMEDIATE
ACTING 3.5 + 12 – 36
Prednisone 4 + 12 – 36
Prednisolone 5 0 12 – 36
Methylprednisolone 5 0 12 – 36
Triamcinolone
LONG ACTING
Paramethasone 10 0 >48
Betamethasone 25 0 >48
Dexamethasone 30 0 >48
Pivalate
P a g e | 33
The dose
Potency of the base, and
Duration of the action of the formulation.
Therapeutic success with fewer side effects is possible with alternate-day therapy.
This stems for the fact that some preparation have slightly longer anti-inflammatory or
immunosuppressive action than their action to suppress the HPA axis.
Stress
Hypothalamus
Corticotropin
Cortisol
The cells of the adrenal cortex secrete endogenous glucocorticoids (cortisol and
corticosterone). The secretion is regulated by hypothalamus-pituitary-adrenal axis (HPAA).
P a g e | 34
See illustration above. Respond to the influence of the circadian rhythm (biological clock)
and the changing stress condition of the animal, the hypothalamus secretes corticotropin –
releasing hormone (CRH), which is brought to the anterior pituitary through the
hypothalamus-pituitary portal system. In the anterior pituitary, CRH stimulate the secretion
of the hormone corticotropin (ACTH). ACTH gets to the adrenal medulla by way of the
blood circulation and stimulate the secretion of glucocorticoids by the cells of the adrenal
cortex.
The secretion of corticotropin by the anterior pituitary is episodic rather than continuous. In
the normal animal, the blood glucocorticoid concentration varies with the circadian rhythm.
The pattern of changing, however differs between species.
For example:
The dog which are generally active during daytime (diurnal), have high blood
glucocorticoid concentration during the day, and low at night
Cats, which are active at night (nocturnal), show the reverse pattern.
The inhibition become irreversible especially with prolonged use and repeated administration
of glucocorticoids. When this happens, the adrenal cortex may not be able to produce
endogenous cortisol resulting in hypoadrenocorticism (Addison’s disease). Some ways to
minimize this adverse effect of glucocorticoids in clinical practice will be discuss later in this
lesson.
Physiological Role of Glucocorticoids
Knowledge of the physiological role of glucocorticoids is essential in the rational and safe
use of these agents in clinical situations. These roles are outlined below.
Regulate the volume and composition of body fluids mainly through their
mineralocorticoids action
Make the animal able to resist the effects of noxious stimuli (such s during
inflammation) and of various form of physical and emotional stress.
P a g e | 35
The pharmacological effects of glucocorticoids are produced by rather than high doses. When
a clinician administers a glucocorticoid to a patient, effect other than those originally desired
may also occur. These side effect may be harmful to the patient and at times mat result in a
condition far worse than the original problem that had required treatment with
glucocorticoids.
Effects of CNS
The effects or glucocorticoid on the CNS are rarely describe in domestic animals, but
may have marked effect on the psyche resulting in a form of mental and physical
dependence. In human beings, glucocorticoids may induce a feeling of euphoria.
The anti-inflammatory of effects are usually what the clinician’s desire when using
glucocorticoids.
Most of these effects however, are nonspecific, that is, they occur regardless of the
nature of the initial insult. When used to suppress tissue graft rejection,
glucocorticoids suppress the initiation and generation of an immune response more
efficiently.
P a g e | 37
Therapy with glucocorticoids (and other steroids for that matter) is not without
danger. One should always remember that serious adverse side effects may occur side
by side with the desire effects.
Glucocorticoids enter cells and bind with specific cytoplasmic receptors, which are found in
vitually all tissues. This interaction causes the receptor to undergo conformation change with
exposes a DNA-binding domain. The steroid-receptor complexes move into the nucleus and
bind to steroid-response elements in the DNA. The effects is either to repress (prevent
transcription of) or to induce (initiate expression) particular gens.
Repression result from inhibition of the action of various transcription factors such as AP-1
and NF-Kb. These transcription factors normally usually switch on the genes for COX-2,
various cytokines, and the inducible from of nitric oxide synthase. Induction involves the
formation of specific mRNAs. Which direct the synthesis of specific proteins.
Much is now known about the anti-inflammatory and immunosuppressive actions of the
glucocorticoids, but their metabolic action are less well understood.
Those with very rapid onset and short Hydrocortisone sodium succinate
duration are used in emergency Hydrocortisone sodium phosphate
situation such as hemorrhagic-shock and Prednisolone sodium succinate
anaphylaxis. These are fast-acting
(within1 min) with short plasma half-
Parenteral life (1-2 h), but are rather expensive
Preparation
Those with short duration are used for Prednisolone - mimics diurnal
acute and chronic condition and for cortisol production in most
alternate –day therapy animals; causes sodium retention
Oral (not significant in dogs and cats)
Preparation
Prednisone- converted to
prednisolone in the liver
Methylprednisolone
After having learned of the various real adverse effects of glucocorticoids, it seems it would
be better for the patient, and for the veterinarian, not to use glucocorticoids at all. Not so!
P a g e | 39
When used properly and with diligent monitoring of patient under therapy, glucocorticoids
can be a valuable therapy of important clinical conditions.
The safety glucocorticoids use may be improved if specific answer to these question can be
made before starting therapy.
1. How serious is the underlying disorder?
2. How long will therapy be required?
3. Is the patient predisposed to any of the complication of steroid therapy?
4. What is the anticipated glucocorticoids dose?
5. Which glucocorticoids preparation should be used?
6. Have other modes of therapy been utilized to minimize thee steroid dosage, and to
minimize the side effects?
7. Is alternate day regimen indicated?
For dogs – give once a day every morning: for cats – give once a day every evening.
P a g e | 40
Supplemental dose given in moderate stress- increase the dose 2-5x; in severe stress-
increase dose 5-20x
Dosage
Hydrocortisone or cortisone 0.2-1 mg/kg
Prednisone or prednisone 0.1-0.2 mg/kg
Dosage:
Prednisolone:
For induction 1.1 mg/kg divided oral, bid until desired effect is
attained
For maintenance 0.55 - 2.2 mg/kg oral every 2 days
Methylprednisolone:
1.1 mg/kg IM, SC every o1-3 weeks
Dosage
Prednisolone (for induction ) 2.2-6.6 mg/kg divided bid until effects
Dexamethasone (for maintenance) 2-2.2 mg/kg every 2 days
Prednisolone (for maintenance) 0.33-1.1 mg/kg
However, the following signs might suggest that further prolongation of the weaning
process is necessary;
Dullness
Mental depression
Increase fatigability
Incoordination
Unthriftiness and weight loss
Diarrhea
Behavioural changes
Hyper function of the adrenal cortex is rare, although cushing’s disease has been reported in
the dog and horse.
Adrenal blocking agents, which would have some clinical indication under this condition are:
1. o,p’DDD
A relative of DDT, is 1,2- dichloro-2,2 (p – chlorophenyl) ethane. This
compound has been found to suppress adrenal cortical activity in the dog, but
it is less successful in other species. Rats and monkeys are unaffected.
P a g e | 43
Prolonged therapy with o,p’DDD in goats caused atrophy of the cortex and
transitory cytopathologic effects of adrenal cortical cells of calves
2. Amphenome B
Is (3, 3- bis p-aminophenyl butane). It inhibits 17 - ketosteroid production in
human and dog
3. Metyrapone
Effectively blocks cortisol production but not corticosterone production inn
young pigs
IMMUNOSUPPRESSANT AGENTS
Most immune suppressant drug act in the induction phase of the specific immunological
response (see Lesson 1) resulting in the reduction of lymphocytes proliferation. There are
also some effects on the effector phase.
Mechanism Example
Inhibitors of interleukins-2 (1L2) Cyclosporin
Tacrolimus
Rampamycin (a.k.a sirolimus)
Inhibitor of cytokine gene expression Glucocorticoids
Cytotoxic agents Cylosphosphamide
Chlorambucil
inhibitors of purine and pyrimidine Azathioprine
synthesis Mycophenolate mofecil
Blockers of T-cell molecule involved in Immunoglobulins (monoclonal and
signaling polyclonal antibodies)
Clinical Uses
The immunosuppressant agents are used commonly in therapy of autoimmune disease, and
prevent and treat transplant rejection. Its use, however may entail some adverse effects, most
common of which are (a) decrease response to infection, and (b) facilitation of the emergence
of malignant cells.
Specific Drugs
Cylosporin A
It is one of the 9 cylosporin A through, each is a cyclic peptide. Cylosporin is very
lipophilic and hydrophobic and must be solubilized before administration
complex then binds with calcineurin (a phosphatase). This binding inhibits the
calcium-stimulated phosphatase activity leading to failure to dephosphorylate
regulatory proteins. Dephosporylation of these proteins is essential to their
translocation into the nucleus where they serve as subunits of transcription factors.
Translocated regulatory proteins set to motion the transcription of gene for 1L-2.
Synthesis of 1L-2 is therefore inhibited in the presence of cyclosporin.
Cyclosporine is given orally or intravenous. Absorption from the gut is generally poor
but more absorbable preparation have been developed.
Rapamycin (sirelinus)
Rapamycin, another macrolides, also binds with FKBP. However, the rampamycin-
FKBP complex does not bind with calcinuerin nor does it effects IL-2 transcription. It
interferes with signal transduction pathway blocking the cell cycle of activated T cells
in G1 phase by inhibiting a novel kinase called mTOR
Glucocorticoids
Immunosuppression by glucocorticoids involves both their effects on immune
response and their anti-inflammatory actions (discussed earlier). Like cyclosporine,
glucocorticoids restrain the clonal proliferation of T cell through decreasing
transcription of the gene for IL-2 and many other cytokine genes including of those
TNFa, INFy, IL-1 and many other interleukins in both the induction and effector phase
of immune response
Cyclophosphamide
It is an alkylating agent with particular action on lymphocytes. As a
immunosuppressant it effects the clonal proliferative phase of the immune response
and reduce both antibody-mediated and cell-mediated immune reactions.
Chlorambucil
It is another alkylating cytotoxic agent similar to cyclophosphamide
P a g e | 45
Azathioprine
It is used particularly for tissue rejection in transplant surgery. It is metabolized to
mercaptopurine, a purine analogue that inhibits DNA synthesis. Both cell-mediated
and antibody-mediated immune reactions, and are depressed by this drug since it
inhibits clonal proliferation in the induction phase of the immune response by a
cytotoxic reaction on dividing cells.
Mycophenolate mofetil
A semi-synthetic derivative of a fungal antibiotic, it is converted to mycophenolic
acid, which restrains the proliferation of both B and tT lymphocytes and reduces the
production of cytotoxic. T cells by inhibiting inosine monophosphate dehydrogenase.
This pathway is essential for de novo purine synthesis B and T lymphocytes are
particularly dependent on this enzyme.
P a g e | 46
Lesson 03
Drugs Acting on
the Respiratory
System
Most of the clinical signs of disease of the respiratory system can be attributed to anorexia or
hypoxia.
Anoxia
Specifically refers to the complete failure of the tissues to get adequate supply of
oxygen
Hypoxia
P a g e | 47
Refers to the reduce supply of oxygen, and is the more appropriate term for the usual
condition accompanying respiratory disorder.
Types of Hypoxia
Anoxic hypoxia
Refers to defective oxygenation of blood in the pulmonary circuit and is
usually caused by a primary disease of the respiratory tract
Anemic hypoxia
Due to deficiency of haemoglobin per unit volume of blood but the blood
oxygen tension is normal
Stagnant hypoxia
Caused by a reduction of the rate of blood flow to and from the capillaries
Histotoxic hypoxia
Result from the failure of the tissue oxidation system in the presence of fully
oxygenated blood.
When anorexia develops rather slowly, the body compensates for the effect oxygen
lack on tissues. The depth and rate of respiratory movement increase (hyperpnoea).
This increase is mediated by the chemoreceptors in the carotid and aortic bodies. The
spleen contracts to express reserve blood into the circulation. Thus, momentarily
increasing the number of circulating red blood cell (RBC) to carry oxygen to tissue
later, the bone marrow produces more RBC (erythropoiesis), as a response to increase
oxygen demand. More red blood cells are produce by, which may eventually result in
polycythemia (increase number of RBC). The heart rate and stroke volume also
increase to maximize the delivery of oxygen to tissues.
Antimicrobial therapy
Bacterial infections involving the respiratory system are treated with
appropriate antimicrobial drugs. Since they are not specific respiratory drugs,
antimicrobial agents are taken up later lessons
Surgery
Obstruction of upper respiratory tract may require tracheotomy in some cases
Oxygen therapy
Admission of oxygen is not often adopted in the treatment of large animals,
but may be used more frequently in small animals. Oxygen therapy, however,
is useful only in cases of anoxia and when the respiratory tract is not
completely obstructed
Artificial respiration
This may be instituted by intermittent – positive pressure ventilation (IPPV)
during open – thoracic surgery or paralysis with a muscle relaxant drugs or
poisons
Supportive therapy
Artificial feeding, correction of acid – base abnormalities, fluid therapy, etc.,
Included in this lesson are agents used in the symptomatic treatment of respiratory disease.
Remember that treatment of respiratory disease will seldom be successful unless the principal
cause of the disease is removed.
Alteration in the quantity and quality of respiratory secretions accompanies many respiratory
disorders. In general, an increase in the volume of secretion is a beneficial protective
response in many respiratory disorders as it dilutes and facilitates removal of offending
agents from the respiratory tract. Gross overproduction of respiratory secretions, however,
may lead to obstruction of the airways to the point that asphyxia (a condition of hypoxia
combined with hypercapnia) may occur. Respiratory secretions may at times be abnormally
reduced. Drugs may be used to increase or decrease the volume of respiratory secretion.
P a g e | 49
This group includes the expectorants. Expectorants are drugs that increase the volume
and fluidity of secretions in the respiratory tract. Production of more fluid secretion
encourages the movement of respiratory debris towards the mouth, at the same time
soothes irritated mucous membranes. Expectorants may act directly or indirectly on the
respiratory mucosa.
Expectorants
Type Mode of Action Examples
Expectorants are not too dramatic in their effect and there have been questions as to
whether they are of value at all. They are probably of some use when there is actual
drying of the mucous membrane and in the removal of inflammatory debris. The soothing
action of expectorants on irritated mucous membranes exerts some degree of antitussives
(cough – suppressing) action (see below).
P a g e | 50
The inhibitors of respiratory secretion are used in the management of acute and chronic
rhinitis. The ɑ - adrenergic stimulants are almost always the best choice.
These are used to degrade and liquefy inflammatory debris. Because of this, mucolytics
are often included with expectorants (see above). Inflammatory materials such as mucus
P a g e | 51
plugs and necrotic debris accumulate in many respiratory disorders and can potentially
lead to obstruction of the respiratory tract. Certain drugs have been used in an attempt to
liquefy the debris so it may be expelled readily by coughing.
D. Antitussives
Antitussives are drugs used to suppress cough. Cough is a protective reflex initiated by
the presence of foreign materials in or by irritation of the respiratory tract. The reflex
which involves the cough center in the brain is characterized by rapid contraction of the
diaphragm and the intercostal muscles, and by a forceful expulsion of air from the
respiratory tract.
P a g e | 52
Antitussives
Categories Mode of Action Mode of action and remarks
Centrally – acting antitussives
Non – selectively depress the cough center
Codeine in the medulla oblongata which may also
Diheydrocodeine lead to depression of other parts of the CNS
Dihydrocodenone (narcosis)
Morphine Codeine and Hydrocodeine seem to have
Narcotic
(analgesic and antitussive) fairly high degree of selectivity for the
antitussives
cough center while opiates are highly
efficacious for they are addicting and are
subject to dangerous drug control
Opiates are relatively toxic in cats
Antitussives should be used only in cases of non – productive cough to prevent trauma to
the respiratory tract and allow the patient to rest. They may be contraindicated in cases of
productive cough. The combination of an expectorant and an antitussive may seem
irrational but does not seem to cause untoward effect in human as well as in animals.
P a g e | 53
E. Bronchodilators
Bronchodilators cause relaxation of the bronchial smooth muscles and increase the
functional capacity of the respiratory tree. Their effect is not too pronounced in the
normal respiratory system but they have dramatic ability to counteract pathological
bronchoconstriction. A wide variety of endogenous substances mediate
bronchoconstriction. These include acetylcholine, histamine, serotonin, bradykinin,
and prostaglandins. The major components responsible for bronchodilation are the β₂
- adrenergic receptors.
Bronchodilators
Drugs Remarks
Methylxanthines
(Theophylline, Potent bronchodilators which act in the manner
Aminophylline, and similar but not identical to the sympathomimetics
Caffeine) They can be used when refractoriness (or tolerance)
develops with prolonged use of sympathomimetics
(see below)
P a g e | 54
DRUGS DOSAGES
EXPECTORANTS
Guaifenesin Horse 0.1 to 0.2 g/50 kg b.w. oral qid
ANTITUSSIVES
Dog 0.55 mg/kg oral
Butorphanol
0.055 mg/kg SC
Codeine Dog 0.1 to 0.3 mg/kg oral tid
Dextromethorphan Dog/Cat 1 – 2 mg/kg oral bid
MUCOLYTICS
50 ml/hr (of 10% aqueous solution) for
Dog
30 – 60 min bid by nebulization
BRONCHODILATORS
Dog 10 mg/kg oral q6 – 8h
Cat 6 mg/kg oral q12h
Aminophylline
Horse 5 – 10 mg/kg oral TID
Calf: 0.5 – 1 g/kg oral QID
Dog 2 mg/kg oral q8h
Ephedrine Cat 1 mg/kg oral q8h
Horse 0.7 mg/kg oral BID
0.5 – 1.5 ml (of 1:10,000 sol’n) IV
Dog/Cat
repeat 30 min for
anaphylaxis
Epinephrine 4 – 8 mg/450 kg IM, SC, (IV only for
Horse
emergencies such as
anaphylaxis)
Sheep 1 – 3 ml of 1:10,000 sol’n IM, SC
Dog 6 – 11 mg/kg oral, IM, IV, tid – qid
Cat 4 mg/kg oral tid
Theophylline 0.1 mg/kg IM, IV tid
Horse 1 mg/kg oral qid
Ruminants 28 mg/kg oral once a day
Albuterol Dog 0.02 - - 0.04 mg/kg oral OD – TID
Clenbuterol Horse 0.8 – 3.2 µg/kg oral BID
0.8 µg/kg IM or slow IV BID
Terbutaline Dog 2.5 mg/kg oral, SC, tid
Cat 1.25 mg/cat oral bid or
0.625 mg/ cat oral, SC, bid
P a g e | 57
Lesson 04
Cardiovascular
Drugs
A
P a g e | 58
Short review of the essential points of cardiac electrophysiology will help us understand the
actions of drugs affecting cardiovascular functions.
The resting membrane potential (RMP) of myocardial cells is controlled mainly by the
difference between the intracellular and extracellular concentrations of potassium ions (K ⁺).
the usual intracellular and extracellular concentrations of potassium are: [K ⁺]ᵢ = 150 mM and
[K⁺]ₒ = 4 mM, respectively. Because of the steep potassium concentration gradient across the
cell membrane, some K⁺ slowly flow outward through the K⁺ leak channels and leaving the
inside more negative in relation to the outside. As the positively charged potassium ions go
out, the membrane becomes polarized with negative and positive side.
The myocardial cell membrane may undergo a partial depolarization either by decreasing the
outflow of K⁺ (reducing the negativity of the cytoplasmic side of the membrane) or by letting
a little Na⁺ to flow inward through the K⁺ leak channel (making the membrane inside more
positive or less negative). Either way, the membrane becomes partially depolarized until
reaching a certain level called threshold potential which then causes the opening of the
sodium channels and becomes activated.
These channels are said to be voltage – dependent for they are activated only when there is an
initial change (toward positive) in the membrane potential. When the sodium channels are
opened or activated, more sodium ions rush in. this movement is easy because both the
concentration and electrical gradients of sodium are directed inward. That means Na ⁺ flows
from outside where it is more concentrated to the inside where it is less concentrated and
since it is positively charged, it is attracted to the negative inside. The rushing in of sodium
ions passes over the threshold potential and consequently produces an action potential
(impulse). In the process, K⁺ is repelled by the increasing positivity or reduced negativity of
the intracellular space. K⁺ then is forced to flow out.
The presence of sodium inside the cell activates the sodium pump or Na ⁺ - K ⁺ - ATPase.
This membrane – bound enzymes pumps out 3 sodium ions in exchange for 2 potassium ions.
The activation of the sodium pump tries to restore resting membrane polarity. Once the
resting membrane potential is restored, the condition is again made conducive to the outward
K⁺ flow as before, and another round of depolarization occurs.
The impulse – conducting system of the heart consists of the sino – atrial (SA) node,
atrioventricular (AV) node, purkinje fibers, and myocardial cells. These specialized cells
have resting membrane potentials in increasing order; lowest (least negative inside) in the SA
node and highest (most negative inside) in the myocardial cells. The one with the least RMP,
P a g e | 59
the SA node, is the most excitable and therefore sets the pace of activation of the heart, thus
serving normally as the pacemaker of the heart. The RMP in any of these calls can be altered
by pathological disturbances such as hypoxia (due to myocardial infarction) and electrolyte
imbalances (e.g. hypokalemia or hyperkalemia).
The myocardial muscle fibers may be grouped into fast – response fibers and slow –
response fibers. This classification, however, is not permanent in the sense that a fast –
response fiber may become a slow – response fiber, or the other way around, depending upon
the pathological conditions of the fibers. For example, a fast – response fiber may become a
slow – response fiber if it subjected to hypoxia. It may then revert to a fast fiber when
hypoxia is corrected.
Have RMP of – 90 mV
Fast – response fibers When depolarized, produce rapid upstroke (phase 0) of action
potential
Impulse conduction along these fibers is rapid (0.3 – 3 m/sec)
Once a myocardial cell is activated (stimulated) by an appropriate stimulus, the rate of rise
(slope) of phase 0 of the action potential, and the velocity of conduction of this action
potential (impulse) is directly proportional the value of the initial RMP. In other words, the
more negative the RMP, the greater is the rise of phase 0 and the faster is the rate of impulse
conduction. The RMP of a fast – response fiber may be reduced under ischemic condition
(due to hypoxia or blood supply); conduction of impulse along these fibers is slowed down
and constitutes a “heart block”.
Phase 0 (depolarization)
Have 2 components:
The fast component
Mediated by the movement of Na⁺ through the “fast channels”
(in myocardium and purkinje fibers) that is voltage –
dependent. These fast channels are fully activated between -70
and -50 mV. As the depolarization progresses, these channels
are inactivated. The time course for inactivation is about 0.5
P a g e | 60
Phase 2 (plateau)
Due to the algebraic sum of the inward Ca²⁺ and the outward K⁺ current,
calcium channel are also voltage – dependent and is activated at -40 mV. The
time course for inactivation is 50 milliseconds. When calcium current ceases,
potassium outflow continues to cause phase 3
Phase 4 (diastole)
Characterized by a restored intracellular K ⁺ concentration caused by Na ⁺ - K ⁺
- ATPase pump. The RMP remains constant in the myocardial cells and
purkinje fibers. However, spontaneous depolarization to threshold potential
occurs in the sinus and AV nodal cells due to progressive decrease in K⁺
permeability (or to slow inward movement of Na⁺ and Ca ⁺⁺). In these tissues,
the RMP becomes progressively less negative. The threshold potential is
eventually reached and an action potential is produced. The frequency of
discharge of action potential is determined by: (a) the distance between the
maximal diastolic potential (phase 4) and the threshold potential and, (b) the
slope of the diastolic depolarization (phase 4), that is the greater the slope, the
earlier the threshold potential is reached and thus more frequent firing.
With the foregoing short review, we are now ready to discuss specific drugs acting on the
heart. If you feel these are not enough, you may consult any reference on cardiovascular
physiology or your old notes in physiology for a more detailed discussion.
Digitalis Glycosides
The digitalis glycosides, also known as cardiac glycosides, are important drugs used for the
treatment of congestive heart failure. They have the ability to increase the force of
myocardial contraction and to increase cardiac output in failing heart.
All digitalis glycosides are derived from plants of similar species which are collectively
called “fox gloves” by the shape of their flowers. Shown below are active principles and their
plant species of origin.
PLANT SOURCES ACTIVE GLYCOSIDES
Digitalis purpuria Digitoxin, gitoxin, and gitalin
Digitalis lanata Digitoxin, digoxin, and gitoxin
Strophantus kombe Strophantin
Strophantus gratis Ouabain
The skin glands of some species of toad contain cardioactive poisons similar to cardiac
glycosides but they are of toxicological importance only.
The cardiovascular response to digitalis glycosides results from a complex of direct and
indirect actions on the heart.
The direct action of digitalis glycosides produces a positive inotropic effect (i.e. increased
force of heart contraction).
This positive effect of digitalis doesn’t seem too dependent upon adrenergic
stimulation; it is believed to be a consequence of inhibition of the sodium pump.
Digitalis binds with the sodium pump (Na⁺/K⁺-ATPase) at the binding site of K⁺ thus
preventing the active transfer of K⁺ back to the inside cell. Consequently Na⁺ is not moved
from the inside to the outside, at least not yet. How can this change lead to a positive
inotropic effect? Three explanation have been offered:
The following repolarization the sodium pump (Na⁺/K⁺-ATPase) returns Na⁺ to the outside
and K⁺ to the inside of the cell. A secondary sodium-calcium exchange mechanism is also
present in the cell membrane and removes intracellular sodium. When the sodium pump is
inhibited by digitalis, the calcium-sodium exchange becomes the predominant mechanism for
P a g e | 62
extruding sodium from the intracellular calcium increases causing stronger myocardial
contractions in the failing heart.
In a resting (unexcited) cell, the intracellular calcium is normally exchanged for extracellular
sodium (that is sodium in; calcium out). Since intracellular sodium is increased when the
sodium pump is inhibited, there is lesser tendency for more sodium to flow in and for calcium
to flow out. Calcium then accumulates inside.
Most of the accumulated calcium is stored in the sarcoplasmic reticulum. Consequently,
greater amount of calcium is released from the sarcoplasmic stores during each about of
action potential, resulting in stronger force of myocardial contraction.
By a mechanism not clearly defined, increased inward calcium current occurs at phase 2 of
myocardial action potential.
The indirect action of digitalis glycosides is made through the parasympathetic autonomic
nervous system. This action effect the electrical activity of the purkinje, atrial and ventricular
fibers, which in turn influence the excitability and automaticity of the cardiac tissues.
Digitalis glycosides have the following effects on the myocardium that influence
excitability:
Decreased action potential duration (ADP)
Decreased duration of phase 2
Increased slope of phase 4
Increased refractory period of atrial and ventricular myocardium
Less negative resting membrane potential (RMP); decreased amplitude of the action
potential; decreased rate of conduction.
At toxic levels of digitalis glycosides, the RMP becomes markedly reduced (becomes much
less negative than usual) causing a further decrease in amplitude of action potential.
Remember that relationship between RMP and the rate of rise of action potential is that as
RMP decreases the rate of rise of action potential is also reduced (see above). This then leads
to reduced conduction velocity. Since under this condition the difference between RMP and
threshold potential is narrow, the myocardial fibers then become more excitable and take on
the properly of slow-response fiber.
Automaticity is the term to describe the behavior by which cardiac tissues spontaneously
generate action potential. The extracellular concentration of potassium ions influences the
action of cardiac glycosides on automaticity.
This action potential then leads to an extra heartbeat called an “ectopic beat” (beat outside of
the normal cardiac rhythm).
Most of the clinically important effect of a digitalis glycoside on the rate of formation of
impulses by the SA and AV nodes are due to its indirect action through the vagal
(parasympathetic) influences on the heart.
The indirect vagal (parasympathetic) action specifically on the atria and ventricles is
believed to mediate the early effects of digitalis on conduction and refractoriness. This action
of digitalis on the atria and the ventricles also leads to a decrease in contractility (a negative
inotropic effect). This is indirect contrast to the desired effect of digitalis on failing heart.
Yes, this in true but the direct effect on the sodium pump is far greater that the indirect effect.
The direct positive inotropic effect, therefore, overshadows the indirect negative inotropic
effects on atria and the ventricles. It is clear then that the observed positive inotropic effect of
digitalis in a failing heart is the net effect of its direct and indirect actions.
Aside from its effect on the heart, digitalis glycosides also affect other organs. Their effect on
other organs are considered side effects, which may or may not be of benefit to the patient.
P a g e | 64
Digitoxin is more readily absorbed from the gut than digoxin. The rate of absorption
may be influenced by the specific pharmaceutical preparation of the drug. Example,
as digoxin tablets have onset of action of 15-30 min, peak in 6-8 hours. An alcoholic
solution or elixir of digoxin has a similar onset of action as tablet but peaks in 1-2
hours Elixir generally has poor shelf.
Distribution
P a g e | 65
In the dog, digitoxin is bound to plasma protein to about 5 times greater than digoxin.
There is a greater likelihood of drugs interaction with digitoxin than with digoxin.
Digitalis glycosides cross-placental barrier but do not usually cause ill effects on the
fetus.
Elimination
Elimination of digitoxin and digoxin in dog is quite different from its elimination in
human beings. In human, digoxin is eliminated by direct renal excretion while
digitoxin is by hepatic metabolism. In dogs, both digoxin and digitoxin are eliminated
by both direct real excretion and hepatic metabolism. Digoxin and digitoxin have
similar half-lives in the dog, but have different half-lives in humans.
Clinical Uses
Pre-surgery use
Digitalis may be used prophylactically in older animals to be operated on to pre-empt
possible heart failure under general anesthesia. This practice is controversial and is
not generally recommended.
Toxicity
Digitalis glycosides are one of the most dangerous drug groups in clinical practice,
having a relatively narrow safety margin. The dose for digitalization is about 40% of
the lethal dose. Cardiac arrhythmias are the most severe toxic effect especially
ventricular fibrillation. Hypokalemia is a common side-effect of digitalis. Important in
the pathogenesis of digitalis-induced cardiac arrhythmias is the depletion K⁺ from
cardiac cells. K⁺ depletion is exacerbated by factors that deplete body K⁺ such as
diuretics. Corticosteroids and prolonged diarrhea. Other factors increasing sensitivity
to digitalis are acidosis, hypoxia, hypothyroidism and catecholamines. Stopping
medication usually reverse usually reverses toxicity. Oral or parenteral K⁺ and an
appropriate antiarrhythmic agent may be needed in some cases.
Dosage
There is no fixed dose for the administration of cardiac glycosides (Digitalis). The
treatment of patients for congestive heart failure should be individualized and the
P a g e | 66
recommended doses should be regarded as guide only. The dose may be decreased or
increased depending upon the clinical response of the patient. In view of the more
recent pharmacological studies the older techniques of rapid digitalization (loading)
now appear unwarranted. The therapeutic serum levels of digoxin may be attained in
the dog given the maintenance dose only as early as 8 hours after the second dose.
Hospitalization and rapid digitalization becomes unnecessary. To hasten digitalization
in more severe cases, however, a loading dose may be given. Digitalization is usually
accomplished in 7 to 14 days.
MAINTAINANCE
ROUTE DRUG LOADING DOSE (mg/kg) DOSE
(mg/kg)
0.11-0.22; ⅟4 of total dose given
Oral Digoxin 0.022 daily
q12h
0.44; ⅟4 of the total dose given
Oral Digitoxin 0.11 daily
q12h
0.022-o.o44 given in 3 divided
IV Digoxin Oral digoxin 0.011 q12h
doses over 24h
0.022-0.033 in divided doses
IV Ouabain Oral digoxin 0.01 q12h
over 24 h
The dose of digitalis glycosides may be based on body surface area (BSA) expressed in units
of m, rather than on body weight. To convert kg to BSA, use the formula: BSA (m) – kg
(0.112). The doses of digoxin for dogs based on BSA are (a) for loading dose: 0.66 mg/m and
for maintenance: 0.22 mg/m.
Horse
for supraventricular tachycardia: DIGITOXIN- oral 0.033-0.066 mg/kg;
DIGOXIN- oral 0.66 mg/kg IM, IV 0.022 mg/kg; OUABAIN- IM, IV 0.0132-
0.022 mg/kg
Cattle
For supraventricular tachycardia: DIGITOXIN- IM 0.031 mg/kg; DIGOXIN-
IM, IV 0.0132-0.022 mg/kg
ACE Inhibitors
One of the complications of congestive heart failure is hypertension, which may be treated
with hypotensive agents.
1. Captopril
The first ACE inhibitor to be developed but enalapril is the most commonly
used in animals. Other ACE inhibitor now available include lisinopril,
ramipril, perindopril and trandolapril (Note: captopril and lisinopril unlike
the others are not prodrugs and are therefore directly acting.)
2. Enalapril
A prodrug that is converted to the active enalapril, which competes with
Angiotensin I for ACE. Enalapril reduces the workload of the heart as a result
of both arterial dilation and reduced fluid retention. Enalapril is well absorbed
in the gut but has a slow onset of action (4-6h) with duration of 12-14h that
allows 2 times daily dosing. The dose must be reduced accordingly in case of
congestive heart failure and renal failure. Enalapril is indicated for treatment
of heart failure in dogs, and hypertension in dogs but not in cats. It is
contraindicated in dogs with known ACE inhibitor hypersensitivity.
Dose of Enalapril
Antiarrhythmic Drugs
Arrhythmia is any deviation from the normal heart rate and rhythm. Not all arrhythmias are
pathologic. Some may be present but may not require treatment. Many are fatal. Arrhythmias
occur more frequently than you think, especially in small animals (dog and cats). If an
arrhythmia is of a type that need to be treated, then knowledge of important prerequisites to
effective therapy is in order. These include: accurate diagnosis (more of this will be learned
in Medicine courses), and good knowledge of antiarrhythmic drugs (our concern in
Pharmacology).
P a g e | 68
The impulse must be sinus origin. That is, the sino-atrial node must be the pacemaker.
The frequency of sinus impulse must be within an optimal range. For example, 80-
120 beats/minute in dogs.
The rate of contraction must occur at regular interval. However, dogs normally show
group of regularly spaced impulses separated from similar group by longer interval.
This separation occurs regularly in association with breathing. This is termed as
“regularly irregular: rate.
Every sinus impulse must be transmitted through the AV node
There must be a normal intraventricular transmission.
Any deviation from this normality constitutes an arrhythmia.
Diagnosis of Arrhythmia
Screening for arrhythmia may be done by auscultation (listening to body sounds with
stethoscope).
Listen for abnormal heart sounds and correlate their occurrence with respiration. Each
heartbeat should cause a femoral pulse. Look for any pulse deficit (heartbeat without
corresponding femoral pulse). A jugular pulse, when present, is abnormal.
Electrocardiography (ECG) is the most specific way to diagnose cardiac arrhythmias.
a. Form of arrhythmia with similar ECG profile may have different causes
b. Pharmacological actions of anti-arrhythmic drugs vary
c. Anti-arrhythmic drug may cause arrhythmia themselves, and
d. The effects of theses drug depend in part upon the underlying state of cardiac cells.
Causes of Arrhythmia
The term automaticity is used to describe the behavior by which cardiac fibers
spontaneously generate action potential. It is a feature common to all cardiac
conduction tissue (including the myocardium), and is believed to be related to
the slow inward movement of calcium ions. Abnormal impulse generation
maybe one of four types.
Abnormal Automaticity
Triggered Activity
Sick-sinus syndrome
Conduction
Refers to the progression movement of an action potential from on area of
myocardium to another. A unidirectional block is required for the re-entry mechanism
to occur.
Slow impulse
Unidirectional block
Re-entering impulse
Cardiac stretch and local tissues hypoxia may reduce the speed of impulse conduction
through segments of myocardium, resting in the re-excitation of recently depolarized, but no
P a g e | 70
longer refractory, tissue. This phenomenon, known as re-entry, is responsible for coupled
beats (called bigeminy) and AV nodal and ventricular tachycardia.
Class I
Membrane Stabilization drugs Divided into three Subclasses
Lidocaine
Members of this class increase the outward K⁺ Phenytoin
current and bring about the following: Tocainide
Increased conduction velocity Mexiletin
Increased threshold potential Aprindin
Hyperpolarization of cells having reduced RMP’s
Subclass IB such as due to hypoxia and anemia
Decreased slope of phase 4
A major effect on abnormal automaticity
A minimal decrease in effective refractory period
and a minimal shortening of action potential
duration
Encanide
Membrane of this class have action and effects as Lorcainide
those in subclasses IA and IB but with very little Flecainide
Subclass IC
effect on refractoriness and on action potential Propafenone
duration
Class II
The antiarrhythmic effect of this group results from Propranolol
a selection ꞵ-adrenergic blocking action (can also Timolol
increase the outward K⁺ current and decrease the Alprenolol
inward Na⁺ current). Member of this class produce Pindolol
the following effects: Metotrolol
Decrease conduction velocity at very high doses
Competitive blockade of enhanced automaticity
resulting from adrenergic stimulation.
Increased the effective refractory period and the
P a g e | 71
1. QUINDINE SULFATE
A dextro-stereoisomer o quinine, an antimalarial drug
Direct action
Decreases maximal rate of phase 0
Decreases slope of phase 4
Decrease spontaneous depolarization of Purkinje fibers, but spares the
automaticity of the SA node (controls automaticity)
Prolongs the effective refractory period of atrial and ventricular myocardium
without affecting the normal pacemaker cells (controls atrial fibrillation)
Indirect action
Atropine-like vagolytic action
Contributes to the effectiveness of treating atrial tachyarrhythmias
May increase ventricular rate (especially when given IV)
Pharmacokinetics
P a g e | 72
Clinical use
For atrial fibrillation in dogs and horses: also for ventricular premature beats.
Adverse effects
Cardiac depression- AV block, windened QRS complex
Decreased cardiac output, hypotention
Extra-cardiac effect- gastrointestinal upset, CNS depression, anaphylactic-like
reaction has been reported in the horse
2. PROCAINAMIDE
Action
Has direct actions similar to quinidine but the indirect effects are not
pronounced
More effective in controlling ventricular arrhythmias
Pharmacokinetics
Absorbed rapidly from GI tract
Liver metabolites may be cardioactive; excreted rapidly in urine
Clinical Use
For ventricular ectopic beats or ventricular tachyarrhythmias.
Adverse Effects
Similar to those of quinidine, but with less GI effects.
Pharmacokinetics
Absorbed well when given orally, but cause severe GI disturbance
Given as bolus IV injection 2% solution for immediate effect (lasts for 10-20
min.), then by slow drip for more sustained effect
Biotransformed in the liver
4. PHENYTOIN (Diphenyldantoin)
Action
Similar to lidocaine; also used as inti-septic.
Pharmacokinetics
Absorbed slowly from GI tract
Rapid biotransformation in dogs
Clinical use
P a g e | 73
5. PROPRANOLOL
Action
Antiarrhythmic action due to adrenergic blocking effect (decreased
spontaneous firing in SA node)
Pharmacokinetics
Well absorbed orally, extensive first-pass effect (hepatic metabolism)
Clinical uses
for severe atrial fibrillation unresponsive to digitalis
sinus tachycardia associated with anesthesia
ventricular arrhythmias
hypertrophic cardiomyopathy
Adverse effects
Bradycardia, hypotension, bronchoconstriction; Dangerous to use in heart
congested heart failure.
DOSAGE
Lesson 05
Diuretic Agents
D iuretics are drugs that increase urine output, with enhanced excretion of
electrolytes and water. They are used to remove edema fluids of various causes.
Glomerular filtration
In a normal nephron there is a net force of about 25 mmHg that tends to push fluid out
of the glomerular capillaries into the glomerular capsule. About 99 percent of the
glomerular filtrate is normally reabsorbed across the renal tubular epithelium. It is
therefore a rather inefficient attempt to produce a diuretic effect by influencing the
glomerular filtration rate (GFR). Certain drugs, however, produce diuresis by virtue of
their action on glomerular filtration. Examples of such drugs are the xanthine
derivatives (theophylline and caffeine) and the cardiac glycosides (digoxin, digitoxin),
which produce diuresis by their influence on renal blood flow.
Tubular reabsorption
Tubular reabsorption of sodium is the most important process influencing the final
volume and concentration of urine. The proximal convoluted tubules are responsible
for 66 percent of the sodium glomerular filtrate reabsorbed by the kidney.
Sodium ion diffuses across the luminal membrane of the proximal convoluted tubular
epithelium. It is subsequently transported actively across the basolateral membrane by
the sodium pump into the interstitial fluid, from which it diffuses into the capillaries.
Chloride ion and water follow Na + to maintain ionic and osmotic equilibrium. Aside
from diffusion with chloride (quantitatively the most important), sodium reabsorption
is also facilitated by co-transport with uncharged substances or acidic anions, and by
counter-transport with hydrogen ions. Reabsorption of bicarbonate, glucose, amino
acids, and other organic solutes also occurs in the proximal convoluted tubules.
The cells of the descending loop of Henle are relatively impermeable to sodium,
chloride, and potassium. Water is reabsorbed in the thin portion of the descending
limb of the loop of Henle due to the osmotic forces generated in the hypertonic
medulla interstitium. The ion transports in the ascending loop of Henle are critical
for proper function of the countercurrent mechanism in the renal medulla. The
ascending loop tubule is not permeable to water, and the fluid in the lumen becomes
progressively diluted. The counter-transport between sodium and hydrogen ions does
not appear to occur in the loop of Henle, and little if any bicarbonate is processed.
In the distal convoluted tubules, reabsorption of Na+ also occurs but little or no
water is reabsorbed. The rate of sodium reabsorption at this site is less than in the
ascending limb. Na+ is reabsorbed in exchange for K + at the collecting duct. This
exchange transport is driven by aldosterone, a mineralocorticoid secreted by the
adrenal cortex. Here, there is a direct relationship between Na + reabsorption and the
secretion of H- and K+. Calcium is actively reabsorbed at this site under the influence
of the parathyroid hormone (PTH).
The wall of the collecting duct becomes permeable to water in the presence of
antidiuretic hormone (ADH or vasopressin, secreted by the posterior pituitary). When
P a g e | 76
Tubular secretion
Tubular secretion is an active process, which occurs mostly in the proximal tubule.
There exist separate secretory mechanisms for organic acids and organic bases.
Inhibition of these mechanisms may affect drug excretion, but is not important in the
action of diuretics.
Atrial natriuretic hormone (ANH), first described in 1984, is involved in the control
of ECF. It is synthesized from a prohormone and stored as a peptide in granules of in
atrial myocardial cells. Concentration increases above baseline when ECF expands,
blood pressure increases, or dietary salt intake increases. Renal blood flow and
glomerular filtration are subsequently increased. Sodium excretion also increases,
presumably by a direct tubular action of ANH. Peripheral vasodilation can result in
decreased blood pressure. Effects occur rapidly but are not sustained, suggesting that
ANH is a mechanism that can store equilibrium rapidly.
Diuretics are used to remove edema fluids caused by various disorders. Three
strategies exist for movement of inappropriate fluid accumulation (edema).
Classification of Diuretics
P a g e | 77
Diuretics are classified by their mechanisms of action and include loop diuretics, carbonic
anhydrase inhibitors, thiazides, osmotic diuretics, and potassium-sparing diuretics. Rational
selection of diuretics is based on the appreciation of their mechanisms of action. All diuretics,
except the osmotic diuretics and carbonic anhydrase inhibitors, target sodium or chloride
reabsorption by tubular cells, effectively preventing the normal ion gradient by renal tubular
cells. Diuretics that increase net urinary excretion of sodium chloride or sodium bicarbonate
are referred to as natriuretic.
OSMOTIC DIURETICS
Mannitol
Mannitol is the most useful osmotic diuretic. Following intravenous injection,
mannitol is filtered at the glomerulus. Since it is not reabsorbed by the renal
tubules, it remains in the renal tubular fluid and osmotically attracts large
volumes of water, which is flushed through the kidney.
Xanthine derivatives have other actions not directly related to diuresis. CNS
stimulation, cardiac stimulation, and bronchodilation.
Digitalis glycosides exert their diuretic effect by improving the blood circulation in
patients with failing heart. They do not cause diuresis in patients without congestive
heart failure.
Carbonic anhydrase inhibitors reduce the rate of production of aqueous humor in the
eye and are useful in treating glaucoma. They also have some anti – epileptic action
but are not used for this purpose. The major clinical use of carbonic anhydrase
inhibitors is to relieve increased intra – ocular pressure in glaucoma. For certain
edematous state, they are used as an adjunct to a more powerful diuretic.
Acetazolamide has also been used to remove udder edema in dairy cows. When
injected, it causes minor pain at injection site.
THIZIDES
Examples
Chlorthiazide Hydrochlorthiazide
Hydroflumethiazide Bendroflumethiazide
Benzithiazide Trichlomethaizide
Methychlorthiazide Metholazone
Quinethazone Chlorthalidone
Polythiazide Cyclothiazide
Thiazides are believed to inhibit Na+ and Cl- reabsorption in the early
distal tubule of the nephron. The mechanism of this inhibition is not
fully known but his action may be related to inhibition of ATPase
activity. They produce pronounced diuresis which the carbonic
anhydrase inhibitors is independent of the acid – base status. Thiazides
have a tendency to develop hypokalemia due to large loss of
potassium. Hyperglycemia (presumably due to inhibition of insulin
secretion), hypomagnesemia, and hypercalcemia are also observed
with thiazides.
LOOP DIURETICS
Examples:
Furosemide Ethaycrynic acid
Bumetanide Muzolimine
Loop diuretics inhibit Na+ and Cl- reabsorption throughout the nephron but especially
in the ascending loop of Henle. They produce extremely potent and sustained diuresis.
The diuretic effect is seen within 30 min. after oral administration and peaks in 1 – 2
h; within 10 – 15 min. after IM or IV, peaks in 30 – 45 min. the effect may last for 6 –
8 h. About 30% - 40 % of Na+ filtered is lost in urine. Large Cl- loss may lead to
P a g e | 79
Loop diuretics are used to treat most edematous conditions but may have adverse
effects in the form of volume and electrolyte disturbances and ototoxicity (deafness).
Because of the latter, these drugs should not be used with known atotoxic agents such
as the aminoglycoside antibacterial.
Spironolactone
Triamterene
Acts on the collecting duct but its action is not associated with aldosterone. It
is a weak diuretic when used alone but when combined with a thiazide, the
effect is more effective than that caused by either drug.
Amiloride
DOSAGES
Lesson 06
Drugs Affecting
Blood Coagulation
T here are two general classes of drugs affecting blood clotting – hemostatic and
anti – hemostatic. Hemostatics are drugs used to prevent or attenuate bleeding
which can be given topically or systematically. Anti – hemostatics are used to
limit the formation of thrombi and are categorized into:
Anti – coagulants drugs that prevent blood coagulation
Fibrinolytics (thrombolytics) are drugs that increase the activity of plasmin
(fibrinolysin), the endogenous compound that is responsible for dissolving clots; and
Anti – thrombotics (anti – platelet agents) are drugs that control platelet activity
Hemostasis is the spontaneous arrest of bleeding from damaged blood vessels. I’m sure you
have experienced bleeding from a minor cut from the skin. Granting that you are not a
hemophiliac (bleeder), you would notice that bleeding from a minor skin cut eventually stops
even though you did not do anything about it. Save for a mild inflammation, you’d be alright.
At first, the damaged blood vessels go into spasm (severe constriction) in an attempt to close
the cut ends blood vessels and thus reduce further blood loss. Then the platelets (also called
thrombocytes) stick to the exposed collagen of the blood vessels to form a plug. This platelet
plug is not very stable so that it needs to be reinforced with fibrin, forming blood clot.
Fibrin formation occurs in three stages:
1. Activation of factor X
P a g e | 82
a. Damage to blood vessels exposes the negatively charged collagen. If this may
sound familiar, it’s because I’ve mentioned this in a previous lesson. You
recall that Factor XII or Hageman Factor is activated to XIIa upon exposure of
the exposed collagen and other surface factors. (Note: “a” means activated). It
is also activated by the action of Kallikrein (ka) in the presence of high
molecular weight kininogen (HMW – K) or Fitzgerald Factor.
2. Formation of Thrombin
Factor Xa with factor V (Proaccelerin), Ca2+ and phospholipid, activates Factor II
(Prothrombin) to IIa (Thrombin).
3. Formation of Thrombin
c. XIIIa then promotes cross – linking of the fibrin monomers into insoluble
fibrin clot (Factor I’’)
What has just been described is the intrinsic system of blood coagulation which has
all the necessary factors contained within the circulation. Another system, the
extrinsic system, operates when blood is exposed to tissue factors outside of the
circulation during tissue damage.
The extrinsic system is faster in forming blood clot than the intrinsic system. During
tissue injury, when blood escapes from the blood vessels faster blood clotting occurs.
The formation of Xa takes several minutes in the intrinsic system while it takes much
less time in the extrinsic system.
a. (Proconvertin) is activated to VIIa by Xia and ka, which are all from the
intrinsic system
b. VIIa, in the presence of Factor III (Tissue Thromboplastin) and Ca2+,
activates X to Xa
c. The next steps are identical to that of the intrinsic system
P a g e | 83
In our normal day – to – day, blood vessels are subjected to some forms of stress such
as from increased blood flow and pressure, pressure from sitting too long on a hard
surface, ischemia due to transient lack of blood flow, etc. these factors may cause
some slight damage to vascular endothelium (but without escape of blood), which
triggers the intrinsic system leading to coagulation. The resultant intravascular
clotting, an attempt to repair the damage, is very well regulated.
Thrombus (pl. thrombi) when formed can block blood flow to tissues. Small pieces
of the thrombus may chip off (now called embolus; pl. emboli), be carried away in
circulation to block tiny vessels in vital organs. This is really life – threatening. The
body has intrinsic mechanisms to regulate blood clotting and prevent thrombus
formation.
Urikinase
Prepared from culture of human renal cells
These three enzymes are used in the treatment of wounds that do not respond to:
a. Antibacterial therapy
b. Burns
c. Ulcers,
d. Chronic eczemas
e. Ear hematomas,
f. Otitis externa,
g. Osteomyelitis,
h. Chronic sinusitis
i. And other chronic lesions.
Cost and lack of refined doses and selectivity have limited the use of
thrombolytic drugs for treatment of thromboembolism in dogs and cats (and
other animals).
Normal blood
Fresh serum is indicated for emergency treatment in cases of acute –
hemorrhagic syndrome due to deficiency of clotting factors and in
thrombocytopenia.
Vitamin K
Vitamin K is required (as a cofactor) for the ϓ-carboxylation of glutamic acid
residues in Factors II, VII, IX and X. without this carboxylation, these
coagulation factors are not functional. The absorption of vitamin K may be
inhibited by intestinal disorders but can be enhanced by fat. Its utilization in
P a g e | 85
the synthesis of the clotting factors is impaired in liver disease. The coumarin
anti – coagulants including bishydroxy – coumarin and warfarin interfere
with the role of vitamin K in the synthesis of active clotting factors II, VII, IX,
and X.
Forms of Vitamin K
1) Vitamin K1 (Phyloquinone or phylotonadione)
If plant origin; the only natural vitamin K available for
therapeutics use; can be given orally or parenterally. It
is more effective than other analogues. Anaphylactic
reactions have been observed with intravenous
administration, perhaps due to the carrier substance
such as polysorbate 80, a known histamine releaser.
2) Vitamin K2 (Menaquinone)
A natural metabolite of gut microorganisms.
3) Vitamin K3 (Menadione)
The synthetic form and must be metabolized to active
form. It is usually absorbed too slow to be used
effectively in acute conditions, but it can be used for
chronic therapy once the acute crisis has been resolved.
Protamine sulfate
A low molecular weight protein found in certain fish. It forms stable salt with
acidic heparin, and is used only to antagonize heparin-induced hemorrhages.
When used otherwise, protamine has anticoagulant properties interfering with
the reaction of thrombin and fibrinogen.
Desmopressin
A synthetic analogue of vasopressin used to treat central diabetes insipidus,
also transiently elevates serum concentration of von-Willebrand factor (VWF).
[Also called Factor VIII-related antigen, VWF circulates as a complex with
Factor VIII, and mediates platelet adhesion to subendothelial surfaces – the
first step in clot formation]. Desmopressin causes the release of preformed
VWF from endothelial cells and macrophages. However, repetitive
administration results in depletion of storage pools and loss of procoagulant
effect. The peak response to desmopressin is 1-2 hours after treatment.
LOCAL HEMOSTATICS
2. Artificial matrices
3. Astringents, and
4. Local vasoconstrictors
Examples are:
o Thromboplastin
o Thrombin, and
o Fibrinogen.
Astringents
Act locally by precipitating proteins. These agents do not penetrate tissues and
thus are restricted to surface cells. They can be damaging to surrounding
tissues.
Examples are:
o Ferric sulfate
o Silver nitrate and
o Combinations that include tannic acid.
Thromboplastin USP
Used as a local hemostatic in surgery; available as spray or direct application
in sponge.
Fibrin foam
Applied directly to hemorrhagic area
Absorbs blood several times its weight and is applied on bleeding area
following closure of surgical wound. It liquefies in 3 to 5 days and is absorbed
in 4 to 6 weeks. It is used for capillary of venous bleeding.
ANTI - COAGULANTS
Vitamin K antagonists
Are of toxicological importance in veterinary medicine. Some are used
as systemic anti - coagulants in human patients. Warfarin and
bishydroxycoumarin are absorbed in the gut and are 90% protein –
bound. They can be stored in the liver and inhibit the hepatic
utilization of vitamin k. Other preparations include: 4 –
hydroxycoumarin, Phenprocoumon, Acenocoumarol, Dicoumarol,
Indan – 1, 3 – dione, and Anisindione.
Sodium oxalate
As 20% solution: use 0.01mL for each mL of
blood to be collected (2 mg/mL)
P a g e | 88
Edetate disodium
(Disodium *EDTA)
Use 1 mg/mL of blood
*Ethylendiamine
tetra – acetic aid
Note that the toxicity of citrated blood varies with the rate of injection
and the total dose
FIBRINOLYTICS
Fibrinolytic agents increase the activity of plasmin (fibrinolysin), the endogenous
compound that is responsible for dissolving clots.
Urikinase
Prepared from culture of human renal cells
These three enzymes are used in the treatment of wounds that do not respond
to:
a. Antibacterial therapy
b. Burns
c. Ulcers,
d. Chronic eczemas
e. Ear hematomas,
P a g e | 89
f. Otitis externa,
g. Osteomyelitis,
h. Chronic sinusitis
i. And other chronic lesions.
ANTITHROMBOTHICS
These are drugs that control platelet activity. Aspirin and related compounds cause
irreversible and thus long – lasting negative effect on platelet activity which is
clinically manifested as prolonged bleeding time. The antiplatelet effect of aspirin can
be separated from its other actions by administration of a low dose.
DOSAGES
As an antithrombotic
Lesson 07
P a g e | 91
Drugs acting on
the Digestive
System
The GIT responds to drug action even without the drug entering the circulation. The entero –
hepatic re – cycling of absorbed drugs can be exploited in the design of therapeutic regimen.
Drugs may be acted upon by gut microbial flora resulting in activation or activation of certain
drugs. Some drugs (prodrugs) may require microbial action to attain full therapeutic activity.
Oral administration makes drug – to – drug and drug – to – food interactions possible and can
influence the absorption and effectiveness of various therapeutic agents. The gut is also one
of the richest endocrine organs of the body. So, any dysfunction of the GIT somehow
influences the effectiveness of orally administered drug.
Some symptoms may be eliminated by withdrawal of the offending agents and may
not require further pharmacological treatment. Treatment is oftentimes not necessary.
At times, gastrointestinal therapeutics may require interruption of normal
P a g e | 92
Cytoprotectants Substances that form thin layer over skin or mucous membrane in
order to prevent contact with possible irritants
Cathartics Substances that hastens the rate of passage of material through the
GIT and promote defecation
EMETICS
Emetics are indicated whenever there is a need to evacuate the stomach of its contents
such as:
During poisoning with non – corrosive materials to reduce further absorption of the
poison. Induction of emesis should not be attempted when corrosive agents had been
ingested. In this case, an adsorbent and a cathartic may be tried
Emptying the stomach prior to induction of anesthesia. Emesis prior to induction of
anesthesia does not always ensure complete emptying of the stomach. Fasting the
patient for at least 12 hours is best whenever possible.
DOSAGE:
3% Hydrogen peroxide Dog/Cat: 5 mL oral
May induce vomiting in 5 – 10 minutes
DOSAGE:
Dog/Cat: 1 tsp.
Sodium chloride
Placed at the back of the tongue
May cause vomiting
P a g e | 94
Copper sulfate
Zinc sulfate
Freshly ground mustard seed
ANTI – EMETICS
DOSAGE:
Anti - histamines
Metoclopramide
0.1 – 0.3 mg/kg, oral, IM
Dog
0.01– 0.02 mg/kg IV drip for severe emesis
ANTACIDS
Antacids are indicated for hyperchlorhydia and peptic ulcers and for grain
engorgement toxemia in large animals. They may act systematically or locally by
neutralizing acid. Drugs that specifically inhibit gastric acid secretion may also be
considered antacids.
Dosage
Cimetidine Dog/Cat 5 mg/kg oral q6 – 8h
Ranitidine Dog/Cat 2 mg/kg oral q8 – 12h
Dog 5 mg/kg oral, IM, SC once daily
Famotidine
Horse 0.23 mg/kg IV q8h as adjunct to ulcer treatment
ADSORBENTS
These drugs, which are not absorbed from the gastrointestinal tract and either line the
mucosa or adsorb toxic compounds, are often incorporated into anti – diarrheal
mixture, or are used alone.
Usually available in combination with pectin (a protectant)
Generally safe but long term use may cause gastric carcinoma
Kaolin The use in neonatal diarrhea is questionable because there has
(hydrated been indication that it is unable to adsorb bacterial endotoxin
aluminum silicate) causing diarrhea in young animals
It inhibits the absorption of certain drugs such as tetracycline
Do not administer kaolin together with other drugs
Local antacids May be considered as adsorbent because they adsorb gastric acid
CYTOPROTECTANTS
P a g e | 98
These drugs form a coating of the gastrointestinal mucosa that prevents irritation or
erosion caused by potentially harmful substances.
ASTRINGENTS
CARMINITIVES
Include mostly spices which prevent gastrointestinal gas formation or promote gas
expulsion from the gut.
Examples are:
Simethicon
Poloxalene
Turpentine
Camphor
Peppermint, and
Capsicum
P a g e | 99
CATHARTICS
The term “cathartic” is used synonymously with the term “laxative”. Strictly, a
laxative is only a mild cathartic.
Stimulant cathartics
These are thought to stimulate mucosal lining of the GIT and thereby initiate a
myenteric reflex that would enhance intestinal transport
Types of Stimulant
Examples Properties
Cathartic
Available in combination with other
substances such as carbamylcholine
(Carbachol)
Emodin irritant
Of vegetable origin, derived from anthacene
cathartic
but its main ingredient is emodin which is
present in a glycoside form
Emodin acts on the
The onset of action is slow (4 – 8 hours
large intestine
even up to 24 hours)
presumably by Cascara sagrada
Recommended for use in cattle, horses, pigs,
irritating the mucosa
dogs, and cats
Cassia Rhamnus,
Other sources of emodin
Aloe, and Senua
A synthetic analogue of emodin
A slow – acting cathartic (onset is 10 – 12
hours)
Some absorption may occur and discolor
urine reddish violet (alkaline) or yellowish
brown (acidic)
With prolonged use, the myenteric plexus
actually degenerates resulting in loss of
Danthron intestinal motility
Danthron is commonly recommended for
horses
DOSAGE of Danthron
Horse 14 – 40 grams
Cattle 0 – 45 grams
P a g e | 101
Dosage
Foal/Calf/Pig 30-180 mL oral
CASTOR OIL
Dog/Cat 4-30mL oral
LINSEED OIL Horse 0.5-1.0 L oral
Bulk Cathartics
Bulk cathartics have action milder than that of the stimulant cathartics. They
increase the fluidity of intestinal content to facilitate defecation. They also
increase the bulk of intestinal content, which then distend the bowel wall and
bring about reflex peristalsis. There are two types of bulk cathartics: Saline
cathartics and gel bulk cathartics.
Ionic compounds with one or more non-absorbable or
slowly absorbable ions
Magnesium These ions make the intestinal fluid hyperosmotic so
sulfate that fluid is drawn into the gut resulting in catharsis.
SALINE
(Epsom’s salt) 500 grams of these salts retain 12 to 15 liters of water
BULK
in the gut within 1-4 hours.
CATHARTIC
Sodium sulfate With Epsom’s salt the patient may get appreciable
S
(Glauber’s absorption of magnesium that may result in CNS
salt) depression and neuromuscular blockage.
In case of poisoning with Epsom’s salt, give patient
activated charcoal and a stimulant cathartic.
DOSAGES
MAGNESIUM SULFATE Cow 0.24-0.48 kilogram
as purgative Sheep 60-120 grams
Cow 60-120 grams
MAGNESIUM SULFATE Sheep 7.5-15 grams
as laxative Horse 30-60 grams
Pig 15-30 grams
P a g e | 103
Lubricant Cathartics
These cathartics lubricate intestinal wall or intestinal content. Two types of
lubricant cathartics: emollients and fecal softeners.
DOSAGES
Large animals 0.5-2 liters
MINERAL OIL
Small animals 2-50 milliliters
Cattle/Horse 5-10 grams/animal
DOCOSATE SALTS Dog 15-120 mg/animal
Cat 15-30 mg/animal
P a g e | 104
MOTILITY MODIFIER
Inhibitors of motility
These are antispasmodic.
There are two subgroups of motility inhibitors:
1) Anticholinergics and
2) Opiates
Anticholinergics include:
Atropine
Tincture of belladonna
Propantheline
Isopropramide
Methscopolamine
Opiates inhibit the propulsive gut movement but enhance anal sphincter tone
and segmental gut movement. These effects increase the transit time of
ingesta. They also reduce intestinal fluid secretion.
Examples are
Diphenoxylate
Loperamide and
Codeine
P a g e | 105
Dosage of Opiates
DIPHENOXYLATE Dog/Cat 0.1mg/kg q8h
LOPERAMIDE Dog 0.08 mg/kg q8h
CODIENE Dog/Cat 0.25-0.5 mg/kg q6h
These agents used in the treatment of secretory diarrhea, fall under four
categories:
NOTES:
Lesson 08
Fluid Therapy
A Animals do suffer from varying degrees of body fluid loss (dehydration) and
electrolyte, acid-base and nutritional imbalances under various clinical
conditions. Dehydration and electrolyte imbalance can be corrected with fluid
therapy. The treatment of nutritional imbalance of an anorexic patient is not
generally required if the duration of therapy is less than five days.
The total body water is about 60% of body weight in adult; about 80% in the young.
The extracellular fluid (ECF) comprises 30% of the body weight: 5% is plasma; 25%
interstitial fluid. The intracellular fluid (ICF) is also 30% of body weight.
P a g e | 107
PO42- 113 2
Organic acids 0 5
Rate of IV Fluid Administration
Generally the rate of intravenous fluid infusion should not exceed 100 mL/kg/hr for
dogs and cats, but may be faster in large animals. The basis for this limit is that faster
infusion rate that recommended may unduly overload the physiologically “fast-
equilibrating” ECF compartment. When this happens, the patient shows signs of
“overhydration” while still suffering from the effects of fluid deficit. Too fast a rate
administration may result in one or more of the ff. signs: tachycardia, increased urine
formation, pulmonary edema, dyspnea and coughing. Must be reduced or
appropriately adjusted in cases of cardiac and renal failures, shock and during
anesthesia.
Osmolality
Number of particles in solution
Tonicity
Compatibility between ECF and ICF
The maintenance of the body fluids depends upon the appropriate regulation of input
and output via the gut, skin, kidneys and lungs, which is influenced by
neuroendocrine mechanisms. These mechanisms involve vasopressin (antidiuretic
hormone, ADH), renin-angiotensin, mineralocorticoid (aldosterone), and vasomotor
mechanisms. If the fluid output exceeds input, dehydration results.
TYPES OF DEHYDRATION
1. Hypertonic Dehydration Results from the Joss of pure water or loss hypotonic fluid
2. Hypotonic Dehydration Results from the loss of hypertonic fluid
3. Isotonic Dehydration Results from the loss of isotonic fluid
Acid-Base Balance
Simple laboratory tests such as PCV, total plasma proteins, and urine
specific gravity (normal = 1.048) may also be useful.
The replacement volume (= deficit) in liters is equal the body weight multiplied by
the estimated percent dehydration. For estimate of % dehydration; see above the
maintenance volume is approximately 40-60 ml/kg/day. The replacement of
contemporary losses from vomiting, diarrhea, polyuria, large wounds, panting fever,
P a g e | 110
or blood loss estimated on case-to-case basis. The sum of these volumes is the
estimated total volume to be infused within a 24-h period. The subsequent fluid doses
are usually the sum of the maintenance and contemporary loss only.
One of several types may be used in most therapeutic needs. Attempt must be made as
much as possible to replace with fluid similar to what has been lost.
Routes of Administration
The intravenous route is usually employed for replacing sudden and extensive losses.
In small-sized animals, the intraperitoneal route may be used for quick absorption. For
less critical cases, or for maintenance, fluids may be administered subcutaneously
(SC). Subcutaneous compartment, however, has limited volume, and absorption is
greatly limited especially when peripheral vasoconstriction is present. Avoid this
route with D5W because equilibration of ECF with a pool of electrolyte-free
solution may aggravate electrolyte imbalance. Since the fluid does not contain any
electrolyte, intracellular and intravascular electrolytes may be drawn into the
subcutaneously infused D5W. The oral route is ideal for hypertonic, high caloric
density fluids.
P a g e | 111
Rate of Administration
Acid-Base Imbalance
Without laboratory tests, therapy may result in a variety of acid-base, osmolality, and
electrolyte disorders.
The bicarbonate deficit is estimated as the difference between 25 mEq/L and the
actual measured plasma bicarbonate to be administered is equal to the product of the
body weight and the bicarbonate deficit (b.w x bicarbonate deficit). If metabolic
acidosis is obvious of imminent as in cardiopulmonary arrest, a safe approach is to
administer 1 mEq/k IV. Therapeutic attempts to correct acid-base abnormality other
than metabolism acidosis should be directed toward the primary cause.
P a g e | 112
Lesson 09
Introduction to
Chemotherapy
species of microorganisms (bacteria, fungi, yeast, etc.) which inhibits the growth of another
microorganisms. Most antibacterial and a few other anti – parasitic drugs are antibiotic by
definition. Today, the term “antibiotics” include not only those that are naturally produced
but also those that are partly or fully synthesized in pharmaceutical laboratories.
DRUG
1 4
2 3
6
PATHOGEN HOST
Antibacterial Agents
Toxic effects
Adverse reaction with other drugs or with nutrients
Allergy
Drug residues in animal tissues
Bacteriocidal or bacteriostatic effect
c. Sensitivity test takes time and may not be too important in emergency cases. It
is not always advantageous to wait for the result of bacterial culture and of
sensitivity test before starting an antibacterial therapy
4. The dose schedule must be followed for 3 to 5 days or longer to ensure elimination of
pathogens and to prevent relapse. If the patient shows improvement before the end of
the set course of therapy, continue the course of medication
5. The drug must be available at the site of infection in a concentration above the
minimum inhibitory concentration (MIC) of the drug for the bacterial species
involved. The MIC of an antibacterial for one species may not be the same for another
species
6. Supportive therapy and ancillary treatments, nutritional support and nursing care must
be instituted: fluid therapy, warmth rest, and proper nutrition
Transduction
This occurs when a bacteriophage (a virus) carries genetic materials for
antibiotic resistance in a newly infected bacterial cell. The DNA can be
incorporated in the new bacterium and antibiotic resistance passed on to its
progeny. Transduction is particularly important in the transfer of antibacterial
resistance among strains of Staphylococcus aureus where bacteriophages can
carry plasmids (extrachromosomal DNA) that code for multiple antibiotic
resistances.
Transformation
This involves the incorporation into bacteria of DNA that is contained in the
environment. The importance of this method is unknown.
Conjugation
This refers to the passage of resistance genes from cell to cell by direct contact
through sex pilus or bridge. This can occur not only within species but also
between species of bacteria (i.e. from non – pathogenic to pathogenic
bacteria). The DNA transferre is contained in plasmids and consists of two
parts: the first part codes for resistance and is termed R – factor. The second
codes for the transfer apparatus and is termed resistance transfer factor
(RTF). The R – factor often codes for resistance to multiple antibiotics. The
efficiency of this process is low but selective pressure applied through the use
of antibiotics has led to the slow development of plasmids for multiple drug
resistance especially among enteric organisms. The multiple – drug resistant
Enterobacteriaceae are a serious problem creating a constant demand for new
antibiotics.
Examples: Polymyxins
Generally bacteriocidal
Disruptors of cell
Their action is not dependent on the rate of active
membrane
bacterial growth or multiplication and therefore, may
be combined with bacteriostatic agents
Antibiotic targets
Cell
membrane Folate
cofactor
Cell wall
Nucleic
acid Protein
P a g e | 120
Amoxicillin Bacitracin
Ampicillin
Penicillin
Ceftiofur
Cell wall
Colistin
Salinomycin
Bacterial cell
membrane
Furazolidone
Nitrofurazone Norfloxacin
P a g e | 121
Dimeindazole
Trimethoprim
Sulfonamide
Sulfonamides
Folate cofactors
Tylosin Chloramphenicol
Erythromycin Florfenicol
Tilmicosin Lincomycin
Spiramycin Spectinomycin
Kitasamycin Tiamulin
Josamycin Valnemulin
P a g e | 122
Lesson 10
Penicillins
T he penicillins belong to the beta – lactam groups of antibiotics together with the
cephalosporins (see Lesson !!). The Penicillins are derivatives of 6 –
aminopenicillanic acid. Developed in the 1930s, benzyl penicillin (penicillin G)
became available for veterinary use in the late 1940s. Penicillin is produced by
P a g e | 123
Penicillium molds and was the first antibiotic available for clinical use. Today, the penicillin
group of antibiotics represents a large group of compounds. Only the primary ones used in
veterinary medicine are covered in this lesson.
Chemistry
In the early days of penicillin use, when it was relatively impure, it was measured by
bioassay. A unit of penicillin was that amount which gave a standard zone of
inhibition on a standard culture medium. Penicillin G (benzyl penicillin) is still
formulated and dosed based on units of activity.
The dosage and antibacterial potency of the semi – synthetic penicillins are
expressed in terms of weight rather than in units (see below).
Mechanism of Action
The penicillins inhibit the third stage of bacterial cell wall synthesis. The 3 stages of
bacterial cell wall synthesis are:
The penicillins cause death and Lysis of bacterial cells, the mechanism of which
involves the role of penicillin – binding proteins (PBPs) and bacterial autolytic
enzymes. Classically, they will only kill growing and metabolically active bacterial
cells. They have reduced killing effects on “dormant” bacteria or bacteria whose
multiplication is inhibited by another antibacterial agent.
Pharmacokinetics
The rate of absorption of penicillins depends on the pharmaceutical formulation used.
They are weak acids with pKa of about 2.76 and tend to distribute into the
extracellular fluid, pleural fluid, pericardial fluid, and bile. Low concentrations are
attained in the prostate, brain, intraocular fluid, and phagocytic cells. Penicillin G
when given orally is degraded by gastric acid but phenoxymethyl penicillin (penicillin
P a g e | 124
V) is relatively acid – resistant. Ampicillin and amoxicillin are also acid – resistant
and are fairly rapidly absorbed. Oral amoxicillin has greater bioavailability than
ampicillin.
Distribution
The penicillins poorly penetrate certain body parts such as the eye, prostate, and
cerebrospinal fluid. However, inflammation enhances the penetrability of the
meninges and may be useful in the treatment of brain infections.
PENICILLINASE –
Their use is generally restricted to infections caused
REISTANT PENICILLINS
by Penicillinase – producing bacteria (e.g.
Staphylococcus)
Examples:
Less active against other Gram – positive bacteria than
Cloxacillin, Oxacillin,
Penicillin G
Dicloxacillin, Flucloxacillin,
Not active against Gram – negative bacteria
Methicillin, Quinacillin
P a g e | 125
Penicillinase
An enzyme produced by certain bacteria which specifically destroys penicillin
antibiotics. It is a form of a β – lactamase enzyme. However, there is no
evidence that some bacteria have shown or developed resistance to the
penicillinase – resistant penicillins.
Benzyl Penicillin G
Good
Aerobic all beta – hemolytic streptococci (S. agalactiae, S. canis, S.
zooepidemiicus, S. dysagalactiae, S. suis, S. uberis): Bacillus
anthracis, most corynebacteria (C. pseudotuberculosis, C. renale):
Erysepelothrix rhusopathiae, Listeria monocytogenes, anaerobic
Clostrdium sp., Fusobacterium sp. And some Bacteroides sp.
Variable
Staphylococcus aureus, S. intermedius
Moderate
P a g e | 126
Resistance
Enterobacteriaceae (except a few Proteus sp.), Bacteroides fragilis,
Bordetella sp., most Campylobacter sp., Nocardia
Aminobenzyl penicillins
Good
As for benzyl penicillin but also good vs. Borrelia sp., Leptospira sp.,
Moraxella sp.
Variable
Actinobacillus sp., E. coli., P. mirabilis, and Salmonella and
widespread resistance by Enterobacteriaceae (though they are
technically susceptible)
Moderate
As for benzyl penicillin and Campylobacter sp., R. equi, and
enterococci
Resistance
Bacteroides fragilis, B. bronchiseptica, Citrobacter sp., Enterobacter
sp., Klebsiella sp., other Proteus sp., P. aeruginosa, Serratia sp.,
Yersinia enterocolitica
Ureidopenicillins
Carboxypenicillins
The direct toxicity of penicillins is generally low. Allergic reactions ranging from
delayed hypersensitivity skin reaction to acute anaphylaxis may occur. Species most
susceptible to allergic reactions are hamsters and guinea pigs.
G. Nephrotoxicity has been reported with the use of methicillin in human but may
also occur in animals.
Superinfection is most likely to occur with broad – spectrum penicillins. Mild cases
results in diarrhea and overgrowth of toxin – producing bacteria can cause death.
Penicillins are contraindicated in herbivore such as guinea pigs, hamsters, rabbits, and
chinchillas for this reason.
Acid stable
can be given orally
Cloxacillin food interferes with absorption
expensive
use only as last resort
Procaine penicillin
25K IU/kg IM q12 – 24h
Ticarcillin
50 – 75 mg/kg IM, IV q6 – 8h
Lesson 11
Cephalosporins
Chemistry
The cephalosporins, like the penicillins, contain a β – lactam ring but the adjacent ring
is a 6 – membered instead of 5 – membered. The active nucleus of most
cephalosporins is 7 – aminocephalosporanic acid. Substitutions at 3 different places in
its structure modify its pharmacokinetics and antibacterial activity.
Mechanism of Action
P a g e | 130
Generation Properties
Third Generation
Introduced in the early 1980s
Primarily with Gram – negative spectrum
They have greatly enhanced resistance t Gram – negative β –
lactamase
Increased activity against Gram – negative bacteria including
strains resistant to the first and second generations
They have some activity against isolates of Pseudomonas (but
activity varies among compounds)
P a g e | 131
Some second and third generation cephalosporins have peculiarities in their spectrum
which are different from others in the group. While these peculiarities can be
important in human medicine, they are of little significance in veterinary medicine
because very few second and third generation cephalosporins are used in animals.
Pharmacokinetics
Only the first and second generations are adequately absorbed when administered
orally. All the other cephalosporins must be administered parenterally. Some may be
given subcutaneously. They are all absorbed when given intramuscularly.
Cephalosporins are distributed similarly as the penicillins. They distribute well to the
extracellular fluid reaching therapeutic levels in most tissues and fluids including the
pericardial fluid, bile, and infected bones. In the absence of acute inflammation,
cephalosporins, with a few exceptions, do not reach useful levels in the CSF.
Cefuroxime, Moxalactam, Cefotaxime, and Ceftizoxime penetrate into the CSF in
sufficient concentrations to treat meningitis.
Most cephalosporins are excreted unchanged in the urine by glomerular filtration and
tubular secretion. A few are reabsorbed significantly such that renal clearance is less
than glomerular filtration rate corrected for protein binding. A few (such as
cephalothin, cephapirin, and Cefotaxime) are metabolized by the liver through de –
acetylation. Cefoperazone and Moxalactam are excreted primarily in the bile.
Adverse Reactions
In general, cephalosporins are safe drugs with low incidence of adverse reactions.
Hypersensitivity reactions are most common which can be an immediate reaction
(anaphylaxis, bronchospasm or urticaria), or a more delayed reaction such as skin
rash. Cephalosporins may have variable cross – reactivity with penicillin
hypersensitivity. However, if the reaction to penicillin was mild, there is a low risk of
anaphylactic reaction to cephalosporins. If the reaction to penicillin was severe, be
very cautious in administering this drug.
P a g e | 132
Individual Drugs
Examples:
ORAL PARENTERAL
Cefadroxil Cephaloridine Cephadrine
Cephalexin Cephapirin Cephalonium
Cephaglycin Cephalothin Cephalexin
Cephradin Cefazoline
Cephacetril
Examples:
ORAL PARENTERAL
Cefaclor Cefuroxime
Cefuroxime Ceforanide
Cefoxitin
Cefonicid
Cephoxazole
Cefamandole
Examples:
ORAL PARENTERAL
Ceftriaxone Cefotaxime
Ceftazidime Cefoperazone
Cefmenoxime Cefsulodine
Cefixime Moxalactam
Ceftiofur
Ceftizoxime
P a g e | 133
Examples:
Cefepime
Useful Dosages
Lesson 12
P a g e | 135
Bacitracin,
Vancomycin, and
Polymyxins
B ACITRACIN
Mechanism of Action
Bacitracin is a bacteriocidal that inhibits the second stage of bacterial cell wall
synthesis by inhibiting the phospholipid carriers that transfer mucopeptide building
blocks to the growing cell wall.
Bacitracin is not absorbed orally and may be used for gut sterilization before
gastrointestinal surgery. It is mainly used for topical application for superficial
infection of the skin and mucous membranes, and is often combined with polymyxin
B or neomycin for this purpose.
Although used to prevent and treat Clostridium perfringens enteritis in pigs and
poultry, it is more commonly employed for improvement of growth rate, feed
conversion efficiency, and egg production in poultry, and as a growth promotant in
cattle and pigs. However, the use of any antibiotic for this purpose in food animals is
strongly discouraged.
Resistance to bacitracin develops rarely in sensitive strains but most Gram – negative
organisms are inherently resistant.
Pharmacokinetics
Bacitracin is poorly absorbed orally through the skin, mucous membrane, or wound
but is absorbed considerably from IM sites. It is widely distributed if given
parenterally but is seldom if at all administered by parenteral routes because of
toxicity.
VANCOMYCIN
Vancomycin is bacteriocidal to most Gram – positive aerobic cocci and bacilli while
Gram – negative bacteria are resistant. Like bacitracin, resistance to Vancomycin is
rare but can occur.
Pharmacokinetics
Vancomycin is very toxic and highly irritating to tissues and can cause
thrombophlebitis, deafness, and nephrotoxicity. In humans, the adverse effects
include hypersensitive skin reaction, pain at intramuscular injection sites, histamine –
like reactions (when administered by rapid IV) and ototoxicity. There is not much
information on toxicity in domestic animals.
POLYMYXINS
The polymyxins are polypeptide antibiotics isolated from soil bacteria on 1947 to
1950. Although there are many polymyxins, only two are clinically useful:
Polymyxin B (from Bacillus polymyxa) and Polymyxin E also called Colistin (from
B. colistinus)
Chemistry
The polymyxins are simple, basic polypeptides with molecular weight of around
1,200 and act as cationic detergent and are readily water – soluble. 1 mg of polymyxin
P a g e | 138
Mechanism of Action
The polymyxins bind to phospholipids in cell membrane and alter the osmotic
pressure, selective permeability, and regulatory properties of cell membrane. This
action results in a bacteriocidal effect. Disruptors of cell membrane such as the
polymyxins, unlike the penicillins, may be used in combination with bacteriostatic
drugs since their antibacterial action does not require rapid multiplication of bacteria.
Polymyxins, specifically Colistin, have additional anti – endotoxin and antipyretic
effects. As basic (cationic) drugs, they may combine with endotoxin which is anionic.
Drug Interactions
The major clinical application in veterinary medicine includes (a) oral treatment of E.
coli and Salmonella, and (b) treatment of Pseudomonas infections such as otitis
externa and superficial lip infection. Part of the therapeutic mechanism in Gram –
negative infection may relate to their ability to bind bacterial endotoxins.
Pharmacokinetics
Because of their cationic nature, polymyxins are not absorbed orally and poorly
absorbed from the gut and through the skin and mucous membrane. Absorption is
rapid following IM and SC administration (parenteral polymyxins may cause toxic
effects). The polymyxins distribute within the extracellular fluid only but do not get to
the CSF. Tissue binding is important and bound drug is not active. The polymyxins
bind to kidney, liver, lung, heart, and skeletal muscle and are excreted unchanged by
the kidneys over several days.
Polymyxin B as sulfate is for both oral and parenteral use. Colistin as sulfate is for
oral administration and as sulfomethate for parenteral.
P a g e | 139
Nephrotoxicity
In the form of reduced tubular perfusion may progress to decrease urine
output, and this is the most important adverse reactions to polymyxins. This
adverse effect occurs at therapeutic doses but is reversible if the drug is
discontinued
Neurotoxicity
Occurs in the form of a dose – dependent curare – like paralysis which is
additive with other drugs such as aminoglycosides and muscle relaxants that
act on the neuromuscular junction. This paralysis may result in respiratory
arrest. Some local allergic reactions may occur but rarely.
Colistin
3 mg/kg q12h IM
Sulfomethate
Colistin
Pigs with colibacillosis: 5 – 10 mg/kg oral
Sulfate
Lesson 13
P a g e | 140
Sulfonamides and
Potentiated
Sulfonamides
S SULFONAMIDES
Sulfonamides (or sulfa drugs) were discovered in the early 1930s when
sulfanilamide was observed to be active portion of the azo dye, protonsil. Protonsil
had been shown to protect laboratory animals from bacterial infection. As many as 5,400
derivatives of sulfanilamide were subsequently synthesized and tested but only less than 30
were developed into therapeutically useful drugs. All sulfonamides are derivatives of
sulfanilamide.
While sulfonamides are antimicrobial agents, they are not antibiotics by the true
definition of the term since they are not derived from microorganisms. However, it is now
common to refer to synthetic antibacterial drugs as “antibiotics”.
Common Sulfonamides
Sulfapyridine
Phthalysulfathiazole
For gastrointestinal use Succinylsulfathiazole
(not well absorbed from the Sulfaguanidine
gut) Sulfaquinoxaline
Sulfazalizine
Silver sulfadiazine
(useful for treatment of burns and chronic
For topical use otitis)
Sulfacetamide
Mefenide
Chemistry
(Please refer to the chemical structure of sulfonamide; sorry not included here!)
The amino nitrogen (N4) must be in the para position on the benzene ring and must
be unsubstituted for antimicrobial activity. The amide nitrogen (N1) is the primary
site of substitution. Substitution alters the pharmacokinetics of sulfonamides.
Heterocyclic aromatic substitutions yield the most potent compounds.
Mechanism of Action
They prevent bacteria from utilizing para – aminobenzoic acid (PABA) in the
synthesis of tetrahydrofolic acid by competitively inhibiting the bacterial enzyme
responsible for the incorporation of PABA into dihyropteroic acid, the immediate
precursor of dihydrofolic acid. Dihydrofolic acid (DHF) is further reduced to
tetrahydrofolic acid (THF) in a reaction catalyzed by THF reductase. THF serves
as a cofactor in one – carbon metabolism involved in DNA and RNA synthesis.
Most bacteria cannot utilize exogenous sources of folic acid. This explains the
selective toxicity of sulfonamides for bacteria. However, blood, pus, and tissue
P a g e | 142
Originally (in 1940s) sulfonamides were active against Gram – positive and Gram –
negative bacteria but now many of these organisms are resistant. The correlation
between the in vitro sensitivity for sulfonamides is less than that seen with other
agents. Often, a higher concentration is needed in vivo that is predicted in vitro.
Widespread resistance has greatly limited the effectiveness of sulfonamides in treating
bacterial diseases in animals. Potentiated sulfonamides (trimethoprim – sulfonamide
combinations) have largely replaced sulfonamides as therapeutic agents used in
companion animals.
Pharmacokinetics
Absorption
Sulfonamides are rapidly absorbed with good bioavailability following oral
administration. There is good bioavailability even in ruminants. However, the
rate of absorption in the ruminants is slower than in monogastrics. Exceptions
are the gastrointestinal sulfonamides which are designed to remain in the GIT
(gut active). They are also absorbed following IM and SC administration.
P a g e | 143
Intravenous formulations, however, are very irritating and can cause tissue
damage because of alkaline pH. The drug should not be administered through
this route. Extravascular injection may result in tissue sloughing. Absorption
is variable following topical, intra – uterine, and intramammary (very
irritating) administration.
Distribution
Sulfonamides are distributed widely in the body tissues and fluids including
synovial, peritoneal, ocular, and pleural fluids. The distribution varies with
individual sulfonamides and is dependent upon the lipid solubility, pKa and
protein binding of each sulfonamide. The degree of protein binding varies
from <10% to >90%. There is both drug – and species – dependent variation
in percent protein binding.
Biotransformation
Sulfonamides are extensively metabolized in animals with differences between
animal species and individual drugs. Acetylation is highest in human, then in
decreasing order, ruminant, horse, and cat. Dogs are unable to acetylate
sulfonamides and are therefore susceptible to sulfonamide toxicity.
The metabolites of sulfonamides are much less active than the parent drug.
Many of the acetylated metabolites are less soluble than the parent
sulfonamides and contribute to renal damage caused by these compounds.
Exceptions are the acetylated metabolites of the sulfapyridines. Sulfadiazine,
sulfamethazine, and sulfamerazine which have similar solubility to the parent
compounds sulfamethoxazole and sulfathiazole are the commonly used
sulfonamides with less soluble acetylated metabolites.
Excretion
The kidney is the primary route of elimination for sulfonamides and their
metabolites. The unbound sulfonamides in the blood are filtered by the
glomerulus. Sulfonamides in the renal tubules undergo tubular reabsorption.
P a g e | 144
Pharmaceutical Considerations
Sulfonamides often come as sodium salts to increase solubility. Monosodium salts are
caustic and to be administered by IV only. Disodium salts may be given IM, IP, etc.
sulfonamides are incompatible with calcium – containing solutions.
There is always a potential for crystalluria (renal deposits) with sulfonamides due to
insolubility. Using “triple sulfa” which is a combination of three sulfonamides of
varying solubility partially solves this problem. According to the law of independent
solubility, the presence of one sulfonamide in solution does not affect the solubility of
the other sulfonamides present in the same solution. Thus, in the triple sulfa, the dose
of the individual sulfonamides is proportionately reduced and the combination is less
likely to cause crystalluria.
Adverse Reactions
The major concern in sulfonamide therapy is the potential damage to the kidneys and
urinary tract. Sulfonamides and their metabolites, because of their limited water
solubility, tend to form crystalline aggregates in the urine as their concentration
increases.
The crystalline aggregates which lead to irritation and obstruction of urine flow are
deposited in the:
renal tubules
renal pelvis
ureters, and
urinary bladder
P a g e | 145
This is more of a problem with older less soluble sulfonamides (e.g. sulfamethazine,
sulfamerazine, and sulfanilamide). Decreased urine output and low urine pH enhance
crystallization. Sulfonamides and their acetylated metabolites are less soluble at acid pH
(pH 5.5 and below).
These effects are caused by direct irritation and by suppression of microbial activity in the
ruminant (very common).
Peripheral neuritis and central and peripheral myelin degeneration may also be observed
in some cases as well as decreased egg production in poultry. Rapid IV injection may
lead to ataxia and collapse. Sulfaquinoxaline used to treat coccidiosis has caused death in
puppies. Hemorrhagic diathesis due to antagonistic effect on vitamin K production is
quite common in poultry.
Drug Interactions
Local anesthetics such as procaine, butacaine, and benzocaine contain a PABA
nucleus which when released by hydrolysis may decrease the effectiveness of
sulfonamides. The combined treatment with sulfonamide and procaine penicillin can
lead to such antagonism.
Solutions for IV administration have very alkaline pH and are very irritating if
administered IM or SC. There are a few preparations buffered to neutral pH. Triple
sulfas were designed to reduce the renal toxicity of sulfonamide therapy. While the
solubility of each sulfonamide is independent of that of the others, the antimicrobial
effects of the three sulfonamides in combination are additive. Most triple sulfas for
veterinary use contain sulfamethazine, sulfathiazole, and one several other sulfas
including sulfanilamide, sulfamerazine, and sulfapyridine.
POTENTIATED SULFONAMIDES
There may be combinations of trimethoprim with other sulfonamides but these are the
most rational since these sulfonamides have pharmacokinetics similar to
trimethoprim. To ensure synergism, the sulfonamide and trimethoprim in combination
must be absorbed, distributed, and excreted together. Very short – acting
sulfonamides are excreted early leaving trimethoprim. On the other hand, long –
acting sulfonamides stay long after trimethoprim has been excreted.
Chemistry
Mechanism of Action
Potentiators of sulfonamides were developed to selectively inhibit bacterial
dihydrofolate reductase. Trimethoprim, a structural analogue of the pteridine
portion of dihydrofolic acid reductase, is a competitive inhibitor of
P a g e | 148
Resistance
All mechanisms of resistance to sulfonamides also apply to the sulfonamides
in combination with trimethoprim. The resistance to trimethoprim is both
chromosomal – and plasmid – mediated, and the mechanisms include:
Pharmacokinetics
Absorption
Trimethoprim is well absorbed orally. It is absorbed faster than sulfonamides
and therefore peaks sooner in the blood. It is degraded by rumen fluid and
therefore, is not orally bioavailable in adult ruminants. Absorption is usually
good following SC administration.
Distribution
Trimethoprim has a greater tissue distribution than sulfonamides. Its tissue
levels are greater than blood levels for most tissues (and body fluids). High
P a g e | 149
Drug Interactions
Trimethoprim – sulfa increases the risk of bone marrow suppression when
used with antimetabolites (6 – mercaptopurine and azathioprine) and
antifolates (methotrexate and Pyrimethamine).
P a g e | 150
Lesson 14
Aminoglycosides
and Spectinomycin
A MINOGLYCOSIDES
Compounds
CHEMISTRY
aminocyclitols, these compounds are weak bases with pKa between 7.2 and 8.2
polycationic, polar, and very water soluble.
Mechanism of Action
The aminoglycosides bind with the free 30S ribosomal subunits so that it is made
unable to bind with mRNA and cause misreading of the genetic code. Because of the
misreading of the genetic code, there is failure of initiation of protein synthesis hence
no protein is produced. This leads to a bacteriocidal effect.
Initiation
Occurs at a specific codon AUG which codes for f – met (formyl –
methionine)
Elongation
Addition of more amino acids to the growing peptide chains
Termination
Occurs on three specific codons; UAA, UGA, and UAG appearing. A specific
release factor binds and hydrolyzes the bond holding the peptide to the tRNA
in the P site. The mRNA dissociates from the 70S ribosome which then breaks
up into 30S and 50S subunits for recycling.
The entry of aminoglycosides into bacterial cells involves both passive diffusion
through aqueous pores and an active transport which requires ATP and is inhibited by
acid pH, lack of oxygen, hyperosmolarity, calcium and magnesium. Entry into the cell
is facilitated by the presence of drugs that interfere with the cell wall synthesis; hence,
the classic synergism between aminoglycosides and beta – lactam antibiotics.
Aminoglycosides affect both Gram – positive (limited) and Gram – negative bacteria
(E. coli, Klebsiella, Enterobacter, Proteus, Pseudomonas, and Salmonella) but their
use should be restricted to severe Gram – negative infections. They are not effective
against anaerobic bacteria. Newer aminoglycosides are effective against resistant
P a g e | 153
Gram – negative organisms like Pseudomonas and Proteus. Streptomycin (but not the
other aminoglycosides) is effective against tuberculosis
Resistance
Pharmacokinetics
Absorption
Aminoglycosides are poorly absorbed from the gut (<1% of the oral dose)
because of their polar and polyionic nature. However, patients with impaired
renal function may get toxicity following repeated oral administration.
Significant absorption may occur, however, from inflamed gut, or from large
burns when applied topically. They may be rapidly absorbed systematically
from serosal surfaces of body cavities and from IM and SC injection sites.
Distribution
Aminoglycosides are distributed only in the extracellular fluid. As obligate
polar molecules, they don’t cross the cellular barriers very well. They have
low plasma protein binding and reach therapeutic levels slowly in synovial,
peritoneal, and pleural fluids after repeated doses.
Aminoglycosides poorly penetrate into the CSF, eye, respiratory secretion, and
prostate. Concentrations in the bile reach one – third of the plasma level. They
tend to accumulate in kidney tissues by pinocytosis by the cells of the
proximal convoluted tubules. High concentrations is also attained in the
endolymph and perilymph of the inner ear (hence, predisposition to
ototoxicity).
These antibiotics are inactive in suppurative wounds because they bind to pus
(probably to DNA). Remember polymyxins?!!
P a g e | 154
Excretion
Aminoglycosides are excreted mainly by glomerular filtration with some
tubular secretion and tubular reabsorption. High concentrations of unchanged
aminoglycosides are attained in the urine. Their elimination half – life is short
(about 1.5 – 3 hours) in domestic animals.
Nephrotoxicity
All aminoglycosides are capable of causing renal damage.
Characterized by:
Renal tubular cell necrosis
Albuminuria
Elevated blood urea nitrogen (BUN)
Reduced urine output
Acute tubular necrosis occurs more likely with prolonged therapy (6 – 7 days
or longer). Tubular casts and increased protein in the urine are characteristic
early signs of toxicity. Significant damage is already present when the BUN
and Creatinine are elevated. Gentamicin is the most nephrotoxic of the
aminoglycosides used systemically but all are nephrotoxic.
Ototoxicity
Damage to the auditory portion of the ear leads to deafness which is may be
permanent. Damage to the vestibular portion causes ataxia and incoordination
P a g e | 155
Neuromuscular Blockade
Curare – like paralysis is dose – related and may occur only with high doses.
Aminoglycosides inhibit pre – junctional release of acetylcholine while also
reducing post – synaptic sensitivity to acetylcholine. This may become
significant only when other neuromuscular inhibitors such as muscle relaxants
and anesthetics are concurrently administered. Neostigmine and calcium
antagonize the neuromuscular blockade produced by aminoglycosides and are
used in the treatment of this form of toxicity.
Drug Interactions
Individual Drugs
SPECTINOMYCIN
Chemistry
Mechanism of Action
P a g e | 158
Like the aminoglycosides, Spectinomycin binds with the 30S ribosome and inhibit
CHON synthesis but does not cause misreading of the genetic code. Unlike
aminoglycosides, it is bacteriostatic.
Resistance
Pharmacokinetics
Spectinomycin is poorly absorbed from the gut (<10% in dogs) but rapidly absorbed
when given IM or SC. It has a low volume of distribution limited to extracellular fluid
because of poor lipid solubility.
Spectinomycin
20 – 40 g/kg oral q8h for enteric infection
Horse: 10 – 20 mg/kg IV daily
Lesson 15
Tetracyclines
P a g e | 160
Chemistry
All tetracyclines are amphoteric (capable of acting both as acid and as base) and
crystalline salts can be formed either with strong acids or strong bases. The most
common salt is the hydrochloride, except for doxycycline which is available as
doxycycline hyaclate (doxycycline hemiethanolate hemihydrate). The tetracyclines
are most lipid soluble in the zwitter - -ion form, at their isoelectric point (pH 5.6 for
all tetracyclines except minocycline which is a full pH unit higher). Minocycline and
Doxycycline are most lipid soluble. All tetracyclines are not very stable in aqueous
solution and are heat labile.
Mechanism of Action
The entry of tetracyclines in the Gram – positive bacterial cells requires two
processes:
Passive diffusion through hydrophilic pores in the outer cell membrane
Active transport through the inner cell membrane
Mechanism of Resistance
P a g e | 161
Pharmacokinetics
Absorption
Distribution
All tetracyclines reach high concentration in the kidney, bile, liver, lung, and
sites of active ossification. There is good penetration in synovial fluid, sinuses,
mucosa, and milk. They cross the placenta and enter fetal circulation and
amniotic fluid. The older tetracyclines penetrate the brain, CSF, eye, and
prostate but may accumulate when injected at high doses IV over a period of
time. The newer more lipid soluble tetracyclines distribute to these tissue well.
Tetracyclines are stored in reticuloendothelial cells of the liver, spleen and
bone marrow, in bone, and in the dentine and enamel of unerupted teeth.
Elimination
Tetracyclines are eliminated in the urine and feces. They undergo entero –
hepatic circulation. Most of the dose is excreted unchanged by the kidney via
glomerular filtration. There may be some degree of tubular reabsorption
depending on lipid solubility and urine pH (alkaline urine increases excretion).
Doxycycline and minocycline are exceptions because they are primarily
excreted in the feces by direct mucosal secretion, as inactive conjugate or
P a g e | 162
In general, tetracyclines are non – toxic. Dogs and cattle usually tolerate the drugs but
cats and horses have more problems. Tetracyclines cause gastrointestinal upset due
to alteration of normal GI flora which can result in superinfection with toxin
producing bacteria and fungi, and by direct irritation of the GI tract.
Dogs may vomit or have diarrhea but giving the drug with food can lessen GIT
upset Avoid tetracycline in dogs in the last 3 weeks of pregnancy and the first
month of life
In cats, oral doses can result in diarrhea, colic, vomiting, depression, fever, and
anorexia which severely limit their use in cats
In the horse, therapeutic doses (IV) of tetracycline have been associated with
apathy, anorexia, severe diarrhea, and even death. Tetracyclines should be used
cautiously in the horse.
The degree of effect is dose – dependent. Tetracyclines cause:
Staining of teeth
Enamel hypoplasia
Stunt bone growth in young animals
Hepatotoxicity has been reported in dogs (and people) especially in patients with
impaired renal function.
The vehicle propylene glycol used in the formulation is probably the cause. Rapid IV
administration in cow can precipitate signs of milk fever (hypocalcemia).
Phototoxicity
Hypersensitivity reactions
Anaphylaxis (rare), and
Increased catabolism
Increased blood urea nitrogen (BUN)
P a g e | 163
Individual Drugs
Lesson 16
Chloramphenicol
and its Derivatives
Chemistry
P a g e | 165
Mechanism of Action
Chloramphenicol and its derivatives which are primarily bacteriostatic, inhibit protein
synthesis by binding to 50S ribosomal subunit. This action prevents the binding of the
amino acid – containing end of tRNA to its ribosomal binding site. They can also
inhibit mitochondrial protein synthesis in mammalian cells. The mammalian
erythropoietic cells seem to be particularly sensitive to these drugs.
In humans, its use is restricted to typhoid fever, or other severe infections against
which no other antibacterial agents are effective. In general, chloramphenicol should
probably be used as a second choice antibacterial but may be a first choice for some
deep infections of the eye.
The rapid elimination of chloramphenicol in the horse and frequent dosing needed to
maintain therapeutic level throws some doubt on its usefulness in the horse.
Mechanism of Resistance
P a g e | 166
Resistance to chloramphenicol has increased since the drug was first introduced.
Plasmid – mediated resistance is very important in Gram – negative bacteria. A
specific acetyltransferase acetylates chloramphenicol to an inactive compound. In
Staphylococcus aureus, an inducible form of chloramphenicol acetyltransferase has
been demonstrated.
Pharmacokinetics
Absorption
Absorption of chloramphenicol after oral administration in monogastric
animals is rapid with good bioavailability but absorption after IM or SC is
unpredictable. In adult ruminants, orally administered chloramphenicol is
inactivated by rumen microorganisms.
Distribution
Chloramphenicol has a very wide distribution in the body due to high lipid
solubility. It penetrates intracellular as well as extracellular fluid. Highest
concentrations are attained in the liver, bile, and kidney. Therapeutic
concentration is attained in the brain, CSF, eye, prostate, and milk.
Chloramphenicol crosses the placenta and may affect the fetus. It may
penetrate the eye following topical or subconjunctival administration.
Elimination
Hepatic metabolism is the principal process of elimination of
chloramphenicol. Chloramphenicol glucoronide is the main metabolite
excreted (90%); others are de – acylated or dehalogenated metabolites (2%),
and unchanged drug (8%). There are species variations in the rate of
glucuronidation and his corresponds with variations in elimination half – life.
For example:
In the horse, the half – life is about 1 hour, and
In cat, about 5 hours
Neonates cannot conjugate chloramphenicol as rapidly as adults
and this results in elimination half – life considerably longer than
in adults
Chloramphenicol may:
Reduce appetite
Cause vomiting
Weight loss
Dehydration
CNS depression
P a g e | 167
The effects are seen more frequently in cats than in dogs. Bone marrow suppression
(hypoplasia) occurs in most species given high levels of the drug, and is dose –
dependent and reversible.
It is characterized by:
Anemia
leukopenia, and
thrombocytopenia
Humans are the only species that develop non - dose – dependent irreversible aplastic
anemia as a result of chloramphenicol therapy (or exposure to chloramphenicol
residues in food). Since there is no safe level set for humans, chloramphenicol is
banned for use in food animals in many countries including Philippines. There is a
claim that thiamphenicol and Florfenicol does not cause bone marrow aplasia in
humans.
“Grey Syndrome” occurs in neonatal humans, and in young and adult cats.
This syndrome is characterized by cardiovascular collapse and cyanosis, and is
associated with deficient glucoronide conjugation in neonates and in cats.
Formulations
Lesson 17
Macrolides
Compounds
Chemistry
Macrolides contain a many – membered lactone ring to which are attached to one or
more deoxysugars. They are weak bases with pKa of 7.1 (Tylosin) or 7.8
(erythromycin). They are highly lipid soluble and their salts have good water
solubility.
Gastrointestinal disturbances
Gastrointestinal are the most common reactions and are dose – related. These
are manifested as vomiting and occasional diarrhea in small animals or
diarrhea in the horse. These can also be a problem in guinea pigs, rabbits, and
hamsters.
Individual Drugs
Erythromycin
This is a second – choice antibacterial for most applications. Therapeutic are
used mainly vs. Gram – positive bacteria including penicillinase – producing
staphylococci and streptococci. Some Gram – negative bacteria such as
Bacteroides, Haemophilus, and Actinobacillus. Bordetella is often sensitive.
mycoplasma infections
acute mastitis
Tylosin
This is also a second choice antibacterial for most application. Less active
against bacteria except Brachyspira hyodysenteriae but is more active against
P a g e | 170
Spiramycin
This is several times less active against bacteria than erythromycin. Not as
effective against Mycoplasma as Tylosin and Tiamulin (See next lesson).
Despite relatively poor activity in vitro, it has exceptional ability to
concentrate in tissues. Has same application as Tylosin but may be a drug of
choice against cryptosporidiosis.
Tilmicosin
In cattle, this is used for the treatment of respiratory disease caused by
Pasteurella spp. Produces cardiovascular toxicity in species other than cattle.
Lesson 18
Pleuromutilins
Chemistry
Activity against anaerobic bacteria and mycoplasma is better than that of macrolide
antibiotics. They are used for swine dysentery (Brachyspira hyodysenteriae) and
chronic swine pneumonia and in strategic medication to prevent and treat common
infections of pigs. Their use as growth promoter in swine is no longer encouraged.
P a g e | 172
Although they are not active against most enteric bacteria, they seem to prevent
colonization of E. coli in intestinal mucosa.
Resistance
Pharmacokinetics
Drug Interactions
Drug interactions are likely to be similar to those described for lincosamides and other
macrolides. A definite synergism between the pleuromutilins with any of the
tetracyclines (particularly chlortetracycline) has been well established.
There may be irritation at IM injection sites but no irritation with sesame oil – based
preparation. IV injection has caused neurotoxicity and death in calves. Pleuromutilins
should not be administered to horses and other herbivores with expanded large
intestines (guinea pigs, rabbits) because of danger of destruction of large bowel flora
and predisposition to colitis.
Tiamulin
Pig: 60 ppm (mg per liter) in water for 5 days or
10 – 15 mg/kg IM for 2 – 3 days for swine dysentery
Lesson 19
P a g e | 174
Lincosamides
Chemistry
Mechanism of Action
The lincosamides are 50S bacteriostatic. They inhibit protein synthesis by binding
with 50S ribosomal subunits
The spectrum of activity includes primarily Gram – positive cocci including some
penicillin – resistant staphylococci, anaerobes including Bacteriodes fragilis, some
Clostridium, some Nocardia, and Actinomyces, some Mycoplasma, and Brachyspira.
Enterococci and Gram – negative bacilli are resistant.
Resistance
Pharmacokinetics
P a g e | 175
Lincosamides are widely distributed in the body. They can penetrate intracellular fluid
but not the CSF. High lipid solubility, ion – trapping can result in high concentration
of lincosamides in some tissues and body fluids. High concentrations are achieved in
skin, bones, and joints. Lincosamides may be the drug of choice for osteomyelitis.
High concentrations may also be achieved in milk (4 – 7 x plasma levels) and bile.
Generally, they are high protein bound.
GI disturbances are the major adverse effect, vomiting especially in cats; diarrhea
resulting from pseudomembranous colitis caused by Clostridium difficile. For this
reason, lincosamides are contraindicated in the horse, rabbit, guinea pig, hamsters.
Dogs are relatively resistant to GI disturbances. Gastroenteritis occurs as fatal colitis
in horses and in cattle, oral lincomycin as low as 7 – 10 ppm in feed causes
inappetence, diarrhea, ketosis and reduced milk production. Hypersensitivity reaction
is rare.
Individual Drugs
Lincomycin
Is approve for use in dogs, cats, swine, and poultry, and is available as
injection, oral tablets, soluble powder, and feed premix. It is not a first line
drug in veterinary medicine but it is used to a limited extent. Lincomycin is
used to:
Clindamycin
Is not used very frequently in veterinary medicine but may be used for
treatment of toxoplasmosis in dogs and cats. It is used for Gram – positive
infections. Other antibiotics that are less expensive and have less effect are
usually preferable.
Lesson 20
P a g e | 177
Nitrofurans and
Nitroimidazoles
N ITROFURANS
The Nitrofurans have broad antibacterial activity but their toxicity largely limits
their use to topical application to the treatment of intestinal infections, and to the
treatment of urinary tract infection.
Compounds
Chemistry
The members of the Nitrofurans group are moderately lipophilic and vary in their
water solubility. Nitrofurazone and Nitrofurantoin are weak acids with pKa of 7.1.
Nitrofurazone is water – soluble and Nitrofurantoin is practically insoluble in water.
Mechanism of Action
Pharmacokinetics
Nitrofurantoin is well absorbed after oral administration but is rapidly excreted in the
urine. They may be some metabolism. The blood and tissue concentrations are too
low for treatment of systemic infections. The mean concentrations attained in the
urine are between 50 to 250 micrograms/ml, therefore, it is quite useful in the
treatment of urinary tract infection. Antibacterial activity is favored by acidic urine.
Nitrofurazone and Furazolidone on the other hand are poorly soluble an d are not
absorbed from the gut.
Contraindicatioins:
Individual Dugs
Nitrofurazone
Nitrofurazone has anti – protozoal activity and is primarily used topically to
treat wounds and infections of the eye, ear, skin, and reproductive tract. It is
used as a feed additive in chickens to control coccidiosis. Water – mix and
solution are available to treat intestinal infections, primarily in swine.
Nitrofurantoin
Nitrofurantoin can be use to treat lower urinary tract infections in small
animals but it can be quite toxic and this limits its use.
Furazolidone
Furazolidone is used a feed additive in poultry and swine to control intestinal
infections and coccidiosis. Oral solution is availab le for intestinal infections
in piglets.
P a g e | 179
NITROIMIDAZOLES
The Nitroimidazoles are used in veterinary medicine for their activity against
anaerobic bacteria. They are similar in spectrum of activity and mechanism of action to the
nitrofurans but are not active against aerobic bacteria
Compounds
Chemistry
Mechanism of Action
The Nitroimidazoles are bacteriocidal to most Gram – positive and Gram – negative
anaerobic bacteria. Some (such as metronidazole and Dimetridazole) are effective
against protozoan parasites: Trichomonas fetus, Giardia lamblia, and Histomonas
meleagridis
Resistance is rare among usually susceptible bacteria, a characteristic shared with the
nitrofurans. Resistance involves reduced intracellular drug activation.
Pharmacokinetics
Metronidazole
Well absorbed after oral administration in monogastric species
Distributed widely in tissues (including CNS)
Penetrates the brain and cerebrospinal fluid
Extensively oxidized and conjugated in the liver and excreted in the urine
Lesson 21
Quinolones,
Rifamycins, and
Novobiocin
P a g e | 182
Q UINOLONES
One derivative, Flumequine, is used mainly for enteric infections of food animals.
Addition of flourine atom at position 6 and a piperazenyl or pyrrolidinyl group at position 7
(Flouroquinolones), dramatically increased both the spectrum of activity and potency. (refer
to chemical structures of quinolones).
Mechanism of Action
The quinolones inactivate DNA gyrase enzyme. DNA gyrase is essential in the
process of DNA replication. The active enzyme consists of two pairs of subunits
called subunits A and B. The quinolones inactivate DNA gyrase by inhibiting the
subunit A which is fewer in number than the subunit B.
Pharmacokinetics
Nalidixic acid is almost completely absorbed after oral administration.
It is extensively and rapidly metabolized and is excreted as inactive
conjugate.
Adverse Reactions
Serious CNS stimulation
Seizures in dogs
Hepatic necrosis
Irritation at injection sites
Ciprofloxacin
Enoxacin
Perfloxacin
Ofloxacin
All these drugs belonging to the second and third generation quinolones have a
common 4 – quinolone nucleus and carboxylate constituent at carbon 3, a 6 – flouro
and a 7 – piperazino substituent. All have enhanced activity against DNA gyrase.
Pharmacokinetics
Rapid and adequate absorption after oral administration, distribute widely in body
fluids, penetrate in to CSF in meningitis. The extent of biotransformation varies with
specific drugs. Excretion is partially through the kidney.
Spectrum of Activity
It is very active against Enterobacteriaceae and many other Gram – negative rods
including Pseudomonas aeruginosa. It is lower but therapeutically useful activity
against Gram – positive aerobes and inactive against anaerobes. With the exception of
Flumequine, all have some activity against mycoplasma, rickettsia. Norfloxacin is
less potent than ciprofloxacin, Enrofloxacin, and Danofloxacin.
Adverse Reaction
Gastrointestinal symptoms:
Nausea
Vomiting
Anorexia
This has limited available data on toxicity in animals and may be toxic for dogs and
cats.
RIFAMYCINS
Compounds:
Rifamycin SV
Rifampin
Rifamide
Mechanism of Action
Rifamycins interfere with DNA – dependent RNA polymerase
Prevents formation of mRNA
Bacteriocidal
Pharmacokinetics
Rifamycins are readily but incompletely absorbed from the gut and diffuse
rapidly into tissues reaching concentrations exceeding those in serum. They
are biotransformed to several metabolites and excreted in bile and in urine.
They may be administered by IM or IV routes.
They are active against Gram – positive bacteria, anaerobes, and some
mycobacteria. Rifampin is the most active drug known against Staphylococcus
aureus. Very active against Mycobacterium tuberculosis but most other
mycobacteria are resistant. Gram – negative bacteria are generally resistant but
Brucella and other fastidious organisms are susceptible. It is active against
Chlamydia and use in foals to control Corynebacterium equi – induced
pneumonia. This is often administered in combination with other antibiotics
such as penicillin and erythromycin due to the ready development of
resistance.
Rifampin
Horse: 5 mg/kg bid oral
Foal: 5 mg/kg SID oral
Dog: 5 mg/kg SID oral
P a g e | 185
NOVOBIOCIN
Mechanism of Action
Pharmacokinetics
Adverse Reaction
Lesson 22
Miscellaneous
Antibiotics
V IRGINIAMYCIN
Mechanism of Action
Pharmacokinetics
P a g e | 187
Adverse Reaction
MONENSIN
A polyether ionophore antibiotic
Mechanism of Action
Competes with sodium in the cell membrane
Causes passive transport of potassium ions out of the cell for hydrogen ions
Low intracellular pH kills the cell
Adverse Reaction
Highly toxic to horses and to ruminants at doses 5 – 1 times those
recommended
Fatal when administered together with tiamulin
Salinomycin is used as a growth promoter in swine but may be fatal when combined
with therapeutic concentrations of tiamulin.
FUSIDIC ACID
A lipophilic steroid antibiotic
Mechanism of Action
Prevents protein synthesis by inhibiting the binding of aminoacyl tRNA to the
ribosomal A site
Mechanism of Action
Inhibits bacterial DNA synthesis
P a g e | 188
Adverse Reaction
Bacterial resistance
Overdose retards growth
Hypokalemia due to adrenal cortex dysfunction
ISONIAZID
A hydrazide of isonicotinic acid and is the most potent anti – tuberculosis drug used
in humans. The mechanism of action is unknown.
Adverse Reaction
Neurotoxic effects resulting from a relative deficiency of pyridoxine which is
inhibited by isoniazid
Accidental ingestion of 66 mg/kg by a dog led rapidly to convulsion.
Mechanism of Action
Inhibit the first part of the cell wall synthesis in Gram – positive bacteria by
inhibiting the formation of phosphoenolpoyruvate.
(Review the process of cell wall synthesis)
FLAVOMYCIN (BAMBERMYCIN)
Lesson 23
P a g e | 190
Antifungal Agents
F
ungal infections are classified according to the location of the infection:
Dermatophytic infections
(Dermatophytoses) are the most common fungal (mycotic) infection. They
involve the skin, hair, and nails. Most dermatophytes can be treated with
topical antifungal agents
Mucocutaneous infections
Involve moist skin and mucous membrane (GIT, perianal, and vulvovaginal
areas), and can be treated with topical application of Amphotericin B,
miconazole, clotrimazole, nystatin, and with oral ketoconazole for chronic
infection
Systemic infections
Occurs less frequently, and is usually chronic in nature. They are rather
difficult to diagnose. Treatment of systemic infections is with parenteral
antifungals
a. irrigant solution
b. ointment
c. tablet, and
d. suppository
Benzoic Acid
Is the main ingredients in Whitfield’s Ointment (formula: 6% benzoic acid,
3% salicylic acid). It has fungistatic as well as a keratolytic property
Salicylic Acid
Is used in the treatment of chronic superficial skin infections. It has moderate
fungistatic activity but with good keratolytic action. Included in many topical
antifungal preparations, it requires the presence of water to produce keratolytic
effects. It may cause skin irritation
Ciclopirox Olamine
Has a broad spectrum antifungal activity. It is effective against Candidia
(yeast), Epidermophyton, Microsporum, and Trichophyton but has limited
penetration of epidermis and is available in 1% solution
Tolnaftate
Used against Epidermophyton, Microsporum, and Trichophyton infections. It
has no effect on bacteria and on Candidia spp. It may be administered with
griseofulvin for more immediate topical effect and is available as 1% cream,
solution, or powder
Candidicin
A fungicidal polyene antibiotic and used primarily for moniliasis
Haloprogin
A synthetic antifungal agent used against Microsporum, Trichophyton, and
Candidia and is available as 1% cream or solution
Iodochlorhydroxyquin
P a g e | 192
Has both antifungal and antibacterial activity and is used for localized
dermatophytoses complicated by bacterial infection. It is available as 3%
cream, ointment, or powder
Natamycin
Also known as PIMARICIN. It is a fungicidal polyene antibiotic effective
against a wide range of filamentous and dimorphic fungi and yeasts. It is used
for local application against ringworm, in udder for yeast mastitis, and on the
eyes for mycotic keratitis and is available as 2.5% or 5% solution
Cuprimyxin
A broad spectrum antifungal with antibacterial activity. After application, the
myxin component is released from the copper complex and is effective against
Microsporum, Trichophyton, and Candidia
Nystatin
A polyene antibiotic which is the first antimycotic antibiotic to be discovered.
It binds to sterols in fungal cell membrane resulting in increased membrane
permeability and cell destruction. Its absorption time from the gut is
negligible. It is not available for parenteral administration due to its renal
toxicity. When used topically, it does not cross skin and mucous membrane.
This has fairly broad – spectrum of activity including yeast and various fungi,
and is used against Candidia, Microsporum, and Aspergillus and may cause
local burning and itching sensation in area of application and gastrointestinal
upset
Dose:
Dog 22,000 units/kg/day
Poultry 62.5 – 250 ppm in drinking water
Clotrimazole
An imidazole which is used for topical treatment of Epidermophyton,
Microsporum, Trichophyton, and Candidia, and is available as 1% cream, or
solution
Ketoconazole
Also an imidazole which is a broad spectrum antifungal and is used also for
dermal and systemic fungal infection. It I effective against Coccidiodes
immitis, Cryptococcus neoformans, Histoplasma capsulatum, Blastomyces
dermatitidis, Candidia spp., Aspergillus sp., and Sporothrix spp. It has an
antagonistic effect with amphothericin B. For Blastomycosis in dogs, use 30
mg/kg daily for 2 months
Thiabendazole
A benzimidazole which has antifungal as well as anthelmintic activity and
may reduce aflatoxin formation in infected feed. It is effective against
Blastomyces, Fusarium, Candidia, Penicillum, and Trichophyton
Iodine Preparations
Include Tincture of Iodine, Potassium Iodide, and Iodophores
P a g e | 193
Dyes
Under this are:
Carbol – Fuchsin solution
Composition:
0.3% basic fuchsin
4.5% phenol
10.5% resorcinol
5.0% acetone
10% ethyl alcohol
Application:
Clean the affected area with soap and water, dry and
apply medication once or twice a day for one week
Gentian Violet
Used for candidiasis and other fungal infections. May be
available alone or in combination with Benzathonium chloride
(a disinfectant)
Griseofulvin
An oral fungistatic antibiotic against dermatophytes that inhibits fungal cell
mitosis by binding to microtubules disrupting the mitotic spindles.
Administered orally, it is variably absorbed from the gut. The rate of
absorption depends greatly on particle size and is enhanced by high fat meal.
Absorbed griseofulvin distributes to growing nails and skin, and binds to
keratin to make the cells resistant to fungal infection. The drug is
biotransformed in the liver an excreted in the urine. This is also effective
against Epidermophyton, Microsporum, Trichophyton, and Sporothrix. It must
be given for prolonged period usually for several weeks to months. Adverse
reactions to griseofulvin include occasional diarrhea and nausea, hepatic
toxicity (has been reported), and teratologic effects in cats.
Dose
Horse 10 mg/kg daily for 20 days; oral
Dog/Cat 7 – 20 mg/kg/day for 3 – 4 weeks; oral
Cattle 7.5 mg/kg for 7 days (in feed)
Amphotericin B
Polyene antibiotic for systemic infections which binds to ergosterol, the
principal sterol of the fungal cell membrane and causes cell destruction. It is
active in both growing and resting cells. Additionally, it causes oxidative
P a g e | 194
damage to fungal cell in vitro and enhances cell – mediated immunity in the
host. Not absorbed after oral administration, it is usually administered IV for
systemic effect. Little amphotericin B penetrates the CSF and excretion is
through the urine and bile
It has no activity against protozoa and bacteria and been the most important
drug available for the treatment of systemic fungal infections but its place is
being challenged by ketoconazole.
5 mg/kg IV 3 x a week;
On the first day, dilute the total dose in 20 ml of 5% dextrose and
Dog
5 ml is given. If no acute anaphylactic response develops give
the remainder over 45 second
Flucytosine
A fluorinated pyrimidine derivative converted in sensitive fungi to 5 –
fluorouracil. Mammalian cells do not convert floucytosine to fluorouracil.
This is further biotransformed to 5 – flouro – deoxyuridylic acid, an inhibitor
of thymidylate synthesis, and thus DNA synthesis
It is well absorbed from the gut and with minimal binding to plasma protein, it
penetrates into tissues and the CSF. Largely excreted in the urine unchanged.
P a g e | 195
Amphotericin B which can reduce renal function, may increase the toxicity of
flucytosine when they are used in combination
The majority of yeast isolates from bovine mastitis are resistant. Major
application is for treatment of cryptococcal infection in cats
Dose
Ketoconazole
Inhibits the fungal enzyme sterol 14 – alpha demethylase and therefore
interferes with the biosynthesis of ergosterol and bind to phospholipids in the
fungal cell membrane. This results in cell leakage. It also interferes with
cytochrome C oxidase and with peroxidase resulting in increased in
intracellular peroxide and cell death
Cats are more sensitive and may develop anorexia, depression, diarrhea, and
fever. Ketoconazole may be embryotoxic and teratogenic; not given to
pregnant animals
Dose
Lesson 24
Drugs against
Antiprotozoan
Parasites
P a g e | 197
T he parasitic protozoa differ to some extent from the pathogenic bacteria and fungi.
Many protozoan parasites require arthropod intermediated hosts for complete
development. They are generally closer in biochemical make – up to the
mammalian cells than are the pathogenic bacteria and fungi. This poses a problem
with regard to the selective toxicity of the antiprotozoan agents
The majority of the antiprotozoan drugs interfere with biosynthetic mechanism of the
cells especially nucleic acid synthesis. A few disrupt membrane function. Many affect
energy metabolism. Selective toxicity appears to be due to differences in
permeability. The mechanisms of selective toxicity are more or less well – defined.
Examples of sites of action having no equivalent in vertebrate cells are L – α –
glycerophosphate oxidase (in trypanosomes) and Dihydro – pteroate synthetase (in
sporozoa)
DRUGS INTERFERING WITH ENERGY METABOLISM
Melarsoprol (Mel B) is more commonly used than Tryparsamide that may cause
blindness, which is why it is very seldom used today. Melarsoprol may cause
encephalopathy, abdominal colic, and vomiting
Suramin
Its mechanism of action is obscure but L – alpha glycerol – phosphate oxidase
of the African trypanosomes which is concerned with the oxidation of NADH, is
sensitive to suramin. It is used to treat trypanosomiasis and has a marked
curative effect but is less useful for prophylaxis against Trypanosoma evansi, T.
brucei, T. equinum, and T. equiperdum. It is a synergistic potentiator of
phenanthridines and of aminoquinaldine derivatives (see below)
Dose
Inhibits the respiratory chain phosphorylation and ATPase activity in rat liver
mitochondria. A similar action may occur in coccidian. The basis of selective
toxicity is unknown and is both coccidiostatic and coccidiocidal. It is used for
prevention and treatment of coccidiosis in poultry
Primaquine (8 – AMINOQUINOLONES)
The action of primaquine is very similar to that of naphthoquinones and is
used as an anti – malarial drugs
Dose
Based on doses for humans
Diiodohydroxyquin 500 mg tid for 10 days
Iodochlorhydroxyquin 600 mg tid for 21 days
Amphotericin B
Disrupts cell membrane functions by binding to membrane sterols. The
membrane becomes leaky to cations. It is used for the treatment of cutaneous
leishmaniasis and also an antifungal agent
These ionophores specifically inhibit the transport of potassium ions and are
used for treatment of coccidiosis in poultry. They prevent the first sexual cycle
by inhibiting the development of first generation schizonts and cause
retardation of development of immunity to coccidiosis. They are suitable for
addition to broiler feed but should not be mixed with the feed for laying birds.
They should not be fed to horses and other equines and must not be used
together with other coccidiostat. Ionophores have been observed to cause
death of host animals when used with therapeutic levels of tiamulin and
valnemulin (pleuromutilins)
Dose
Monensin 0.01 – 0..121% in feed
Lasalocid 0.005 – 0.0075% in feed
Salinomycin 0.01% in feed
Narasin 0.007%
Maduromicin 0.0005 – 0.0006%
Amprolium
Is as structural analogue of thiamine or vitamin B1, a cofactor of a number of
decarboxylase enzymes, used as an anti – coccidial by inhibiting chiefly the
first generation schizonts; effective in the early stages of infection. It is used as
a preventive and therapeutic drug against Eimeria tenella and E. acervulina in
broilers and may be used as a preventive anticoccidial in layers. Adverse
reactions include bleeding disorders but addition of vitamin K in the diet
reduces mortality
Dose
Pyrithidium Br 2 mg/kg IM
Radanil (2 – NITROIMIDAZOLES)
See notes under Metronidazole, Dimetridazole, and Ipronidazole below
Dose
Dose
Curative for trypanosomiasis 3.5 mg/kg SC or IM
Cattle 100 – 150 ml of 1% solution for trichomoniasis
Injected into the prepuce, retained for 15 minutes,
Diminazene
and massage the penis and prepuce thoroughly
Aceturate
Cattle and 2.0 mg/kg SC or IM for Babesia bigemina;
Dog 5mg/kg for B. bovis
Horse 5 mg/kg SC or IM
Horse 8.8 mg/kg for babesiosis
Phenamidine
Dog 15 mg/kg for babesiosis
This drug seems to act by displacing Mg²+ and polyamines from the
cytoplasmic ribosomes of trypanosomes causing ribosomal aggregation and
inactivation. It is used for treating trypanosomiasis (T. congolense, T. vivax, T.
equiperdum, and T. equinum). Chloride salt – insoluble in water is used for
curative effect. Methylsulfate salt – soluble in water is used for prophylaxis
and is well – tolerated by ruminants; local and systemic reactions may occur in
horses and dogs
Dose
Clindamycin (LINCOSAMIDES)
(CARBANILIDES)
Nicarbazine (anticoccidial)
Used for prophylaxis against E. tenella, E. necatrix, and E. acervulina.
It has a marked inhibitory effect on the second generation schizonts. It
is suitable for broilers; given for first 12 weeks of chicken life but
unsuitable for layers or breeding stock because of the effect on egg
color and hatchability. Adverse effects include interrupts egg – laying
period; reduces hatchability; mottled egg yolk
Nitrophenide
Acts against E. tenella, E. necatrix. Give at 0.025% in feed. It inhibits
second generation schizonts
Imidocarb dipropionate
Is used for treatment and prophylaxis of babesiosis and anaplasmosis
Dose
Imidocarb
Cattle 1.2 mg/kg SC or IM
For babesiosis Horse 2.4 mg/kg SC or IM
Dog 6 mg/kg SC or IM
For anaplasmosis Cattle 3 mg/kg
Amicarbalide
Cattle 5 mg/kg
For babesiosis
Horse 8 mg/kg
NITROBENZAMINES
Dinitolmide (Zoalene)
An anticoccidial that inhibits the development of second – generation
schizonts but does not inhibit the development of immunity
Nitromide
Also an anticoccidial which is a mixture of 30% N – acetyl – N’ – p –
nitrophenyl sulfanilamide, 25% 3,5 – dinitrobenzamide, and 5% 4 –
hydroxyl – 3 nitrophenylarsonic acid
P a g e | 204
Dose
0.25 mg/kg
Dog
Inhibits cholinesterase in the hosts
Halofuginone
A preventive anticoccidial; given at 0.0003% in feed
COCCIDIOSIS
a. 2 important genera: Eimeria and Isospora
b. Coccidia may enter cells in liver (rabbit) and intestine (cattle, sheep, pigs,
goats, dogs, rabbits, and fowls)
c. A major problem in poultry industry
d. Infection is self – limiting
e. The parasites undergo 2 stages of multiplication:
1. Asexual (schizogony)
2. Sexual (sporogony)
f. The rapid multiplication during sporogony is the most pathogenic stage
g. Repeated infection may cause host immunity
h. Symptoms include:
1. Diarrhea or bloody feces
2. Heavily infected animals become emaciated and anorexic
Animals adopt a drooping posture and often huddle together for warmth
P a g e | 205
LIFE CYCLE
Male
Release 3rd Merozoites
microgametes
generation invade other cells
fertilize the female
schizonts or and develop into
to become
intracellular 2nd generation
infective zygotes
sexual schizonts
(oocysts) that are
gametocytes
discharged in the
host’s feces
o Nitrofurazone
7 – 10 mg/kg daily for 7 days
May be given in feed at 0.0165% or 0.008% in drinking
water
o Amprolium
50 – 62.5 mg/kg in water or feed for sheep
100 mg/kg for 4 days or longer for goats
In Cattle
Infection occurs in all parts of the world
Serious outbreak occurs in dairy herd
P a g e | 206
o Lincomycin HCl
1 g/calf in water for 21 days
o Sulfonamides
Not recommended
In Pigs
Treat with Nitrofurazone 0.44% in feed for 7 days
In Poultry
The problem of coccidiosis is ubiquitous in poultry management
It is difficult to avoid infection completely
The clinical disease is dependent upon the number of oocyst ingested
by individual birds
An outbreak of coccidiosis depends upon the factors that allow oocysts
to sporulate and remain viable
There is no cross – immunity between species of coccidia
Treatment:
o Anticoccidials are used usually in starter ration for broilers
o Protection is more important in fast – growing birds than the
egg – laying type where immunity and caging alter the
demands for anticoccidial drugs
o The emergence of resistant strains presents a major problem
Methods to prevent the development of resistance:
o Switching around the 13 different classes of drugs
o “Shuttle Program” – a planned switch of drug in the middle of
growing period
Curative treatment
Should be given immediately after diagnosis
o Sulfadimidine
0.2% in drinking water for 2 periods of 3 days
separated by 2 days without treatment
P a g e | 207
o Sulfaquinoxaline
In feed at 5%
o Sulfonamides
Are active against schizont stage specifically the
second – generation schizonts
Preventive medication
Prolonged continuous use of coccidiostatic compounds in feed
and water
TOXOPLASMOSIS
Caused by Toxoplasma gondii
Definitive hosts are domestic cat, jaguarondi, ocelot, mountain lion, and
leopard cat
Intermediate hosts are almost all warm – blooded animals
Of zoonotic importance
Most infections are asymptomatic
Complete eradication is by elimination of infected animals
Treatment:
P a g e | 208
Pyrimethamine
Found effective in monkeys and humans
Of little value against cyst forms
Synergistic with sulfonamides
Clindamycin
TRYPANOSOMIASIS
Caused by blood parasites Trypanosoma spp. and with a few exceptions, these
organisms undergo cyclical changes in arthropod vectors
Treatment:
Suramin
Horse: 4 g/45 kg IV
Dog: 0.03 g/kg IV for 6 days
Homidium Br
Cattle: used as 1 – 2.5% solution at 1 mg/kg IM
Protection lasts for about 5 weeks
Pyrithidium Br
Cattle/Sheep/Goat: 0.5 mg/kg deep IM
Protection lasts for 6 – 10 months
LEUKOCYTOZOONOSIS
Treatment is usually not effective
Prophylactic medication
BABESIOSIS
Caused by Babesia spp
Transmitted by arthropods
Acute, often fatal infections characterized by fever and anemia
Treatment:
In addition to injection of babesiacide, supportive therapy is important
Recovery is most likely when treatment is given early in the course of
infection in cattle
Quinoronium sulfate is used in infections caused by all species but
does not eliminate infections
Phenamidine isothionate
Diminazene Aceturate
P a g e | 209
ANAPLASMOSIS
Treatment:
Tetracycline
11 mg/kg for 10 days
Tetracycline TA
22 mg/kg, administer 4 doses, 3 days apart
Imidocarb propionate
4 mg/kg, 2 weeks apart or daily for 3 days at 5 mg/kg
Lesson 25
P a g e | 210
Anthelmintic
Drugs
Qualities of Antiparasitic:
Wide therapeutic index
Examples:
Toxic dose is at least 3x the therapeutic dose
Effective after only 1 dose
Easy to administer
Inexpensive
Does not leave residues
Important in food – producing animals
Mechanism of Action
Benzimidazoles
Examples:
Mebendazaole, Flubendazole
P a g e | 211
Inhibitors of Glycolysis
Examples:
Arsenicals such as Thiacetarsamide that binds to sulfhydryl (-
SH) group of glycolytic enzymes thereby altering the tertiary
structure of proteins and the active sites of enzymes in both
host and parasites
BENZIMIDAZOLES
Examples
Thiabendazole Albendazole Cambendazole
Fenbendazole Flubendazole Mebendazole
Oxfendazole Oxibendazole Netobimin
Febantel Thiophanate
P a g e | 212
General Properties
Low toxicity
Some are teratogenic
Mechanism of Action
Inhibit energy metabolism
All except Flubendazole and Mebendazole inhibit fumarate reductase
Disrupt the function of cell microtubules. This is a slow process and
requires a prolonged contract between drugs and parasites
They are more effective in ruminants and horses than in other species
Divided doses are more effective than a single dose
Oxfendazole (a sulfoxide) is the active metabolite of Fenbendazole and
Febantel
Albendazole, Fenbendazole, Thiophanate, and Febantel are
biotransformed in the liver and re – secreted into the alimentary tract as
active metabolites
Thiabendazole
Well – absorbed and therefore may destroy larval forms in the
tissues
May cause hypersensitivity reaction but this may be due to the
destruction of migrating larvae rather than due to the drug itself
Has antifungal and antiyeast properties also
Doses
Thiabendazole
66 mg/kg
Cattle 110 mg/kg for severe parasitism or
severe infection with Cooperia
Sheep and 44 mg/kg
Goat 66 mg/kg for severe parasitism
44 mg/kg
Horse
88 mg/kg for ascarids
Pigs 75 mg/kg
Laboratory 48 – 100 mg/kg
Animals
In Cattle
Virtually 100% effective against all gastrointestinal
roundworms except Nematodirus, Ostertagia, and Trichuris
P a g e | 213
In Horses
Very effective against large and small strongyles and pinworms
(Oxyuris)
The drug of choice against strongyles in foals
Low activity against Parascaris and immature Oxyuris
Combination with Piperazine or with trichlorfon (an
organophosphate) is effective against these parasites
In Pigs
Effective against strongyles in piglets
May be effective against Hyostrongylus rubidus, Strongyloides
ransomi, and Oesophagustomum dentatum
In Cattle
Cambendazole, Fenbendazole, Albendazole, and Oxfendazole
Effective against adult and immature lungworm Dictyocaulus
Fenbendazole
Effective against inhibited Ostertagia larvae
Parbendazole
Causes death of Ostertagia larvae in abomasal modules
Albendazole
Affects most species of roundworms, tapeworms, and liver
flukes
In Horses
P a g e | 214
Mebendazole
Effective against lungworm at increased dose
Also affects equine tapeworm Anoplocephala
Albendazole
Used against equine lungworm
In Pigs
All are effective against Hyostrongylus, Strongyloides ransomi,
Oesophagustomum dentatum, and Ascaris
Fenbendazole
50 mg/kg/day for 2 – 3 days
Doses:
DRUG HORSE CATTLE SHEEP GOAT PIG DOG/CAT
Cambendazole 20 25 20 - 20 -
Fenbendazole 5 7.5 5 5 5 50
Mebendazole 8.8 - - - 1.25 22
Parbendazole - 30 15 – 30 - 25 – 30 -
Albendazole - 7.5 7.5 - 5 – 10 -
Oxfendazole 10 4.5 5 7.5 3 – 4.5 -
Oxybendazole 10 - - - 15 -
Flubendazole - - 10 - 5 -
THE PROBENZIMIDAZOLES
Febantel
Metabolized to true benzimidazoles similar to Oxfendazole and
Fenbendazole which are responsible for its pharmacological activity
P a g e | 215
In Horses
o Large and small strongyles
o Adult ascarids and adult pinworms
o Effective against Gastrophilus bots (larvae) when
combined with trichlorfon
Thiophanate
Metabolized to Lobendazole (not marketed)
Effective against most nematodes of farm animals
Netobimin
Metabolized to Albendazole sulfoxide by the gut microflora
Doses
Febantel
Horse 6 mg/kg oral
Dogs and Cats 10 mg/kg for 3 days
Puppy and Kitten 15 mg/kg for 3 days
Ruminants 10 mg/kg
Pig 20 mg/kg
Thiophanate
Ruminants 50 mg/kg
Pig 5 – 12 mg/kg for 14 days
Netobimin
Ruminants 7.5 mg/kg
20 mg/kg for inhibited larvae flukes and tapeworms, oral
12.5 mg/kg by injection
IMIDOTHIAZOLES
Levamisole
Has nicotine – like action on the neuromuscular junction and minor ability
to inhibit fumarate reductase. Available for oral (drench, or feed additive)
and for parenteral (SC) administration which has better systemic effect
In Ruminants
Affects all major gastrointestinal roundworms (Haemonchus,
Ostertagia, Cooperia, Trichostrongylus, Bunostomum,
Oesophagostomum), lungworms (Dictyocualus), and eyeworm
(Thelazia)
In Horses
For ascarids, lungworms, and adult pinworms
In Pigs
Controls nodular worms (Oesophagostomum), ascarids,
Strongyloides ransomi, and Metastrongylus lungworm
In Dogs
Affects all stages of heartworm and also ascarids, and
hookworms, but not whipworms
In Chickens
Affects Ascaridia, Heterakis, Capillaria, and Oxyspirura
mansoni
Tetramisole
A racemic mixture of D – and L – isomers
Anthelmintic activity is associated with L – form which is Levamisole
Toxicity is associated equally with both forms
Practically the same as Levamisole except for higher dose required and
greater likelihood of toxic reactions
Doses
Levamisole
8 mg/kg oral or SC
Ruminants and Pigs 1o mg/kg as pour on
5 – 6 mg/kg SC for Thelazia
Horse 10 – 15 mg/kg oral or SC
Dogs and Cats 7.5 mg/kg SC
40 mg/kg in half the daily consumption of
Chicken
drinking water
Tetramisole
15 mg/kg oral or SC
Ruminants and Pigs
Not to exceed 4.5 g in cattle
Chicken 3.6 mg/kg/day for 3 days (for gapeworms)
P a g e | 217
ORGANOPHOSPHATES
Administered orally in various forms, well – absorbed from the gut, and
specifically larvicidal
Examples
Dichlorvos
Trichlorfon (Metriphonate)
Mechanism of Action
Inhibition of cholinesterase resulting in paralysis of adult worms
The mechanism of larvicidal action is unknown
Toxicity
Overdose leads to muscarinic and nicotinic signs
Animals that are susceptible to toxic effects of organophosphates
include:
o Debilitated animals
o Dogs with hookworm
o Cattle with migrating warble fly larvae
o Animals concurrently medicated with succinyl – choline or
similar drugs
o Certain breeds of dogs
o Young animals
o Pregnant animals
Treatment of toxicity:
Atropine sulfate and pralidoxime (2PAM)
In Ruminants
Mainly for worms in the abomasum and upper small intestine and has
little effect on worms in the large intestine
In Horses
Good activity against ascarids, pinworms, and bots, less activity
against strongyles and are also used against stomach bots
In Pigs
Active against ascarids, Oesophagostomum and Trichuris
P a g e | 218
Doses
DICHLORVOS
Horses (feed formulation)
31 – 41 mg/kg
Foal (gel formulation)
20 mg/kg for bots
Pig 11.2 – 21 mg/kg
Dogs 27 – 30 mg/kg
Puppies/Cats 11 mg/kg
TETRAHYDROPYRIMIDINES
Examples
Pyrantel
Morantel
Mechanism of Action
Nicotine – like depolarization of neuromuscular junction leading to
paralysis of the worms
Pharmacokinetics
Poorly absorbed systematically and therefore, has less larvicidal
activity. The tartrate salt, however, is well absorbed in the pig and the
dog. The pamoate salt is less soluble in water and therefore, stays in
the gut
Toxicity
Less toxic than OP’s and Levamisole
In Horses
Effective against tapeworm Anoplocephala, against strongyles,
ascarids, and pinworms with variable effects
In Pigs
For ascarids and Oesophagostomum
In Ruminants
Against Haemonchus contortus (including Thiabendazole – resistant
strains), Ostertagia, Trichostrongylus, Nematodirus, and Cooperia
In Dogs
P a g e | 219
For hookworms and ascarids but not too effective against whipworms
Doses
Pyrantel tartrate
Horse 12.5 mg/kg
Pig 22 mg/kg maximum of 2 g/animal
Ruminants 25 mg/kg
Pyrantel pamoate
Horse 6.6 mg base/kg
Dog 5 mg base/kg for dogs over 2.2 kg, suspension
15 mg/kg for smaller dogs less than 2.2 kg
Morantel tartrate
Sheep 10 mg/kg
Cattle 8.8 mg/kg oral; 2% ointment for Thelazia eyeworm
Morantel fumarate
Sheep 12.5 mg/kg
PHENOTHIAZINE
An older anthelmintic. The development of truly broad – spectrum anthelmintics
has reduced its clinical use
Mechanism of Action
Not known and does not seem to be related to the amount of drug
absorbed by the worms
Toxicity
Safer in sheep than in other species
Photosensitization, conjunctivitis, and corneal opacities in ruminants
Hemolytic anemia and jaundice in horses
Anemia in dogs and cats
In Ruminants
P a g e | 220
In Horses
100% effective against small strongyles and not effective against
ascarids. At present, used only for prevention rather than for
therapeutic purposes
Doses
Phenothiazine
Sheep and Goat 25 – 30 g for weighing over 27 kg
12.5 g for 11 – 23 kg animals
Cattle 10 g/45 (maximum of 70 g)
Horse 3 g/45 kg for small strongyles
5 g/45 kg for large strongyles
(give ½ of the calculated dose daily for 2 days
Chicken 0.5 g/bird
Turkey 1 g/bird
Daily prophylactic dose
Sheep and Goat 0.25 – 0.5 g/animal
Cattle 0.5 g/45 kg
Horse Adult 2 – 5 g
Colt 1g
PIPERAZINE
Available as:
Adipate (active Piperazine content = 37%)
Chloride (= 48%)
Citrate (= 35%)
Dihydrochloride (-50 – 53%)
Hexahydrate (= 44%)
Phosphate (= 42%), and
Sulfate (= 46%)
Mechanism of Action
A non – depolarizing (curare – like) neuromuscular blockade resulting
in the paralysis of the worms
Toxicity
With a wide margin of safety, large doses may produce emesis,
diarrhea, incoordination, and head pressing in dogs and cats
Spectrum of Activity
Administered orally and therefore active only against adult worms in
the gut
In Horses
P a g e | 221
In Pigs
For ascarids and nodular worms
In Chickens
For ascarids
Doses
Piperazine
Dog and Cat 45 – 64 mg/kg
Horse 110 mg/kg
Pig and Ruminants 110 mg/kg
32 mg/kg given in each of 2
Poultry successive feeding or in drinking
water for 2 days
IVERMECTIN
Mechanism of Action
Unique among anthelmintics in that it accentuates endogenous GABA
– induced inhibitory actions in worms to cause paralysis
Pharmacokinetics
Orally and parenterally active, poor penetration of membrane barriers
In Ruminants
Effective against adult and fourth larval forms of Haemonchus,
Ostertagia, Cooperia, Trichostrongylus, Strongyloides, Bunostomum,
Nematodirus, Trichuris, Oesophagostomum, Dictyocaulus, and
Chabertia
P a g e | 222
In Horses
For small strongyles, large strongyles, ascarids, pinworms, stomach
worms, intestinal worms, and lungworms. Also for Gastrophilus bots
and Onchocerca microfilariae
In Pigs
For immature and adult forms of Ascaris, Hyostrongylus,
Strongyloides, Trichuris, Oesophagostomum, Metastrongylus,
Stephanurus dentatus, and the intestinal stages of Trichenella
In Dogs
As a preventive for heartworm, effective against hookworms, ascarids,
and whipworms
Doses
Ivermectin
Cattle 0.2 mg/kg SC
Sheep 0.2 mg/kg oral
Horse 0.2 mg/kg oral or IM
Dog 0.006 – 0.12 mg/kg oral
n – BUTYLCHLORIDE
A liquid given orally in capsule for the control of ascarids and hookworms in dogs
and cats but has no effect on migrating larvae. Less effective against hookworms
than against ascarids and does not have satisfactory activity against parasites of
other domestic animals
Toxicity
Usually non – toxic when given therapeutically
Safe unless the patient is debilitated or too young
Fasting of animal for 12 – 24h prior to administration is required
Give a laxative such as magnesium sulfate 30 minutes to 1 hour after
administration of n – butylchloride
Doses
n – Butylchloride
Dog and Cats
Body weight < 2.25 kg 1 ml
2.25 – 4.5 kg 2 ml
4.9 – 9 kg 3 ml
9 – 18 kg 4 ml
>18 kg 5 ml
P a g e | 223
ANTITREMATODAL DRUGS
Albendazole
Approved for use against mature liver flukes such as Fasciola hepatica in
cattle
With 27 – day withdrawal period
A teratogen
Praziquantrel
For lung flukes in dogs
Also effective against liver flukes but too expensive to use in ruminants
Clorsulon
Most effective against mature and immature F. hepatica cattle
Adverse effects is rare
Mode of action is that it deprives flukes of metabolic energy source
Preslaughter withdrawal period is 8 days and milk withdrawal period is 4 days
PHTHALOFYNE
For oral and intravenous administration; toxic causes gut disturbances and
CNS depression
GLYCOBIARSOL
For oral administration; poor systemic absorption, less toxic than phthalofyne
BUTAMISOLE
Chemically related to Levamisole and used for treatment of whipworm and
hookworm in dogs
Doses
Butamisole
Dog and Cat 24 mg/kg SC
Toxicity
Vomiting, ataxia, tremors, lateral recumbency, and convulsions
Contraindicated in debilitated, or heartworm – positive dogs
Do not use within 24h of bunamidine HCl (an anti – cestodal
treatment)
DISOPHENOL (DNP)
For dogs and cats, there is uncouple oxidative phosphorylation. Given orally or
SC
Affects only adult worms; repeat treatment in 2 – 3 weeks
Toxicity
Hyperthermia, tachycardia, dyspnea, convulsions, cataracts maybe
induced but usually disappears spontaneously
Avoid hot environment and restrict exercise for a couple of days after
therapy
Caution when used in sick and debilitated animals
Dose
Disophenol
Dog 10 mg/kg SC
In an existing case, use adulticide to kill adult worms, followed 6 weeks later by a
microfilaricide. Then give preventive daily during mosquito season and for 2 months
thereafter
ADULTICIDES
Eliminate both immature and adult heartworms
Sodium Thiacetarsamide
An organic arsenical which is very irritating when injected
extravascularly
Given IV for a couple of days to allow gradual kill of worms which
starts to die 5 – 7 days after treatment
Release of decomposing dead worms into pulmonary circulation may
cause pulmonary thrombosis, cough, and fever
Kidney and liver toxicity may be produced
Keep dog confined and quiet 1 – 2 weeks after treatment
Concurrent antibiotics and corticosteroids reduce deleterious effects of
dead worms
In case of extravascular injection, dilute drug by infiltration of
injection site with normal saline solution and glucocorticoids
Topical application of dimethylsulfoxide (DMSO) may also help
P a g e | 225
Melarsomine hydrochloride
A new organoarsenical adulticide with efficacy superior to
Thiacetarsamide
Administered by deep IM rather than IV injection, some muscle and a
few days of mild soreness may develop
The filaricide efficacy of Melarsomine, unlike that of Thiacetarsamide,
can be graded by dose. It is possible to eliminate worms from heavily
infected dogs in stages, thereby distributing the impact of worm emboli
Toxicity of Melarsomine differs from Thiacetarsamide by not causing
any hepatic necrosis
Doses
Thiacetarsamide
Dog 2.2 mg/kg 2x a day for 2 days
Melarsomine
MICROFILARICIDES
Given at 6 weeks after adulticides
Dithiazanine iodide
Given orally continuously until microfilariae disappear from the blood
Give for 1 week then check for microfilariae. If still positive, give
another course of therapy. If still positive, give another course of
therapy at a higher dose
Toxicity
Gut disturbances and potential nephrotoxicity
Dose
Fenthion
Pour on OP insecticide, applied topically on dorsal midline once a week; no
longer used because of toxic fatalities
P a g e | 226
Ivermectin
Dose: 0.5 mg/kg in single oral dose
PREVENTIVES
Diethylcarbamazine (DEC)
Also active against ascarids and hookworms in dogs and cats
Maintenance of effective blood level during mosquito season
Prevents development of larvae following mosquito bite
Given daily during mosquito season and 2 months after season is over
Puppies are started on therapy at weaning
Remember, it is only a preventive
Do not give to animals with existing heartworm infestation. Sudden
massive destruction of microfilariae may produce fatal anaphylactic
reaction. DEC “opsonizes” the microfilariae to the action of antibodies
Ivermectin
Dose: 0.006 mg/kg once a moth prevents infestation
Milbemycin oxime
Related to ivermectin and is used as a heartworm preventive at 0.25 –
0.5 mg/kg. It is effective against canine hookworms and other
roundworms
An effective anticestodal drug must remove both the scolex and the body of the worm
PRAZIQUANTREL
Broad – spectrum
Affects adult and juvenile forms of all species of tapeworms; certain lung and
pancreatic flukes
Dose
Praziquantrel
Dogs 1 mg/kg for Taenia
2.5 mg/kg for Dipylidium
Cattle 10 – 50 mg/kg
Sheep 50 mg/kg
NICLOSAMIDE
A salicylanilide
Uncoupler of oxidative phosphorylation
Effective against Monieza, Taenia, and Dipylidium
Not effective against Echinococcus granulosus
Removes scolex and body of worms
Relatively nontoxic with little systemic absorption
Dose
Niclosamide
P a g e | 227
BUNAMIDINE
Removes whole tapeworm
Effective against all tapeworms of dogs and cats including Echinococcus
Must not be given with cholinesterase inhibitors and can cause ventricular
fibrillation
Toxic when given IV but oral route has been followed by fatalities
Dose
Bunamidine
Dog 25 – 50 mg/kg after 3 – 4h fast
BENZIMIDAZOLES
Mebendazole
Given 22 mg/kg/day for 3 days for Taenia of dogs and cats
20 mg/kg at each of 2 treatments with 48h interval for adult
Echinococcus granulosus of dog
20 mg/kg for Monieza of ruminants
Fenbendazole
50 mg/kg/day for 3 days for Taenia of dogs and cats
15 mg/kg for Monieza of cattle and sheep
HALOGENATED HYDROCARBONS
P a g e | 228
Examples
Carbon tetrachloride
Used in sheep at 1 ml/animal
Hexachloroethane
Tetradichloroethane
Hexachloroparaxylene
SALICYLANILIDES
Brotianide
3.5 mg/kg for 12 week old flukes
7.1 mg/kg for 6 week old flukes
Clioxanide
15 mg/kg for adult flukes
40 mg/kg for immature flukes (6 weeks)
135 mg/kg for immature flukes (4 weeks)
Closantel
2.5 mg/kg SC
Oxyclzanide
15 – 20 mg/kg oral for adult flukes
45 mg/kg for immature flukes (acute fasciolosis)
Rafoxanide
7.5 mg/kg for adult (100% effective), for immature (50% effective)
Available in combination with Thiabendazole
SUBSTITUTED PHENOLS
Bromsalans
100% effective against immature flukes
Dose:
Bromsalans 30 – 60 mg/kg
Hexachlorophene
25 mg/kg oral or SC
P a g e | 229
Bithionol
60 mg/kg oral
Niclofolan
2.6 mg/kg (adult flukes)
6 mg/kg (6 weeks)
8 mg/kg (8 weeks) oral
0.6 – 1 mg/kg SC
Nitroxynil
10 mg/kg SC only
Clorsulon
3.7 mg/kg (adult flukes)
15 mg/kg (6 weeks)
30 mg/kg (8 weeks)
AROMATIC AMIDES
Examples
Diamphenithide affects the youngest stages of liver flukes
Effectivity diminishes as flukes mature
BENZIMIDAZOLES
Albendazole
7.5 mg/kg in sheep
15 mg mg/kg in cattle
Triclabendazole
10 mg/kg has high activity against all stages of liver flukes
Unlike other benzimidazoles, Triclabendazole does not have effects on
roundworms and tapeworms
P a g e | 230
Lesson 26
Pesticides
P
P a g e | 231
esticides are chemical agents used against external parasites or ectoparasites (flies, lice, ticks,
fleas, mites, bots, and grubs).pesticides include insecticides and acaricides. Insecticides are
agents against parasitic insects while Acaricide are agents against parasitic arachnids.
Ectoparasites
Accurate identification of ectoparasites or correct diagnosis is based on clinical signs
prerequisite to selection of the most appropriate agent
Arachnida
Ticks
Affects mainly ruminants but may also affect horses, dogs, and poultry
Mites
Sarcoptidae includes:
o Mange mites (Chorioptes, Notoedres, Otodectes, Psorergetes,
Psoroptes, and Sarcoptes
o Demodicidae includes parasites causing demodectic mange
o Gamasidae includes the northern and red mites of poultry
Insecta
Diptera (flies)
Anopleura (lice)
Siphonaptera (fleas)
Methods of Application of Pesticides
External Application
Dipping
Spraying
Application by hand as dust, ointment, spray, foams, mist spray, aerosol, pour
on, spot – on
By tags and strips
By self – treatment devices
Systemic Application
As feed additive
By injection
By stomach tube
By intranasal administration (for Oestrus ovis)
Pesticide Toxicity
All pesticides are poisons and can be toxic to both warm – blooded and cold –
blooded animals
Proper application of pesticides must be practiced to prevent injury to animals
Acute signs of poisoning usually results from over dosage
Horses show extreme sensitivity to various pesticide formulations
Animals that are in stress, debilitated, very young, or very old must be treated with
pesticides with extreme caution if at all
Cats are very sensitive to effects of pesticides because their grooming behavior and
deficient drug biotransforming capacity
Animals should not be treated in confined and poorly ventilated area
Do not apply pesticides in combination unless specified by the manufacturer
P a g e | 232
CLASSIFICATION OF PESTICIDES
Stomach Poisons
Must be eaten by parasites to be effective
Contact Poisons
Enter the body of parasites through the skin
Fumigants
Released into the atmosphere as a gas or a mist and enters the parasites
via respiratory system
Chlorinated hydrocarbons
These are becoming less popular because of persistence in the
environment
Lindane, bromocyclen, and methoxychlor are still used such as against
mange mites in pigs while DDT and Chlordane are no longer approved
for use
Examples include:
Lindane
Bromocyclen
Methoxychlor
DDT
Chlordane
Organophosphates
Are both contact and Stomach poison
Inhibits cholinesterase enzymes
P a g e | 233
Pyrethrins
o Are naturally occurring
o Extracted from chrysanthemum flower
o With rapid but brief action
o Relatively non – toxic to dogs and cats
o Often mixed with synergist 9see below) to enhance efficacy
Pyrethroids
o Are synthetic compounds with greater potency than pyrethrin
o Examples include:
Cypermethrin Permethrin
Decamethrin Resmethrin
Allethrin Fenvalerate
Rotenone
A naturally occurring compound obtained from derris roots
Largely superseded but still used in mixtures with other pesticides
Used for car mite and demodectic mange in dogs
Formamidines
Offers an alternative for treatment of ticks and mange mites on
livestock where resistance to OP’s has developed
Examples:
Amitraz which is an acaricidal compound that kills parasites by
inhibiting monoamine oxidase
Synergist
P a g e | 234
Repellents
Used in veterinary medicine but not yet well – established
Examples:
Dimethyl phthalate
Deet (N – N – diethyl – m – toluamide)
D - Limonene
Linalool
Crude citrus oil extract
Avermectins
Examples:
Ivermectin
Abamectin
Doramectin
Benzyl benzoate
An acaricide used as an adjunct in the treatment of sarcoptic mange in
dogs
Toxic to cats
Sulfur
For years, was the best external treatment for mange until the advent of
chlorinated hydrocarbons
May be applied as ointments or oily suspension at a concentration of
1:8
Still used for routine baths for dogs
As powders diluted with talc, kaolin, etc., or as dispersible bath
powders combined with soaps and wetting detergents
Examples:
P a g e | 235
Sublimed sulfur
Precipitated sulfur
Monosulfiram
Fipronyl
Acts by a non – competitive inhibition of GABA, an essential
neurotransmitter of the CNS of invertebrates. By binding to a receptor
site inside the chloride channel, Fipronyl inhibits the intracellular flow
of chloride ions, thereby, inducing a rapid and sure death of the
ectoparasite due to hyper – excitation
A contact insecticide
Available as spray or spot – on preparation and may be applied on a
monthly basis