Monday 11 February 2019

Drugs to be avoided in epilepsy and myasthenia Gravis

Drugs to avoid in patients with seizures -

 Although the list of medications that lower seizure threshold is huge, most of these medications cause seizures only in rare occasions. - Example; all cephalosporins have the potential of inducing seizures, however only cefepime was found to be commonly implicated and rest of cephalosporins are rarely associated with seizures. - Here, we included only drugs that known to commonly induce seizures. Group Family Drugs shown to lower seizure threshold Antibiotics 4 th generation cephalosporins Cefepime Carbapenems Imipenem Penicillin Ampicillin – Ampicillin/Sulbactam (Unasyn) Quinolones Ciprofloxacin – Levofloxacin Antipsychotics Atypical antipsychotics Clozapine Typical antipsychotics Chlorpromazine Antidepressants Aminoketones Bupropion Serotonin agonists Buspirone SNRI Venlafaxine Tricyclics Amitriptyline – Nortriptyline – Clomipramine Mood stabilizers Lithium Lithium Analgesics Narcotics Fentanyl – Tramadol – Meperidine Immunosuppressants Calcineurin inhibitor Cyclosporine – Tacrolimus




Drugs to Avoid with Myasthenia Gravis

Antibiotics Heart medications Anesthesia Brain/Nerve Others ampicillin Quinidine Procainamide Lithium Timolol eye drops Amoxicillin Quinine Succinylcholine Phenytoin Cortisones Penicillin Procainamide Curare derivatives Gabapentin Penicillamine Imipenem Statins Botox Iodinated contrast Aztreonam Atenolol Nicotine Magnesium Ofloxacin Metoprolol Methocarbamol Interferon alpha Levofloxacin Sotalol Ciprofloxacin Propranolol Erythromycin Pindolol Clindamycin Nebivolol Azithromycin Nadolol Clarithromycin Labetalol Amikacin Esmolol Gentamycin Carvedilol Tobramycin Bisoprolol Kanamycin Acebutolol Neomycin Amlodipine Streptomycin Verapamil Red: Strong evidence of harmful effect Diltiazem Yellow: Few case reports of harmful effect Nifedipine Blue: Should be avoided in spite of weak clinical evidence Felodipine NB: to make it a patient friendly list, names of all common individual medications were listed instead of pharmacutical group listing so you can 

Pharmacotherapeutic Principles of Neurological and Psychiatric Disorders

Pharmacotherapeutic Principles of Neurological and Psychiatric Disorders 




John A. Schetz Summary The pharmacotherapeutic management of neurological and psychiatric disorders relies primarily on the modulation of central nervous system (CNS) neurotransmission with drugs that intervene at chemical synapses. The receptors, transporters, and enzymes for the dopaminergic, serotonergic, and noradrenergic systems are the most common neuropsychiatric drug targets, because these neurotransmitter systems play a central role in the regulation of a range of cognitive and motor behaviors.

 The key to understanding or anticipating the clinical profile (dose–effect) of a particular drug is to have an appreciation for both its pharmacodynamic and pharmacokinetic properties. Key Words: Psychiatric; neuroscience; pharmacology; pharmacodynamics; pharmacokinetics; synapse; G protein-coupled receptors; dose–effect; theory; disorder; neurotransmission. 



1. INTRODUCTION

 The appropriate, effective, and safe utilization of drugs in the treatment of disease requires a basic understanding of the dose–effect relationships of medications. Relating dose to effect requires a combined appreciation of pharmacodynamic concentration–effect relationships, or what drugs do to the body, and of pharmacokinetic dose–concentration relationships, or what the body does to or with drugs. For this reason, considerable attention is devoted to both the pharmacodynamic and pharmacokinetic aspects of drug therapies.

The aim of this chapter is to provide a theoretical rationale that is necessary for appropriately interpreting the results of basic and clinical neuropharmacology studies and for understanding many of the drug treatment strategies commonly encountered in clinical neurology and psychiatry. Approximately one-fourth of all drugs prescribed worldwide exert their therapeutic actions on CNS targets. Of the top five selling drugs in this category, three are antidepressants and two are atypical antipsychotics

 (1). The relative success of pharmacological intervention is highlighted further when one considers that these 30 Schetz drugs are treating an estimated 7–15% of the population who suffer from one or more of the neurological or psychiatric disorders discussed in this book. Currently, the biogenic amine neurotransmitter systems, and in particular dopaminergic, serotonergic, and noradrenergic receptors, transporters, and metabolic enzymes, cover the vast majority of neuropsychiatric drug targets. The reason for this is that the biogenic amine systems are key modulators of neuronal excitability, and the molecular components of these systems are located at chemical synapses, which are sites that are accessible to intervention by drugs.

2. THE CHEMICAL SYNAPSE AS THE MAIN SITE OF DRUG INTERVENTION

Therapeutic approaches to modulating neuronal excitability at chemical synapses can be categorized as presynaptic and postsynaptic.

 Presynaptic strategies involve altering the levels of neurotransmitter in the synaptic cleft. This can be achieved by changing the amount of endogenous neurotransmitter released or available for release into the synaptic cleft, or by altering the amount of neurotransmitter taken back up (reuptake) into the presynaptic terminal.

 The dopaminergic synapse can be used as a specific example to illustrate these points (Fig. 1).

 For example, monoamine oxidase B inhibitors, such as selegiline, block dopamine (DA) degradation, which makes more DA available for release. Inhibitors of DA synthesis, such as α-methylparatyrosine, reduce the amount of DA available for release. Drugs like reserpine and tetrabenazine decrease vesicular-mediated release by blocking vesicular monoamine transporters, which prevents the storage of neurotransmitter into synaptic vesicles.

 Inhibitors of plasmalemmal DA transporters, such as buproprion or cocaine, block the reuptake of DA from the synapse, and thereby, keep levels of DA in the synaptic cleft high. Certain drugs, like the psychostimulant amphetamine, cause nonvesicular DA release by running the DA transporte Pharmacotherapeutic Principles 31 31 Fig. 1. 32 Schetz agonists, such as DA or the antiparkinsonian drug pergolide, directly activate DA receptors, whereas neuroleptic drugs like thioridazine and haloperidol block DA receptor activation.

3. CLASSIFICATION   OF    DRUGS ON THE BASIS OF THE    RESPONSES   PRODUCE ON   THEIR RECEPTORS 

When a drug reversibly binds to the orthosteric (primary) site on its receptor one of four outcomes are to be expected: the receptor becomes activated, the receptor becomes partly activated, the receptor becomes inactivated, or the receptor is unable to be activated. Consequently,

drugs are generally classified based on their actions.

 A drug is an agonist if it fully activates receptors, a partial agonist if it partly activates receptors, an inverse agonist if it inactivates receptors (and prevents them from being activated), or an antagonist if it only prevents receptors from being activated. For instance, the endogenous neurotransmitter DA is an (full) agonist of DA receptors, and the antiparkinsonian drug bromocriptine is a partial agonist at D2 receptors. Antipsychotics like haloperidol and clozapine may be inverse agonists of D2 DA receptors (4,5), whereas L-741,626 is an (neutral) antagonist.

 Receptor activation is a thermodynamic process, whereby agonist binding induces a conformational change in the receptor and converts it from the inactivated state (agonist low-affinity binding state) to the activated state (agonist highaffinity binding state). Typically, neutral antagonist binding is indifferent to the conformational (affinity) state of its receptor, because it must only occupy the orthosteric site rather than occupy and then induce a change in it.

However, an inverse agonist is a special type of antagonist in that when it binds to a receptor in the activated state it converts it to the inactivated state.

 For this reason, inverse agonists reduce the basal levels of constitutive receptor activity, which corresponds to the (typically small) proportion of receptors that are in the activated state in the absence of agonist. Such distinctions in the molecular mechanisms of action of antipsychotic drugs that act on D2-like dopamine and 5-hydroxytryptamine (5-HT)2- like serotonin receptors may be critical to understanding their unique therapeutic profiles (4,6). Although most drugs bind directly to the orthosteric site of the receptor, other drugs bind at another (secondary) receptor site, called an allosteric site. Ligands that bind to the allosteric site are known as allosteric modulators, because they indirectly modulate the binding of primary ligands to the orthosteric site by remotely altering the orthosteric-binding site.


The modulation is said to be positive if the modulator facilitates a primary ligand’s interaction with the primary site or negative if the modulator attenuates its interaction with the primary site. The extent to which the allosteric site and the orthosteric site are coupled, or their coopera Pharmacotherapeutic Principles 33 range of allosteric mechanisms and corresponding allosteric sites exist for modulating the effects of endogenous and therapeutic agents

 (7). For example, the endogenous tripeptide proline–leucine–glycine (PLG) is a positively cooperative allosteric modulator of agonist binding to D2 DA receptors. Sodium ions are negatively cooperative allosteric modulators of agonist binding to D2 DA receptors, and zinc ions are neutrally cooperative allosteric modulators of antagonist binding to D4 DA receptors.




 4. PHARMACODYNAMICS    OF PHARMACOTHERAPIES 



Chemical agents that have therapeutic actions are referred to as drugs.

 The term pharmacodynamics describes what drugs do to the body. Most drugs exert their actions on the body by interacting with specific sites called receptors. Consequently, pharmacodynamics deals with the interactions of drugs with their receptor sites. The most critical drug–receptor properties concern the strength of their attraction (binding affinity) and functional effects (potency) expressed in units of drug concentration, and the quantity of receptor in the target tissue (receptor density) or the maximal extent of a receptor’s functional effect (efficacy). The density of receptor sites is typically expressed as moles receptor per amount of tissue, whereas the maximal functional effect, which relies in part on receptor density, is usually expressed as receptor activity per unit amount of tissue.

 The functional activity of a receptor can be measured by a variety of endpoints, ranging from changes in biochemical markers to behaviors. Two coupled events occur when a drug interacts with its receptor.


 First the drug binds to its receptor, and second it mediates some functional effect that is transduced by the receptor. Although drug binding and receptor activation are coupled, they are mechanistically distinct molecular processes under the control of unique receptor microdomains and they can be influenced by different factors.

Consequently, there may not be a direct one to one correspondence linking one process to the other.

4.1. Determination of Drug Affinity and Maximal Receptor Density:

 Ligand–Receptor Binding Interactions The reversible (noncovalent) binding of a ligand with its receptor is a dynamic process, which is usually studied in one of two ways. The first way is to measure the kinetics of binding—the rate of approach to or departure from the equilibrium condition.

The second way is to measure the free energy forces of binding under the equilibrium condition. It is helpful to review the general principles of receptor binding theory, in order to know what sorts of experiments to perform to extract kinetic and equilibrium properties of ligand–receptor binding interactions, and additionally, to know how to interpret the meaning of such properties in the context of drug therapies. The theoretical construct that allows one to extract the properties that describe both kinetic and equilibrium types of ligand binding processes and the relationship between them is referred to as the mass action law.

This law assumes that a ligand 34 Schetz reversibly binds to a single homogenous population of receptor sites.

 Because the law is restricted to reversible reactions, which are those that can attain an equilibrium condition, the ligand–receptor interaction can be modeled as an equilibrium reaction. As with all equilibrium reactions, when equilibrium is achieved the rate of the forward and reverse reactions are equal; at equilibrium, the rate of ligand– receptor association equals the rate of ligand–receptor dissociation as shown in Eq. 1. Rate of association (forward reaction) = Rate of dissociation (reverse reaction) (1) The rates at equilibrium can be expressed mathematically in terms of reactants and products as shown in Eqs. 2 and 3. Rate of association = [LIGAND][RECEPTOR]kon (2) Rate of dissociation = [LIGAND·RECEPTOR]koff (3) in which [LIGAND] is the ligand concentration expressed in units of Molarity (i.e., moles/liter) [RECEPTOR] is the total receptor concentration expressed in units of Molarity [LIGAND·RECEPTOR] is the ligand–receptor complex expressed in units of Molarity kon is the association rate constant for the binding of a ligand with its receptor expressed in units of (s-1 M-1) koff is the dissociation rate constant for the separation of ligand from its receptor expressed in units of s-1

A mathematical model of receptor occupancy can thus be formulated from the theoretical expectation at equilibrium by substitution of the equalities in Eqs. 2 and 3 for those in Eq.

1 to yield Eq. 4. [LIGAND][RECEPTOR] kon = [LIGAND·RECEPTOR] koff (4) Equation 4 can be rearranged such that all the concentration variables occur on one side of the equation and all rate constants occur on the other side as shown in Eq.


5. The ratio of the reactants (ligand and receptor) to products (ligand–receptor complex) thus equals the ratio of the rate of complex dissociation over the rate of reactant association. ([LIGAND][RECEPTOR])/[LIGAND·RECEPTOR] = koff/kon = KD (5) These ratios are also equal to the equilibrium dissociation constant (KD), which represents the concentration of ligand required to occupy half of the total number of receptors. The units of KD are Molarity. A series of substitutions and algebraic manipulations to Eq. 5 (8) puts it in the general form of a rectangular hyperbola (Eq. 6) to yield Eq. 7. y = ax/(b+x) (6) [LIGAND·RECEPTOR] = ([RECEPTOR][LIGAND])/(KD + [LIGAND]) (7) Pharmacotherapeutic Principles 35 Separation of the dependent and independent variables allows for the graphing of the data and the extraction of the receptor-binding properties for a ligand that are the constants in the square hyperbola equation (e.g., a = [RECEPTOR] and b = KD). In the laboratory, the amount of ligand that is specifically bound to its receptor ([LIGAND·RECEPTOR]) is measured as a function of various ligand concentrations ([LIGAND]), and then the [RECEPTOR] and KD are solved for graphically (by applying a square hyperbolic math function). A common practice is to introduce a radioactive atom into the ligand (so that it can be detected), incubate various concentrations of this radiolabeled ligand in a solution containing a fixed amount of its receptor until equilibrium is reached, and then rapidly separate (so as not to disrupt the equilibrium condition) the radioligand bound to the receptor ([LIGAND· RECEPTOR]) from the unbound radioligand in solution ([LIGAND]). The radioactivity of the receptor-bound and (unbound) free radioligand is then quantified in a radioactivity counter. The resulting data obtained for such a saturation isotherm type of binding experiment is depicted in Fig. 2. As can be seen from Eq. 5, the equilibrium dissociation constant can also be calculated by measuring the kinetic rates of ligand association and dissociation from its receptor as the binding reaction proceeds toward or away from the equilibrium condition. This can be accomplished by measuring the amount of radioligand bound to its receptor as a function of time. Kinetic determinations of KD require two separate experiments (association and dissociation rates) for each KD determination and provide no information on receptor density. Therefore, they are usually not the method of choice for determining equilibrium dissociation constant values. Fig. 2. Example of saturation isotherm data for [3H]mesurguline equilibrium binding to cloned human serotonin 5-HT2C receptor expressed in COS-7 cells. A saturation isotherm experiment is conducted by keeping all conditions fixed while varying the concentration of radioligand. The KD = 0.24 nM and Bmax = 0.5 pmoles/mg protein. (Copyright John A. Schetz, 2003.) 36 Schetz Although saturation isotherms have the advantage that they are direct measures of affinity (KD) and receptor density (Bmax), relatively few radiolabeled ligands are available, and consequently the binding affinity for most ligands must be determined indirectly. The inhibition constant (Ki ) is an indirect measure of a ligand’s affinity for its receptor that is numerically equivalent to the equilibrium dissociation constant (Ki = KD). In contrast to saturation isotherm experiments, in which the only ligand present is the radioligand, an inhibition type equilibrium binding experiment examines the ability of a nonisotopic ligand to compete with the radioligand for binding to the receptor site. The inhibition binding experiment is performed with a fixed concentration of radioligand and receptor vs increasing concentrations of competing ligand. An inhibition affinity constant for the nonisotopic ligand is derived from its IC50, which is the concentration of nonisotopic (cold) ligand required to displace half of the total amount of radioligand bound to the receptor (Fig. 3). The semilog dose–response curves for competition experiments take on a sigmoidal appearance. The relative IC50 value extracted from the sigmoidal dose–response curve is then converted, by applying the Cheng-Prusoff transformation (9), to an absolute affinity value (Ki ) that is independent of radioligand affinity and concentration. The competitive form of the Cheng-Prusoff equation (Eq. 8) is a measure of receptor occupancy at equilibrium that obeys the law of Fig. 3. Example of competition binding data for raclopride displacement of [3H]methylspiperone from cloned rat dopamine D2 and D4 receptors expressed in COS-7 cells. The IC50 is the concentration of competing ligand, which is needed to displace half of the radioligand occupying the receptors. Note that IC50 values are relative measures that are dependent on the concentration of radioligand employed in the experiment. In order to convert IC50 values to a concentration-independent equilibrium binding constant (Ki ) a correction factor called the Cheng-Prusoff equation must be applied (9). (Copyright John A. Schetz, 2003.) Pharmacotherapeutic Principles 37 mass action, i.e., it assumes that the nonisotopic ligand binds the same receptor in the same manner as the radioligand—a perfectly competitive inhibition at a single homogenous population of receptor sites. Ki = IC50/(1 + ([RADIOLIGAND]/KD)) (8) The KD value in Eq. 8 corresponds to the affinity of the radioligand and the Ki value corresponds to the affinity of the competing ligand. If the interaction is truly competitive then the linear part of the sigmoidal inhibition curve will have a negative slope equal to unity (pseudo Hill slope = 1).

 More shallow slopes can indicate more than one type of receptor, more than one affinity state for a single receptor or a negatively cooperative allosteric interaction. Steeper slopes indicate a positively cooperative allosteric interaction. For example, agonists can bind different conformational states of the receptor (e.g., high- and low-affinity states) with different affinities, and in these cases, the apparent slope will be shallow.

When the slope is different from unity the assumptions of the law of mass action are violated and a true Ki value cannot be determined. In practice many ligand–receptor interactions are not perfectly competitive, which is sometimes indicated by reporting a relative inhibition constant (K0.5). If the difference between high- and low-affinity states are large enough (e.g., approx 100-fold) the binding curve will be clearly biphasic, and in these cases, the binding interaction can be described with a two-state model.


 A simple competitive binding model is usually not appropriate for determining the equilibrium dissociation constants for allosteric modulators, because they are by definition not acting at the same site on the receptor as the primary ligand-binding site. Instead Schild-type null pharmacological methods (10) or complex kinetic methods

 (11) must be used, in order to assign an equilibrium dissociation constant that accurately reflects the binding interaction of the allosteric modulator with its allosteric receptor site.

 4.2. Determination of Ligand Potency and Efficacy: Ligand–Receptor Functional Interactions Although the description of the binding of a ligand to a receptor provides information about the affinity of the ligand for its receptor, it lacks information about what sort of response the ligand induces in the receptor once it is bound. In order to accommodate a response component, additional terms, which describe factors that affect the functional response can be incorporated into the framework of the receptor occupancy model outlined above. For example, the Ariens equation (12) expresses receptor activity as a fraction, Afraction, of the maximal activity, Amax, and equates this activity ratio to the fraction of ligand–receptor complex ([LIGAND· RECEPTOR]) and the total amount of receptors ([RECEPTOR]) as shown in Eq. 9. Afraction/Amax = (α[LIGAND·RECEPTOR])/[RECEPTOR] (9) The term α is a proportionality factor that is an expression of the efficiency of the coupling of the binding of the ligand with its receptor to its subsequent activation of a receptor response. This efficiency of coupling term is an acknowledgment that some agonists, known as partial agonists, promote less than the optimal coupling 38 Schetz that is required to produce a full response. Consequently, even at maximal receptor occupancy the maximal response for a partial agonist will be less than for a full agonist. In other words, the amount of receptor occupancy is not directly proportional to the relative amount of response if the ligand is not a full agonist. The quantity α thus represents the intrinsic activity of a ligand, which is generally defined as equal to one for the endogenous agonist, 0 for an antagonist, and in between 0 and 1 for a partial agonist. Because the endogenous agonist is assumed to be a full agonist, xenobiotic agonists that produce a greater maximal response than the endogenous agonist can have an efficacy greater than unity. Inverse agonists are a special case of negative efficacy. The negative value is owing to the fact that low levels of receptor can, under normal circumstances, assume the activated state in the absence of agonist.


This basal agonist-independent activated state is known as constitutive activity. The concept of negative efficacy is a result of defining the basal state (agonist-independent activity) as the 0 or baseline value for agonist-stimulated activity. Because inverse agonists bind to the activated (high-affinity) state of the unoccupied receptor and convert it to the inactivated (low-affinity) state, they inhibit basal activity and are said to possess negative efficacy. In contrast to an inverse agonist, an antagonist has no effect on basal activity and because it also is incapable of stimulating the receptor to produce a functional response it is said to have no efficacy. From Eq. 9 it can be seen that two important factors controlling the measured functional activity of receptors in response to ligand binding are receptor density ([RECEPTOR]) and stimulus–response coupling efficiency (intrinsic activity, α). Like Eq. 7, which describes a saturation binding reaction, Eq. 9 describing the functional response also can be expressed in the form of a rectangular hyperbola (Eq. 6) to yield Eq. 10. Afraction = (α Amax[LIGAND])/([LIGAND] + (1/ KD)) (10) When plotted on a semilogrithmic scale the rectangular hyperbolic function takes on the form of a sigmoidal curve. Consequently, a plot of the fraction of functional response (Afraction) vs the logarithmic concentration of drug (log[LIGAND]) can be fitted with Boltzman’s equation describing sigmoidal functions (Fig. 4). The maximal function response or efficacy for a given ligand is the point in which the functional response reaches a plateau at higher concentrations of ligand (Fig. 4), although the concentration of ligand that produces half of the maximal response (Afraction/Amax = 0.5 = EC50) is defined as the potency. Both potency and efficacy are relative measures whose values rely in part on receptor density. Examples of the receptor mechanisms underlying the expected functional responses produced by ligands with different functional activities are depicted in Fig. 4. The functional response term EC50 and the competitive ligand binding property IC50 bear some relation to one another, and although it is tempting to try to draw an analogy between them, there are some important distinctions. Both the EC50 and the IC50 are terms that correspond to concentrations of ligand that produce a half maximal measurement (i.e., activity or inhibition of binding). However, the IC50 is Pharmacotherapeutic Principles 39 a measure of the ability of a competing ligand to inhibit the binding of a radioligand to its receptor that is both independent of receptor density and directly proportional to receptor occupancy. The EC50 is a measure of functional effect that is dependent on receptor density and not necessarily directly proportional to receptor occupancy. The reason that the functional response is not always directly proportional to receptor occupancy by ligand is that the strength of coupling between binding and response must be considered. This is not the case for a ligand–receptor-binding interaction because there is no additional coupling component to consider. This difference between binding interactions and functional responses is the molecular explanation regarding why, depending on the ligand’s intrinsic activity and the conditions under which it is tested, a ligand’s affinity value for its receptor may be different from its potency value.


 5. PHARMACOKINETICS OF PHARMACOTHERAPIES 



The clinical evaluation of a drug in vivo concerns dose–effect relationships, but the pharmacodynamic measures of the concentration–effect of drugs, described above, provide only part of the information. Relating dose to effect requires one to consider the dose–concentration relationships of a drug and then associate this with its concentration–effect relationships. A knowledge of pharmacokinetics, which is what the body does to or with a drug once it is administered, is key to understanding the relationship between drug dose and attaining a concentration of drug at the desired target site for an appropriate period of time to produce the intended therapeutic effect. Because the drug targets for neuropsychiatric disorders are embedded in brain structures that are not readily accessible, drugs cannot be easily applied directly to Fig. 4. Examples of responses for ligands with agonist, partial agonist, inverse agonist and (neutral) antagonist functional properties. The EC50 is the concentration of ligand that produces a half maximal effect, while the efficacy corresponds to the relative level of maximal effect, which can be denoted as intrinsic activity (α). (Copyright John A. Schetz, 2003.) 40 Schetz the target tissues. Rather neuropsychiatric drugs must be introduced into the body at some distal site and then travel to their target sites in the brain. Of great importance to dosing is what happens to a drug once it is administered and en route to its target site. Although some drugs are applied intravenously in clinical trails, once their effectiveness is established most drugs are formulated for oral dosing. The oral administration of drugs is the preferred route of administration for clinical applications, because it eliminates safety concerns associated with the use of needles and it facilitates outpatient treatment. Following oral administration and on its way to its target site,

a drug will encounter various biological barriers, metabolic tissues, and nontarget tissue deposition sites. The collective effect of these factors largely determines the amount of intact drug that is free to interact with the intended receptor target within a given time frame after dosing. Some critical pharmacokinetic parameters to consider for a drug are the time and concentration of its maximal blood levels, its apparent volume of distribution, its rate of clearance and its half-life.


These parameters depend on the processes of drug absorption, distribution, metabolism, and excretion. 

5.1. Absorption of Orally Administered Drugs and the Time and Amount of Maximal Drug Levels in Blood For orally administered drugs, absorption begins with the transport of a drug from the gut to portal blood, continues as the drug passes through the liver, and ends when the drug reaches systemic circulation. If the drug is metabolized by the liver or its passage across the gastrointestinal barrier is incomplete,
 then the drug has reduced bioavailability. Bioavailability is defined as the fraction of intact drug that reaches the systemic circulation relative to the administered dose. With the exception of replacement strategies, such as L-DOPA treatment for Parkinson’s disease, most drugs cannot utilize existing active transport mechanisms utilized by endogenous agents, and consequently, their transport properties are largely determined by passive diffusion across biological barriers. The rate and extent of oral drug absorption depends strongly on the physiochemical characteristics of the drug,

 the formulation state of the drug, and the gastric composition. Because the gut– blood barrier is comprised of cells with lipid membranes and aqueous interiors, the passive transport properties of a drug correlates well with partition coefficient measures of its preference for octanol (a lipophilic environment) over water

(a Pharmacotherapeutic Principles 41 The drug formulation is another factor that can affect absorption of a drug. For example, oral formulations for the antiparkinsonian drug Sinemet® (L-DOPA plus carbidopa) can range from a rapidly absorbed liquid to a slowly absorbed capsule and even more slowly absorbed controlled release tablet.

 For many therapeutic applications it is desirable to gradually increase and then maintain steady blood levels, as rapid rises in blood levels of drugs can desensitize receptor responses or produce significant adverse side effects
(e.g., nausea in the case of DA receptor agonists), and large changes in blood levels can result in fluctuating therapeutic responses. The blood adsorption characteristics of a drug that are usually of most interest are its maximal blood concentration and the time at which this maximum is achieved. 5.2. Distribution of Absorbed Drugs and Apparent Volume of Distribution Distribution is a process involving the exchange of drug in systemic blood with tissues that it comes in contact with as it travels throughout the body.

 Circulating drugs can either remain soluble in the aqueous blood phase or they can be carried by blood components. Usually the carrier components in blood are proteins, but in rare instances, such as for the mood stabilizer sodium valproate, lipids can be the carrier. In many cases, the drug is not very tightly bound to blood components and it will prefer to associate with a tissue with which it comes in contact.

Once the drug has transferred from blood to a tissue it is said to have been distributed or deposited. In certain cases, a drug may bind so tightly to carrier proteins that it cannot readily dissociate and interact with other tissues, and the blood proteins then act as a nontarget tissue deposition sites. Such tightly protein-bound drugs are usually therapeutically inactive in vivo.

Distribution can be a complex process requiring passage across more than one barrier that separates biological compartments. For example, neuropsychiatric drugs must cross the bloodbrain barrier (central nervous system compartment) before they can cross the cellular membrane barriers (cellular compartment) surrounding their target tissues in the brain. Some drugs can redistribute themselves to the periphery once deposited in the brain, but this effect is rarely significant for neuropsychiatric drugs.

 More relevant to the pharmacokinetics of neuropsychiatric drugs is the distribution of antipsychotic and antidepressant drugs into lipophilic stores such as fat. 

The reason for this is that antipsychotic and antidepressant drugs tend to be very lipophilic owing to having a number of aromatic rings. Such antipsychotic or antidepressant drugs can remain intact when stored in fatty tissues, and they can be slowly released over time, which can account for their sometimes long washout period. 

On the other hand, the antimania drug lithium is a very water-soluble elemental ion that distributes in a manner similar to bulk water. Lithium is also unique among neuropsychiatric agents in that it is not protein bound, and it is primarily transported into cells via passage through voltage-dependent sodium channels. 


Once inside cells, lithium is only slowly released, because it does not substitute for sodium for active transport through the sodium-potassium pump. 42 Schetz A useful parameter for describing drug distribution is the volume of distribution (Vd), which is an apparent measure of the accessible space in the body that is available to contain a drug. It can be defined as the ratio of the amount of drug in the body to the concentration present in the aqueous portion of blood (blood water) as shown in Eq. 11. Vd = amount of drug/concentration of drug in blood water

(11) Vd is only an apparent value, because it often does not relate to the real volume of the body. Instead, Vd is an operational definition that relates to a volume that would be required to homogeneously contain drug at the concentration found in blood. Large volumes of distribution indicate that the amount of drug measured in the blood is low as a result of distribution of the remaining drug into various tissues.

Drugs that are not highly bound to blood constituents and that readily distribute into body tissues will have larger volumes of distribution. Note that the Vd values apply to intravenously administered drugs, unless an orally administered drug is completely or almost completely bioavailable, otherwise Vd values for an orally administered drug must be estimated by multiplying Vd by drug bioavailability. 5.3. Termination of Drug Responses A drug response is terminated by excreting the drug from the body or by metabolically inactivating it. The excretion of a drug from the body depends on its clearance.

 Systemic clearance

 is a process by which the portion of drug that it is not metabolized and not protein-bound is removed from systemic circulation by hepatic excretion into the bile and/or by renal excretion into the urine. Renal excretion is common for small or polar drugs.

 For the majority of neuropsychiatric drugs at the doses utilized in clinical settings, the clearance is assumed to be a first-order process and is constant.

Another parameter that describes the elimination of drug is the elimination rate constant (Ke). The constant Ke is the fraction of drug excreted at any instant in time, and it is a function of clearance and volume of distribution as shown in Eq. 12. Ke = systemic clearance/Vd (12) The elimination half-life (t1/2) is the time needed to eliminate half of the drug from the body. The Ke, or its related clearance and Vd values, can be used to estimate the elimination half-life (t1/2) of a drug as shown in Eq. 13. Elimination half-life = t1/2 = ((ln(2))(Vd))/systemic clearance = (ln(2))/Ke (13) The value ln(2) in Eq. 13 is the proportionality constant for the first-order elimination of half of the drug. The elimination half-life value can be utilized to estimate drug-dosing regimens, the time needed to achieve steady-state levels, and the time needed to wash out the drug following the last dose. In general, the time needed to attain steady-state drug levels, or to approximate the drug wash out period is estimated to be greater than five elimination half-lives. Pharmacotherapeutic Principles 43 The dependence of the elimination half-life on Vd is as a result of the fact that only drugs that are in systemic circulation and in contact with organs of elimination (e.g., liver and kidney) can be cleared, although drugs distributed into other tissues cannot. The clearance of many drugs relies on the rate of blood flow to the organs of elimination. In these cases, the functional status of the heart, as a result of age, disease, or drugs that alter cardiac function, can significantly affect clearance, because blood flow rate is altered.

 The functional status of the major organs of elimination, owing to disease or age, for example, can also affect clearance rates, and consequently, the elimination half-life of drugs that are cleared by these organs.

 For example, impaired renal function, which is common in the elderly populations, can essentially double the elimination half-life of lithium as it is primarily cleared by the kidney

(13). Although clearance is often the predominant factor in the termination of responses for drugs with low-molecular weights or significant polarity, most drugs used to treat neuropsychiatric disorders tend to be lipophilic and to have relatively large molecular volumes.

Thus, the majority of such drugs must undergo biotransformation to more polar metabolites before they can be effectively excreted. 

The production of more polar metabolites can occur by enzymatic reactions that either induce or unmasked polar functional groups (phase I reactions), or that conjugate endogenous polar groups like sugars and polar amino acids (phase II reactions), or both. For example, desimipramine metabolism involves hydroxylation followed by glucuronidation.

Although many drug metabolites are biologically inactive, some retain activity or have modified activity. A variety of drugs used to treat neuropsychiatric disorders have active metabolites. For example, desimipramine is an active metabolite of the tricyclic antidepressant imipramine, and norfluoxetine is an active metabolite of the selective serotonin reuptake inhibitor fluoxetine; in both cases the metabolites have the same targets as the parent drug.

 In other cases, the activity profile of the metabolites is significantly different from the parent drug. For example, buproprion selectively blocks the DA transporter over the norepinephrine (NE) transporter, although one of its hydroxylated metabolites gains significant affinity for the NE transporter (14,15). In another example, the antipsychotic drug loxapine is metabolized to the antidepressant amoxapine, which converts it from a D2 DA receptor-blocking drug to a drug with significantly more norepinephrine transport blocking activity. Consequently, the metabolism of drugs can either terminate their actions, by forming inactive metabolites, or when active metabolites are formed, metabolism can be an underlying reason for their unique pharmacological effects. 6. RECEPTOR RESPONSIVENESS AND TIME OF ONSET OF THE THERAPEUTIC ACTIONS OF DRUGS The relationship between drug concentration and functional effect described in the sections above is for a single challenge of drug at a naïve receptor. Following prolonged or repeated occupancy, most receptors undergo changes in responsive- 44 Schetz ness or density that protects them from excessive stimulation or blockade. Such adaptive responses to the repeated application of drugs can have significant consequences with respect to their actions. For example, attenuated responsiveness may limit the effective therapeutic use of a drug, it may result in tolerance to side effects, or it may be the underlying cause for their effectiveness.


 Persistent activation as a result of persistent receptor occupancy by agonists leads to a reduction in receptor responsiveness. In the case of G protein-coupled receptors (GPCR), such as DA receptors, NE receptors, and most serotonin receptors, attenuated responsiveness is characterized by three types of temporally and mechanistically distinct adaptive processes (16). Persistent receptor stimulation by acutely administered agonists results in GPCR desensitization followed by internalization. Receptor desensitization is the result of an uncoupling of the GPCRs from their G proteins. This uncoupling involves a phosphorylation-dependent (e.g., by kinases) blocking by cytoplasmic accessory proteins (e.g., arrestins) of intracellular portions of the GPCR that interact with G proteins (e.g., the intracellular loops and cytoplasmic tail).

Desensitized receptors then undergo internalization whereby GPCRs are redistributed from plasma membranes to intracellular membranes via endocytosis. In some case, the internalized receptors are resensitized by dephosphorylation in clathrin-coated vesicles and recycled back to the plasma membrane.


Under conditions of chronic stimulation, internalized GPCRs are not resensitized; rather, they are downregulated, which leads to a reduction in receptor density owing to proteolytic degradation. In some cases, chronic stimulation is additionally associated with a reduction in the amount of newly synthesized receptor. Although most GPCRs display attenuated responsiveness following persistent activation, the rate and extent of this effect can vary considerably depending on the receptor subtype and drug pharmacokinetics. In contrast to persistent activation, persistent blockade of GPCRs can lead to receptor supersensitivity or receptor upregulation. Neurotransmitter transporters and metabolic enzymes can also display changes in responsiveness as a result of persistent occupancy, but the details of the molecular mechanisms are distinct from those described for GPCRs (17,18). The general expectation is that the onset of drug action will be a function of how long it takes for a drug to reach its target tissue and then act on its receptor, which in most cases is rapid. For instance, intravenous bolus injection of the appropriate dose of phenobarbital into the tail vein of a rat produces sedation in less than 1 minute. However, the rate of onset of the therapeutic actions of drugs used to treat neuropsychiatric disorders can vary considerably. The anti-attention deficit hyperactivity disorder effect of psychostimulants, like D-amphetamine and methylphenidate, produce dramatic changes in behavior that closely parallels the expected dose–effect relationship. In contrast, the onset of action of chronically administered antipsychotic or antidepressant drugs can be much longer, requiring weeks for a full therapeutic effect to be achieved. In these cases, the large disparity between the expected and actual time course of the therapeutic effect implies that clinical efficacy is not as a result of acute effects on the target receptor; rather, it is because of chronic compensatory changes in the target receptor (e.g., up- or downregulation of recep- Pharmacotherapeutic Principles 45 tor density) or some other receptor system whose function is linked to the target receptors. For instance, the therapeutic effect of chronic antidepressant treatment may be as a result of desensitization of presynaptic autoreceptors, such as somatodendritic serotonin 5-HT1A receptors (19) or terminal serotonin 5-HT1B receptors (20), and/or downregulation of serotonin transporters (21). The end result of each of these effects is an increase in the level of synaptic serotonin. A chronic elevation in synaptic serotonin could be signaling changes in the levels of nuclear transcription factors, which then regulate the expression of genes related to neurotransmission, and this might also account for the delay between the onset of drug treatments and their therapeutic effect.



7. THE MEANING OF DRUG SELECTIVITY 




When the term “selectivity” is used to describe a drug it can take on a variety of contextual meanings. Selective effects of drugs can be as a result of differences in potency, efficacy, or pharmacokinetic accessibility.


 However, drug selectivity usually refers to the binding affinity for one receptor (or a subfamily of receptors) over others.

 The most important factors to consider are the relative frame of reference and the magnitude of the drug selectivity. Although it may be possible to accurately measure a fivefold difference in the affinity for one drug over another in isolated tissue fractions or when dealing with cloned receptor systems, for whole tissue in vitro or in vivo work, in which a large number of potential receptor sites are available, at least a 200-fold difference in affinity is usually required to elicit a truly selective response.



 A selectivity window of this size allows for dosing that will result in a maximal occupancy of the intended receptor target with little or no occupancy at nontarget receptors. An important caveat with respect to drug selectivity is that the selectivity of any drug may be difficult to rigorously define,


 because it is not feasible to screen all known related receptor sites and a drug may bind to receptor sites that have yet to be discovered or pharmacologically characterized.

The term “frame of reference” refers to the number of competing targets for a particular drug. For instance, a compound like NGD 94-1 has an affinity that is over 500-fold higher for the D4 subtype of DA receptor than for any of the other DA receptor subtypes (D1, D2, D3, and D5).

It also has over a 500-fold higher affinity for the D4 receptor than for other GPCRs (e.g., serotonin, sigma, and adrenergic receptors) for which it has been evaluated (22). Thus, within the frame of reference of receptor sites that were tested, it can be said that NGD 94-1 is a DA D4 receptorselective drug. However, drugs this selective for a particular receptor subtype are not available for many key receptor systems.

The antipsychotic haloperidol is a more prototypical example as it binds with high affinity to cloned D2, D3, and D4 receptors (Ki = 1.2, 4.1, and 1.6 nM, respectively). Because haloperidol has less than a four-fold lower affinity for the D3 subtype, its in vivo selectivity over the D2 and D4 subtypes is negligible.

 However, if the comparison is expanded to include the entire DA family of receptors, then it can be said that haloperidol is D2-like selective, as it binds with higher affinity to all members of the D2-like subfamily 46 Schetz (i.e., D2, D3, D4) than to the D1-subfamily (D1 and D5, Ki = 63 and 124 nM, respectively). If our frame of reference is among serotonin 5-HT1A, 5-HT2A, and 5-HT2C receptors (Ki = 2425, 54, and 4475 nM, respectively),

 then haloperidol can be said to be 5-HT2A selective. If we extend our frame of reference to include both these serotonin receptor subtypes and the entire family of DA receptors, then haloperidol’s selectivity can be said to be mixed and would thus more accurately be defined as being 5-HT2A/D2-like receptor selective.

For these reasons, quantitative in vitro tissue or in vivo studies often must be interpreted with caution, especially if one neglects to selectively block, with other selective drugs, known sites that are not of interest.

For instance, low concentrations of the high-affinity serotonin receptor selective antagonist mianserin and the high-affinity D1-like selective antagonist SCH23390 could be added to block 5-HT2A and D1-like sites in brain tissue when using [3H]haloperidol as a radioligand to detect D2-like sites. By analogy, in vivo chemical lesioning with the neurotoxin 6-hydroxydopamine (6-OH) is usually performed in the present of a norepinephrine transporter inhibitor, like imipramine, to permit uptake (via catecholamine transporters) into dopaminergic, but not noradrenergic neurons.



 8. TARGETED     PHARMACOTHERAPEUTIC MANAGEMENT OF SELECTED   SYMPTOM   MODALITIES 


Drugs that target dopaminergic and serotonergic, and to a lesser extent noradrenergic systems, are the ones most often encountered in the pharmacotherapeutic management of the neurological and psychiatric disorders discussed throughout the following chapters. This may seem odd given the vast array of unique clinical symptoms observed for the different neuropsychiatric disorders, but sense can be made of this by realizing that the pharmacotherapies for neuropsychiatric disorders are largely palliative, and usually, they are designed to provide relief for only one of a range of symptom modalities encountered for each disorder. 

For example,

antipsychotic drugs are prescribed for the treatment of disorders as divergent as autism, Tourette’s syndrome, and schizophrenia, but their application is designed to alleviate different symptoms associated with each: 

aggression and self-injurious behavior for autism, repetitive motor behaviors for Tourette’s, and psychosis for schizophrenia. Utilization of similar treatments for different neuropsychiatric symptom modalities is thus possible because of the key roles that the various dopaminergic pathways play in the modulation of a range of cognitive and motor functions.





 ACKNOWLEDGMENTS The author thanks Drs. Michael Oglesby, Robert Luedtke, and Anna Ratka for insightful discussions and helpful comments. This work was supported in part by grant R01 MH063162-01 awarded to J.A.S. Pharmacotherapeutic Principles 47 REFERENCES 1. Jones BJ, Blackburn TP. The medical benefit of 5-HT research. Pharmacol Biochem Behav. 2002;71:555–568. 2. Xu Y, Ito A, Arai R. Immunohistochemical localization of monoamine oxidase type B in the taste bud of the rat. Neurotoxicology. 2004;25:149–154. 3. Mannisto PT, Kaakkola S. Catechol-O-methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors. Pharmacol Rev. 1999;51:593–628. 4. Hall DA, Strange PG. Evidence that antipsychotic drugs are inverse agonists at D2 dopamine receptors. Br J Pharmacol. 1997;121:731–736. 5. Wilson J, Lin H, Fu D, Javitch JA, Strange PG. Mechanisms of inverse agonism of antipsychotic drugs at the D(2) dopamine receptor: use of a mutant D(2) dopamine receptor that adopts the activated conformation. J Neurochem. 2001;77:493–504. 6. Weiner DM, Burstein ES, Nash N, et al. 5-hydroxytryptamine2A receptor inverse agonists as antipsychotics. J Pharmacol Exp Ther 2001;299:268–276. 7. Schetz JA. Allosteric modulation of dopamine receptors. Mini-review. Med Chem. 2004; in press. 8. Limbird LL. Identification of receptors using direct radioligand binding techniques. In: Cell Surface Receptors: A short course on Theory and Methods. Martinus Nijhoff Publishing, 1986:51–96. 9. Cheng Y-C, Prusoff WH. Relationship between the inhibition constant (Ki ) and the concentration of inhibitor which causes 50 percent inhibition (IC50) of an enzymatic reaction. Biochem Pharmacol. 1973;22:3099–3108. 10. Ehlert FJ. Estimation of the affinities of allosteric ligands using radioligand binding and pharmacological null methods. Mole Pharmacol. 1988;33:187–194. 11. Christopoulos A, Kenakin T. G protein-coupled receptor allosterism and complexing. Pharmacol Rev. 2002;54:323–374. 12. Ariens EJ. Affinity and intrinsic activity in the theory of competitive inhibition. Arch Int Pharmacodyn Ther. 1954;99:32–49. 13. Ritschel WA. Pharmacokinetics in the aged. In: Pagliaro LA and Pagliaro AM, eds. Pharmacologic aspects of aging. Mosby, 1983. 14. Ascher JA, Cole JO, Colin, J-N, et al. Bupropion: a review of its mechanism of antidepressant activity. J Clin Psychiatry. 1995;56:395–401 15. Bondarev ML, Bondareva TS, Young R, Glennon RA. Behavioral and biochemical investigations of bupropion metabolites. Eur J Pharmacol. 2003;474:85–93. 16. Tsao P, Cao T, von Zastrow M. Role of endocytosis in mediating downregulation of G-proteincoupled receptors. Trends Pharmacol Sci. 2001;22:91–96. 17. Torres GE, Gainetdinov RR, Caron MG. Plasma membrane monoamine transporters: structure, regulation and function. Nat Rev Neurosci. 2003;4:13–25. 18. Kumer SC, Vrana KE. Intricate regulation of tyrosine hydroxylase activity and gene expression. J. Neurochem. 1996;67:443–462. 19. Hensler JG. Regulation of 5-HT1A receptor function in brain following agonist or antidepressant administration. Life Sci. 2003;72:1665–1682. 20. Blier P. Pharmacology of rapid-onset antidepressant treatment strategies. J Clin Psychiatry. 2001;62 Suppl 15:12–17. 21. Benmansour S, Owens WA, Cecchi M, Morilak DA, Frazer A. Serotonin clearance in vivo is altered to a greater extent by antidepressant-induced downregulation of the serotonin transporter than by acute blockade of this transporter. J Neurosci. 2002;22:6766–6772. 22. Tallman JF, Primus RJ, Brodbeck R, et al. NGD 94-1: identification of a novel, high-affinity antagonist at the human dopamine D4 receptor. J Pharmacol Exp Ther. 1997;282

Fundamentals of how we choose stimulant or depressant Drug Therapy in Neurological disorders and diseased

Fundamentals of Drug Therapy in Neurology


 fundamentals of drug therapy in neurology provides a readable synopsis of how the classic pharmacology concepts such as pharmacodynamics and pharmacokinetics are related to therapy in neurology. This section also discusses the pathogenesis of potentially serious side effects such as skin rashes and liver disorders, and the role of pharmacogenetics in both drug effect and side effects.



 Overview



Physical therapists and occupational therapists are non-prescribing healthcare practitioners. You may be asking yourself, "Why do I need to know this?" If that is your mindset, I hope that we can help you to see why it's important that you become competent with basic pharmacological principles. In all 50 states, physical therapists have autonomous practice ability. Some states won't allow long-term treatment without a physician referral, but they will allow evaluation. With autonomous practice, you have a professional responsibility to screen your patients' medications and be able to point out potential issues with those medications.
Additionally, medications significantly influence your patients' ability to participate in physical therapy. When I was beginning in home health, I had been a stay-at-home mom for several years. I was working in a patient's home, and I called their physician because I was concerned that my patient had a very low heart rate. It turns out the patient was on beta blockers and I had no idea that was why their heart rate had decreased. That's one of the reasons why I decided to become a champion for physical therapists to be able to understand and implement knowledge of medication into their practice.
Medicines can significantly influence a patient's ability to participate in therapy. Conversely, therapy can significantly influence the action of certain medications. Sometimes, therapy can hasten medication metabolism. Sometimes it can slow it. Sometimes it can impact how it is metabolized. As therapists, we need to be aware of that. If we have knowledge in that area, we can optimize both the pharmacological intervention and the physical therapy intervention.
First, I will provide a brief overview of pharmacology. Next, we will discuss the most common ways in which medications are used for central nervous system disorders. We will take a look at some case examples of patient problems to apply our knowledge and discuss as a group. I will try to point out some commonalities among diagnoses and medications. Finally, I will show you how to find reliable websites that provide accurate drug information.

Pharmacology Review

Pharmacology is the study of medications. Pharmacology can be divided into two subcategories: pharmacotherapeutics and toxicology. Pharmacotherapeutics is the study of the beneficial aspects of medication. Toxicology is the study of the harmful aspects of medication, those which may even lead to death. For the purposes of today's presentation, we will solely be looking at pharmacotherapeutics, which can be divided further into two more categories: pharmacokinetics and pharmacodynamics.
Pharmacokinetics is the study of what the body does to the drug. The drug is administered, and then it is absorbed into the bloodstream. It's distributed into certain systems within the body. It is metabolized and eventually excreted. The body is trying to get rid of the drug from the moment it's administered.
Pharmacodynamics is the study of what the drug does to the body. The drug exerts an effect on the cellular level, and the combined cellular level effects cause a systemic effect that can exert an influence throughout the body.
The primary mechanism of action is the means by which a drug produces an alteration in function. That usually occurs at a cellular level, with some type of interaction with a cellular receptor. The drug binds to a cellular receptor and causes a biochemical reaction that alters the cell function. If you remember back to physiology class, we have a lipophilic bipolar cell membrane, and embedded in the membrane are glycoproteins. The glycoproteins have communication with the extracellular space and also within the cell. Often, neurotransmitters will bind to these receptors and they cause some change. The drug can also bind to the receptor to cause some action. 
To reiterate and summarize pharmacotherapeutics, it is helpful to look at the following flowchart. On the upper half of Figure 1, we can see the process of pharmacokinetics: what the body does to the drug. First, a dose of a drug is administered either enterally (i.e., by mouth) or parenterally (i.e., going around the digestive system). Parenteral administration is given through an IV or via some intramuscular or subcutaneous method. After administration, the drug is absorbed and eventually makes its way into the bloodstream. After absorption, we want the medication to be distributed to particular tissues so that it can have some pharmacological effect. As this distribution is going on, the drug is also being metabolized and excreted. This administration, absorption, distribution, metabolism, and elimination is all part of pharmacokinetics.
Figure 1. Processes of pharmacokinetics and pharmacodynamics.
The bottom portion of Figure 1 shows the process of pharmacodynamics: what the drug does to the body. When the drug reaches the site of action in the body in a sufficient concentration, it should have a pharmacological effect and provide some clinical response. For example, if I administered a medication for high blood pressure, the clinical response would be lowered blood pressure. Such a response as lowered blood pressure would be an efficacious response to that medication. Hopefully, our medications have a high level of efficacy and give the desired response, but medications are known to also give side effects and potentially toxic effects. That entire process is pharmacodynamics.
There are many different drugs for neurological disorders. I've tried to provide a structure for you to look at these drugs. If you encounter an unfamiliar drug, you can refer back to this structure, so you can compare and contrast the new drug with the drugs that you're familiar with.
Drugs are commonly given for central nervous system disorders for one of four purposes:
  1. To minimize secondary damage acutely, immediately after a traumatic event, such as:
    • Spinal cord injury (SCI)
    • Ischemic cerebrovascular accident (CVA)
    • Traumatic brain injury (TBI)
  2. To manipulate neurotransmission (either to increase or decrease neurotransmission) in cases such as:
    • Parkinson's disease (PD)
    • Alzheimer's disease (AD)
    • Psychiatric diseases
  3. To try to slow the disease progression, in cases such as:
    • Multiple sclerosis (MS)
    • Amyotrophic lateral sclerosis (ALS)
  4. To minimize signs/symptoms and secondary problems that may develop with neurological disorders:
    • Hypertone/spasticity

Minimize Secondary Damage Following Acute Event

The central nervous system is composed of the brain and the spinal cord. Everything else, including the autonomic nervous system, is considered part of the peripheral nervous system. An acute event, usually traumatic, can occur in the central nervous system, the most common of which is an ischemic cerebrovascular accident. The next most common type is a traumatic brain injury, followed by traumatic spinal cord injury.
An acute event occurs that causes damage somewhere in the central nervous system. The damage is not ongoing. It's a static event that causes primary cell death and some secondary damage, but it's not progressive. You could contrast that to Parkinson's disease, which is a progressive disease (i.e., the damage keeps occurring). The central nervous system disorders that occur following an acute event usually are more of a static lesion. It occurs once; it's not going to keep occurring over time.
After a central nervous system injury, there is a series of events that occur (Figure 2). If a person receives a blow to the head, for example, that will result in a central core of cell death. That person will suffer primary cell death. In addition to that primary cell death, those neurons that are in communication with those now dead cells are very susceptible to secondary injury. The areas adjacent to initial injury that may die secondarily is called the penumbra. In fact, any cell that communicates with that core area is susceptible to later cell death. Any cell, whether it's in the other hemisphere of the brain or if it's further down in the spinal cord, is susceptible to cell death. Those interconnected areas that may eventually die are called the diaschisis.
Figure 2. Events after a CNS injury.
Even if the initial area of core cell death is relatively small, that area grows over time. The penumbra is the area that's adjacent to the initial injury; because of the interconnectivity in the central nervous system, a significant lesion may grow from the initial central core death. This is not only true in the brain, but also true in a spinal cord injury. A traumatic injury may cause a central core, but over time (hours or days), that lesion could grow significantly.
The traumatic event causes an area where some cells are not getting the oxygen that they need. The trauma also causes the release of excitatory amino acids, which activate receptors that cause a major influx of calcium. All of this leads to excitotoxicity. A further cascade of events occurs that lead to inflammation in the area of injury, all of which leads to further cell death through necrotic pathways and apoptotic cell death, which causes an increase in the lesion's size. As the lesion size increases, it leads to an increase in the patient's functional deficits. You might start with a small lesion and end up with a very large lesion and a lot of functional deficits.
The pharmacological management after these central nervous system acute injuries is to try to decrease the excitotoxicity. If we can decrease this excitotoxicity and decrease the inflammation, we won't cause growth in the lesion and we'll have fewer functional deficits. That's the pharmacological premise in trying to give drugs that will help to decrease that secondary cell death.
Next, we will review some different diagnoses and take a look at some of the medications that are used for each diagnosis.

Acute Spinal Cord Injury 

Research conducted in the 1990s by Bracken, et al in the New England Journal of Medicine looked at using a glucocorticoid (a steroid) in the acute management of spinal cord injury. They particularly looked at methylprednisolone (MP). This study showed such a significant improvement in those patients who were acutely given MP that they stopped the clinical trial, and this was implemented across the country as the standard procedure of giving a high dosage bolus IV of MP, followed by an infusion over the next 23 hours. It was found that this significantly decreased the inflammatory effects and decreased the secondary cell death. They also found that this must be given very early. The maximum window of opportunity is within the first eight hours after a spinal cord injury.

Acute Traumatic Brain Injury 

Initially, glucocorticoids and MP were also considered for use with cases of acute TBI. Although it showed great promise in animal models, it led to a higher mortality rate in humans. The appropriate pharmacological mechanism to prevent secondary damage after acute TBI is a little bit different than that of acute SCI. After a TBI, the two main pharmacological goals are to maintain an optimal blood pressure and to normalize intracranial pressure. After a TBI, you don't want the blood pressure hypertensive (too high), but it's equally if not more important to not have the blood pressure get too low. If the blood pressure is too low, the brain is not getting the perfusion that it needs, leading to increased cell death. Research has shown that for every 10-point increase in systolic blood pressure, there is a decrease of almost 20% in the adjusted odds of death. It's very important to maintain normal blood pressure so that we can have appropriate perfusion in the brain. The most common medication used for that purpose is a sympathomimetic drug, phenylephrine HCL, which actually increases blood pressure. For those patients whose blood pressure is low, they will administer this medication. It increases peripheral vascular resistance in the periphery, but it does not do that in the blood vessels, and so we get better perfusion in the brain. It's administered intramuscularly or through IV.
With acute TBI, we also want to maintain normal intracranial pressure (ICP). Due to the nature of a TBI, an increase of intracranial pressure is very common, which can result in significantly further brain damage. The two agents that are most commonly used to control ICP are osmotic agents and barbiturates. Mannitol is the gold standard for normalizing ICP, keeping it at less than 15 mm Hg. Mannitol is an osmotic diuretic that has an effect on intracranial pressure. It enhances cerebral blood flow and brain metabolism. Initially, it can be a good medication to lower intracranial pressure. Use of Mannitol does require an intact blood-brain barrier. As such, it may not be appropriate to use with someone who has sustained significant head trauma. One of the side effects of Mannitol is pulmonary edema, which can later predispose the patient to pneumonia, hypotension and renal injury, as it is metabolized through the kidneys.
It should be noted that there has not been a large randomized controlled trial of Mannitol against a placebo because to do so would be to withhold appropriate treatment for patients with TBI.
Barbiturates are another common classification of medication used to lower the intracranial pressure, by altering the vascular tone. Barbiturates are a potent GABA facilitator. GABA is an inhibitory neurotransmitter. By inhibiting excitatory neurotransmission, barbiturates have the effect of suppressing brain activity. Suppression of brain activity will also lead to sedation, amnesia, and loss of consciousness. As such, barbiturates are used to initiate a medically induced coma. There are two medications (Pentobarbital and Secobarbital) that are frequently used for this purpose. Pentobarbital is short-acting and Secobarbital is medium-acting. Once these barbiturates do get into the system, it takes quite a while for them to be cleared, and for a patient to wake up from a medically induced coma.

Acute Ischemic Stroke 

First, I want to differentiate between an ischemic stroke and a hemorrhagic stroke. An ischemic stroke occurs as a result of an obstruction within a blood vessel supplying blood to the brain. If no blood can get through, there is a hypoxic insult and some brain tissue dies. With a hemorrhagic stroke, a weakened blood vessel ruptures, resulting in hypoperfusion. Blood spills into or around the brain and creates swelling and pressure, damaging cells and tissue in the brain. The treatment for ischemic stroke is very different from hemorrhagic stroke. In fact, treatment for an ischemic stroke, if given to someone who is having a hemorrhagic stroke, will usually be fatal. The first step in the emergency room is to differentiate whether the patient is having an ischemic stroke or a hemorrhagic stroke.
For an ischemic stroke, blood pressure control is very similar to TBI, except that patients with an ischemic stroke often present with hypertension and not low blood pressure. Secondarily, they want to break down any clot that is causing the blood vessel occlusion using fibrinolytic drugs, and then give anticoagulants to prevent further damage. By doing that, that will help us to save and prevent secondary damage and save that penumbra and the diaschisis, so the patient has minimal or no functional deficits.
Fibrinolytic agents (commonly known as "clot busters") need to be administered within three hours of symptom onset. There are two fibrinolytic agents that are used frequently: streptokinase and tissue plasminogen activator (tPA). Tissue plasminogen activator is the more expensive of the two drugs. Streptokinase is a lot less expensive, however, it has a shorter shelf life. Hospitals have found that if streptokinase sits on their shelf, it expires and has to be discarded. tPA has a longer shelf life. Even though it's more expensive, they don't need to waste it as often. For that reason, more facilities use tPA than use streptokinase. The mechanism of action of both of these drugs is to activate plasminogen bound to fibrin. It breaks up a clot, which is what's usually causing the occlusion in the blood vessel. The fibrinolytic agents can be given through an IV. They also have been given directly in the brain, which requires a specialized facility.
The second line of defense to minimize secondary damage is to prevent further clots from forming, and that can be done through the use of anticoagulants. One of the most common anticoagulants administered parenterally (through an IV) is heparin. The most common drugs administered subcutaneously are Lovenox and Arixtra. Orally, the most common anticoagulant drug used is a vitamin K antagonist known generically as warfarin, or under the brand name, Coumadin.

Rehab Implications

Medications that are given acutely to prevent secondary damage do have some rehab implications. Acutely, you probably won't be working with a patient until they're deemed medically stable. If you are a working with a patient who may not be medically stable, you likely will be doing some passive, dependent activities (e.g., turning, maintaining mobility, working with respiration, preventing skin breakdowns).



The appropriate, effective, and safe utilization of drugs in the treatment of disease requires a basic understanding of the dose–effect relationships of medications. Relating dose to effect requires a combined appreciation of pharmacodynamic concentration–effect relationships, or what drugs do to the body, and of pharmacokinetic dose–concentration relationships, or what the body does to or with drugs. For this reason, considerable attention is devoted to both the pharmacodynamic and pharmacokinetic aspects of drug therapies. The aim of this chapter is to provide a theoretical rationale that is necessary for appropriately interpreting the results of basic and clinical neuropharmacology studies and for understanding many of the drug treatment strategies commonly encountered in clinical neurology and psychiatry. Approximately one-fourth of all drugs prescribed worldwide exert their therapeutic actions on CNS targets. Of the top five selling drugs in this category, three are antidepressants and two are atypical antipsychotics (1). The relative success of pharmacological intervention is highlighted further when one considers that these

neurological disorders continues to expand steadily and the aim of this book is to provide a perspective for neurologists, psychiatrists, and other clinicians on how they can be used effectively. This perspective includes information about the pathophysiology of disease as well as drug interactions with the nervous system and the rest of the body. This introductory chapter deals with the fundamental concepts that are common to more specific treatment issues covered in detail in the rest of the book (Table 1–1). First we discuss some major molecular targets of drug action in the nervous system, an area that has expanded greatly as a result of the use of molecular genetic and electrophysiological techniques. The interaction of drugs with these targets is generally known as pharmacodynamics (PD) or the study of mechanisms of action and the relationship between drug concentration and effect. Then we discuss the absorption, distribution, metabolism and elimination of drugs, which determine their pharmacokinetics (PK). Taken together, a drug’s PD and PK characteristics define its therapeutic window or the range of dose and/ or blood level at which it has optimal effect with minimal side effects. For many drugs these characteristics are under genetic control and this is the major focus of the field of 


note

pharmacogenetics. Genetic variation in the distribution and metabolism of drugs as well as in targets of drug action such as ion channels and neurotransmitter receptors are being recognized at an increasing rate. Pharmacogenetic factors can influence the dose of a drug required for treatment of individual patients, as well as its efficacy if a mutation is present in its molecular target of action. Another important factor that strongly influences drug therapy in neurology is the toxicity of these drugs. Because they target the nervous system, these drugs are prone to produce potentially disabling effects on cognition and other brain functions and this often influences the choice of drugs for individual patients. Finally we discuss the influence of ageon effects of nervous system drugs, especially on fetuses, pregnant women, and elderly individuals.



CLASSIFICATION OF DRUGS ON THE BASIS OF THE RESPONSES THEY PRODUCE ON THEIR RECEPTORS

 When a drug reversibly binds to the orthosteric (primary) site on its receptor one of four outcomes are to be expected: the receptor becomes activated, the receptor becomes partly activated, the receptor becomes inactivated, or the receptor is unable to be activated. Consequently, drugs are generally classified based on their actions. A drug is an agonist if it fully activates receptors, a partial agonist if it partly activates receptors, an inverse agonist if it inactivates receptors (and prevents them from being activated), or an antagonist if it only prevents receptors from being activated. For instance, the endogenous neurotransmitter DA is an (full) agonist of DA receptors, and the antiparkinsonian drug bromocriptine is a partial agonist at D2 receptors. Antipsychotics like haloperidol and clozapine may be inverse agonists of D2 DA receptors (4,5), whereas L-741,626 is an (neutral) antagonist. Receptor activation is a thermodynamic process, whereby agonist binding induces a conformational change in the receptor and converts it from the inactivated state (agonist low-affinity binding state) to the activated state (agonist highaffinity binding state). Typically, neutral antagonist binding is indifferent to the conformational (affinity) state of its receptor, because it must only occupy the orthosteric site rather than occupy and then induce a change in it. However, an inverse agonist is a special type of antagonist in that when it binds to a receptor in the activated state it converts it to the inactivated state. For this reason, inverse agonists reduce the basal levels of constitutive receptor activity, which corresponds to the (typically small) proportion of receptors that are in the activated state in the absence of agonist. Such distinctions in the molecular mechanisms of action of antipsychotic drugs that act on D2-like dopamine and 5-hydroxytryptamine (5-HT)2- like serotonin receptors may be critical to understanding their unique therapeutic profiles (4,6). Although most drugs bind directly to the orthosteric site of the receptor, other drugs bind at another (secondary) receptor site, called an allosteric site. Ligands that bind to the allosteric site are known as allosteric modulators, because they indirectly modulate the binding of primary ligands to the orthosteric site by remotely altering the orthosteric-binding site. The modulation is said to be positive if the modulator facilitates a primary ligand’s interaction with the primary site or negative if the modulator attenuates its interaction with the primary site. The extent to which the allosteric site and the orthosteric site are coupled, or their cooperativity, can be weak or strong. Noncompetitive interactions, which result in a complete occlusion of the orthosteric site leading only to a decrease in the maximum density of sites with no change in affinity, are also allosteric in nature but they are a special case of neutral cooperativity. Within the DA receptor family, for example, a diverse





Central Nervous System (CNS) Depressants and Stimulants

Prescription Drugs and Pain Medications: Part 2 of 3

Central Nervous System (CNS) Depressants 
CNS depressants slow normal brain function. In higher doses, some CNS depressants can become general anesthetics. Tranquilizers and sedatives are examples of CNS depressants. CNS depressants can be divided into two groups, based on their chemistry and pharmacology:Brain IllustrationBarbiturates, such as mephobarbital (Mebaral) and pentobarbitalsodium (Nembutal), which are used to treat anxiety, tension, and sleep disorders.
Benzodiazepines, such as diazepam (Valium), chlordiazepoxide HCl (Librium), and alprazolam (Xanax), which can be prescribed to treat anxiety, acute stress reactions, and panic attacks. Benzodiazepines that have a more sedating effect, such as estazolam (ProSom), can be prescribed for short-term treatment of sleep disorders.
There are many CNS depressants, and most act on the brain similarly—they affect the neurotransmitter gamma-aminobutyric acid (GABA). Neurotransmitters are brain chemicals that facilitate communication between brain cells. GABA works by decreasing brain activity. Although different classes of CNS depressants work in unique ways, ultimately it is their ability to increase GABA activity that produces a drowsy or calming effect. Despite these beneficial effects for people suffering from anxiety or sleep disorders, barbiturates and benzodiazepines can be addictive and should be used only as prescribed.
CNS depressants should not be combined with any medication or substance that causes sleepiness, including prescription pain medicines, certain over-the-counter cold and allergy medications, or alcohol. If combined, they can slow breathing, or slow both the heart and respiration, which can be fatal.
Discontinuing prolonged use of high doses of CNS depressants can lead to withdrawal. Because they work by slowing the brain’s activity, a potential consequence of abuse is that when one stops taking a CNS depressant, the brain’s activity can rebound to the point that seizures can occur. Someone thinking about ending their use of a CNS depressant, or who has stopped and is suffering withdrawal, should speak with a physician and seek medical treatment.
In addition to medical supervision, counseling in an in-patient or out-patient setting can help people who are overcoming addiction to CNS depressants. For example, cognitive-behavioral therapy has been used successfully to help individuals in treatment for abuse of benzodiazepines. This type of therapy focuses on modifying a patient’s thinking, expectations, and behaviors while simultaneously increasing their skills for coping with various life stressors.
Often the abuse of CNS depressants occurs in conjunction with the abuse of another substance or drug, such as alcohol or cocaine. In these cases of polydrug abuse, the treatment approach should address the multiple addictions.
Stimulants 
Stimulants increase alertness, attention, and energy, which are accompanied by increases in blood pressure, heart rate, and respiration.
Historically, stimulants were used to treat asthma and other respiratory problems, obesity, neurological disorders, and a variety of other ailments. As their potential for abuse and addiction became apparent, the use of stimulants began to wane. Now, stimulants are prescribed for treating only a few health conditions, including narcolepsy, attention-deficit hyperactivity disorder (ADHD), and depression that has not responded to other treatments. Stimulants may also be used for short-term treatment of obesity and for patients with asthma.
Stimulants such as dextroamphetamine (Dexedrine) and methylphenidate (Ritalin) have chemical structures that are similar to key brain neurotransmitters called monoamines, which include norepinephrine and dopamine. Stimulants increase the levels of these chemicals in the brain and body. This, in turn, increases blood pressure and heart rate, constricts blood vessels, increases blood glucose, and opens up the pathways of the respiratory system. In addition, the increase in dopamine is associated with a sense of euphoria that can accompany the use of stimulants.
Research indicates that people with ADHD do not become addicted to stimulant medications, such as Ritalin, when taken in the form and dosage prescribed.1 However, when misused, stimulants can be addictive.
The consequences of stimulant abuse can be extremely dangerous. Taking high doses of a stimulant can result in an irregular heartbeat, dangerously high body temperatures, and/or the potential for cardiovascular failure or seizures. Taking high doses of some stimulants repeatedly over a short period of time can lead to hostility or feelings of paranoia in some individuals.
Stimulants should not be mixed with antidepressants or over-the-counter cold medicines containing decongestants. Antidepressants may enhance the effects of a stimulant, and stimulants in combination with decongestants may cause blood pressure to become dangerously high or lead to irregular heart rhythms.
Treatment of addiction to prescription stimulants, such as methylphenidate and amphetamines, is based on behavioral therapies proven effective for treating cocaine or methamphetamine addiction. At this time, there are no proven medications for the treatment of stimulant addiction. Antidepressants, however, may be used to manage the symptoms of depression that can accompany early abstinence from stimulants.
Depending on the patient’s situation, the first step in treating prescription stimulant addiction may be to slowly decrease the drug’s dose and attempt to treat withdrawal symptoms. This process of detoxification could then be followed with one of many behavioral therapies. Contingency management, for example, improves treatment outcomes by enabling patients to earn vouchers for drug-free urine tests; the vouchers can be exchanged for items that promote healthy living. Cognitive-behavioral therapies, which teach patients skills to recognize risky situations, avoid drug use, and cope more effectively with problems, are proving beneficial. Recovery support groups may also be effective in conjunction with a behavioral therapy.
References:
1 Nora Volkow, et al., Dopamine Transporter Occupancies in the Human Brain Induced by Therapeutic Doses of Oral Methylphenidate, Am J Psychiatry 155:1325–1331, October 1998.
National Institute on Drug Abuse (NIDA)
National Institutes of Health (NIH)
U.S. Department of Health & Human Services
For more information on addiction to prescription medications, visit http://www.drugabuse.gov/drugpages/prescription.html.










Classifying drugs by their effect on the central nervous system

 Page last updated: 2004
Drugs can be classified in many ways. For example, they can be classified according to:
  • uses (medicinal or recreational)
  • effect on the body (the specific effect on the central nervous system)
  • source of the substance (synthetic or plant)
  • legal status (legal/illegal)
  • risk status (dangerous/safe).
One of the most common and useful ways of classifying a drug is by the effect that it has on a person's central nervous system. The brain is the major part of the central nervous system, and this is where psycho-active drugs have their main effect.

The below sub-section summarises the major classifications of drugs including stimulants, depressants and hallucinogens. The group 'others' includes those psycho-active drugs that do not fit neatly in any other category. Some drugs can be classified in a number of categories, e.g. cannabis and ecstasy.

Classifying drugs by their effect on CNS

Stimulants

Tend to speed up the activity of a person's central nervous system (CNS) including the brain.

These drugs often result in the user feeling more alert and more energetic.

Examples include:
  • Amphetamines
  • Cocaine
  • Pseudoephidrine (found in medications such as Sudafed, Codral Cold and Flu)
  • Nicotine
  • CaffeineTop of page

Depressants (also known as relaxants)

Tend to slow down the activity of the CNS, which often results in the user feeling less pain, more relaxed and sleepy.

These symptoms may be noticeable when a drug is taken in large amounts.

It is important to note that the term 'depressant' is used to describe the effect on the CNS, not mood.

CNS depressants are more likely to result in euphoria than depression, especially in moderate use.

Examples include:
  • Alcohol
  • Major tranquillisers
  • Benzodiazepines (e.g. Valium, Temazepam) Opioids (heroin, morphine)
  • Volatile substances (can also be classified as 'other' (glue, petrol, and paint).

Hallucinogens

Have the ability to alter a user's sensory perceptions by distorting the messages carried in the CNS. A common example is LSD (trips).

Hallucinogens alter one's perceptions and states of consciousness.

Examples include:
  • LSD
  • Psilocybin (magic mushrooms)
  • Mescaline (peyote cactus)

Other

Includes psycho-active drugs that do not fit neatly into one of the other categories, but which are clearly psycho-active, such as antidepressants (e.g. Zoloft) and mood stabilisers (e.g. Lithium).

Examples include:
  • MDMA (ecstasy)*
  • Cannabis*
  • Volatile substances (petrol, glue, paint)
* Both ecstasy and cannabis can produce hallucinations, especially in cases of heavy use, or inexperienced users. However they are usually considered primarily as CNS stimulants and depressants respectively, as these effects are almost always present



 Naming drugs

 Page last updated: 2004
Drugs can be named in a range of different ways from chemical formulae to street terms. The following demonstrates how the same drug can be named in different ways.

Cannabis (generic name) is also known as:
  • Chemical name: Delta 9 - tetra hydro cannabinol (THC)
  • Brand name: N/A
  • Common term: Marijuana
  • Street name: Pot/mull
Alcohol (generic name) is also known as:
  • Chemical name: Ethanol
  • Brand name: Victoria Bitter
  • Common term: Beer
  • Street name: Grog
Temazepam (generic name) is also known as:
  • Chemical name: 7-chloro-1, 3-dihydro-1-methyl-5-phenyl-2H-1, 4-benzodiazepin-2-1
  • Brand name: Normison
  • Common term: Sedatives
  • Street name: Pills
Young people may often have their own names (sometimes called street names) for particular drugs and these names can differ from area to area. The list in the following exercise lists the street names of some of the common drugs. Street names will change over time, and can be specific to a particular region or local area. If you want to find out the names that are commonly used in your local area, you could contact your local health service or speak with some young people.

Exercises

Top of page

Workplace learning/brainstorm acitivty

Question - What are street names in your local area for:
  • Cannabis
  • Inhalants
  • Benzodiazepines
  • Ecstasy
  • Amphetamines
  • Hallucinogens
  • Opioids
  • Cocaine

Street names for drugs

Task - writing exercise

Write in any additional street names for each drug that is used in the area in which you work.

Cannabis:
  • Possible street names: Pot, grass, weed, reefer, joint, spliff, Mary-Jane, Acapulco Gold, rope, mull, cone, dope, skunk, bhang, ganja, hash, chronic, wacky tobacky
  • Names in my area:
Inhalants:
  • Possible street names:
    • Nitrous Oxide: laughing gas, whippits, nitrous
    • Amyl Nitrate: snappers, poppers, pearlers, rushamies
    • Butyl Nitrate: locker room, bolt, bullet, rush, climax, red gold
  • Names in my area:
Benzodiazepines:
  • Possible street names: Pills, downers, benzos, rohies, normies, vals, serries
  • List names in my area:
Ecstasy:
  • Possible street names: E, eccy, love drug, eggs, point, paste base, zip
  • Names in my area:
Top of pageAmphetamines:
  • Speed, uppers, ice, crank, meth, crystal, whiz, snow, goee, shabu
  • Names in my area:
Hallucinogens:
  • Possible street names:
    • LSD: acid, trips, wedges, windowpane, blotter, microdot
    • Psilocybin: mushies, blue meanies, magic mushrooms, gold tops
    • PCP: angel dust, hog, loveboat
  • Names in my area:
Opioids:
  • Possible street names:
    • Heroin: horse, hammer, H, dope, smack, junk, gear, boy
    • Methadone: done ('doan')
  • Names in my area:
Cocaine:
  • Possible street names:
    • Cocaine: coke, flake, snow, happy dust, Charlie, gold dust, Cecil, C, freebase, toot, white girl, Scotty, white lady
    • Crack: rock, base, sugar block
  • Names in my area:
While it can be very helpful to get to know some of the commonly used street names for drugs in the community in which you work, it is important to clarify which drug a young person is referring to by using universally understood terms such as heroincannabis and ecstasy, to eliminate any possible confusion.

3.3 The legality of drugs

 Page last updated: 2004
Drugs can also be classified in terms of their legal status. There are a variety of reasons why some recreational drugs are legal while others are illegal. The legal status of drugs is often due to historical and political factors rather than their harmful nature. For example, penalties are imposed for the use of some drugs, such as heroin, which only a small proportion of the population use, while others of apparently equal or greater danger are widely used, accepted and promoted, such as alcohol.

Formal sanctions such as laws that prohibit the use of certain substances can deter people from using those drugs. However, they do not necessarily stop use altogether. Prescriptions are another way of influencing the use and availability of drugs.

Task - writing exercise

Write the names of at least two or more psycho-active drugs in each category below.
  • Legally available to adults e.g. Over the counter painkillers
  • Legal with prescription e.g. Ritalin
  • Illegal to use e.g. Cocaine

Summary

  • The most useful system of classifying drugs is by their effect on the central nervous system
    • Stimulants speed up the CNS
    • Depressants slow down the CNS
    • Hallucinogens distort the message carried in the CNS
    • Other - those drugs that do not easily fit into the other groups.
  • Some drugs such as cannabis and ecstasy can fit into more than one category.

Distance learners

(A good point for student to contact facilitator.)

Distance learners should take time now to reflect on their learning, check in with their facilitator and determine their progress

4.1 The effect of drugs on the central nervous system (CNS)

 Page last updated: 2004
Drugs produce their effect on the body through two major processes. The first is the effect of the chemical properties of the drug on the central nervous system (CNS) which includes the brain and the spinal cord. This process is called pharmacodynamics. The second is how the drugs enter, are metabolised, and absorbed by the body. This process is known as pharmacokinetics. These two processes work together to produce a certain effect.

Pharmacodynamic processes

Neurons

A psycho-active drug must find its way to the bloodstream to have an effect on the brain. Once the drug reaches the brain, it can lodge on to specific receptor sites on the neurons which are sensitive to particular types of drugs. Each drug affects specific neurons in a number of parts of the brain. There are 13 billion neurons or nerve cells in each person's brain.

Neurotransmitters

Many drugs seem to imitate neurotransmitters, the natural chemicals that facilitate or inhibit the transfer of electrical impulses between neurons. For example, opiate drugs such as heroin are thought to exert their drug action by mimicking endorphins which are naturally occurring proteins that reduce pain.

Drug action

Like neurotransmitters, drugs can speed up (CNS stimulants) or slow down (CNS depressants) the transfer of electro-chemical messages between neurons in the brain. Messages between neurons can also be distorted when hallucinogenic drugs are taken.

Pleasure centre

In addition to affecting the transfer of messages between neurons, drugs appear to act directly on 'pleasure centres' in the brain, which may explain the euphoria experienced by users of many different types of drugs. It is believed that the effect on the pleasure centre is highly rewarding for many young people and is crucial to the development of drug dependence.

Review Quiz

Top of page

Task - writing exercise

Complete the following quick quiz which reflects your learning so far.
  1. Pharmacology is :

    1. a branch of science that deals with emotions
    2. the study of how drugs work
    3. the study of living things
  2. Circle the drug that is not psycho-active.

    1. alcohol
    2. petrol
    3. antibiotic
  3. A neuron is a:

    1. chemical in the brain
    2. gap between nerve cells
    3. nerve cell
  4. Drugs work by:

    1. imitating neurotransmitters
    2. destroying brain cells
    3. creating dysfunctional neural pathways
  5. The euphoria (good feeling) that drug use promote is caused by:

    1. the distortion of electrochemical messages between neurons
    2. stimulation of pleasure centres in the brain
    3. elimination of withdrawal symptoms
  6. Drugs can be classified by their effect on the CNS. What are the three major groups called? Provide two examples of drugs that fit in each of these categories.

    • Group 1
      • Group Name -
      • Example 1 -
      • Example 2 -
    • Group 2
      • Group name -
      • Example 1 -
      • Example 2 -
    • Group 3
      • Group name -
      • Example 1 -
      • Example 2 -
  7. Why is it necessary to have a fourth group called 'others'?