Drug/Application Antibiotics Clinical Uses Drugs used to treat infectious agents Dosing: Antibiotics with low PAE – optimize the duration of time the serum is > MIC; Those with high PAE – peak conc. is important Used in three ways: Empirical therapy 1. Determine if infected 2. Identify site of infection 3. Ascertain possible pathogens 4. Predict susceptibility 5. Obtain specimens for testing Definitive therapy Prophylactic therapy Possible routes of administration: IM, SC, IV, topical Mechanism Pharmacokinetics: successful therapy depends on the drug’s ability of killing the pathogen without causing harm Distribution: Difficult sites to access: brain, eye, prostate – contain permeability barriers Elimination: Most eliminated by kidney unless otherwise noted Post-antibiotic effect (PAE): suppression of bacterial growth after removal of antibiotic. PAE usually seen in gram (+) bugs, sporadically in (-) bugs. Antibiotics that inhibit cell wall synthesis (β-lactams and glycopeptides) have minimal or no PAE against gram (-) bugs Aminoglycosides- PAEconcentration-dependent killing -lactams- no PAE-time above MIC determines efficacy Drug Interactions/ Side Effects Direct effects: the result of direct interaction b/w the drug and/or one its metabolites and a specific tissue or organ in the body. Hypersensitivity – Type I HS rxn, Coomb’s (+) hemolytic anemia, serum sickness, and Stevens-Johnson syndrome Changes in microbial flora all antibiotics may lead to pseudomembranous colitis Drug interactions – esp. with Warfarin – enhances anticoagulation Host factors – genetics, age Pregnancy Comments Minimum inhibitory conc. (MIC) – the smallest conc. of the drug that inhibits the growth Minimum bactericidal conc. (MBC) – the conc. of the drug that will kill the bacterium Bactericidal: achievable blood concentration is > MBC Bacteriostatic: achievable concentration > MIC, but below MBC 1 Drug/Application INHIBITORS OF CELL WALL SYNTHESIS -lactams Bactericidal Penicillin * Standard penicillins Clinical Uses Mechanism Drug Interactions/ Side Effects Comments Non-competitive inhibitors of transpeptidases (penicillinbinding proteins-PBPs) Resistance mechanisms: Inactivation of antibiotics by β-lactamase – most common mechanism, those produced by S. aureus, hemophilus, and E. colo are relatively narrow in substrate specificity and woll hydrolyze penicillins but NOT cephalasporins Modification of target PBP’s – mech for MRSA and penicillin resistance in pneumococci Impaired penetration of drug to target PBP’s – presence of impermeable membram (gram neg) Presence of an efflux pump Possess a four member nitrogencontaining beta lactam ring Inhibit bacterial growth by interfering with a specific step in bacterial cell wall synthesis. Β-lactam antibiotics are structural analogs of the natural D-Ala-D-Ala substrate and are covalently bound by PBP’s at the active site. Binding leads to inactivation of the transpeptidase reaction – inhibiting peptidoglycan synthesis. High therapeutic index Type I – mediated by IgE – urticaria, rhinitis, angioedema, conjuctivits, or systemic anaphylaxis Type II – rare, interaction of IgG or IgM with antigen complement activiation, eg. Hemolytic anemia Type III – most common – serum sickness, arthralgia, rash, fever, lymphadenopathy, or vasculitis Type IV – CMI: cutaneous eruptions or thrombocytopenia Cell wall is composed of a complex cross-linked polymer, peptidoglycan, consisting of polysaccharides and polypeptides PBP in bacteria catalyze the transpeptidase reaction that removes the terminal alanine to form cross link with a nearby peptide Beta-lactam ring fused to a 5member, sulfur containing thiazolidine Modification of side chain differing properties Inhibitor of cell wall synthesis Penicillin allergy occurs in 0.5% of patients Probenecid-inhibits tubular secretion of penicillinused therapeutically Targets gram (+) 2 Drug/Application *Penicillin G (benzylpenicillin - IV) (crystaliine penicillin G – IM) (Benzathine penicillin G – long lasting – 1 mo) *Penicillin V (phenoxymethylpenicillin) *Antistaphylococcal penicillins (Methicillin – allegic intestinal nephritis) (Nafcillin – IV, preferred to above, metabolized by liver) (Oxacillin) *Dicloxacillin *Aminopenicillins *Amoxicillin *Ampicillin Clinical Uses Mechanism Drug Interactions/ Side Effects Comments Route – IV, IM Syphilis – treated with Benzathine penicillin G Rheumatic fever – treated with Benzathine penicillin G Neurosyphilis and meningitis due to S. Pneumoniae and Neisseria meningitides. Meningococcal infection Streptococci (S. pnemo) Route - Oral Pharmacokinetics – penetrates CNS in high doses Elimination: Primarily renal (10% by glomerular filtration and 90% tubular secretion) HS reaction: rash, serum sickness, and rare anaphylaxis Hematologic – Coombs (+) hemolytic anemia, rare Neuromuscular irritability/seizures – associated with high dose penicillin therapy in patients with renal failure Probenecid - inhibits tubular secretion, can be used to increase blood concentration and prolong the half-life Resistance – in S. pneumoniae and meningococci in some parts of the world Penicillin resistant staphylococci (S. aureus) Treat methicillin-sensitive staphylococcal infections (not active against MRSA) Stable to staphylococcal betalactamase Antistaphylococcal penicillin Structural analog of oxacillin Well absorbed after oral administration Addition of an amino group to the penicillin side chain Inhibitor of cell wall synthesis Not -lactamase resistant Distribution – Similar to other betalactams, can reach CSF in presence of inflamed meninges Elimination – primarily kidney, biliary excretion also occurs Hematologic – esinophilia Neutropenia may occur with long course (>21d) of nafcillin Hepatic dysfunction with high dose oxacillin More stable in the presence of acid than Penicillin G Minor staphylococcal infections Enhanced activity against gram (-) bacilli Some streptococci Enterococci and L. monocytogenes – has more activity against than Pen G E. coli, proteus mirabilis, H. in fluenza, samonella, and shgella Broad spectrum Route – oral Route – IV Meningitis Aminopenicillin Aminopenicillin Large dose – enters CSF Macropapular rash in patients with mononucleosis, Chronic Lymphocytic Leukemia, or on allopurinol Spectrum: Gram (+) Gram (-) bacilli Formulated with betalactamase inhibitors to increase the spectrum of activity 3 Drug/Application *Antipseudomonal penicillins -lactamase inhibitors *Clavulanate (Augmentin) *Sulbactam (Unasyn) Clinical Uses Same spectrum as aminopenicillins plus additional activity against gram (-) bacilli including Pseudomonas aeruginosa (used in combo with aminoglycoside) IV Available only in fixeddose combination with beta-lactamase sensitive penicillins Inhibit beta-lactamases produce by staphylococci, gonococci, H. influenza, B. fragilis, and some enterobacteriaceae Used in the treatment of intra-abdominal infections, bite wound infection and infected cutaneous ulcers Used in combination with AMOXICILLIN See above for uses Used in combination with AMPICILLIN See above for uses Mechanism Drug Interactions/ Side Effects Comments Exteded spectrum aminopenicillins Not stable against beta-lactamase Inhibitor of cell wall synthesis Subclasses Carboxypenicillins: ticarcillin Ureidopenicillins: piperacillin Beta-lactamase inhibitor Contain beta-lactam ring Covalently bind bacterial lactamase w/o intrinsic antibiotic activity Non-competitive inhibition Other combinations: Ticarcillin/clavulanate Piperacillin/tazobactam Beta-lactam antibiotic with a betalactamase inhibitor Beta-lactam antibiotic with a betalactamase inhibitor 4 Drug/Application Cephalosporins First Generation Cephalosporin *Cefazolin *Cephalexin (Keflex) Clinical Uses Activity against gram (-) bacteria increases from 1st gen. to 3rd gen. Antistaphylococcal activity decreases from 1st gen. To 3rd gen. No loss of antistreptococcal activity Activity against enterobacteriaceae superior to that of aminopencillins Resistant to most lactamases Activity against Streptococci Staphylococci E. coli P. mirabilis Klebsiella pneumoniae Useful against skin and soft tissue infections due to streptococcus pyogens or S. aureus Prophylaxis against infection following surgical procedures IV only Skin/soft tissue infections Oral Mechanism Beta-lactam ring fused to a sixmember sulfur-containing dihydrothiazine ring Individual cephalosporins are created by side-chain substitutions Inhibitor of cell wall synthesis Drug Interactions/ Side Effects Comments Approximately 10% crossallergenicity between cephalosporin and penicillin Avoided in patients who show IgE-mediated penicillin allergy Classified into different generations based on their spectrum Used as alternatives to penicillins in penicillin=allergic individuals 1st generation 1st generation 5 Drug/Application *Second Generation Cephalosporin *Third Generation Cephalosporin - look for the “t” for third *Cefotaxime *Ceftriaxome *Ceftazidime Clinical Uses Genrally used against mixed aerobic/anaerobic infections Against B. fragilis (ONLY 2nd gen) Activity against H. influenzae (β-lactamase producers or not) Community-acquired respiratory tract infections due to S. pneumoniae or H. influenzae Against enterobacteriaceae Children infections: streptococci, S. aureus, and H. influenzae Increased activity against aerobic gram (-) bacilli Esp. against Enterobacteriaceae and H. influenza Reduced activity against S. aureus Treat nosocomial pneumonia: gram - bacilli IV Activity against meningeal pathogens (S. pneumoniae, N. meningitidis, and H. influenzae) Activity against meningeal pathogens (S. pneumoniae, N. meningitidis, and H. influenzae) Active against P. aeruginosa Mechanism Drug Interactions/ Side Effects Comments Not used for meningitis Cefuroxime Cefprozil Stable to beta-lactamases that are produced by H. influenza and N. gonorrhoeae, and many of those produced by enterobacteriaceae 3rd generation Enters CNS NOT effective against type I chromosome mediated inducible cephalosporinase produced by Enterobacter sp., citrobacter freundii, serratia marcescens, and P. aeruginosa Oral form is available but has reduced activity, only used for enterobacteriaceae 3rd generation Enters CNS 3rd generation 6 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects Comments *Fourth Generation Cephalosporin Excellent activity against enterobacteriaceae and P. aeruginosa Good activity against S. aureus Carbapenems Broadest spectrum of all antibiotic Beta-lactam ring fused with a 5member carbon containing penem ring Inhibitor of cell wall synthesis Very broad spectrum IV only Used against Streptococci Enterobacteriaceae P. aeruginosa Hemophilus species Anaerobic bacteria, inc. B. fragilis Has better penetration and access (through pore channels) to the periplasmic space in gram (-) bacteria Resistant to -lactamases Pharmacokinetics: low oral bioavailability, well distributed to most tissue, and excreted by the kidneys Seizures Elimination: broken down by the kidney by human betalactamase (dehydropeptidase1) to a nephrotoxic metabolite Monocyclic beta-lactam – single ring structure attached to a sulfonic acid group Only binds transpeptidases of gram (-) Essentially nonallergic *Imipenem ‘Rambocillin- blows everything away, including your kidney’ Monobactams *Aztreonam ‘A bullet through an AZ tree is a negative thing’ Targets aerobic gram (-) bacilli inc. P. aeruginosa No activity against gram (+) bacteria or anaerobes IV only Cefepime – IV NO activity against: MRSA Enterococci Listeria B. fragilis Meropenum – similar activity as imipenem but does not produce toxic metabolite and is slightly more active against aerobic gram (-) bacilli and less active against gram (+) cocci ALWAYS co-administered with Cilastatin, a dehydropeptidase-1 inhibitor NOT active against enterococcus faecium, legionella, mycoplasma, or chlamydia sp. 7 Drug/Application Glycopeptides *Vancomycin - Bactericidal ‘Vanquishes gram (+)’ Clinical Uses Targets gram (+) bacteria -esp. those resistant to lactams MRSA β-lactam resistant strains of coagulase (-) staphylococci, enterococci, and pneumococci Serious infections with S. aureua, enterococci in pts. intolerant of β-lactam antibiotics Given orally for C. dificile Used as an IV Endocarditis prophylaxis for selected GU or GI procedures in β-lactam intolerant pts. Mechanism Inhibits cell wall synthesis Covalently binds terminal two Dalanine residue at the free carboxyl end of pentapeptide Sterically hinders the elongation of peptidoglycan backbone Unable to penetrate the cell membrane of gram (-) bacteria Low oral availability Excreted unchanged by the kidneys, half-life = 6 hrs Diffuses across serous membranes into pleural, pericardial, synovial, and ascitic fluid Enters CNS only at high doses Unaffected by beta-lactamase production or PBR alteration Drug Interactions/ Side Effects No cross-reactivity between beta-lactams and vancomycin Patients with anuria, half-life = 7 days Nephrotoxicity Ototoxicity ‘Redneck’ or ‘red-man’ syndrome- histamine release w/ rapid infusion-slow infusion better Comments 15% of enterococci resistant to vancomycin Vancomycin-resistant enterococci (VRE) is often resistant to all other antibiotics incurable VanA – Genes are carried on Transposable elements thant encode enzymes responsible for resistance D-Ala-D-Ala terminal is converted to D-Ala-Dlactate on resistant enterococci S. aureus strains gaining enterococcal transposon 8 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects Comments INHIBITOR OF PROTEIN SYNTHESIS Ribosomes are the site of protein synthesis in both prokaryotic and eukaryotic The differences in ribosomes between bacteria and humans provide a useful target for antibiotics Selective toxicity for bacteria *Aminoglycosides Bactericidal for aerobic gram (-) bacteria, staphylococci (in combo), and mycobacteria (in combo) Used only in serious infections due to enterobacteriaceae and P. aeruginosa and in a hospital setting IV only Once daily dosing/ single large dose – conc. dependent killing therefore the peak conc./MIC ration is the best predictor of bacterial killing Significant post antibiotic effect (PAE) Synergistic w/ penicillins Irreversible inhibitors of protein synthesis Crosses outer membrane through porin channels (passive diffusion) Then actively transported across the cell membrane by an oxygen dependent mechanism Binds IRREVERSIBLY to the 30S ribosomal subunit Blocks initiation of protein synthesis Blocks further translation and elicits premature termination Incorporation of incorrect amino acid Absorbed poorly for GI Highly polar compounds – don’t enter cell readily Low intracellular conc. except in proximal renal tubule Do NOT enter CSF Excreted by kidneys – glomerular filtration Half-life = 2-3 hr Consist of two or more amino sugars linked by glycosidic linkage to a hexose nucleus Aminoglycoside Nephrotoxicity – concentrates in renal tubules Reversible - Increased when combined with another nephrotoxic drug Ototoxicity (auditory and vestibular) - permanent Risk of toxicities are dose and duration dependent Control with MONITORING Neuromuscular paralysis Half-life in renal impairment = 24 – 48 hr Ribosomes: Bacteria – total 70S Small: 30S = 16S + 21 proteins Large: 50S = 23S 5S rRNA’s + 21 proteins Humans – total 80S Small: 40S Large: 60S Resistance: Bacteria produce transferases enzymes that inactivate the aminoglycoside Mutation of porin or of the 30S subunit Aminoglycosidemodifying enzymes (AMEs) differ among aminoglycosides – so bacteria may be resistant to one but not to another Anerobic bacteria are innately resistant to aminoglycosides - lack the oxygen dependent transport Other aminoglycoside: Amikacin – contains structural change that prevents its inactivation by many bacterial enzymes - Neomycin ‘A mean guy hits his opponent in the ear and kidney. He drops, paralyzed.’ *Streptomycin Activity against mycobacterium tuberculosis 9 Drug/Application *Gentamicin *Tobramycin *Macrolides Clinical Uses Hospital use Anti-Tb drug Used in combo with penicillin, ampicillin or vancomycin to treat enterococci and Listeria monocytogenes IV Opthalmic prep available Most active against P. aeruginosa IV Opthalmic prep.available Treat respiratory tract infections Alternate to penicillin to treat streptococcal pharyngitis Atypical strains of TB Mild community acquired pneumonia Treat hospitalized community acquired pneumonia (with IV 3rd gen. cephalosporin), Active against: - Pneumococci - M. Pneumoniae - C. pneumoniae - Legionella species Drug of choice to treat pertussis and Legionella Mechanism Drug Interactions/ Side Effects Comments Most active aminoglycoside for synergy in combo therapy Aminoglycoside Inhibits prokaryotic protein synthesis Do not affect the donor acceptor transfer reaction Prevent translocation of peptidyl tRNA from the acceptor site to the donor site, thus halting protein synthesis Macrocyclic lactone ring to which deoxy sugars attach SEE BELOW Resistance: Reduced permeability or reduced active efflux of drug Production of esterases that hydrolyze macrolides Ribosomal binding site mutation NOT active against enterobacteriaceae or P.aeruginosa 10 Drug/Application *Erythromycin ‘Someone with diarrhea and whooping cough laying a wreath on a Legionnaire’s cross (gram +) Clinical Uses Mechanism Group A streptococci Staphylococci Bordetella pertussis Corynebacterium Diphtheriae Campylobacter jejuni Mycoplasma pneumoniae Legionella species Chlamydia species Macrolide Contains two sugars moieties attached to 14-atom lactone ring Administered with enteric coating due to destruction by stomach acid Partly metabolized by the liver by N-demethylation Mainly excreted unchanged in the bile Only 5% excreted in the urine Do not penetrate the CSF Macrolide Semi-synthetic derivatives of erythromycin – addition of a methyl group Increased absorption Increased half-life Metabolized in the liver to 14hydroxyclarithromycin (also has antibacterial activity) Eliminated via hepatic metabolism and urinary excretion of intact drug Macrolide Semi-synthetic derivatives of erythromycin – adds methylated nitrogen to the lactone ring Increased absorption Increased half-life – slow elimination Produces lower serum conc. Has a large volume of distribution, tissue conc exceeds serum conc by 10 to 100-fold Penetrates most tissue Does NOT penetrate CSF Better absorption Eliminated in urine and feces *Clarithromycin Increase spectrum of activity See above plus against H. influenza Active against Mycobacterium avium complex (pathogen in pts. with AIDS) Very active against H. pylori *Azithromycin Increase spectrum of activity See above plus against H. influenza Active against Mycobacterium avium complex (pathogen in pts. with AIDS) Treat urethritis and cervicitis caused by C. trachomatis Once daily therapy Drug Interactions/ Side Effects Comments High incidence of GI upsetbinds to motilin receptors Poorly tolerated - dyspepsia, nausea, and vomiting Elevates serum theophylline Prolong QT interval when combined with non-sedating antihistamines- caution b/c of Torsades de Pointe risk Interferes with the metabolism of other drugs Less GI effects than erythromycin Prolongs QT interval – see above Available as erythromycin base and various salts and esters Weak activity against H. influenza Less GI effects than erythromycin Fewer drug interactions Primarily hepatic metabolism Spectrum: extended beyond erythromycin 11 Drug/Application Clinical Uses C. Lincosamides *Clindamycin *Tetracyclines ‘T-A-G 30S’ Oral or IV administration Active against: Streptococci Staphylococci (not MRSA) Anaerobic bacteria, inc. B. fragilis Several protoza Used as an alternative to beta-lactams Inhibits prokaryotic protein synthesis Chlamydiae Mycoplasmas Spirochetes Rickettsial infections Legionella Brucella *Tetracycline Administered qid *Doxycyline Once – twice a day administration Mechanism Drug Interactions/ Side Effects Comments Inhibits prokaryotic protein synthesis Binds to the 50S ribosome at the same site as macrolides Prevent translocation of peptidyl tRNA from the acceptor site to the donor site, thus halting protein synthesis Do not affect the donor acceptor transfer reaction Metabolized by liver Higher risk of pseudomembranous colitis by C. difficile Resistance due to mutation of the ribosomal binding site Causes cross-resistance to macrolides NO activity against enterococci or aerobic gram (-) bacilli Binds 30S rRNA, directly blocking the binding of the AA-charged tRNA to the acceptor site of the ribosome-mRNA complex Excreted by kidneys and in the bile Do not penetrate CSF Strong affinity for developing bone and teeth - Yellowbrown discoloration GI adverse effects – nausea, vomiting, and diarrhea Photsensitization Vestibular reactions (with minocycline) CONTRAINDICATION: pregnancy, children < 8 yrs Chelated by divalent or trivalent cations - ↓ absorption with Ca, Mg, Alcontaining antacid, dairy products, Ca supplements or sucralfate See above Resistance- due to changes in the transport mechanism decrease accumulation of tetracycline in the bacteria Widespread use resistance among pneumococci, group A streptococci, and staphylococci See above Binds 30S rRNA, directly blocking the binding of the AA-charged tRNA to the acceptor site of the ribosome-mRNA complex Short acting Binds 30S rRNA, directly blocking the binding of the AA-charged tRNA to the acceptor site of the ribosome-mRNA complex Long half-life 12 Drug/Application *Chloramphenicol ‘Like pouring chlorine bleach on organisms/baby/bones’ *F. Streptogramins *Oxazolidinones- Linezolid Clinical Uses Mechanism Inhibits prokaryotic protein synthesis NOT used as 1st line therapy in US Alternative tx. for bacterial meningitis Used for meningitis (S. pneumoniae, N. meningitidis, and H. influenzae) Treat brain abscess Broad spectrum IV Twice daily dosing Gram (+) bacteria Against Staphylococci (inc. MRSA), streptococci, and enterococcus facecium (inc. resistant forms) Binds to the 50S subunit at the peptidyltransferase site to prevent transpeptidation Enters CSF and brain parenchyma Conjugated in liver to its inactive form glucuronide (neonates are less able to conjugate) Serious toxicity virtually obsolete in US Gray baby syndrome - ↑ serum chloramphenicol: abdominal distension, vomiting, cyanosis, and circulatory collapse. Myelosuppression – dose dependent (reversible) Irreversible aplastic anemia (rare but fatal) Huge use in 3rd world Not effective against enterobacteriaceae B form binds to the 50S ribosome at the same site as macrolides Prevent translocation of peptidyl tRNA from the acceptor site to the donor site, thus halting protein synthesis Do not affect the donor acceptor transfer reaction Streptogramins A and B act synergistically Rapidly cleared by nonrenal mechanisms Large PAE Inhibitor of bacterial ribosomal protein synthesis Binds to the 50S subunit near its interface with the 30S subunit preventing the formation of the 70S subunit 100% oral bioavailability Distribution volume of >0.8L/kg Good tissue penetration Clearance is nonrenal and nonhepatic Metabolized by nonenzymatic oxidation Types: Quinpristin/dalfopristin (Synercid) – IV To be approved Resistance is infrequent Gram (+) pathogens Inc. MRSA, penicillinresistant pneumococci, macrolide-resistant streptococci, and vancomycin-resistant enterococci Saved for the treatment of resistant bugs Drug Interactions/ Side Effects Comments 13 Drug/Application INHIBITORS OF DNA SYNTHESIS *Fluoroquinolones Bactericidal *Ciprofloxacin ‘Flock of sinners gyrating’ *Metronidazole Bactericidal ‘An underground metro-no air’ Clinical Uses Mechanism Drug Interactions/ Side Effects Comments Oral – use in outpatient Broad-spectrum Against aerobic gram (-) bacilli Enterobacteriaceae Hemophilus species Moraxella catarrhalis Respiratory fluoroquinolones (newer) – improved activity against S. pneumoniae, inc. penicillin resistant strains The addition of fluorine atom to the quinolone nucleus enhances activity against gram (-) bacteria Inhibits both: DNA gyrase in gram (-) by binding to DNA-enzyme complexes – interfering with DNA replication Topoisomerase IV- gram (+) High oral bioavailability Divalent cations interfere w/ absorption Large volume of distribution – penetrate prostate, lung, bile, and ascitic fluid Penetrates CSF – but little clinical use for meningitis Elimination by kidneys Others: Levofloxacin, moxifloxacin, and gatifloxacin Overuse has eroded utility Resistance due to 1) mutations in DNA gyrase and Topoisomerase IV 2) decrease permeability DNA-gyrase – responsible for introducing neg. supercoils into the DNA Topoisomerase IV – separates interlinked daughter DNA molecules following replication See above Generally well-tolerated Common: nausea, vomiting, and diarrhea Occasional: HA, dizziness, insomnia, skin rash Photosensitivity Abnormal liver function test Hallucinations/Delirium Seizures, increase risk with NSAIDS Prolonged QT Damage growing catilage Arthropathy/tendonitis CONTRAINDICTION: pregnancy, younger then 18 Inhibits eukaryotic topoisomerase II at high conc. Increase serum theophylline levels No prolonged QT Nausea and vomiting Disulfiram-like reaction with ethanol (avoid EtOH up to 48 hrs after administration) Psychosis w/ disulfiram Aerobic bacteria – inherently resistant, lack nitroreductase Acquired resistance can develop decreased uptake or decreased nitroreductase production Oral Broad spectrum Pseudomonas aeruginosa Anthrax Anaerobic bacteria, esp. against B. fragilis Choice for C. difficile associated diarrhea/colitis Certain protozoa: giardiasis, ambiasis, and trichomoniasis Acne rosacea Nitroimidazole Enters bacterial cell by passive diffusion Reduced by nitroreductase Produce short-lived intermediate compounds or free radicals that interact with DNA and possibly other macromolecules leading to the disruption of DNA and inhibition of nucleic acid synthesis Large volume of distribution Enter CSF and brain Metabolized by liver, metabolite has antibacterial activity Excreted primarily in urine Weak activity against S. pneumoniae 14 Drug/Application INHIBITORS OF RNA SYNTHESIS *Rifamycins *Rifampin *Antifolates Sulfonamides (sulfa drugs) Bacteriostatic Trimethoprim Bacteriostatic Clinical Uses Mechanism Drug Interactions/ Side Effect Comments Inhibit bacterial RNA synthesis by inhibiting DNA-dependent RNA polymerase Metabolized by liver See above Enters CSF Potent inducer of hepatic microsomal enzymes Impart an orange color to all body fluids, esp. urine Resistence: Mutation in the DNA-dependent RNA polymerase Hepatotoxicity Flu-like syndrome/ Drug fever Drug Interactions: increases the metabolism of many drugs, e.g. glucosteroids, oral contraceptives, quinidine, phenytoin, barbiturates, theophylline, clarithromycin, ketoconazole, intraconazole, cyclosporine, warfarin Most allergenic of all antibiotics-macropapular rash Greater risk in HIV pts. Life Threatening Stevens-Johnson syndrome: flu-like syndrome involving rash, cardiac, renal, GI, and pulmonary complications Toxic epidermal necrolysis Resistance: organisms that use pre-formed folic acid, altered DHPS, increased PABA production, decreased permeability Anti-tuberculous therapy Activity against staphylococci, N. meningiditis, H. influenza, and Legionella pneumophila Eliminate nasal carriage state of N. meningiditis and S. aureus Used orally, IV form is also available Rarely used due to high rates of bacterial resistance and superiority of other drugs Active against Enteriobacteriaceae Rarely used alone Antagonize folate syntetic pathway by 1) inhibition of dihydropteroate synthetase (DHPS) 2) or dihydrofolate reductase (DHFR) Antifolates Inhibits dihydropteroate synthase (DHPS)- blocking bacterial dihydrofolate synthesis from PABA Excreted unchanged by kidney Antifolate Inhibit dihydrofolate reductase Excreted unchanged by kidney 15 Drug/Application *SulfamethoxazoleTrimethoprim (TMP/SMZ) Bacteriocidal Other *Nitrofurantoin Anti-TB drugs Clinical Uses Oral and IV form UTI Prevention and treatment of Pneumocystis carinii Enterobacteriaceae H. influenzae Many strains of streptococci and staphylococci Oral UTI Active against most Enteriobacteriaceae and enterococci Use in combination Duration of therapy - Isoniazid + rifampin = 9 mo course - Addition of pyrazinamide for 1st two mo, permits shortening of total duration to six months If drug resistant organism, use 4 drugs (ethambutol or streptomycin) If sputum (+) after 3 mo, use DOT, and conduct susceptibility studies Mechanism Drug Interactions/ Side Effects Comments Sulfonamide and Trimethoprim Combination targets both steps in folate synthesis Results in antibacterial activity that is 20 –100 x greater than sulfonamide alone Excreted unchanged by kidney Penetrates CNS – but not used See sulfonamides Not active against P. aeruginosa or anaerobic bacteria Mechanism of action unknown High oral bioavailability Low blood conc. – not adequate to treat infection High urinary and renal conc. Activity reduced in an alkaline pH Empiric therapy: isoniazid, rifampin, pyrazinamide, streptomycin, and ethambutol Isoniazid and rifampin are the most active Susceptibility testing is performed, then: Pyrazinamide (1st 2 mos.), rifampin, isoniazid (6 mos) if susceptible No cross-resistance Long-term use peripheral neuropathy and pulmonary fibrosis Hemolytic anemia in pts. with G6PD deficiency Not active against P. aeriginosa Not effective to treat a UTI caused by proteus species due to alkaline pH Difficulties with treating Tuberculosis: Mycobacteria are slow growing organisms Very resistant to antibiotics Have a lipid rich mycobacterial cell wall, very impermeable Organism mostly resides intrcellularly, poor penetration Develop drug resistance 16 Drug/Application *Isioniazid (INH) Bactericidal *Rifampin Bactericidal *Pyrazinamide (PZA) *Ethambutol Bacteriostatic Clinical Uses -M. Tuberculosis Less active against atypical mycobacteria As a single agent used for latent infections (prophylaxis) Prevention of TB in close contacts of active cases of pulmonary TB Single drug therapy Access poorly accessible organisms, such as intracellular organisms and those sequestered in abscesses and lung cavities See antibacterials First-line drug used in conjunction with INH and rifampin in short course (6 mo) regimens Kills intracellular M. tuberculosis Enhances uptake of rifampin into bacteria Mechanism Inhibits mycolic acid synthesis, which are a componet of the cell wall Acts on actively growing TB Penetrates all tissues, including CSF Penetrate phagocytic cells, so active against extracellular and intracellular organisms Prodrug- activated by mycobacterial catalase-peroxidase (katG) Metabolized by liver Nacetyltransferase Inhibits RNA synthesis Inhibit bacterial RNA synthesis by inhibiting DNA-dependent RNA polymerase Readily penetrates most tissues and into phagocytic cells Mechanism of action is unknown Activated by mycobacterial pyrazinamidase Taken up by macrophages and exerts its activity against intracellular organisms residing here Inhibits mycobacterial arabinosyl transferase, which is involved in the polymerization reaction of arabinoglycan Essential component of mycobacterial cell wall Alters cell wall permeability Well absorbed, eliminated in feces and urine in unchanged form Drug Interactions/ Side Effects Comments Genetics determines if rapid/slow acetylators adjust dose INH-induced Hepatitis Peripheral neuropathy at high doses or in slow acetylators and in pts with predisposing conditions (associated with pyridoxine deficiency) – treated with pyridoxine Resistance develops with mutations in the katG enzyme and in pther proteins Single drug therapy 10 – 20% prevalence of isoniazid resistance in clinical isolates Metabolized in liver, turn body fluids red-orange Hepatotoxicity Flu-like syndrome Induces microsomal Cytochrome p450- increases metabolism of many drugsprotease inhibitors, etc. Hyperuricemia that may precipitate acute gout (common) Hepatotoxicity No cross-resistance to other antimicrobial drugs, but cross-reactivity to other rifamycin Resistance: altered RNA Pol Dose-related: Retrobulbar (optic) neuritis which causes loss of acuity and red-green color blindness Monitoring at high doses Resistance develops quickly, so always administered in combination Resistance due to mutations that impair conversion of PZA to its active form No cross-resistance with INH or other antimycobacterial drugs 17 Drug/Application *Streptomycin Clinical Uses Active against extracellular TB Antivirals * Oseltamavir/Zanamavir Anti-influenza *Amantidine/Rimantidine Anti-influenza No activity against Influenza B. Prophylactic and lessen severity of symptoms Must be started within 48 hrs. of onset Mechanism Drug Interactions/ Side Effects Aminoglycoside antibiotic (see under AB section) Binds IRREVERSIBLY to the 30S ribosomal subunit Blocks initiation of protein synthesis Blocks further translation and elicits premature termination Incorporation of incorrect amino acid Penetrates cell poorly Inhibit steps in viral replication: 1. absorption to and penetration into susceptible host cell 2. uncoating of viral nucleic adic 3. synthesis of early regulatory proteins 4. synthesis of RNA or DNA 5. Synthesis of late, structural proteins 6. Assembly of viral particles 7. release from cell Neuraminidase inhibitor for both influenza A & B-prevents binding Ototoxic Nephrotoxic Inhibit un-coating of viral RNA of influenza A by binding M2 protein. GI intolerance CNS complaint – nervousness, difficulty in conc., lightheadedness Reduced dose in pts > 65 and in pts with renal insufficiency Comments serf Non-HIV viral infections Influenza Herpes virus (HSV, VZV, CMV) Hep C, RSV DNA viruses: ASH B (adenovirus, small pox, herpes, Hep. B) Prophylaxis Lessen duration of illness by 1 day 18 Drug/Application Clinical Uses *Acyclovir Antiherpes agent Oral, IV and topical formulation Primary and recurrent genital herpes ORAL: Shortens duration of symptoms, time of viral shedding, and time of resolution of lesions IV: tx for herpes simplex encephalitis and neonatal HSV infection Higher doses for VZV – shortens duration and lesions *Ganciclovir Anti-herpes IV and oral Potent activity against cytomegalovirus (CMV) IV use: CMV retinitis, colitis, and esophagitis in AIDS pts. IV use: Reduces incidence of symptomatic CMV disease of administered before organ transplantation IV use: CMV pneumonitis in immunocompromised pts. in combo w/ CMV Ig Mechanism Acyclic guanosine derivative that lacks a 3’-OH on the side chain Converted to monophosphate derivative by virus-specified thymidine kinase – therefore only active in infected cells Then cellular enzyme convert it to the di and tri- compounds Inhibits viral DNA syn. by 2 mechanisms: 1) Competitive inhibition of deaxyGTP for the viral DNA polymerase 2) Chain termination following incorporation into the viral DNA Elimination: Glomerular filteration and tubular secretion Enters CNS Acyclic guanosine analog Phosphorylated (activated) by CMV protein kinase phosphotransferase or HSV thymidine kinase Competitively inhibits viral DNA polymerase Incorporated into DNA- chain terminator Enters CNS Clearance related to creatine clearance Drug Interactions/ Side Effects Comments Generally well-tolerated Nausea, diarrhea, and HA IV infusion associated with renal insufficiency or neurologic toxicity (may include tremors and delirium) Resistance - via mutation of viral thymidine kinase or DNA Pol. Valcyclovir – L-valyl form of acyclovir – achieves plasma levels 3 –5 times higher Myelosuppression, particularly neutropenia 19 Drug/Application Anti-HIV (p.44) *Nucleoside Reverse Transcriptase Inhibitors (NRTI) Clinical Uses Multi-drug regimens essential b/c resistance develops Active against HIV-2 and HIV-1 Important component of multi-drug regimens *AZT (Zidovudine) / Didanosine (ddI), Zalcitabine (ddC), Abacavir, 3TC Nucleoside ReverseTranscriptase Inhibitors (NRTI) *Nonnucleoside Reverse Transcriptase Inhibitors (NNRTI) Specific activity against HIV-1 *Protease Inhibitors Combination therapy is necessary to combat resistance Mechanism Drug Interactions/ Side Effects Comments Substrate for reverse transcriptase, which converts viral RNA into proviral DNA before its incorporation into the host cell chromosome Pro-drug –phosphorylated by cell enzymes Incorporation into DNA terminates chain elongation See above Chain terminators; viral RT is ‘sloppier’ than host enzymes, more susceptible Bind directly to a site on the viral reverse transcriptase (not same as NRTI) Blockade of RNA- and DNAdependent DNA polymerase activities Metabolized in liver by CYP450 enzymes Bind reversibly to the active site of HIV protease, preventing cleavage of gag-pol polyproteins into mature products Prevent a new wave of infection High affinity for HIV proteases Poor systemic bioavailability Undergo oxidative metabolism by CYP3A4 – occurs in liver Lactic acidosis Severe hepatomegaly with steatosis See Above Multiple drug interactions, including protease inhibitors Rapid resistance develops – need to use as a multiregimen No cross resistance b/w NNRTIs and NRTIs or protease inhibitors Nausea, vomiting, diarrhea, and paresthesias Glucose intolerance Diabetes Hypercholesterolemia Hypertriglyceridemia Prolonged administration – Cushingoid syndrome: fat redistribution, esp. central fat accumulation, buffalo hump, breast enlargement, and subcutaneous lipomas Drug interactions occur with other protease inhibitors, NNRTIs and non-HIV drug Protease is essential for the production of mature infectious virions during HIV replication Drugs: Saquinavir/Indinavir/Nelfi navir/Ritonavir/Amprenavi r/Lopinavir 20 Drug/Application Clinical Uses *Antifungals Systemic drugs (oral or IV) for systemic infection Oral drugs for mucocutaneous infections Topical drugs for mucocutaneous infection *Amphotericin B Flucytosine IV (colloid suspension w/bile salt, and deoxycholate) First line therapy for lifethreatening mycotic infection Broad spectrum, inc. Candida albicans and Cryptococcus neoformans Organisms causing endemic mycoses Pathogenic molds Used for systemic infections Intrathecal therapy for fungal meningitis Lipid drug formulation (Liposomal Amphotericin B) – created to decrease toxicity ($$$) Active only against yeasts Griseofulvin Fungicidal “Ampho-terrible” Drug Interactions/ Side Effects Comments MODE OF ACTION (4 classes): - Polyene macrolides that lead to an alteration of membrane funtion - DNA and RNA synthesis inhibitors - Azole derivatives that inhibit sterol 14-α-demethylase, a key enzyme in ergosterol biosynthesis - 1,3-β-glucan synthase inhibitors Binds ergosterol (cell membrane sterol specific for fungi) and alters the permeability of the cell by forming pores Leads to cell death Poorly absorbed from GI Large tissue distribution 2-3% reach CNS Mostly metabolized, excreted slowly in urine Half-life = 15 days No adjustment required in hepatic/renal failure Binds to human membrane sterols toxicity Immediate infusion reaction: fever, chills, vomiting, HA, muscle spasms (prevented w/ slowing infusion rate of decreasing dose OR by premedication with antipyretics or meperidine) Renal toxicity, presents with renal tubular acidosis, severe K and Mg wasting Resistance occurs when ergosterol binding is imparied Initial therapy for serious fungal infection, then replaced w/ azole drug Facilitates entry of flucytosine Converted to 5-fluoruracil Myelosuppression Concentrates in keratinized epithelium, blocking fungal microtubules and preventing growth. Prevents infection of new cells. Resistance: Mutant permease prevents entry into cell ‘A grease covering the skin, blocking further infection’ - Nystatin (topical/ ‘swish and swallow for thrush’ Mechanism 21 Drug/Application *Azoles *Ketoconazole Clinical Uses Broad spectrun Candida species Cryptococcus neoformans Endemic mycoses Aspergillus infections Limited use due to toxicity Mechanism Reduction of ergosterol synthesis by inhibiting 14--demethylase (cytochrome P450 dependent enzyme system), inhibiting growth Increased affinity for fungal cP450 See above Less selective for fungal P450 Drug Interactions/ Side Effects *Fluconazole Polyenes Oral Treatment and secondary prophylaxis of cryptococcal and coccidiodal meningitis Alters fungal cell membrane permeability See above Enters CNS Good bioavailability Binds ergosterol, disrupting membrane structure; causes leakage and cell death Relatively non-toxic GI: nausea and vomiting Abnormal liver enzymes Clinical hepatitis (rare) CP450 metabolism Inhibition of human cP450 interferes with biosynthesis of adrenal and gonadal steroid hormones Endocrine effects: Gynecomastia, infertility, and menstrual irregularities Alter the metabolism of other drugs Other drugs can influence its blood levels (H2 blockers and rifamycins) Less drug interactions Least effect on human cytochrome P450 system Comments Other: Itraconazole Poor activity against aspergillus 22 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects Anesthetics Depth of anesthesia: ↑ BP indicates patient is feeling pain (too little anesthesia), ↓ BP indicates excessive anesthesia (respiratory depression). Inhaled anesthetics Organ effects: CNS: - Depressed - ICP with intracranial masses Respiration: - ventilation CV: - Depressed - Dose-related, (-) inotrophic effects - Peripheral vasodilation - Decreased blood pressure - Nodal arrhythmia - PVC’s Uterus: - Inhibit contraction Hepatic/Metabolic: - Blood flow is diminished - Decreased renal fxn. Nausea and vomiting Nausea and vomiting temperature Nitrous oxide Non-volatile (anesthetic) Quickly gets in, quickly gets out via lungs. Important actions at sites in the brain and the spinal cord. Act at the synapse. Interact with the hydrophobic regions of the membrane protein. Inhibition of AA GABAA and glycine receptors by interacting with specific sites in the transmembrane domain 2 and 3 of the receptor protein. The most hydrophobic anesthetics are the most potent. Blood-gas partition coefficient: if low, less affinity for blood, gets into brain more easily (lower MAC). Also leads to faster emergence from anesthesia. Oil-gas partition coefficient: if high, more affinity for CNS, less likely to remain in blood (lower MAC). Little effect on emergence. MAC = 105 Blood-gas coefficient = 0.4 Oil gas coefficient = 1.4 Comments General anesthetic: produces unconsciousness Objectives of general anesthesia: (1) Unconsciousness (2) Analgesia (3) SKM relaxation (4) ↓ reflexes (gag, etc.) Anesthetic activity depends on partial pressure. Volume % = partial pressure/ atmospheric pressure Minimum alveolar concentration (MAC): conc. of anesthetic in alveolar gas that prevents the response to pain in 50% of patients. 23 Drug/Application Clinical Uses Mechanism Halothane/ Enflurane/Isoflurane/ Desflurane/Sevoflurane Volatile IV anesthetics Induction or maintenance of anesthesia Barbiturates Most commonly used: thiopental and methohexital Enhance inhibitory neurotransmission or inhibit excitatory neurotransmission IV: depends on renal/hepatic clearance. Specific antagonists may be available. GABAA receptor agonist *Sodium pentothal Methohexital Thiopental Most commonly used IV induction of anesthesia Administered by infusion for cerebral protection Oxybarbiturate Administered rectally for induction in children Highly alkaline – tissue damage if injected extravenously Drug Interactions/ Side Effects Comments Halothane- Bradycardia Other volatile – mild tachycardia Desflurane and isofluraneexcite CNS when inhaled amount is sharply increased HTN and tachycardia Malignant hyperthermia: Produces SER dumping of Ca++, ↑ metabolism/ contraction leads to excessive heat generation, ↑ CO2 leads to acidosis. Fever, tachycardia, tachypnea, dysrhythmia, muscle rigidity, mottled skin, cyanosis, CV instability Treatment: dantrolene (muscle relaxant). Dose-dependent respiratory depression Cause apnea for 30 – 90 sec after induction of anesthesia All volatile gases have much lower MAC’s than NO, this means less drug is needed for induction Cardiovascular depression by ↓ myocardial contractile and ↓ systemic vascular resistance May precipitate episodes of acute intermittent porphyria ‘Truth serum’ Used for cerebral protection 24 Drug/Application Benzodiazepines *Midazolam *Etomidate *Propofol ‘Milk of amnesia’ *Ketamine Clinical Uses Mechanism Most common: Midazolam Anxiolytic Sedation GABAA receptor agonists Administered intramuscularly Administered orally as a sedative premedication in pediatric patients Used for intravenous induction in pts. With CV compromise Water soluble – less local irritation At physiological pH, it rearranges to a more active lipid soluble form Induction and maintenance of anesthesia or for sedation Antiemetic Rapid emergence and a feeling of well-being Ambulatory surgery Intramuscular induction in children or in uncooperative patient. Dissociative anesthetic used in veterinary medicine 2,6- diisopropylpylphenol GABAA receptor agonist Carboxylated imidazole derivative Pain on injection GABAA receptor agonist Structurally related to PCP NMDA (glutamate) receptor blocker Stimulates the sympathetic NS and, therefore, stimulates the CV system Direct myocardial depressant Drug Interactions/ Side Effects Comments Cardiovascular depression by ↓ myocardial contractile and ↓ systemic vascular resistance Diazepam and Larazepam also used Indirectly cause myoclonic movements during induction Produces no CV depression in HEALTHY patients CV depression in compromised pts compensatory ↑ in sympathetic activity Adrenal suppression Pain on injection – egglecithin emulsion Cardiovascular depression by ↓ myocardial contractile and ↓ systemic vascular resistance Used in cardiaccompromised patients Increases cerebral metabolism and intracranial pressure Changes in mentation Catatonia, hallucinations Bronchodilation CONTRAINDICTION: - pts. at risk for cerebral ischemia - Intracranial hypertension - Careful: critically ill pts. with depleted catecholamine reserves, pts. with CAD Cerebroprotective action by preventing excitatory AA-mediated neurotoxicity through its blockade of the NMDA receptor Little respiratory depression 25 Drug/Application Neuromuscular blockers *Succinylcholine Non-depolarizing Mivacurium Clinical Uses Mechanism Muscle relaxants Tracheal intubation When rapid onset is required Depolarizing neuromuscular blocker, acts at NMJ Initially causes fasciculation Desensitization of Ach receptor Broken down by plasma cholinesterase Onset within 1 min. Clinical duration 2-3 min. Full recovery 10-12 min. Selected on 1. Onset and duration 2. Route of metabolism and/or elimination 3. Cardiovascular side effects. Competitively inhibits Ach receptor Partial to complete blockade occurs over the narrow range of 75% 100% of receptor occupancy. Short acting Motor end plate acetycholine receptor antagonist Duration of action 20 min. Metabolized by plasma cholinesterase Motor end plate acetycholine receptor antagonist Duration of action 30 – 45 min. Atracurium and cis-atracurium are metabolized by a nonenzymatic mechanism (Hoffman elimination) and by ester hydrolysis Motor end plate acetycholine receptor antagonist Atracurium, cis-Atrcurium, Vecuronium, and rocuronium Intermediate acting Rocuronium – most rapid onset and succinylcholine is contraindicated *d-tubocurarine (curare), pancuronium, doxacurium, and pipecuronium Long-acting Drug Interactions/ Side Effects Comments Bradycardia + asystole: rare Myalgias 24 to 48 hours after administration IOP, ICP Hyperkalemia – in pts. with UMN and LMN lesions, major crush injuries, burns or prolonged immobility Phase II block- prolonged neuromuscular block Metabolism and elimination is important in pts. with impaired organ clearance CV effects: - Vagolytic effects ↑ HR - Autonomic block ↓ HR - Histamine release ↑ HR and hypotension - or NO CV effects Histamine release Cholinesterase SNPs or long-term succinylcholine infusion can lead to phase II block Vecuronium, cis-Atrcurium, rocuronium: NO cardiovascular effect Atracurium: Histamine relaease Atracurium and cisatracurium are independent of hepatic or renal function Pipecuronium: NO cardiovascular effects Pancuronium: Vagolytic effect D-tubocurarine: Autonomic ganglionic blockade and Histamine release Antagonized by neostigmine. Neostigmine side effects blocked by atropine Blockade is monitored by electrical stimulation of nerve 26 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects Comments Produce a transient and reversible loss of sensation in a circumscribed area of the body Topical anesthesia Blockade of peripheral nerves Spinal or epidural blockades Intravenous regional anesthesia Systemic uses: Antiarrhythmic, pain syndromes Interfere with nerve conduction, by blocking the influx of Na+ ions, thereby preventing the depolarization of the nerve Binds to sodium channels at site exposed during activity - More active neurons are more quickly blocked - Blocks both sensation and motor function - Conformational change 80% block required for anesthesia Does not alter resting membrane potential or threshold potential Systemic Toxicity Initial symptoms: CNSlightheadness, dizziness, seizures, tinnitus Higher serum conc.: ↓ myocardial contractility and systemic vasodilatation hypotension (blocks sympathetic conduction) High systemic conc.: Cardiac arrhythmia (inc. VF) Mostly hydrolyzed by plasma cholinesterase *Procaine Route – infiltration, spinal Chloroprocaine Route – epidural Peripheral nerve block Tratrcaine Route – topical, spinal Allergenic reactions can occur - Skin rashes - Bronchspasms Allergic reactions: RARE Stable in solution Local anesthetics Esters Amides *Lidocaine Route – topical, infiltration, spinal, epidural Peripheral nerve block IV regional anesthsia Sodium channel blocker Slow onset Short duration Sodium channel blocker Fast onset Short duration Sodium channel blocker Slow onset Long duration Metabolized in the liver Sodium channel blocker Fast onset Moderate duration PHYSIOCHEMICAL CHARACTERISTICS: The more lipid soluble, the more potent The lower the pKa and the greater the lipid solubility greater rate of onset The lower the pKa more anesthetic in unionized form The uncharged form (base) diffuses more readily than charged form (acid) Duration of action correlates with the degree of binding to generalized proteins in vitro 27 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects Opioids Narcotic analgesic Opium-like drugs Cross tolerance occurs to all members of opioid class Opioid poisoning: coma, pinpoint pupils, respiratory depression *Morphine Pain management Given parenterally, but in large doses it is effective orally Hospice Mix – morphine + flavored syrup Antitussive (rarely used) Antidirrheal (rarely used) Major active ingredient in opium opioid receptor agonist Receptors located on nerve cells in the brain, spinal cord, digestive system, etc Low oral bioavailability Metabolism – inactivated by glucuronide formation in the liver then excreted in urine 0.02% of dose crosses BBB Crosses placenta Ion trapping in stomach Duration of action: 4 –6 hours Effects: - CNS: Inhibitory: Analgesia, Euphoria, Drowsiness, Respiratory depression, Depression of cough reflex, Decreased sympathetic outflow Stimulatory: Stimulation of CTZ, Release of PRL and ADH, Miosis - Cardiovascular: Bradycardia, Decrease BP, Postural hypotension, Release Histamine peripheral vasodilatation, Cerebral vasodilatation due to increased pCO2 Increased ICP -Constipation, Inhibits gastric emptying, Inhibits flow of bile, Enhances absorption of fluid OPIOID POISONING Triad: Respiratory Depression, coma, miosis Nausea and vomiting Constipation Orthostatic hypotension Dependence develops after long term therapy Opioid withdrwal syndrome - 8-12 hr: lacrimination, rhinorrhea, yawning, sweating - 10-16 hr: Restless sleep - 16-24 hr: anorexia, dilated pupils, restlessness, tremor, gooseflesh - 48-72 hr: PEAK- anorexia, severe sneezing, irritability, insomnia, diarrhea, chills/sweats, abdominal cramps, orgasm, muscle pain, leg muscle spasms, waves of gooseflesh - 7-10 days: recovery form acute phase of withdrawal - Months: Narcotics huger Comments Physical dependence: desire to avoid withdrawal Psychological dependence: preference for drugged state Alcohol extract of opium – Laudanum Deodorized camphorated tincture of opium – Paregoric Acute withdrawal state can be stopped with an opioid agonist 28 Drug/Application Clinical Uses *Heroin Not medically used *Codeine Antitussive Antidiarrheal Pain management – not as effective as morphine *Meperidine (Demerol) Analgesics Methadone (Dolophine) Oral analgesic Antitussive Heroin addict rehab – prevents withdrawal Mechanism Diacetylmorphine Opioid receptor agonist 3 – 5 times more potent then morphine Crosses BBB much more effectively than morphine, where it is converted rapidly to morphine in brain. Methylmorphine Opiod receptor agonist 1/6 – 1/10 as potent as morphine Demethylated in brain to form morphine Orally active – resists hepatic metabolism Phenylpiperidine derivative Opioid receptor agonist 10% as potent as morphine Short duration of action (2 –4 hr) Not antitussive or constipating Diphenylheptane derivative Opioid receptor agonist Orally bioavailable Does not induce euphoria Drug Interactions/ Side Effects Comments High abuse potential Low abuse potential Respiratory depression Convulsions – toxic metabolite (normeperidine) Does NOT suppress cough reflex NON-constipating Abuse potential = morphine Antitussive Constipation Withdrawal – more rapid time course than for morphine and heroin Rehabilitation- eliminates needle fixation, prevents withdrawal for 24+ hrs, given in doses large enough to induce tolerance to heroin, allows for stabilization, pts become addicted to methadone vs. heroin 29 Drug/Application Other Opioid Agonist d-Propoxyphene (Darvon) Fentanyl (Sublimaze) *Dextromethorphan (Romilar) Diphenoxylate and Loperamide Opioid Antagonist *Naloxone (Narcan) Clinical Uses Analgesic Anesthesia Epiduraly following abdominal or pelvic surgery Antitussive (OTC) Antidiarrheal Reverse effects of opioid agonist drugs Reverse opioid agonist effects during an overdose Naltrexone (Trexan) Prevent opioid readdiction in detoxified patients Opioid Agonist-Antagonist Analgesics Pentazocine (Talwin) Analgesic *Buprenorphine (Temgesic) Analgesic Treatment for opioid dependent persons Mechanism Drug Interactions/ Side Effects Comments Weak opioid agonist Strength between aspirin and morphine Potent opioid receptor agonist 80x more potent than morphine Short duration of action Moderate abuse potential High abuse potential Specific opioid receptor agonist Not analgesic Opiod receptor agonist Vey low abuse potential receptor competitive antagonist Contains a bulky allyl group substitution at the nitrogen function of the piperidine ring Poor oral bioavailability Short duration of action (1 – 4 hrs) receptor competitive antagonist High oral bioavailability Antagonist effects at opioid receptor Agonist actions at opioid receptor Weak antagonist effects at opioid receptor Agonist actions at opioid receptor Non-toxic – but must be administered slowly to avoid triggering a opiod withdrawal syndrome IV Less abuse potential than opioids Respiratory depression at low dose BUT has ceiling effect i.e. not dose dependent Low abuse potential High doses – dysphoria and psychomimetic effects May precipitate withdrawal symptoms in a person with opioid tolerance dependence Potent partial opioid agonistantagonist Schedule III drug Does NOT replace methadone therapy 30 Drug/Application Anti-inflammatory Drugs NSAIDS Clinical Uses Mechanism Analgesic (esp. HA and Somatic pain) Antipyretic Anti-inflammatory (alleviate symptoms) COX-1 and COX-2 inhibitors Anti-inflammatory/Antipyretic: blocks synthesis of TNF- and IL-1, which normally act on anterior hypothalamus to synthesize PGE2. PGE2 raises the ‘set-point’ and causes edema. Analgesic: Somatic pain results from inflammation that triggers paininducing PGs. Drug Interactions/ Side Effects Due to the inhibition of COX-1 Blocking PGI2: GI intolerance/ulceration- PGs maintain blood flow and reduce acid secretion Blocking PGI2 and PGE2: Renal insufficiency/ hypertension- PGs increase renal blood flow; can lead to fluid retention. Caution in those with hypertension or CHF using ACE inhibitors or diuretics. Blocking PGF2: Inhibits uterine contraction-prolongs pregnancy, increases perinatal mortality and risk of postpartum hemorrhage (contraindicated in 3rd trimester) Blocking TXA2: Decreases platelet aggregation Aspirin induced asthma/allergy: due to shunting to LOX pathway Reye’s syndrome- associated febrile illness/ viral infection, do not give salicylates to children Comments General Properties of NSAIDS: Most are effective against fever, pain, and inflammation Gastrointestinal upset is a frequent side effect Most show cross sensitivity in aspirin sensitivity Most are highly bound to plasma proteins Symptoms of inflammation are affected more than the underlying cause. 31 Drug/Application Clinical Uses Mechanism *Aspirin Reduce fever Diminish pain (dull, aching pain) Anti-inflammatory RA Ibuprofen Irreversibly acetylates COX-1 (which is constituently expressed) Blocks COX-2 (responsible for inflammatory actions) ↓edema: Decreased pathological movement of fluid and cellular elements from the vasculature Decreased migration of leukocytes into tissue by decreasing adhesion molecule expression, and, therefore, decreased release of lytic enzymes Antipyretic: Leukocytes release IL1, which causes the formation of PGE2 by the COX enzyme (blocked by aspirin )that acts on the thermoregulatory region of the anterior hypothalamus, raising the set point. Suppress the production of TNF and IL-1B Analgesic: Inhibition of the synthesis of PG’s, mainly peripheral pain Anti-inflammatory: Blocks expression of leukocyte adhesion molecules. Induces synthesis of lipoxins and adenosine release (both anti-inflammatory). Metabolized in liver; 1st order at low conc., zero order kinetics at high conc. COX-1 and COX-2 inhibitor *Celecoxib (Celebrex) Arthritic conditions Selective COX-2 inhibitor Rofecoxib Selective COX-2 inhibitor Drug Interactions/ Side Effects Comments See above Most effective anti-platelet NSAID because of irreversible acetylation Gastrointestinal toxicity Reversible renal failure in pts. with intrinsic renal disease who are > 65, HTN, CHF, using diuretics and ACE inhibitors No gastrointestinal side effects Use with misoprostol (PG analog) – provide gastrointestinal protection Not used prophylatively against MI Not used prophylatively against MI 32 *Acetaminophen (Tylenol) Analgesic Antipyretic Less effective in reducing inflammation Not effective in reducing peripheral inflammation or producing endogenous antipyretics Inhibits COX-3, located in brain tissue 3 Metabolic Pathways: Metabolized by glucuronidation (60%), sulfation (35%), and oxidation mediated by cytochrome P450s (3%) Oxidation products are toxic but are detoxified by glutathione (limited amount in liver) Corticosteroids Glucocorticoid activityincrease glycogen deposition, antiinflammatory Mineralocorticoid activitymimics aldosterone: sodium retention USES: anti-inflammatory, Immunosuppresion Oral Inhalation: Asthma IV or IM Topical Endogenous cortisol- Hypothalamus secretes CRH, stimulates ACTH in anterior pituitary, regulated by negative feedback from cortisol. Carbohydrate metabolism – Increase gluconeogenesis by converting AA to glucose and glycogen; induce hepatic enzymes that increase formation of glucose from pyruvate Lipid metabolism – redistribution that causes buffalo hump and moon face Anti-inflammatory: 1. Promotes transcription of the gene encoding IκBα, which inhibits transcription of inflammatory cytokines (IL-6 and IL-8) by binding to the transcription factor NF-κB. 2. Produce peptides that stabilize membranes and reduce release of arachidonic acid. Anti-inflammatory activity: 1 Mineralocorticoid activity: 1 Short duration of action Hydrocortisone (cortisol) Well tolerated Hepatic injury (centrilobular necrosis) with doses > 4g/day Interactiion with ethanol: Acutely: EtOH induced cytochromes (no effect because cytochromes are occupied with EtOH met.) Chronic followed by abstinence: Now cytochromes metabolize acetaminophen →toxic metabolite Exacerbated by glutathione depletion seen in alcoholics Muscle atrophy due to gluconeogensis Glycogen deposition in liver Exacerbates peptic ulcers Painless perforation CNS toxicity: Insomnia, changes in mood, irritability, psychopathy Immunosuppressive Glaucoma, lens opacities Osteopenia/osteonecrosis Acute adrenal insufficiency after abrupt withdrawal Diabetes mellitus Treat hepatotoxicity with acetylcysteine- helps replenish glutathione Considerations when selecting a steroid: - Anti-inflammatory effects - Sodium retention - Deposition of liver glycogen Doesn’t stop RA disease progression Prototype natural glucocorticoid 33 Drug/Application Clinical Uses Mechanism Dexamethasone Asthma AI disorders Inhalation, oral Anti-inflammatory activity: 25 Mineralocorticoid activity: 0 Long duration of action Betamethasone *Prednisone Migraine-treatment *Ergotamine and DHE Ergot based Chronic use of NSAIDs can lead to headache syndrome Moderate to severe Migraine treatment Anti-inflammatory activity: 5 Mineralocorticoid activity: 1 Intermediate duration of action Anti-inflammatory activity: 25 Mineralocorticoid activity: 0 Long duration of action Anti-inflammatory activity: 25 Mineralocorticoid activity: 0 Long duration of action Vascular phase: triggered by changes in serotonergic brain stem function constriction then dilation of vessels (due to spreading cortical excitation-aura) activation of CN V nocieoceptive pathways with neurogenic inflammation Complex 5-HT, DA, NE partial agonist/antagonist Constriction of arteries and veins Drug Interactions/ Side Effects Comments Prototype synthetic glucocorticoid Cushing’s Disease test: Pituitary (ACTHproducing) tumor is cortisol resistant: cortisol, ACTH but ACTH inhibited by exogenous (high dose) dexamethasone Adrenal (cortisolproducing) tumor: cortisol, significantly ACTH- will show no further ACTH drop Ectopic ACTH-producing tumor: cortisol, ACTH, non-responsive to dexamethasone challenge Nausea Induce abortion Contraindication in pregnancy, HTN, coronary, cerebral, and peripheral vascular disease DHE is a more efficacious vasoconstrictor 34 Drug/Application *Sumatriptan (Imitrex) Prophylactic therapy *Methysergide Clinical Uses Mechanism Moderate to severe Migraine treatment Available in US: subcu, oral (IV elsewhere) Selective agonist 5HT-R1 located in extracranial circ. to cause vasoconstriction. More than 2-3 attacks/mo Last longer than 48 hrs Significant functional impairment Inadequate relief for tx or have unacceptable side effects Attacks occur after a prolonged aura Prophylactic prevention of migraine Given intermittently with monitoring for fibrotic complications Drugs used: Beta-blockers TCA Calcium channel blockers Anticonvulsants Combo of above MAO inhibitors – interactions with many foods (COUNSEL!!!) Ergot derrivative Complex 5-HT, DA, NE agonist/antagonist Drug Interactions/ Side Effects Comments Anaphylaxis Contraindication in pregnancy, CAD, postmenopausal, men >40, diabetes, obesity, HTN, smokers, hyperlipidemia, family hx. of vascular disease First does given under medical supervision Retroperitoneal fibrosis with ureteral obstruction Contraindiction in CAD, gastritis, HTN, connective tissu disease and pregnancy 35 Drug/Application Anti-Rheumatic Drugs *NSAIDS *Corticosteroids (Low dose) Clinical Uses Rheumatoid Arthritis: morning stiffness >1 hour Swelling in hands and 3 other joints Erosion on X-ray Rheumatoid nodules/pannus: inflammatory exudate overlying synovium Rheumatoid factor (IgM IgG) See anti-inflammatory RA See anti-inflammatory RA - low dose (< 7.5 mg/d) - Intra-articular injections can be used for individual joint flares Mechanism Drug Interactions/ Side Effects Comments Goals of therapy: 1. Relieve pain and inflammation 2. Prevent joint destruction 3. Maintain function Does not affect disease progression GI toxicity common Renal complications CNS toxicity Does not affect disease progression Tapering and d/c of use is often unsuccessful Low dose result in skin thinning, ecchymoses, and Cushinoid appearance Osteopenia, osteoporosis, osteonecrosis Endocrine: Adrenal insufficiency, hyperglycemia, DM and hypercholesterolemia GI: PUD with NSAIDS Muscle: Myopathy COX-1 and COX-2 inhibitor Help control inflammation and pain Improve mobility, flexibility, ROM Improve quality of life Relatively low-cost Modify gene transcription of inflammatory and noninflammatory mediators Bridge gap between initiation of DMARD therapy and onset of action Anti-inflammatory and immunosuppressive effects 36 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects Anti-inflammatory Slow inflammatory disease progression Myelosuppression *Methotrexate Folic acid analog Antimetabolite Inhibits dihydrofolate reductaselimiting ‘active THF’ productionalso directly blocks DNA, RNA, and protein synthesis Dosing: initial- 7.5 mg/wk,then increase, max dose 20mg/wk Short half-life, renal tubular secreted non-metabolized in urine Sulfasalazine Mild-moderate RA Broken down to 5-aminosalicylate (anti-inflammatory properties) and sulfapyridine (which has some immune modulatory affects) Dosing: Maintenance dose – 1 gm BID, max. is 4 gm/day Azathioprine Used for moderate to severe RA Prodrug form of 6-mercaptopurine Inhibits nucleic acid synthesispurine analog Pulmonary pneumonitis and/or fibrosis GI: nausea, anorexia, dirrhea, LFTs ↑ Skin: mucositiis Heme: ↓ blood counts, bone marrow suppression Other: nodulosis, opportunistic infections Renal tubular dysfunction Hepatic cirrhosis in RA dosing Contraindication: EtOH abuse, pregnancy, hepatitis, severe hepatic, renal, hematologic or interstitial lung disease Contraindication: Documented allergic rxn to sulfa drugs or aspirin, severe hepatic or hematologic disease. GI: nausea, vomiting, anorexia, LFTs Skin: rash, urticaria, StevenJohnsons syndrome (rare) Heme: neutropenia, anemia, thrombocytopenia DMARDs (Disease-modifying antirheumatic drugs) The “Gold Standard” Psoriasis Rheumatoid arthritis Graft vs. host disease Wegener’s granulomatosis Comments Choice of DMARD: Cost, convenience of administration, convenience of toxicity monitoring, compliance, comorbid disease, toxicity, severity and prognosis of patient Monitoring: Initial – CBC, LFTs, alb, hepatitis B and C serology, consider CXR, PFT’s F/U – CBC, LFTs, alb q 6-8 wks, creatine periodically Gold standard DMARD S-phase specific, but effects on protein and RNA synthesis may ‘self-limit’ this effect Monitoring: Initial – CBC, LFTs, G6PD F/U – CBC, LFTs, every 6 –8 wks. Not to be used in conjunction with allopurinol Rarely used 37 Drug/Application Clinical Uses Mechanism Hydroxychloroquine Mild RA, alone or in combination with other drugs (esp. Methotrexate) Dosing: Start 200 mg BID and keep under 5.0 mg/kg Not shown to prevent erosion New generation DMARD RA Taken orally Inhibits dihydroorotate dehydrogenase, an intermediate in the pyrimidine synthetic pathway (decrease synthesis) Reduces pain and inflammation Retartds structural damage evidence by x-ray, e.g. erosions and joint space narrowing Selectively targets autoimmune lymphocytes (T helper cells) Early onset of action Binds to TNF-α to prevent binding to receptor Slow radiologic progression of joint damage (more than MTX?) Newer DMARDs *Leflunomide *Etanercept *Infliximab RA Given every 2 wks RA Given once a month Chimeric IgG 1K monoclonal antibody Binds specifically to human TNFα Neutralizes TNFα biologic activity by binding both soluble and transmembrane forms, thus inhibiting binding of TNFα with its natural receptor Shown to prevent erosion (w/ MTX) Drug Interactions/ Side Effects Comments Contraindication: Preexisting retinopathy GI: nausea, vomiting, anorexia CNS: irritability, nervousness Heme: anemia Eye: retinopathy, macular atrophy Hepatotoxicity gastrointestinal Monitoring: Initial – Ophtamological exam, G6PD (if suspected) F/U: Ophth exam q 6 – 12 mo. Multiple sclerosis Aplastic anemia Systemic infection/sepsis Worsening of CHF Reactivation of prior granulomatous disease Long-term effects unknown Multiple sclerosis Aplastic anemia Systemic infection/sepsis Worsening of CHF Reactivation of prior granulomatous disease Long-term effects unknown Fast acting (~ 4 weeks) Expensive Better than methotrexate 38 Drug/Application *Anakinra Clinical Uses RA Gout treatment Gout Characteristics: Acute attacks of crystal induced arthritis Chronic deposits of monosodium urate in and around joints and cartilage and in the renal parenchyma Uric acid kidney stones Hyperuricemia *Colchicine Acute gout – no longer used Prophylaxis – low oral dose Mechanism Recombinant human IL-1ra produced in E. coli Identical to human IL-1ra except with an additional 1 n-termainal methionine Half-life = 6 hrs Reduce radiologic progression of joint damage Hyperuricemia is secondary to increased uric acid production or decreased renal clearance. Causes: Increased ProductionGout Lesch-Nyhan syndrome Lymphoproliferative disease Cytotoxic drugs Sickle cell anemia Decreased Renal ClearanceIntrinsic renal disease Renal failure Gout Lead nephropathy Decreased excretion due to competition Organic acids Increased lactate Increased ketone bodies Other- HTN, CAD Interrupts the inflammation triggered by the phagocytosis of urate crystals by inhibiting the release of a chemotactic glycoprotein from PMN and synoviocytes Spindle poison Metaphase arrest Microtubular poison Drug Interactions/ Side Effects Comments Multiple sclerosis Aplastic anemia Systemic infection/sepsis Worsening of CHF Reactivation of prior granulomatous disease Long-term effects unknown Oral: Nausea Vomiting Diarrhea Severe abdominal pain IV: Increased morbidity and mortality 39 Drug/Application Clinical Uses Mechanism NSAID: Acute Gout Prophylaxis COX-1 and COX-2 inhibitors Decrease the inflammation associated with gout *Allopurinol Gout prophylaxis Overproducers Oxypurinal Gout prophylaxis Overproducers Competitive inhibitor of xanthine oxidase Xanthine oxidase converts hypoxanthine to xanthine and converts xanthine to uric acid Lowers production of uric acid, lowers serum uric acid, and decreases uric acid excretion Also inhibits de nova production of purines Metabolite of allopurinol Inhibitor of xanthine oxidase Also inhibits de nova production of purines *Sulfinpyrazone/ *probenecid Gout prophylaxis Under-excretors Diminish renal tubular reabsorption of uric acid Promotes uricosuria Lowers serum uric acid Drug Interactions/ Side Effects Comments Newer drugs have fewer side effects Do NOT use aspirin or other salicylates Immunologic Bone marrow depression Skin rash Vasculitis Drug Interaction 6-mercaptopurine and azathioprine – cancer drug that is metabolized by xanthine oxidase Opioids and intra-articular glucocorticoids are useful in persons who cannot take NSAIDs Transiently increases serum uric acid levels as uric acid is mobilized- can lead to attack of acute gout Immunologic Bone marrow depression Skin rash Vasculitis Drug Interaction 6-mercaptopurine and azathioprine – cancer drug that is metabolized by xanthine oxidase Contraindication in hyperuricemia associated with uric acid nephropathy Transiently increases serum uric acid levels as uric acid is mobilized- can lead to attack of acute gout Transiently increases serum uric acid levels as uric acid is mobilized-can lead to attack of acute gout 40 Drug/Application Ethanol *Ethanol Clinical Uses Mechanism Used socially Treat methanol poisoning (IV) Treat ethylene glycol intoxication Mechanism of action: Bind to GABA type A receptors facilitating the entrance of Cl- into the cell thereby causing cellular depression Metabolism: In the liver 1) Metabolized by alcohol dehydrogenase to acetaldehyde 2) Metabolized by aldehyde dehydrogenase to acetate 3) which is activated to acetyl CoA in peripheral tissues Both reactions are redox reactions that increases the NADH/NAD+ ration, affecting other metabolic pathways that require NAD+: glycolysis, CAC, pyruvate dehydrogenase, fatty acid oxidation, and gluconeogenesis Distribution: Tissues with a high water content and that are highly perfused receive the most amount of EtOH Drug Interactions/ Side Effects Side Effects Glucose intolerace Alcoholic hypoglycemia Alcoholic ketoacidosis Lactic acidosis CNS: Behavioral syndrome, cerebellar syndrome, Wernick-korsakoff syndrome, cerebellar atrophy, central pontine myelinosis, demylination of corpus callosum, mamillary body destruction GI: local irritant, stimulate gastric acid production, gastric and duodenal ulcers, vomiting, acute and chronic pancreatitis, hepatic steatosis, hepatitis, cirrhosis Withdrawal syndrome: psychologic and physical – treat physical with benzodiaepines Interactions Acute – Metabolized by CYP2E1, so increases sensitivity of drugs metabolized by this enzyme Chronic – Induces CYP2E1 so the metabolism of drugs is increased in the absence of EtOH Alcoholics - develop metabolic tolerance due to increased CYP2E1 Contraindications Pregnancy – FAS Breast feeding Comments Factors influencing the BAC: Conc. of EtoH Irritant properties Type of beverage Blood flow at site of absorption Rate of ingestion Food 41 Drug/Application Clinical Uses Mechanism Drug Interactions/ Side Effects *Methanol Metabolized to formaldehyde and formic acid Visual damage Acidosis *Pyrazole Methanol intoxication *Disulfiram Aversion therapy for alcoholism *Ethylene glycol Ingredient in antifreeze Drugs of Abuse Motives for Using Obtain perceived therapeutic benefit (not abuse) Improve performance Obtain a rewarding subjective effect Hallucinogens AKA: Psychedelics and psychotomimetic *Lysergic Acid Diethlamide (LSD) AKA: Acid, boomers, yellow sunshine Important neurotransmitters: NE, dopamine, serotonin (5-HT) Induce altered states of consciousness Antagonist and agonist effects on 5-HT receptors- Synesthesia 5-HT2 on post-synaptic side, 5-HT1A on both pre and post synaptic neurons Raphe nuclei – involved in sleep, exerts presynaptic actions at 5-HT1A receptor to decrease function of 5HT neuron Agonist effects on presynaptic 5HT1A autoreceptors to decrease release of 5-HT Inhibits alcohol dehydrogenase Decreases the metabolism of EtOH Inhibits aldehyde dehydrogenase Leads to the build up of acetaldehyde to the point of causing noxious effects ADH mediates its metabolism to glycolic and oxalic acids Comments Treated with hemodialysis, ADH inhibitor, or IV ethanol Nausea Sweating Vomiting Increased heart rate Severe CNS depression Renal damage (due to build up of oxalic acid) Meabolic acidosis (due to build up of glycolic acid) Psychological dependence – results from craving for the drugged state in preference to the undrugged state Physical dependence- the appearance of a withdrawal syndrome in the absence of the drug Produces altered perception without clouded consciousness Perceptual sphere: visual hallucinations Affective sphere: extreme emotional lability Cognitive sphere: variation in logic Revelation: special insight Chronic effects – flashbacks which are transient, chronic abuse → depression/suicide Treatment: “Talk-downs”, maintenece of a quiet environment, subdued lighting; Diazepam, haloperidol, risperdone PRN Treated with hemodialysis, ADH inhibitor, or IV ethanol 42 Drug/Application Dimethyltryptophan Bufotenin Clinical Uses Synthetic hallucinogen AKA: Business man trip (BMT) Dose: 1mg Rarely seen in US Pilocybin Magic mushrooms of Mexico Psychomotor stimulants *Amphetamine Management of ADD or hyperkinesis in children – has paradoxical calming effect Mechanism Drug Interactions/ Side Effects Comments Same as above Short duration of action (30 min) Lethal: 10 – 15 mg/kg Same as above 10 times less potent than LSD Same as above – acts on S-5HT1A receptor 10 times less potent than LSD Release of newly synthesized monoamines: epinephrine, dopamine, and NE from brain neurons Rewarding effects are mediated through the mesolimbic and mesocortical dopamine systems Alertness Wakefulness Increased vigilance Increases physical and mental energy Tolerance develops rapidly Large dose: induces euphora, improved self-confidence, increased speech activity, and improved ability to concentrate Amphetamine-induced paranoia Secondary depression Physiological effects: increased BP, increased temp, and appetite suppression Toxicity: Seizures, cardiac arrhythmias, CVA, HTN and severe anxiety Treatment of overdose: Induce emesis, gastric lavage, urine acidification, haloperiodol Similar drugs: mescaline, dimethoxymethylampheta mine, and methylene dioxy amphetamine 43 Drug/Application Clinical Uses Mechanism *Methylated Amphetamine (MDMA) AKA: Adam, ecstasy, XTC, clarity, lover’s speed Previously used for psychotherapy Street form: MDA Acts at alpha receptors – stimulating both adrenergic and noradrenergic neurons Increases 5-HT release and blocks uptakes MDA- metabolite *Cocaine Routes of administration: topical- nasal mucosa, IV, smoked Local anesthetic Ingredient in Brompton’s mixture for cancer pts Causes a massive release of dopamine at the synaptic cleft Prevents the re-uptake of dopamine, NE, and serotonin Acts on pleasure centers of brain Rapid onset of action when inhaled *Phenyclidine (PCP) AKA Angel dust Often used as an adulterant to marijuana or sold as LSD, mescaline, etc. Chemically related to ketamine Psychotomimetic Open channel blocker: Noncompetitive antagonist at the NMDA glutamate receptor (blocks the excitatory NT) Reabsorbed through the enterohepatic circulation Persist for days to weeks Drug Interactions/ Side Effects Stimulant and psychedelic effects Neronal toxicity in animal models – destroys dorsal neurons Muscle tension Nausea Blurred vision Chills MDA – Serenity, joy, insight, and self-awareness; toxicity: profuse sweating, skin reactions, and confusion Intense euphoria Increased self-confidence Increased energy Chronic use: disruption of sleeping and eating, psychological disturbance Strong psychological dependence Toxicity: HTN< tachycardia, diaphoresis, mydriasis, vasospasms MI and CVA Euphoria Hallucinations (often auditory) Can induce (+) and (-) symptoms of schizophrenia R – rage E – erythemia D – dilated pupils D – delusions A – amnesia N – horizontal nystagmus E – excitation S – dry skin Comments Treatment: talk down, and diazepam (rarely used) Brompton’s mixture: cocaine, methadone, and alcohol Treatment: Acute anxiety with Diazepam, seizures with chlordiazepoxide, cocaine psychosis with haloperidol Treatment: Stabilization of CV and respiratory system and protection for selfinflicted harm. HTN – diazoxide; Convulsions – Diazepam; mechanical respiratory assistance; activated characoal Clinical presentation: HTN, convulsions With alcohol – respiratory depression 44 Drug/Application Clinical Uses Mechanism Marijuana Prepared from the leaves and flowering tops Anti-emetic in cancer Decrease IOP Appetite stimulation Chemotherapeutic agents Cell cycle specific (CCS): schedule-dependent cytotoxicity (repetitive/ prolonged administration better) -Majortiy are active in S phase OR Cell cycle non-specific Active ingredient: deltatetrhydrocannabinol (THC) Acts on cannabinoid receptor which is a G-protein coupled to cyclase THC receptors are concentrated in the hippocampus (memory), substantia nigra (reticulata), and cerebellum (motor and balance) No receptors in the medulla Fat soluble Slow excretion - half-life = 56 hrs, can be detected up to 120 hrs after last use. More rapidly growing tumors (embryonal>lymphomas>sarcomas> squamous cell carcinomas>adenocarcinomas) are more susceptible to chemotherapeutic agents Larger tumors have central ischemia- less blood-borne delivery of anti-cancer agents; less O2 to form radicals from radiation Antiemetic Prevent nausea and vomiting Antiemetic Antiemetic Antiemetic *Ondansetron Prochlorperazine Lorazepam and Dexamethasone Drug Interactions/ Side Effects Comments Improve mood and alter cognitive ability Exaggerated perception of time Sense of well-being or euphoria Vivid visual imagery with novel sight and sound Munchies Motivational syndrome with chronic use Chronic use - ↓ imuune system Withdrawal syndrome: restlessness, agitation, insomnia, sleep ECG disturbance, nausea, cramping Each individual treatment can reduce the tumor burden by 99% Prevent alopecia by scalp cooling- vasoconstriction leads to less drug delivery Bone marrow: Leukopenia and lymphocytopenia, immunosuppression, thrombocytopenia GI: Nausea and vomiting, oral and intestinal ulcerations, diarrhea Hair: Alopecia Gonads: Ammenorrhea, infertility, impaired spermatogenesis, sterility Kidney: Depends on cancer Fetus: Teratogenesis Nadir- low point of myelosuppression Majority produce: - Myelosuppression - Alopecia (reversible) 5-HT-3 inhibitor Dopamine antagonist CNS-targeting agent 45 Drug/Application Alkylating Agents *Mechlorethamine *Cyclophosphamide (Cytoxan) *Carmustine (Nitrosurea) Platinum Analogs *Cisplatin *Carboplatin Clinical Uses Mechanism Drug Interactions/ Side Effects Lymphomas, esp. Hodgkin’s disease IV Alkylation of DNA Nitrogen mustard Solid tumors (lung, breast) Hematologic cancers (myeloma, acute leukemia, non-Hodgkin’s) IV or PO Also used as a DMARD Alkylating agent- cross-links guanines-CCNS Metabolized to active form by cytochrome P450 Hepatically cleared, slowly excreted in urine CNS tumors Multiple myeloma IV Alkylating agent (carbonium ions)-CCNS CNS penetration Metabolized to active form by cytochrome P450 Toxic products: -DNA alkylating corbonium – crosslink DNA - Isocyanates- inactivate proteins (not anti-tumor) Excretion via hepatic inactivation with urinary elimination Platinum analog-alkylating agentCCNS Binds DNA nucleophiles following the loss of Cl- side group to the low Cl- intracellular environment – intrastrand guanosine crosslinks most toxic Platinum analog-alkylating agentCCNS Binds DNA nucleophiles following the loss of carboxyl side group Testicular cancer Ovarian cancer Head and neck cancer Limited myelosuppression Ovarian cancer (Head and neck) Slowly activated Phlebitis Nausea and vomiting Alopecia Myelosuppression (rapid nadir, granulocytes and platelets) Bladder toxin: Acrolein, a toxic metabolite, accumulates and causes fibrosis, acute ulceration, ↑ risk for cancer Myelosuppression – rapid Nadir, ↓ thrombocytopenia Alopecia – transient Emetic potential – low - mod Hepatotoxicity Profound and delayed myleosuppression – 6 wk. Nadir, 8 wk recovery Phlebitis due to ethanol diluent Heavy metal damage to renal tubular epithelium due to reduced creatinine clearance and BUN elevation Severe emesis Peripheral neuropathy Rare HS: ototoxicity No neurotoxic effects Greater myelotoxicities – thrombocytopenia Decreased vomiting Comments To prevent acrolein toxicity: Hydrate – high urine flow Frequent voiding Nucleophilic thiols to conjugate acrolein Mesna forms monomer that inactivates acrolein Avoid renal damage by: High urine flow Diuretic use 46 Drug/Application Clinical Uses DNA Intercalating Agents Doxorubicin (Adriamycin) Broad spectrum Heamatologic cancers Solid tumors, esp. sarcomas and breast cancer Topoisomerase Inhibitors Small cell lung cancer Refractory lymphomas Testicular cancer *Etoposide *Irinotecan Pro-drug Refractory solid tumors Non-small cell lung cancer Mechanism DNA binding antibiotics –CCNS Derived from streptomoycetes Plana chromophore slides b/w stacked nucleotides in DNA helix – distorts helix Anthracycline Antibiotic Intercalation of DNA blocks both DNA and RNA synthesis Quinone moiety promotes radical formation Inhibition of DNA Topoisomerase II – Accentuates strand breaking activity of topo II while inhibiting ligase activity, leading to cell lysis Long half life (24 –36 hrs) Biliary secretion Epipodophyllotoxin derivative DNA Topoisomerase II inhibitor CCS for G2 phase Results in the production of DNA ds breaks Topoisomerase I inhibitor CCS for late S/early G2 phase Stabilizes TOPI –I nicked DNA ‘cleavable complex’ causing strand breaks to persist Hydrolyzed to active product Half-life: 11 hrs Glucuronidated in liver Drug Interactions/ Side Effects Comments Myelosuppression Myelosuppression – short Nadir (8 – 10 d) Moderate nausea and vomiting Alopecia – prevent w/ scalp cooling Congestive heart failure-heart has low antioxidant activity – prevent w/ continuous drug infusion Colors urine red Myelosuppression: granulocytopenia, short nadir (8 – 10 d) Diarrhea Myelosuppression: Neutropenia Topiomerase I – alter the topology of DNA during transcription and replication Schedule-dependent toxicity – frequent administration to increase toxicity 47 Drug/Application Clinical Uses Antimetabolites *6-Mercaptopurine Oral adminstration to organ allograft pts. Oral admin: remission of ALL in children *Cladribine (2-cda) Hairy cell leukemia – curative Non-Hodgkins lymphomas CLL Infusion for 7 d Remission induction and maintenance of acute myelogenous leukemia Infuse for 5 – 10 d *Cytarabine *Gemcitabine (Gemzar) Palliative care of pancreatic cancer – reduces pain, maintenance of wt, increased energy level *5-Fluorouracil Colon cancer Solid tumors: breast, head and neck IV – constant infusion Mechanism Structurally similar to DNA and RNA Needs metabolic activation Schedule dependent toxicity Dose-limiting myelosuppression Purine antimetabolite-CCS for S phase Inhibits de novo purine biosynthesis Blocking the amination of phosphoribosyl pyrophosphate by glutamine DNA miscoding by direct insertion of metabolite of 6-MP into DNA/RNA Inactivated by xanthine oxidase New purine antimetabolite- CCS for S phase Inhibits adenosine deamininase leads to lethal accumulation of deoxyribonucleosides in cells Cytidine (pyrimidine) analog-CCS for S phase Inserted in growing DNA chain DNA chain terminator Inactivation by deaminase (liver) Cytidine (pyrimidine) analog-CCS for S phase Inhibit deoxycytidin synthesis Insertion into chain – premature DNA chain termination Uracil (pyrimidine) analog- partially CCS for S phase Inhibits DNA synthesis by inhibiting thymidine biosynthesis – forms complex: FdUMP-reduced folate-thymidylate synthetase RNA chain terminator by insertion Plasma half-life: 10-20 min Drug Interactions/ Side Effects Comments Granulocytopenia Do not use w/ allopurinol Azathioprine is prodrug form of mercaptopurine Myelosuppression Myelotoxicity Conjunctivitis – acute Cerebellar dysfunction – chronic Well-tolerated – low toxicity Does not increase survival Myelosuppression – minor with continuous infusion, major w/ intermittent tx Stomatitis Diarrhea 48 Drug/Application Clinical Uses Mechanism *Imatinib (Gleevec) Chronic myelogenous leukemia Gastrointestinal stromal tumors Dose and schedule dependent *Methotrexate Cancers: ALL, osteogenic sarcoma, breast, head and neck, cervical Non-malignant: psoriasis, RA, GVHD, Wegener’s Can be administered directly into CNS *Leucovorin Treat MTX toxicity Microtubular Inhibitors *Vincristine Hematologic cancers *Vinblastin Solid tumors Inhibits t(9;22) bcr-abl tyrosine kinase, stopping cell proliferation and inducing apoptosis Metabolized by CYP3A4, eliminated in feces Half life 18 hr (parent), 40 hr (metabolite) CCS for S phase – self limited Folic acid antagonist Inhibit DHFR – blocks conversion to THF (active form) Inhibition of thymidylate synthetase – blocks DNA syn. Half-life: 2-4 hrs Renal tubular secretion Fully reduced folate Given 24 – 48 hrs after high dose MTX Vinca alkaloid CCS- M-phase microtubule inhibitor, binds in S phase Bind tubulin subunits, capping the microtubule and preventing elongation, disrupting chromosome segregation Long half-life – 30 hrs Excreted in bile Vinca alkaloid CCS- M-phase microtubule inhibitor, binds in S phase Bind tubulin subunits, capping the microtubule and preventing elongation, disrupting chromosome segregation Long half-life – 30 hrs Excreted in bile Drug Interactions/ Side Effects Unusually well-tolerated Nausea Fluid retention Muscle cramp Chronic tx: suppression of WBC and platelets Comments Resistance: Point mutation or c-abl overexpression Bcr-abl tyrosine kinase – only in tumor cells Lethal myelosuppressionreversed by Leucovorin – fully reduced folate – given 24 – 48 hrs. high dose MTX Mocositis Dermatitis Renal tubular dysfuction Daily doses: cirrhosis Resistance: DHFR gene amplification Peripheral neuropathy – Parethesis, ↓ reflexes, paralytic ileus, jaw pain SIADH – rare Management- delay or reduce next dose NOT myelotoxic Tubulin fxn: 1) Intracellular solute transport 2) Cell movement 3) Nuclear scaffolding 4) Formation of mitotic spindles Myelotoxic: granulocyte and platelet suppression Short Nadir: 8-10 d Infection and bleeding Tumor pain – rare Management – Antibiotics or platelet transfusion 49 Drug/Application Clinical Uses *Paclitaxel (Taxol) Solid tumors Advanced and refractory ovarian cancer IV Steroid Hormones Breast cancer Suppressive but notcurative in estrogenresponsive tumors May be efficacious in tumors that are estrogen receptor negative (rarely) *Tamoxifen *Megestrol Breast cancer *Anastrozole Breast cancer *Leuprolide Prostate cancer Palliative care inc. decreased bone pain Subcu pellet in abdomen Monoclonal Antibodies HER2-/Neu (+) Breast cancers Highly effective in combo with cytotoxic drugs *Trastuzumab (Herceptin) *Rituximab (Rituxan) B cell non-Hodgkin’s lymphoma Mechanism Drug Interactions/ Side Effects Comments CCS- M-phase microtubule inhibitor Binds complete microtubules, causing clumping and disrupting mitosis Short half-life 6-8 hrs Excreted in bile Anti-estrogen-CCNS Breast estrogen receptor antagonist Bone, endometrium estrogen receptor agonist HS rxn to Cremophor EL – bronchospsms, dyspnea, hypotension Myelosuppression – short Nadir (10-14d) Peripheral neuropathy Cremophor EL (vehicle) – premedicate with dexamethasone, diphenhydramine, or ranitidine, cimetidine Acute disease flare Menopause symptoms inc. hot flashes Hormonally dependent breast cancers: Post menopausal ↑ estrogen and progesterone receptors Response to hormonal manipulation Long disease-free period after 1º tx Progestin agent Decreases Estrogen receptors Enhances estrogen metabolism Reversible selective aromatase inhibitor – blocks estrogen syn. GnRH derivative Acutely: agonist - ↑ FSH and sex hormones Chronically: antagonist - ↓ FSH and sex hormones (castration levels) Humanized murine Ab binds to erb-b2 receptor (HER-2/Neu), member of EGFR family. Leads to antibody dependent cellular cytotoxicity Humanized murine monoclonal Ab binds to CD20 Blocks cell signaling (proliferation) → apoptosis Appetite and weight gain Fluid retention Vaginal bleeding Lethargy Adrenal insufficiency Dermatitis Well-tolerated Hot flashes Give w/ dexamethasone and mineralocorticoids First infusion rxn – fever, chills, and rigors Enhanced antracyclineinduced cardiotoxicity Erb-b2 receptor –overexpressed in 20-25% of breast cancers- poor prognosis First infusion rxn CD-20: on immature B cells 50 Drug/Application Clinical Uses Mechanism Decrease Parkinson signs Initial improvement Chronic use: effectiveness decreases due to progression of disease Dopamine precursor - restores dopamine conc. in the basal ganglia Low bioavailability due to decarboxylation and MAO in gut *Pergolide Parkinson Used in combo w/ levdopa Dopamine receptor agonist – mimics endogenous dopamine *Seligiline Effective in early Parkinson Can be used in combo with levodopa *Benztropine Useful in patients taking neuroleptics Selectively inhibits MAO-B, in dopaminergic neurons Increases Dopamine by preventing metabolism Muscarinic receptor antagonist *Amantidine Parkinson’s Benefit decreases with long-term use Parkinson’s Drugs *Levodopa (w/ Carbidopa) Antiviral drug Releases endogenous dopamine (?) Drug Interactions/ Side Effects Choreiform movements of face and limb – limits dose On-off effect Nausea and anorexia – treated w/ peripheral dopamine antagonist Hypotension High doses: psychotic effects – confusion, insomnia, and nightmares Paranoia Hallucinations Confusion and Nightmares Dyskinesia Vomiting Less wine-cheese effect Fatal hyperthermia in combo w/ meperidine, fluoxetine, or cocaine Dry mouth Heat stroke Impaired vision Urinary retention Constipation Drowsiness Confusion Minor Comments Administered with peripheral dopa decarboxylase inhibitor (carbidopa) Decreases levadopa side effects Does not retard neurodegenerative process 51 Drug/Application Anti-anxiety Drugs *Buspirone *Diazepam Clinical Uses Anti-anxiety Takes 1 wk to work Oral Anti-anxiety and insomnia Sedation in mania Spastic due to CP or tetanus Anesthesia – milk of amnesia EtOH detox Seizures – status epilepticus Block sedating effects of benzos Schizophrenia Intractable hiccups Huntington’s chorea Ballism Tourette’s syndrome Acute psychotic depression Mechanism Mechanism unknown Serotonin 1a receptor agonist (?) Half-life 2-4 hrs Metabolized in liver Long-acting benzodiazepine GABA-A receptor agonist – binds to alpha-2 subunit plus others to increase Cl channel opening thereby hyperpolarizing the cell Half-life 20-80 hrs Penetrates CNS Liver metabolism, renal excretion Active metabolite Benzodiazepine receptor antagonist Act mainly on D2 receptors, with the atypicals also acting on serotonin receptors Also have limited action at αadrenergic, cholinergic, histamine Higly lipophilic Metabolism by cP450 Drug Interactions/ Side Effects Mild sedation No withdrawal syndrome Gradual withdrawal to avoid seizure Alpha-2 subunit – binding here causes anti-anxiety effects Other subunits are responsible for anesthetic effects *Haloperidol (+) Symptoms of schizophrenia Typical anti-psychotic Blocks D2 receptors in the limbic and mesocortical areas – need to block > 75% High potency Sedation Ataxia Dependence Death when combined with EtOH Amnesia Withdrawal effect: seizures and agitation Crosses placenta Precipitate withdrawal symptoms SEE NOTES Occupancy > 85% → extrapyramidal movements due to increased sensitization of unoccupied receptors to dopamine Cross placenta Neuroleptic malignant syndrome – needs immediate care Extra-pyramidal movements Sedation *Chlorpromazine Schizophrenia Typical anti-psychotic Block D2 receptors Low potency Photosensitivity Large autonomic effects Sedation Extrapyramidal movements *Flumazenil Anti-psychotics Comments α-adrenergic – control excitement, postural hypotension Cholinergic – atropine side effects Histamine - sedation 52 Drug/Application Clinical Uses Mechanism *Thioridazine Schizophrenia Typical anti-psychotic Block D2 receptors Low potency *Clozapine (-) and (+) Symptoms of schizophrenia *Dantrolene Neuroleptic malignant syndrome Malignant hyperthermia Serotonin syndrome Depression Panic or phobic disorders OCD Enuresis Anorexia and bulimia Treat for 1 – 6 mo Depression Atypical anti-psychotic Block Serotonin receptors and D2 receptors Low potency – blocks 35% of D2 receptors Direct muscle relaxant Anti-depressants *Fluoxetine (Prozac) Depression: Dysregulation of NE and 5-HT leads to alterations in the NE and 5-HT receptors Antidepressants re-regulate receptor sensitivity Takes 2 wks for re-regulation SSRI Potent blockers of serotonin reuptake Active metabolite w/ half-life of 128 hrs. Drug Interactions/ Side Effects Comments Pigmentary degeneration of retina – browning of vision Large autonomic effects Abnormal cardiogram – prolong Q-T Sedation Mild Extrapyramidal movements Agranulocytosis in 1% Large autonomic effect Sedation No extrapyramidal movement All characterized by having a labile system Non-compliance due to oversedation, anticholinergic side effects, or sexual dysfunction Drug selection is based on past response to drug, susceptibility to side effects, compatibility w/ other drugs pt. is taking Nausea, HA, nervousness, and insomnia Some sedation Anorganism, impotence, ↓ libido (30-40%) Fatal interaction w/ MAOI → Serotonin Syndrome Treat serotonin syndrome with Dantrolene, avoid by spacing out tx. switch (4wk. For Prozac) 53 Drug/Application Clinical Uses Mechanism *Tranylcypromine Depression MOAI Prevent the metabolism of monoamime NT – increasing the conc. at nerve terminal *Setraline (Zoloft) Depression SSRI Potent blockers of serotonin reuptake *Imipramine Depression TCA – tertiary Blocks reuptake of norepinephrine and serotonin Drug Interactions/ Side Effects Wine-cheese effect Dry mouth, constipation, difficult urination (no direct anticholinergic effects) Sedation Weight gain Sexual dysfuction CV: orthostatic hypotension Monoclonic jerks in sleep Serotonin syndrome w/SSRI Enhances depressive effects of EtOH Nausea, HA, nervousness, and insomnia Some sedation Anorganism, impotence, ↓ libido (30-40%) Fatal interaction w/ MAOI → Serotonin Syndrome Anticholinergic – dry mouth, blurred vision, constipation, urinary retention and speech blocking Postural hypotension Tachycardia, arrhythmia, ECG abnormalities, A-V block Sedation – take QHS Weight gain Tremor and akathisia Lowers seizure’s threshold Sexual dysfunction: resolves Comments Wine-cheese effect: Tyramine, usually broken down by GI MAOI’s, is taken into nerve endings where it causes massive NE release → HTN crisis, tachycardia, HA< chest pain, dilated pupils, nausea and sweating Treat w/ α-adrenergic blocker Treat serotonin syndrome with Dantrolene, avoid by spacing out tx. switch (2 wks) Treat tremor w/ propranolol Used for SUICIDE 54 Drug/Application Clinical Uses Mechanism *Lithium Mood stabilizer Acute mania Manic episode of bi-polar disorder Slow onset – intiate tx. w/ benzo or anti-psychotic Severe recurrent depression Substitutes sodium in generating action potentials Pumped out slowly → accumulates → partial ndepolarization Enhancement of serotonin effects Inhibition of NE and dopamine release Augmentation of Ach synthesis 2nd messenger systems → reduced response to muscarinic and αadrenergic stimulation Renal clearance Anti-convulsants Steady plasma conc. must be reached before changing dose *Ethosuximide Uncomplicated absence seizures Epilepsy is a chronic brain disorder 10-30% don’t respond to therapy General site of action: Sodium channels, GABA Inhibiting low threshold voltage dependent (T-type) calcium channels in thalamocortical neurons *Topiramate Adjunctive or monotherapy for all types of seizures *Lamotrigine Adjunctive or monotherapy for all seizures 3 mechanisms of action: Na channel blocker Enhancement of GABA at GABA-A receptors AMPA receptor antagonist Inhibits release of excitatory amino acids Drug Interactions/ Side Effects Comments Narrow therapeutic index (0.8-1.2 meq/L) – check serum conc. regularly Tremor – alleviate w/ propranolol Decreased thyroid function Polydipsia and polyuria Edema ECG abnormalities depressed T wave Excreted in breast milk Therapeutic overdose – treat w/ hemodialysis, peritoneal dialysis Physical work-up and renal function before prescribing Drowsiness GI distress: nausea and vomiting HA Somnolence and fatigue Dizzyness Difficulty concentrating Speech disorders and ataxia Some dizziness, ataxia, somnolence, and fatigue Rash in 5% Increased elimination when taken with carbamazepine, phenytoin, and pheno Decreased elimination with valproic acid 55 Drug/Application Clinical Uses Mechanism *Phenobarbital Suppress seizure activity, elevate seizure threshold, and limit spred from a focus Generalized tonic/clonic Partial seizures Status epilepticus Barbiturate Enhance GABA inhibition by increasing Cl channel opening Bind to BZD binding → increase the frequency of opening but not the time *Phenytoin (Dilantin) Generalized tonic/clonic Partial seizures Status epilepticus Oral Na channel blocker Maintains Na channel in inactive state for a prolonged perion Follows Zero-ordered kinetics – small changes in dose → large changes in plasma level Metabolized in liver, half life is 24 hrs. *Carbamazepine Generalized tonic/clonic Partial seizures Status epilepticus Focal epilepsy in children *Valprocic Acid Absence seizures Myoclonic seizures Partial seizures: complex Na channel blocker Increase central NE transmission Adenosine partial agonist (?) Metabolism: 95% hepatic, half life is 12 hrs Induces own metabolism by increasing microsomal enzymes Inhibits metabolism of GABA Prolongs recovery of Na channels Some inhibition of low threshold voltage dependent (T-type) calcium channels Drug Interactions/ Side Effects Comments Sedation and impaired cognitive function Ataxia Withdrawal syndrome Osteomalacia Paradoxical hyperactivity in some children Agitation and confusion in elderly pts. May WORSEN absence seizures Dizzyness, ataxia, nystagmus, diplopia Gingival hyperplasia Hirsutism Skin rash Mental confusion Altered vit. D and Ca metabolism Tetratogenic Drowsiness Disequilibrium Blurred vision Ataxia Used in children Anorexia Nausea Vomiting Hand tremor at high doses Alopecia Weight gain Hepatoxic Neural tube defects Increase Phenobarbitol levels in blood Minmal drowsiness and general depression Use w/ caution in liver disease Fosphenytin – used for Status epilepticus (IV) Need to increase dose over time 56 Drug/Application Clinical Uses *Gabapentin Partial seizures Neuropathic pain Psychiatric conditions *Tiagabine Adjunctive therapy in adults and children (>12) for tx of partial seizures Absence seizures Myoclonic seizures Atonic seizures *Diazepam Mechanism Unknown Indirectly increases CNS GABA levels, ↑release (?) Absorption shows saturability from GI Renal elimination Interfers with GABA reuptake 96% protein bound, half-life is 7-9 hrs Benzodiazepine Enhances GABA actions by increasing the frequency of Cl channel opening → hyperpolarized Drug Interactions/ Side Effects Comments No drug interactions Mild: somnolence, fatigue, ataxia, and dizzyness Drugs effecting micrsomal enzymes will increase its metabolism Side effects may lessen w/ long term use Impaired concentration Somnolence and fatigue Dizzyness and nervousness CNS depression Drowsiness Ataxia Dysarthria Respiratory depression and bronchial hypersecretion 57