Term
Approximately how long is the development and pre-clinical testing process? |
|
Definition
|
|
Term
How long are Phase I clinical trials? |
|
Definition
|
|
Term
How long are Phase II clinical trials? |
|
Definition
|
|
Term
Which of the two (phase I or phase II) has more participants? |
|
Definition
|
|
Term
How long are Phase III trials? |
|
Definition
|
|
Term
Approximately how many participants are in Phase III trials? |
|
Definition
|
|
Term
What is the average cost for the development of a drug? |
|
Definition
|
|
Term
How long do patents last? |
|
Definition
|
|
Term
How many marketed drugs manage to recoup the cost of development? |
|
Definition
|
|
Term
What are some reasons drugs fail? |
|
Definition
Clinical safety, efficacy, formulation, PK/bioavailability, commercial, toxicology, cost of goods |
|
|
Term
Generally, where is drug discovery performed? |
|
Definition
|
|
Term
Generally, where is drug development performed? |
|
Definition
|
|
Term
What determines the biological activities of compounds? |
|
Definition
|
|
Term
What chemical properties affect biological activity? |
|
Definition
Physiochemical (lipophilicity, ionization, solubility), Electronic (resonance, inductive effects, bonding potential), and spatial (molecular dimension, stereochemistry) |
|
|
Term
How are new drugs created from old drugs? |
|
Definition
Alteration of side chains to improve something (PK, dosage form, etc.) Example: Captropil. |
|
|
Term
How is biological information used to create drugs? |
|
Definition
Study of indigenous medications, study of warfare chemicals (nitrogen mustards), observations in in vivo testing (vinca alkaloids), or observations of clinical side effects (Viagra) |
|
|
Term
What is the modern approach to drug development? |
|
Definition
Use knowledge of the biology of the disease to identify targets that can be manipulated by drugs, then use that knowledge of target protein to design new inhibitors. |
|
|
Term
What are the steps in a drug development project? |
|
Definition
Target selection/validation, assay development, identification of a lead compound, lead optimization, preclinical evaluation, and clinical trials |
|
|
Term
What is the most critical stage of drug development? |
|
Definition
Target selection/validation. One wants targets that are relevant and selective |
|
|
Term
What are traditional strategies for target selection? |
|
Definition
Study disease biology, key regulatory proteins, response to previous drugs, and engineered models |
|
|
Term
Most drug targets are and ? |
|
Definition
|
|
Term
Why are other targets, like DNA or nuclear receptors, not as common? |
|
Definition
|
|
Term
What is functional genomics? |
|
Definition
Genetic analyses for disease genes and protective genes, by DNA microarray analyses. Strategy for target selection. |
|
|
Term
Methods for target validation? |
|
Definition
Biology of disease, gene knock-out/in, constitutively-active/domiant-negative mutants, antisense, ribozymes, RNA interference |
|
|
Term
What is RNA interference? |
|
Definition
Selectively decreases levels of expression of specific mRNA in a cell. Mimics the ‘perfect’ drug. |
|
|
Term
What is important in assay development? |
|
Definition
Relevance, selectivity, sensitivity, reproducibility, speed, and cost. |
|
|
Term
Activities of signaling proteins are affected by what? |
|
Definition
|
|
Term
What are the two basic approaches to lead compound identification? |
|
Definition
Computation modeling and screening |
|
|
Term
|
Definition
Substrate analogs to act as enzyme inhibitors |
|
|
Term
Computer-aided drug design downside? |
|
Definition
Need to know structure of target |
|
|
Term
|
Definition
Testing collections of compounds for biological activity, don’t need to know structure. ‘Hits’ = CxD |
|
|
Term
|
Definition
Chemical diversity of compound collection. |
|
|
Term
What are some sources of compounds for lead discovery? |
|
Definition
Natural products, synthetic compounds |
|
|
Term
What is combination chemistry? |
|
Definition
Assembly of molecular building blocks. Uses high-throughput synthesis. Requires high-throughput screening for lead discovery |
|
|
Term
If there are 5 sites for substitution, and 20 building blocks, how many potential compounds can be created? |
|
Definition
|
|
Term
What is a benefit of robotic screening systems? |
|
Definition
Decreased time, decreased error |
|
|
Term
What are the goals of lead optimization? |
|
Definition
Increase target selectivity (decrease adverse effects), increase potency (decrease size), simplify synthesis ( increase ease of manufacture), optimize chemical and metabolic stability, and optimize in vivo activity (therapeutic efficacy). |
|
|
Term
|
Definition
|
|
Term
What is the pharmacophore? |
|
Definition
3D arrangement of chemical determinants that are necessary for binding to the receptor |
|
|
Term
What is traditional lead optimization? |
|
Definition
Synthesize homologous series, or use ‘chemical insight’ or bioisterism rules. Synthesis of few compounds. Low-throughput screening, high cost, slow progress |
|
|
Term
Lead optimization of combinatorial chemistry? |
|
Definition
Synthesis of ‘focused libraries’, may/may not require high-throughput screening, high start up costs, but fast and efficient. |
|
|
Term
Lead optimization by computer-aided drug design? |
|
Definition
Analysis of receptor on pharmacophore. Allows for QSAR analyses and drug design without knowledge of protein structure |
|
|
Term
|
Definition
a. Quantitative structural activity relationships. b. Mathematical relationship between biological activity and chemical properties and allows for prediction of activity of new compounds |
|
|
Term
|
Definition
QSAR, biological activity = f . Limits the number of compounds that are made. |
|
|
Term
|
Definition
Predicts solubility, oral bioavailability, biodistribution, and metabolism |
|
|
Term
What is tested in in vitro studies? |
|
Definition
Chemical purity and stability, formulation, mechanism of action, and cellular pharmacology |
|
|
Term
What is tested in in vivo studies? |
|
Definition
Efficacy in animals, short-term and long-term toxicity, carcinogenicity, metabolism, PK, and biodistribution |
|
|
Term
What is the final step in drug development? |
|
Definition
|
|
Term
What is a clinical trial? |
|
Definition
A prospective study comparing the value of intervention against a control in humans |
|
|
Term
What is required of a clinical trial? |
|
Definition
May not be retrospective, must employ one or more intervention techniques, must contain a sufficienctly similar control group, approval for the study must be obtained from the FDA, and an Investigational New Drug application must be submitted |
|
|
Term
What must be included in the IND application? |
|
Definition
Descriptions of the manufacture of the drug, its pharmacology and toxicology in animals, and a detailed protocol for clinical trials. |
|
|
Term
What department accepts or rejects INDs? How long does the process take? |
|
Definition
The CDER. If, after 30 days, no rejection is received, the study has implicit approval to proceed. |
|
|
Term
What are the goals of Phase I clinical trials? |
|
Definition
Primarily to establish maximum tolerated dose in humans. Secondarily to examine pharmacokinetics in humans. Phase I is not for treating the disease. |
|
|
Term
Who are the subjects in Phase I trials? |
|
Definition
20-50 Mostly healthy volunteers or patients who did not respond to other treatments (especially in cancer and AIDS cases) |
|
|
Term
What is the classical procedure for determining the maximum tolerated dose in humans? |
|
Definition
3 subjects per dose level. Starting dose is 1/10 of the LD10 for mice, or 1/3 of the toxic dose in dogs. If none of the 3 experience a toxic reaction, 3 different people are treated at a higher dose. If one of the three has a reaction, three different people are treated with the same dose. This is continued until 2 of the three have a reaction. |
|
|
Term
What are the goals of phase II clinical trials? |
|
Definition
To determine the efficacy of the drug and to continue monitoring for toxicity. |
|
|
Term
Who are the subjects in Phase II trials? |
|
Definition
50-300 patients with carefully matched disease |
|
|
Term
What is the procedure of a Phase II trial? |
|
Definition
Two-stage design. Drug given at dose determined by Phase I. Blinded design. May be uncontrolled in cancer drug studies |
|
|
Term
What are the goals of Phase III? |
|
Definition
To verify efficacy and safety |
|
|
Term
Who are the subjects in Phase III trials? |
|
Definition
100s-1000s of patients with carefully matched disease |
|
|
Term
What is the procedure of a Phase III trial? |
|
Definition
Often multi-arm studies in which the drug is compared with the standard drug for that disease and can involved cross-over and double-blind studies. Drug given at dose used in Phase II. Response can be increased survival, improvement of a biomarker, or improved quality of life |
|
|
Term
Who conducts the final review process? How long does it take? |
|
Definition
The CDER. Less than one year. |
|
|
Term
|
Definition
Continued monitoring of the drug post-marketing |
|
|
Term
In the past, drug discovery was essentially what kind of process? |
|
Definition
Random, observational and relied on finding a molecule that worked and figuring out how it worked later |
|
|
Term
Who discovered penicillin? |
|
Definition
Alexander Fleming in 1929 |
|
|
Term
|
Definition
Biomolecules whose functions can be artificially modulated |
|
|
Term
What technologies enabled the new methods of drug discovery? |
|
Definition
Molecular biology, in vitro assays, and 3D structural models of drugs and their receptors. |
|
|
Term
In drug design and discovery, what is the difference between high-throughput screening and computer prediction models? |
|
Definition
a. High-throughput tests large numbers of molecules (10^6) and has a typical hit rate of <1% b. Computer prediction tests the compounds on the computer first, thus decreasing the number of molecules actually tested and increasing the hit rate. |
|
|
Term
What forces drive states of matter and the 3D conformation of macromolecules? |
|
Definition
|
|
Term
What are interatomic forces? |
|
Definition
Covalent interactions (-50 to -100 kcal/mol), and noncovalent interactions (electrostatic -80 to -120, van der walls -0.5 to -1, hydrophobic effects -0.5 to -1, and hydrogen bonding -3 to -6) |
|
|
Term
What are hydrogen bonding interactions? |
|
Definition
It occurs when two electronegative atoms compete for the same hydrogen, and create an electron deficient hydrogen. |
|
|
Term
What are some H-bond donors? Some H-bond acceptors? |
|
Definition
a. Donors: NH, OH b. Acceptors: C=O, OH |
|
|
Term
Why are hydrogen bonds important in drug discovery? |
|
Definition
Influence interactions between drug and solvent, drug and target, and drug-to-drug |
|
|
Term
What is the hydrophobic effect? |
|
Definition
It is a major driving force in binding of drugs to receptors. Enzyme active sites are hydrophobic in nature. It occurs to maximize contact between nonpolar parts of drug and receptor and contributes to bonding. |
|
|
Term
|
Definition
Complementarity between ligand and receptor – like a lock and key. |
|
|
Term
What are the two types of complementarity? |
|
Definition
Shape complementarity, in which the 3D shape of the drug matches that of the receptor, and chemical complementarity, in which groups on the drug can interact with appropriate groups on the receptor. |
|
|
Term
The affinity of a drug is described as what? |
|
Definition
The binding energy, which is the sum of the forces between the drug and the receptor |
|
|
Term
What is the equation for binding energy? |
|
Definition
dG = dH – TdS, where H is enthalpy and S is entropy. |
|
|
Term
Where do enthalpy and entropy come from? |
|
Definition
Enthalpy results from forces within and between molecules. Entropy comes from solvent molecules and movement and rotation of bonds |
|
|
Term
What is protein X-ray crystallography and how is it used in drug design? |
|
Definition
X-rays shined through molecule. Diffraction finds regions with high concentrations of electrons. These regions are used to assign positions to atoms. |
|
|
Term
What is molecular modeling (computational chemistry)? |
|
Definition
Allows one to create 3D models of molecules by calculating their properties and minimizing their energy. Can model conformations of ligands and receptors and their interactions and their binding energy |
|
|
Term
What is the pharmacophore? |
|
Definition
The features of a drug necessary to ensure optimal interactions with a biological target and to trigger or block it’s response |
|
|
Term
How is virtual (computer) screening useful? |
|
Definition
Matches the 3D structure of potential drug with the shape of the receptor |
|
|
Term
What is high-throughput docking? |
|
Definition
Fitting the molecule into the receptor pocket and then scoring it based on interactions in 3D structure. |
|
|
Term
What are the goals of substrate metabolism? |
|
Definition
Increase water solubility and polarity, deactivation and excretion, or activation. |
|
|
Term
What are some common sites of biotransformation? |
|
Definition
The liver (main), intestinal mucosa, kidney, and lungs |
|
|
Term
What types of reactions are Phase I? |
|
Definition
Oxidation, hydroxylation, reduction, and hydrolysis |
|
|
Term
What are the goals of Phase I reactions? |
|
Definition
Introducing a new functional group, modifying an exisiting functional group, or exposing an existing functional group (more than one may occur). |
|
|
Term
What are the Enzymes involved in Phase I reactions? |
|
Definition
Cytochrome P450 and Flavin-containing monooxygenases |
|
|
Term
What are Phase II reactions? |
|
Definition
Conjugations: acetylation, glucuronidation, sulfation, and amino acid conjugation |
|
|
Term
What is the goal of Phase II reactions? |
|
Definition
To conjugate an endogenous molecule to a functional group to increase polarity or water solubility to facilitate excretion |
|
|
Term
What is the most common reaction in xenobiotic metabolism? Which enzyme performs is? |
|
Definition
|
|
Term
Where is the highest concentration of CYP450s? |
|
Definition
|
|
Term
Where are the CYP450s located? |
|
Definition
Most are on microsomes, some are on mitochondria. |
|
|
Term
What type of proteins are CYP450s? |
|
Definition
Heme proteins with an iron protoporphyrin ring. |
|
|
Term
What components are required for CYP450 systems? |
|
Definition
Oxygen, NADPH, NADPH-CYP450 reductase, and a lipid environment |
|
|
Term
Describe the catalytic pathway for CYP450. |
|
Definition
The CYP has a Ferric Iron (Fe3+). The drug binds to the CYP. P450 reductase reducese the iron to the Ferrous (Fe2+) state. Oxygen binds to the Fe2+ iron. Radical on oxygen oxygenates the drug. Fe4+ is formed, oxygen binds to the drug and drug leaves the complex oxidized. |
|
|
Term
What are some hydroxylation mechanisms catalyzed by CYP450? |
|
Definition
Aromatic hydroxylation, aliphatic hydroxylation, deamination, O-Dealkylation, N-dealkylation, and sulfoxidation |
|
|
Term
What does decreasing metabolism do? |
|
Definition
It decreases toxicity. Example: in decreasing toxicity and metabolism levels: methoxyflurane, halothane, enflurane, isoflurane, desflurane. |
|
|
Term
What is particularly interesting about haloflurane? |
|
Definition
At two doses, can cause haloflurane hepatitis |
|
|
Term
Which are the most predominant CYP genes? |
|
Definition
|
|
Term
What does CYP2A1 generally metabolize? |
|
Definition
|
|
Term
What is CYP2E1 specific for? |
|
Definition
Small molecules, ethanol, anesthetics, small organic solvents |
|
|
Term
Which isoforms metabolize most drugs? |
|
Definition
|
|
Term
Family 1 metabolizes what? |
|
Definition
Environmental carcinogens, in particular aromatic hydrocarbons and aryl amines |
|
|
Term
|
Definition
Aromatic hydrocarbon hydroxylase, primarily expressed in extrahepatic tissues (small intestine, lung, placenta). Inducible. Variation. |
|
|
Term
|
Definition
Expressed in liver, stomach, and intestine. Inducible. Variation. |
|
|
Term
|
Definition
Found in liver, lung, and nasal epithelium. Metabolizes drugs (coumarin), procarcinogens, and nicotine. Polymorphisms. |
|
|
Term
|
Definition
Includes CYP2C8 (liver and skin), CYP2C9 (Liver and intestine), and CYP2C19 (liver and intestine). Metabolize 25% of clinically important drugs. Represent 20% of total CYP in liver. Polymorphisms. |
|
|
Term
|
Definition
Polymorphisms. Metabolizes 21% of clinically important drugs. 3% of total CYP in liver. Not inducible. Prefers lipophilic amines |
|
|
Term
|
Definition
Minimal drugs. Expressed in liver, kidney, intestine, and lung. Inducible by alcohol, diet, and diabetes. Polymorphism in Chinese people. |
|
|
Term
|
Definition
Most abundant CYP in liver. Found in liver, intestine, kidney, brain, uterus, and placenta. Metabolizes 33% of clinically important drugs. Inducible. Inhibited by erythromycin. Activates aflatoxin B1 and Benzo[a]pyrene. Prefers lipophilic drugs. |
|
|
Term
List the CYPs in order of drug metabolisms |
|
Definition
|
|
Term
What factors can affect CYP drug metabolism? |
|
Definition
Interindividual variation, induction, inhibition, genetic polymorphisms |
|
|
Term
What is the consequence of induction of CYP450? |
|
Definition
Altered PK and PD, increased rates of metabolism, enhanced activation of procarcinogens |
|
|
Term
|
Definition
Phenobarbital, rifampin, cigarette smoke, brussel sprouts, cabbage, cauliflower, alcohol, St. John’s Wort, omeprazole, bile acids, Vitamin D |
|
|
Term
What receptor does Omeprazole induce? |
|
Definition
Aryl Hydrocarbon Receptor (AHR) |
|
|
Term
What does Phenobarbital induce? |
|
Definition
a. Constitutive androstane receptor (CAR) |
|
|
Term
119) What does Rifampin induce? |
|
Definition
a. Pregnane X Receptor (PXR) |
|
|
Term
120) What does bile acid induce? |
|
Definition
a. Farnesoid X receptor (FXR |
|
|
Term
121) What does Vitamin D induce? |
|
Definition
a. Vitamin D Receptor (VDR |
|
|
Term
122) What do fibrates induce? |
|
Definition
a. Peroxisome proliferators activated receptor (PPAR) |
|
|
Term
123) What does all-trans-retinoic acid induce? |
|
Definition
a. Retinoic acid receptor (RAR) |
|
|
Term
124) What does 9-cis-retinoic acid induce? |
|
Definition
a. Retinoid X Receptor (RXR) |
|
|
Term
125) Which compounds cause reversible CYP inhibition? |
|
Definition
a. Fluroquinolones, cimetidine, antifungals, and quinidine |
|
|
Term
126) What compounds cause irreversible CYP inhibition? |
|
Definition
a. Macrolide antibiotics (trolendomycin, erythromycin, and clarithromycin), chloarmphenicol, cyclophosphamide, and spironolactone |
|
|
Term
127) What are molecular mechanisms of genetic polymorphisms? |
|
Definition
a. Single nucleotide polymorphism substitution (SNPs) and Indels |
|
|
Term
128) What are the three types of SNPs |
|
Definition
a. Coding nonsynoymous, coding synonymous, and noncoding |
|
|
Term
129) What is a coding nonsynoymous SNP? |
|
Definition
a. Change in nucleotide sequence with a change in amino acid composition. Change in affinity or activity |
|
|
Term
130) What is a coding synonymous SNP? |
|
Definition
a. Change in nucleotide sequence without a change in amino acid composition. Changes in transcript stability or splicing. |
|
|
Term
131) What is a noncoding SNP? |
|
Definition
a. Change in nucleotide sequence, change in promoter or other regulatory regions. |
|
|
Term
|
Definition
Insertion/deletion polymorphisms. Same effects as SNPs. Can be short repeats or larger deletions/insertions. |
|
|
Term
133) Which CYPs are polymorphic? |
|
Definition
|
|
Term
|
Definition
a. Metabolizes nicotine to conicotine. Better to be a poor metabolizer |
|
|
Term
135) What do polymorphisms in 2C19 cause? |
|
Definition
a. Large variability in therapeutic response to mephenytoin |
|
|
Term
136) What happens to Benzo[a]pyrene? |
|
Definition
a. It can be oxidized by many different routes. If it becomes the 4,5-dihydrolol or an ortho-quinone, it is non-toxic. However, if it becomes (+)benzo[a]pyrene-7,8-dihydrolol-9,10-epoxide, it is toxic. |
|
|
Term
|
Definition
a. Flavin-containing monooxygenases. 5 different isozymes. NADPH and oxygen dependent. Located in smooth endoplasmic reticulum. Most xenobiotics that are substrates for CYP450 are also substrates for FMO but not vice versa. |
|
|
Term
138) What types of compounds do FMOs oxygenate? |
|
Definition
a. Nitrogen compounds and sulfur compounds |
|
|
Term
139) What are MAOs and where are they found? |
|
Definition
a. Monoamine Oxidases, found almost all tissues. There are two forms, A and B, and most tissues contain both. Placenta contains only A and Platelets and lymphocytes contain only B. They are flavin containing. |
|
|
Term
140) What are the substrates for MAOs? |
|
Definition
a. Primary, secondary, and tertiary amines. |
|
|
Term
141) What are two nonspecific inhibitors for MAOs? What inhibits MAO-A specifically? What inhibits MAOB specifically? |
|
Definition
a. Nonspecific: phenelzine and tranylcypromine. Specific for A: clorgyline. Specific for B: pargyline and selegiline. |
|
|
Term
142) Describe alcohol dehydrogenase |
|
Definition
9 subunits that form heterodimers and homodimers. NADH dependent. |
|
|
Term
143) What are the substrates for alcohol dehydrogenase? |
|
Definition
a. Diverse primary and secondary alcohols. They convert alcohols to aldehydes. |
|
|
Term
144) Where are the highest concentrations of alcohol dehydrogenases? |
|
Definition
a. Liver, kidney, lung, and gastric mucosa |
|
|
Term
145) What is responsible for the difficulty processing alcohol in some Asian people? |
|
Definition
|
|
Term
146) Describe aldehyde dehydrogenase |
|
Definition
a. 12 different forms. Cystolic, mitochondria, and microsomes. NADH dependent. Reversible |
|
|
Term
147) What are the substrates for aldehyde dehydrogenase? |
|
Definition
|
|
Term
148) What are involved in hydrolysis? |
|
Definition
a. Carboxylesterases – cholinesterase, pseudocholinesterase, arylcarboxyesterase, liver microsomal esterase. |
|
|
Term
149) What are the substrastes of the hydrolysises? |
|
Definition
a. Carboxylic acid esters, amide, and thioesters. Note: amides more resistant to metabolism than esters. |
|
|
Term
150) What are the conjugation reactions? |
|
Definition
a. Glucuronidation, sulfation, amino acid conjugation, acetylation, glutathione conjugation, methylation |
|
|
Term
151) What are the consequences of conjugation reactions? |
|
Definition
a. Increase water solubility and polarity. Activation (morphine and minoxidil) and inactivation, toxicity. |
|
|
Term
152) What functional groups are conjugated and what can be done to them? |
|
Definition
a. Hydroxyls are sulfated, glucuronidated, and methylated. Amines are acetylated, sulfated, and glucuronidated. Carboxyls are amino acid conjugated and glucuronidated. |
|
|
Term
153) What is the major and most common conjugation reaction? |
|
Definition
|
|
Term
154) What does glucutonidation require? |
|
Definition
a. UDP=glucuronosyl transferase and uridine diphosphate glucuronic acid. |
|
|
Term
155) Where is glucuronidation located? |
|
Definition
a. Microsomes of liver and other tissues, near CYP450s |
|
|
Term
156) What endogenous substances are glucuronidated? |
|
Definition
a. Thyroxine, bilirubin, steroids |
|
|
Term
157) What is involved in sulfation? |
|
Definition
a. Sulfotransferases and phophoadenosine phosphosulfate (PAPS |
|
|
Term
|
Definition
a. Cytolsol and other tissues |
|
|
Term
159) What are the substrates of sulfation? |
|
Definition
a. Ones that contain hydroxyl, amines, or N-oxides |
|
|
Term
160) What endogenous substances are sulfated? |
|
Definition
a. Steroids, catecholamine, thyroxine |
|
|
Term
161) What is SULT 2 specific for? What is SULT 1 specific for? |
|
Definition
|
|
Term
162) What is the deciding factor between glucuronidation and sulfation? |
|
Definition
a. Sulfation has a higher affinity but lower capacity than glucuronidation. |
|
|
Term
163) What is the rate limiting step in sulfation? |
|
Definition
|
|
Term
164) What does amino acid conjugation do and what does it do it to? |
|
Definition
It adds amino acids (mostly glycine, glutamine, and taurine) to substrates that contain a carboxylic acid group. Drug is conjugated to its coenzyme thioester. |
|
|
Term
165) What are the substrates of acetylation? |
|
Definition
a. Primary aliphatic or aromatic amines on amino acids, hydrazines, sulfonamides. |
|
|
Term
166) Polymorphisms in NAT1 and NAT2 can cause what? |
|
Definition
a. Toxicity. Examples: Isoniazid and Dapsone – peripheral neuropathy. Hydralazine and procainamine – lupus. Sulfasalazine – hematologic disorders. |
|
|
Term
167) What is glutathione? |
|
Definition
a. Cytoprotectant tripeptide. Gamma-glutamyl-cysteinyl-glycine. |
|
|
Term
168) What are the two glutathione transferases? |
|
Definition
a. GTSM1 (anticancer drugs) and GSTT1 (small organic molecules, solvents, halocarbons, electrophiles) |
|
|
Term
169) What are the substrates in glutathione conjugation? |
|
Definition
a. Electrophiles (like epoxides) |
|
|
Term
170) What does glutathione conjugation result in? |
|
Definition
a. Mercapturic acid derivative |
|
|
Term
171) Describe methylation. |
|
Definition
a. Common rxn for endogenous compounds. Minor for xenobiotics. It can increase lipophilicty and pharmacological activity. Metabolizes endogenous neurotransmitters, like norepinephrine. |
|
|
Term
172) Describe O-methylation. |
|
Definition
a. Can decrease water solubility. Catechol-o-methyltransferases – catecholamines, steroids. Hydroxyindole-o-methyltransferase – serotonin |
|
|
Term
173) Describe N-methylation. |
|
Definition
a. Phenyethanolamine-N-methyltransferase – norepinesphrine. Histamine-N-methyltransferase – histamine. Amine-N-methyltransferase – serotonin, dopamine, amphetamine |
|
|
Term
174) Describe S-methylation |
|
Definition
a. Microsomal. Disulfram, propylthiouracil, captopril, penicilamine, 6-mercaptopurine. |
|
|
Term
175) What molecular weight determines if something is enteropathically cycled? |
|
Definition
|
|
Term
176) Describe intestinal metabolism |
|
Definition
a. CYP450 (esp CYP3A4), glucuronidation, sulfation, glutathione s-transferases. The further into the GI tract, the less metabolism. |
|
|
Term
177) Describe lung metabolism. |
|
Definition
a. CYP2E1, FMO, conjugation reactions |
|
|
Term
178) Describe nasal metabolism |
|
Definition
a. Very active CYP450s, FMO, carboxylesterase, conjugation reactions. |
|
|
Term
179) What are the principles of toxicology? |
|
Definition
a. Spectrum of toxic dose, duration and frequency of exposure, local versus systemic, reversible and irreversible, delayed toxicity, spectrum of undesired effects, chemical allergic reactions, and idiosyncratic reactions. |
|
|
Term
180) What are the mechanisms of toxicology? |
|
Definition
a. High affinity binding to macromolecules, biotransformation to reactive intermediate |
|
|
Term
181) How many hospitalized and outpatients have ADRs? |
|
Definition
a. ~15% of hospitialized patients and an estimated 15% of outpatients have ADRs |
|
|
Term
182) How many deaths per year are attributed to ADRs? |
|
Definition
a. Approximately 106,000 deaths per year. |
|
|
Term
183) What are the predictable ADRs? |
|
Definition
a. Overdose, known side effects, and secondary effects. |
|
|
Term
184) What are the unpredictable ADRs? |
|
Definition
a. Intolerance, idiosyncratic, allergic, and pseudoallergic |
|
|
Term
185) Of the two, which ADRs are most troublesome? |
|
Definition
|
|
Term
186) When do drug hypersensitivity reactions NOT occur? |
|
Definition
a. When drug dose is less than 10 mg/day |
|
|
Term
187) How heavy must compounds be to illict a response? Why do small molecules illict a response? |
|
Definition
a. 1000-2000 Daltons. The active metabolite or the drug are binding to biomolecules and iliciting a response through haptenation. |
|
|
Term
188) What causes a protein to be recognized as foreign? |
|
Definition
a. A change in conformation |
|
|
Term
189) What are the functional groups capable of causing immuno-hypersensitivity reactions? |
|
Definition
a. Ring-strained beta-lactams, epoxides, alpha,beta-unsaturated carbonyl and imino compounds capable of undergoing Michael reactions, anilines, and thiols. |
|
|
Term
190) What reacts with beta lactams? |
|
Definition
a. Nucleophiles, such as amino groups on proteins, can form a new acyclic amide bond. |
|
|
Term
191) What results in epoxides? |
|
Definition
a. Aromatic hydroxylation reactions |
|
|
Term
191) What results in epoxides? |
|
Definition
a. Aromatic hydroxylation reactions |
|
|
Term
192) What can epoxides react with? |
|
Definition
a. Cellular nucleophiles such as NHR or SH |
|
|
Term
193) What is the aromatic anticonvulsive syndrome? |
|
Definition
a. Phenytoin, Phenobarbital, and carbamazepine all undergo aromatic hydroxylation via an epoxide. A patient who has a reaction to one has a 40-60% chance of having a reaction to the other two. This is called cross-reactivity. |
|
|
Term
194) What is an example of an alpha,beta-unsaturated carbonyl or imino group capable of undergoing a Michael reaction? |
|
Definition
a. The addition of an electrophilic SH (cysteine) or NH2 (lysine) to uroshiols that causes poison ivy. |
|
|
Term
195) What is special about anilines? |
|
Definition
a. Almost all primary aromatic amines given at dosages of 10mg/day or more will have a significant incidence of immune mediated hypersensitivity reactions, regardless of the rest of the structure. |
|
|
Term
196) What are some drugs that are known to be mutagens? |
|
Definition
a. Epoxides, alkylating agents, alkanesulfonates, nitroso compounds, planar polycyclic aromatic hydrocarbons (via intercalation), analogs of DNA bases, and anilines. |
|
|