Pharmaceutical Chemistry – Drug Discovery/Drug Metabolism – Flashcards

Unlock all answers in this set

Unlock answers
question
Approximately how long is the development and pre-clinical testing process?
answer
6.5 Years
question
How long are Phase I clinical trials?
answer
1 year
question
How long are Phase II clinical trials?
answer
2 years
question
Which of the two (phase I or phase II) has more participants?
answer
Phase II
question
How long are Phase III trials?
answer
3 years
question
Approximately how many participants are in Phase III trials?
answer
1,000 to 3,000 people
question
What is the average cost for the development of a drug?
answer
$2.5 billion
question
How long do patents last?
answer
20 years.
question
How many marketed drugs manage to recoup the cost of development?
answer
~30%
question
What are some reasons drugs fail?
answer
Clinical safety, efficacy, formulation, PK/bioavailability, commercial, toxicology, cost of goods
question
Generally, where is drug discovery performed?
answer
Academic settings
question
Generally, where is drug development performed?
answer
Business settings
question
What determines the biological activities of compounds?
answer
Their chemistry
question
What chemical properties affect biological activity?
answer
Physiochemical (lipophilicity, ionization, solubility), Electronic (resonance, inductive effects, bonding potential), and spatial (molecular dimension, stereochemistry)
question
How are new drugs created from old drugs?
answer
Alteration of side chains to improve something (PK, dosage form, etc.)
Example: Captropil.
question
How is biological information used to create drugs?
answer
Study of indigenous medications, study of warfare chemicals (nitrogen mustards), observations in in vivo testing (vinca alkaloids), or observations of clinical side effects (Viagra)
question
What is the modern approach to drug development?
answer
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.
question
What are the steps in a drug development project?
answer
Target selection/validation, assay development, identification of a lead compound, lead optimization, preclinical evaluation, and clinical trials
question
What is the most critical stage of drug development?
answer
Target selection/validation. One wants targets that are relevant and selective
question
What are traditional strategies for target selection?
answer
Study disease biology, key regulatory proteins, response to previous drugs, and engineered models
question
Most drug targets are and ?
answer
Receptors and enzymes
question
Why are other targets, like DNA or nuclear receptors, not as common?
answer
Drugs not as selective
question
What is functional genomics?
answer
Genetic analyses for disease genes and protective genes, by DNA microarray analyses. Strategy for target selection.
question
Methods for target validation?
answer
Biology of disease, gene knock-out/in, constitutively-active/domiant-negative mutants, antisense, ribozymes, RNA interference
question
What is RNA interference?
answer
Selectively decreases levels of expression of specific mRNA in a cell. Mimics the ‘perfect’ drug.
question
What is important in assay development?
answer
Relevance, selectivity, sensitivity, reproducibility, speed, and cost.
question
Activities of signaling proteins are affected by what?
answer
Subcellular localization
question
What are the two basic approaches to lead compound identification?
answer
Computation modeling and screening
question
Rational drug design?
answer
Substrate analogs to act as enzyme inhibitors
question
Computer-aided drug design downside?
answer
Need to know structure of target
question
Screening?
answer
Testing collections of compounds for biological activity, don’t need to know structure. ‘Hits’ = CxD
question
What is “D”?
answer
Chemical diversity of compound collection.
question
What are some sources of compounds for lead discovery?
answer
Natural products, synthetic compounds
question
What is combination chemistry?
answer
Assembly of molecular building blocks. Uses high-throughput synthesis. Requires high-throughput screening for lead discovery
question
If there are 5 sites for substitution, and 20 building blocks, how many potential compounds can be created?
answer
20^5 > 3,200,000
question
What is a benefit of robotic screening systems?
answer
Decreased time, decreased error
question
What are the goals of lead optimization?
answer
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).
question
What do SARs define?
answer
The pharmacophore
question
What is the pharmacophore?
answer
3D arrangement of chemical determinants that are necessary for binding to the receptor
question
What is traditional lead optimization?
answer
Synthesize homologous series, or use ‘chemical insight’ or bioisterism rules. Synthesis of few compounds. Low-throughput screening, high cost, slow progress
question
Lead optimization of combinatorial chemistry?
answer
Synthesis of ‘focused libraries’, may/may not require high-throughput screening, high start up costs, but fast and efficient.
question
Lead optimization by computer-aided drug design?
answer
Analysis of receptor on pharmacophore. Allows for QSAR analyses and drug design without knowledge of protein structure
question
What is QSAR?
answer
a. Quantitative structural activity relationships.
b. Mathematical relationship between biological activity and chemical properties and allows for prediction of activity of new compounds
question
The Hansch Approach?
answer
QSAR, biological activity = f . Limits the number of compounds that are made.
question
Computational ADME?
answer
Predicts solubility, oral bioavailability, biodistribution, and metabolism
question
What is tested in in vitro studies?
answer
Chemical purity and stability, formulation, mechanism of action, and cellular pharmacology
question
What is tested in in vivo studies?
answer
Efficacy in animals, short-term and long-term toxicity, carcinogenicity, metabolism, PK, and biodistribution
question
What is the final step in drug development?
answer
Clinical trials
question
What is a clinical trial?
answer
A prospective study comparing the value of intervention against a control in humans
question
What is required of a clinical trial?
answer
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
question
What must be included in the IND application?
answer
Descriptions of the manufacture of the drug, its pharmacology and toxicology in animals, and a detailed protocol for clinical trials.
question
What department accepts or rejects INDs? How long does the process take?
answer
The CDER. If, after 30 days, no rejection is received, the study has implicit approval to proceed.
question
What are the goals of Phase I clinical trials?
answer
Primarily to establish maximum tolerated dose in humans. Secondarily to examine pharmacokinetics in humans. Phase I is not for treating the disease.
question
Who are the subjects in Phase I trials?
answer
20-50 Mostly healthy volunteers or patients who did not respond to other treatments (especially in cancer and AIDS cases)
question
What is the classical procedure for determining the maximum tolerated dose in humans?
answer
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.
question
What are the goals of phase II clinical trials?
answer
To determine the efficacy of the drug and to continue monitoring for toxicity.
question
Who are the subjects in Phase II trials?
answer
50-300 patients with carefully matched disease
question
What is the procedure of a Phase II trial?
answer
Two-stage design. Drug given at dose determined by Phase I. Blinded design. May be uncontrolled in cancer drug studies
question
What are the goals of Phase III?
answer
To verify efficacy and safety
question
Who are the subjects in Phase III trials?
answer
100s-1000s of patients with carefully matched disease
question
What is the procedure of a Phase III trial?
answer
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
question
Who conducts the final review process? How long does it take?
answer
The CDER. Less than one year.
question
What is Phase IV?
answer
Continued monitoring of the drug post-marketing
question
In the past, drug discovery was essentially what kind of process?
answer
Random, observational and relied on finding a molecule that worked and figuring out how it worked later
question
Who discovered penicillin?
answer
Alexander Fleming in 1929
question
What are drug targets?
answer
Biomolecules whose functions can be artificially modulated
question
What technologies enabled the new methods of drug discovery?
answer
Molecular biology, in vitro assays, and 3D structural models of drugs and their receptors.
question
In drug design and discovery, what is the difference between high-throughput screening and computer prediction models?
answer
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.
question
What forces drive states of matter and the 3D conformation of macromolecules?
answer
Interatomic forces
question
What are interatomic forces?
answer
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)
question
What are hydrogen bonding interactions?
answer
It occurs when two electronegative atoms compete for the same hydrogen, and create an electron deficient hydrogen.
question
What are some H-bond donors? Some H-bond acceptors?
answer
a. Donors: NH, OH
b. Acceptors: C=O, OH
question
Why are hydrogen bonds important in drug discovery?
answer
Influence interactions between drug and solvent, drug and target, and drug-to-drug
question
What is the hydrophobic effect?
answer
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.
question
What drives binding?
answer
Complementarity between ligand and receptor – like a lock and key.
question
What are the two types of complementarity?
answer
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.
question
The affinity of a drug is described as what?
answer
The binding energy, which is the sum of the forces between the drug and the receptor
question
What is the equation for binding energy?
answer
dG = dH – TdS, where H is enthalpy and S is entropy.
question
Where do enthalpy and entropy come from?
answer
Enthalpy results from forces within and between molecules. Entropy comes from solvent molecules and movement and rotation of bonds
question
What is protein X-ray crystallography and how is it used in drug design?
answer
X-rays shined through molecule. Diffraction finds regions with high concentrations of electrons. These regions are used to assign positions to atoms.
question
What is molecular modeling (computational chemistry)?
answer
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
question
What is the pharmacophore?
answer
The features of a drug necessary to ensure optimal interactions with a biological target and to trigger or block it’s response
question
How is virtual (computer) screening useful?
answer
Matches the 3D structure of potential drug with the shape of the receptor
question
What is high-throughput docking?
answer
Fitting the molecule into the receptor pocket and then scoring it based on interactions in 3D structure.
question
What are the goals of substrate metabolism?
answer
Increase water solubility and polarity, deactivation and excretion, or activation.
question
What are some common sites of biotransformation?
answer
The liver (main), intestinal mucosa, kidney, and lungs
question
What types of reactions are Phase I?
answer
Oxidation, hydroxylation, reduction, and hydrolysis
question
What are the goals of Phase I reactions?
answer
Introducing a new functional group, modifying an exisiting functional group, or exposing an existing functional group (more than one may occur).
question
What are the Enzymes involved in Phase I reactions?
answer
Cytochrome P450 and Flavin-containing monooxygenases
question
What are Phase II reactions?
answer
Conjugations: acetylation, glucuronidation, sulfation, and amino acid conjugation
question
What is the goal of Phase II reactions?
answer
To conjugate an endogenous molecule to a functional group to increase polarity or water solubility to facilitate excretion
question
What is the most common reaction in xenobiotic metabolism? Which enzyme performs is?
answer
Oxidation CYP450s
question
Where is the highest concentration of CYP450s?
answer
In the liver
question
Where are the CYP450s located?
answer
Most are on microsomes, some are on mitochondria.
question
What type of proteins are CYP450s?
answer
Heme proteins with an iron protoporphyrin ring.
question
What components are required for CYP450 systems?
answer
Oxygen, NADPH, NADPH-CYP450 reductase, and a lipid environment
question
Describe the catalytic pathway for CYP450.
answer
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.
question
What are some hydroxylation mechanisms catalyzed by CYP450?
answer
Aromatic hydroxylation, aliphatic hydroxylation, deamination, O-Dealkylation, N-dealkylation, and sulfoxidation
question
What does decreasing metabolism do?
answer
It decreases toxicity. Example: in decreasing toxicity and metabolism levels: methoxyflurane, halothane, enflurane, isoflurane, desflurane.
question
What is particularly interesting about haloflurane?
answer
At two doses, can cause haloflurane hepatitis
question
Which are the most predominant CYP genes?
answer
CYP3A4/5, 2C, and 1A2
question
What does CYP2A1 generally metabolize?
answer
Environmental compounds
question
What is CYP2E1 specific for?
answer
Small molecules, ethanol, anesthetics, small organic solvents
question
Which isoforms metabolize most drugs?
answer
3A4/5, 2D6, and 2C8/9
question
Family 1 metabolizes what?
answer
Environmental carcinogens, in particular aromatic hydrocarbons and aryl amines
question
Describe CYP1A1
answer
Aromatic hydrocarbon hydroxylase, primarily expressed in extrahepatic tissues (small intestine, lung, placenta). Inducible. Variation.
question
Describe CYP1A2
answer
Expressed in liver, stomach, and intestine. Inducible. Variation.
question
Describe CYP2A6
answer
Found in liver, lung, and nasal epithelium. Metabolizes drugs (coumarin), procarcinogens, and nicotine. Polymorphisms.
question
Describe CYP2C
answer
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.
question
Describe CYP2D6
answer
Polymorphisms. Metabolizes 21% of clinically important drugs. 3% of total CYP in liver. Not inducible. Prefers lipophilic amines
question
Describe CYP2E1
answer
Minimal drugs. Expressed in liver, kidney, intestine, and lung. Inducible by alcohol, diet, and diabetes. Polymorphism in Chinese people.
question
Describe CYP3A4
answer
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.
question
List the CYPs in order of drug metabolisms
answer
CYP3A4, CYP2CX, CYP2D6.
question
What factors can affect CYP drug metabolism?
answer
Interindividual variation, induction, inhibition, genetic polymorphisms
question
What is the consequence of induction of CYP450?
answer
Altered PK and PD, increased rates of metabolism, enhanced activation of procarcinogens
question
What can induce CYP450?
answer
Phenobarbital, rifampin, cigarette smoke, brussel sprouts, cabbage, cauliflower, alcohol, St. John’s Wort, omeprazole, bile acids, Vitamin D
question
What receptor does Omeprazole induce?
answer
Aryl Hydrocarbon Receptor (AHR)
question
What does Phenobarbital induce?
answer
a. Constitutive androstane receptor (CAR)
question
119) What does Rifampin induce?
answer
a. Pregnane X Receptor (PXR)
question
120) What does bile acid induce?
answer
a. Farnesoid X receptor (FXR
question
121) What does Vitamin D induce?
answer
a. Vitamin D Receptor (VDR
question
122) What do fibrates induce?
answer
a. Peroxisome proliferators activated receptor (PPAR)
question
123) What does all-trans-retinoic acid induce?
answer
a. Retinoic acid receptor (RAR)
question
124) What does 9-cis-retinoic acid induce?
answer
a. Retinoid X Receptor (RXR)
question
125) Which compounds cause reversible CYP inhibition?
answer
a. Fluroquinolones, cimetidine, antifungals, and quinidine
question
126) What compounds cause irreversible CYP inhibition?
answer
a. Macrolide antibiotics (trolendomycin, erythromycin, and clarithromycin), chloarmphenicol, cyclophosphamide, and spironolactone
question
127) What are molecular mechanisms of genetic polymorphisms?
answer
a. Single nucleotide polymorphism substitution (SNPs) and Indels
question
128) What are the three types of SNPs
answer
a. Coding nonsynoymous, coding synonymous, and noncoding
question
129) What is a coding nonsynoymous SNP?
answer
a. Change in nucleotide sequence with a change in amino acid composition. Change in affinity or activity
question
130) What is a coding synonymous SNP?
answer
a. Change in nucleotide sequence without a change in amino acid composition. Changes in transcript stability or splicing.
question
131) What is a noncoding SNP?
answer
a. Change in nucleotide sequence, change in promoter or other regulatory regions.
question
132) What are Indels?
answer
Insertion/deletion polymorphisms. Same effects as SNPs. Can be short repeats or larger deletions/insertions.
question
133) Which CYPs are polymorphic?
answer
a. 2A6, 2C9, 2D6, 2C19
question
134) What does 2A6 do?
answer
a. Metabolizes nicotine to conicotine. Better to be a poor metabolizer
question
135) What do polymorphisms in 2C19 cause?
answer
a. Large variability in therapeutic response to mephenytoin
question
136) What happens to Benzo[a]pyrene?
answer
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.
question
137) Describe FMOs.
answer
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.
question
138) What types of compounds do FMOs oxygenate?
answer
a. Nitrogen compounds and sulfur compounds
question
139) What are MAOs and where are they found?
answer
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.
question
140) What are the substrates for MAOs?
answer
a. Primary, secondary, and tertiary amines.
question
141) What are two nonspecific inhibitors for MAOs? What inhibits MAO-A specifically? What inhibits MAOB specifically?
answer
a. Nonspecific: phenelzine and tranylcypromine. Specific for A: clorgyline. Specific for B: pargyline and selegiline.
question
142) Describe alcohol dehydrogenase
answer
9 subunits that form heterodimers and homodimers. NADH dependent.
question
143) What are the substrates for alcohol dehydrogenase?
answer
a. Diverse primary and secondary alcohols. They convert alcohols to aldehydes.
question
144) Where are the highest concentrations of alcohol dehydrogenases?
answer
a. Liver, kidney, lung, and gastric mucosa
question
145) What is responsible for the difficulty processing alcohol in some Asian people?
answer
a. ADH2*2 (atypical ADH)
question
146) Describe aldehyde dehydrogenase
answer
a. 12 different forms. Cystolic, mitochondria, and microsomes. NADH dependent. Reversible
question
147) What are the substrates for aldehyde dehydrogenase?
answer
a. Aldehydes.
question
148) What are involved in hydrolysis?
answer
a. Carboxylesterases – cholinesterase, pseudocholinesterase, arylcarboxyesterase, liver microsomal esterase.
question
149) What are the substrastes of the hydrolysises?
answer
a. Carboxylic acid esters, amide, and thioesters. Note: amides more resistant to metabolism than esters.
question
150) What are the conjugation reactions?
answer
a. Glucuronidation, sulfation, amino acid conjugation, acetylation, glutathione conjugation, methylation
question
151) What are the consequences of conjugation reactions?
answer
a. Increase water solubility and polarity. Activation (morphine and minoxidil) and inactivation, toxicity.
question
152) What functional groups are conjugated and what can be done to them?
answer
a. Hydroxyls are sulfated, glucuronidated, and methylated. Amines are acetylated, sulfated, and glucuronidated. Carboxyls are amino acid conjugated and glucuronidated.
question
153) What is the major and most common conjugation reaction?
answer
a. Glucuronidation.
question
154) What does glucutonidation require?
answer
a. UDP=glucuronosyl transferase and uridine diphosphate glucuronic acid.
question
155) Where is glucuronidation located?
answer
a. Microsomes of liver and other tissues, near CYP450s
question
156) What endogenous substances are glucuronidated?
answer
a. Thyroxine, bilirubin, steroids
question
157) What is involved in sulfation?
answer
a. Sulfotransferases and phophoadenosine phosphosulfate (PAPS
question
158) Where is sulfation?
answer
a. Cytolsol and other tissues
question
159) What are the substrates of sulfation?
answer
a. Ones that contain hydroxyl, amines, or N-oxides
question
160) What endogenous substances are sulfated?
answer
a. Steroids, catecholamine, thyroxine
question
161) What is SULT 2 specific for? What is SULT 1 specific for?
answer
a. Alcohols. Phenols.
question
162) What is the deciding factor between glucuronidation and sulfation?
answer
a. Sulfation has a higher affinity but lower capacity than glucuronidation.
question
163) What is the rate limiting step in sulfation?
answer
a. PAPS
question
164) What does amino acid conjugation do and what does it do it to?
answer
It adds amino acids (mostly glycine, glutamine, and taurine) to substrates that contain a carboxylic acid group. Drug is conjugated to its coenzyme thioester.
question
165) What are the substrates of acetylation?
answer
a. Primary aliphatic or aromatic amines on amino acids, hydrazines, sulfonamides.
question
166) Polymorphisms in NAT1 and NAT2 can cause what?
answer
a. Toxicity. Examples: Isoniazid and Dapsone – peripheral neuropathy. Hydralazine and procainamine – lupus. Sulfasalazine – hematologic disorders.
question
167) What is glutathione?
answer
a. Cytoprotectant tripeptide. Gamma-glutamyl-cysteinyl-glycine.
question
168) What are the two glutathione transferases?
answer
a. GTSM1 (anticancer drugs) and GSTT1 (small organic molecules, solvents, halocarbons, electrophiles)
question
169) What are the substrates in glutathione conjugation?
answer
a. Electrophiles (like epoxides)
question
170) What does glutathione conjugation result in?
answer
a. Mercapturic acid derivative
question
171) Describe methylation.
answer
a. Common rxn for endogenous compounds. Minor for xenobiotics. It can increase lipophilicty and pharmacological activity. Metabolizes endogenous neurotransmitters, like norepinephrine.
question
172) Describe O-methylation.
answer
a. Can decrease water solubility. Catechol-o-methyltransferases – catecholamines, steroids. Hydroxyindole-o-methyltransferase – serotonin
question
173) Describe N-methylation.
answer
a. Phenyethanolamine-N-methyltransferase – norepinesphrine. Histamine-N-methyltransferase – histamine. Amine-N-methyltransferase – serotonin, dopamine, amphetamine
question
174) Describe S-methylation
answer
a. Microsomal. Disulfram, propylthiouracil, captopril, penicilamine, 6-mercaptopurine.
question
175) What molecular weight determines if something is enteropathically cycled?
answer
a. 300-500 MW
question
176) Describe intestinal metabolism
answer
a. CYP450 (esp CYP3A4), glucuronidation, sulfation, glutathione s-transferases. The further into the GI tract, the less metabolism.
question
177) Describe lung metabolism.
answer
a. CYP2E1, FMO, conjugation reactions
question
178) Describe nasal metabolism
answer
a. Very active CYP450s, FMO, carboxylesterase, conjugation reactions.
question
179) What are the principles of toxicology?
answer
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.
question
180) What are the mechanisms of toxicology?
answer
a. High affinity binding to macromolecules, biotransformation to reactive intermediate
question
181) How many hospitalized and outpatients have ADRs?
answer
a. ~15% of hospitialized patients and an estimated 15% of outpatients have ADRs
question
182) How many deaths per year are attributed to ADRs?
answer
a. Approximately 106,000 deaths per year.
question
183) What are the predictable ADRs?
answer
a. Overdose, known side effects, and secondary effects.
question
184) What are the unpredictable ADRs?
answer
a. Intolerance, idiosyncratic, allergic, and pseudoallergic
question
185) Of the two, which ADRs are most troublesome?
answer
a. The unpredictable.
question
186) When do drug hypersensitivity reactions NOT occur?
answer
a. When drug dose is less than 10 mg/day
question
187) How heavy must compounds be to illict a response? Why do small molecules illict a response?
answer
a. 1000-2000 Daltons. The active metabolite or the drug are binding to biomolecules and iliciting a response through haptenation.
question
188) What causes a protein to be recognized as foreign?
answer
a. A change in conformation
question
189) What are the functional groups capable of causing immuno-hypersensitivity reactions?
answer
a. Ring-strained beta-lactams, epoxides, alpha,beta-unsaturated carbonyl and imino compounds capable of undergoing Michael reactions, anilines, and thiols.
question
190) What reacts with beta lactams?
answer
a. Nucleophiles, such as amino groups on proteins, can form a new acyclic amide bond.
question
191) What results in epoxides?
answer
a. Aromatic hydroxylation reactions
question
191) What results in epoxides?
answer
a. Aromatic hydroxylation reactions
question
192) What can epoxides react with?
answer
a. Cellular nucleophiles such as NHR or SH
question
193) What is the aromatic anticonvulsive syndrome?
answer
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.
question
194) What is an example of an alpha,beta-unsaturated carbonyl or imino group capable of undergoing a Michael reaction?
answer
a. The addition of an electrophilic SH (cysteine) or NH2 (lysine) to uroshiols that causes poison ivy.
question
195) What is special about anilines?
answer
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.
question
196) What are some drugs that are known to be mutagens?
answer
a. Epoxides, alkylating agents, alkanesulfonates, nitroso compounds, planar polycyclic aromatic hydrocarbons (via intercalation), analogs of DNA bases, and anilines.
Get an explanation on any task
Get unstuck with the help of our AI assistant in seconds
New