1 Flashcards
1) What are the types of new drugs?
- New drug for novel use: New discovery about a disease state that may have no treatment before or this drug represents a new approach in treatment 2. A new drug that represents a new generation of another group of drugs already in use. 3. A new drug that is just another variation of a known drug (me-too)
2) What are the different approaches to target selection?
- Hypothesis driven (traditional approach): identify disease -> understand disease ->devise approach -> identify drugs that fit approach 2. Genomic approach: - Analysing the entire genetic information of an organism in the context of healthy vs disease state - Understanding genotype-phenotype correlation to identify disease target genes - Eg. Molecular basis of CML: Philadelphia chromosome encodes constitutively active tyrosine kinase, BCR-ABL. Imatinib blocks ATP binding and thus inhibits BCR-ABL kinase activity. 3. Post genomics approach-proteomics: Separation and characterization of proteins in an organism. Compare protein expression in normal vs disease state to identify novel target proteins.
3) How do you know that a gene/protein is targetable?
RNAi screens can be done. This is carried out by ‘knocking down’ individual genes to find genes that regulate key disease processes. This allows identification of targets by function. There are in vitro and in vivo approaches to this.
4) How is target validation carried out?
- In vitro: cell based assays to verify that the target gene/protein is involved in disease progression at the cellular level. Cell lines can be engineered to display loss of function (using siRNA/shRNA) or gain of function (overexpression). This allows analysis of specific roles of the target. 2. In vivo assays: Animal disease models that can be used to verify that your target gene/protein contributes to disease progression in a more complex environment. To study loss of function: inject mice with tumor cells, use shRNA to knock down a potential oncogene and observe tumor progression. To study gain of function, overexpress normal mice with potential oncogene and observe if tumors form.
5) Define a 1) HIT 2) LEAD 3) preclinical development candidate.
• HIT A compound that interacts with the chosen target at a given concentration (usually in the micromolar range) • LEAD A compound with drug-like properties, initial SAR and a promising IP position • Preclinical Development Candidate NCE with optimized pharmacological and pharmacokinetic properties and a secure IP position
6) What are the methods of lead identification? What are their benefits/drawbacks?
(full answer on p2) 1. Rational drug design: Understanding structure activity relationship. This requires coordination of structural biology and organic chemistry, and drug design is based on the pharmacophore of endogenous ligand/substrate. Designed to bind active site and block receptor activation or enzyme activity. Eg. BCR-ABL inhibition: Identification of c-ABL autoinhibitory mechanism. Myristoylation of N-Term of c-ABL causes binding of myristate moiety into deep hydrophobic pocket of kinase domain. This results in a 90 degree bending of the a-1 helix of the C-term and autoinhibition. BCR-ABL lacks N-term myristoylation site, but it can be replaced with allosteric inhibitors. 2. High throughput screening: Often relies on cell free/cell based assays– Target-specific effects are measured quantitatively by a reporter assay Ex: fluorescence, luminescence, cell shape, cell metabolism, color formation. Drug candidates are evaluated for ability to block activity. Formats: 96-well and 384-well plates (high-throughput), 1,536-well plates (ultra high-throughput (UHTS)). SAR is difficult to do without knowing the exact molecular target and mode of drug interaction.
7) What are the cell based/cell free approach to HTS? What is a robust screening assay?
Cell free: based on isolated target molecule (can be whole or active fragment). Examples are binding and enzymatic activity assays. Cell based: Cell based reporter gene assay. Disadvantage is that the drug hit may be acting either directly on target or indirectly be interfering pathway (up or downstream of target) Z’=1–(3s +3B)/(μs-μB) Z’>0.8 (very good) Z’>0.6 (good) Z’<0.5 (not robust for screening)
8) How does fragment based screening work?
Run diverse set of structures and identify those that bind to the target (does not have to be a perfect fit. Custom building the drug based on fragments that bind.
9) How are compound libraries created? What are the requirements for a good library?
– Acquisition from external vendors – Generation from chemical library synthesis • Random libraries • Focused libraries – Generation from medicinal chemistry efforts • Targeted synthesis • Combinatorial synthesis • A good library should be – Large – Diverse – Examples of libraries: FDA-approved drugs, Natural product libraries – Containing only “lead-like” or “drug-like”compounds • Non-reactive • No known toxic moieties • Following Lipinski’s Rule-of-5 • Aqueous soluble
1) What is Lipinski’s rule of 5?
- Fewer than 5 hydrogen bond donors 2. Fewer than 10 hydrogen-bond acceptors 3. A molecular weight of less than 500 daltons 4. A partitioning coefficient (logP) of less than 5
2) What is lipophilicity and how is it calculated?
Lipophilicity is the ability of a compound to partition between lipophilic organic phase (octanal) and polar aqueous phase (water) LogP =[Conc]octonal/[Conc]water LogP <1: poor permeability 1-3: moderate permeability 3-5: high permeability >5: high permeability
1) What are the key issues to be addressed in lead optimization?
- Efficacy 2. Potency (target affinity and PK parameters) 3. Adverse effects/toxicity profile 4. Route of administration (stability, absorption, distribution) 5. Onset and duration of action
2) What are the key criteria for a lead series?
• Binding/functional potency in primary assay (IC50 < 100nM) • Potency in secondary assay (cell proliferation GI50 <500nM) • Meets Lipinski rules (of 5) (MW<500, cLogP<5) • In vitro ADME liabilities (tó >60min) • Synthesis in less than 10 steps • Multiple points of modification • Patentable
3) Explain chemical modification.
The goal of chemical modifications is to determine which functional groups are important for biological activity. The procedure is to alter or remove functional groups using chemical synthesis and test the activity of the altered molecule. Bioisosteric replacement involves substitution of atoms or groups of atoms in a the parent molecule to produce compounds with broadly similar biological properties to the parent with structural diversity.
4) What are the factors that affect absorption and permeability?
- Route of administration: Oral administration is the most convenient and cost-effective. Absorption takes place mostly from the small intestine. 2. Rate of dissolution (tablet, capsule, suspension or solution): 3. Speed of uptake by GI tract: Dependent on the lipophilicity and extent of ionization of the drug. 4. Drug complex with dissolved food.
5) What happens after the drug is absorbed?
It passes through the portal vein and enters the liver, where is may be metabolized.
6) What factors affect solubility and stability?
Solubility requires adherence to Lipinski rule of 5. Stability is measured at different pH and temperatures. Eg. Orally available gemcitabine: prodrug mediates oral-mediated absorption of gemcitabine with less toxicity. Minimal hydrolysis of prodrug to gemcitabine at low pH.
7) What is bioavailabilty and why is it important? How is it calculated?
Bioavailabilty is the fraction of unchanged drug that enters systemic circulation. It should be studied as early as possible because a lack of desired response may be due to lack of bioavailability (not reaching the required drug concentration). Compounds can be suitably modified to maximize bioavailability. F=[AUC(test) x D(iv)]/[AUC(iv) x D(test)] X 100%
8) How does drug distribution affect drug response?
A drug can be distributed to tissues/organs from the bloodstream. Different drug concentrations are attained in different tissues/organs. A drug may be preferentially distributed to its target tissue/organ or not at all.
9) Explain clearance and metabolism.
• Drugs may be eliminated either unchanged (as the parent drug) or as metabolites depending on the lipophilicity • Most drugs are eliminated through the kidneys which can excrete only relatively polar substances • Thus lipophilic drugs must be metabolized into more polar metabolites for elimination • Drugs are metabolized to different extent mostly in the liver • Metabolism mostly lead to inactivation of a drug but many drugs have active metabolites • Therefore important to study the metabolism of a drug under development in order to know the impact it may have • First studied in liver microsomes • CYP enzymes inhibition – Drug-drug interactions
10) What is the process of liver metabolism?
Phase I (Functionalization): Functional groups are altered through monooxygenase reaction via CYP enzymes, leading to a loss of activity. Eg. Paclitaxel undergoes metabolic modifications before it can be renally excreted. Phase II (Conjugation): Addition of highly polar conjugates to drugs to increase their hydrophilicity. Eg. Irinotecan is metabolized to SN38, an active metabolite. SN-38 is inactivated by UGTs via the addition of glucuronic acid. UGT1A1*38 polymorphism inactivates UGT, making SN-38 difficult to be inactivated, leading to increased toxicity.
11) What are the in vitro ADME assays?
- In vitro: -Microsome metabolism. Incubate animal/human microsome with lead drug candidate, incubate over a timecourse and analyse by LC/MS. Higher percentage of parent compound remaining indicates higher metabolic stability. -PAMPA assay: A well within a larger well, lipid membrane in the inner well. Lead molecules in the inner well, identify those that pass through lipid membrane.
12) What are the in vivo ADME assays?
- Animal based models: -validate in vivo biomarkers for drug efficacy -required for efficacy and toxicity drug evaluation -in vivo evaluation of PK/PD in normal/disease animal models -Dynamic evaluation of drug efficacy: Histological analysis, tissue sample analysis (RNA,DNA,Protein), in vivo imaging of disease progression. 2. Human chimera mice: These are mice that contain transplanted human hepatocytes. It is a more accurate preclinical model than regular mice in terms of ADME properties. It allows evaluation of disease in human liver model (eg. Hepatitis viral infection) and evaluate new drug efficacy (ie. Antiviral drugs)
14) What other evaluations can be done before selection of a preclinical candidate?
Gross pathology, Histopathy, immunohistochemistry, molecular pathology hematology, immunology.
1) What are the non-GLP CMC studies?
- Chemical development: Improvement of the synthesis to reduce cost, increase output, safety and quality (purity and consistency). 2. Salt and formulation: - Finding best salt to balance solubility and lipophilicity of the drug (eg. Co-solvents, emulsions, pH adjustment, salt formation etc) - Finding best formulation for the chosen route of administration (eg. Tablet, capsule, solution, controlled release etc)
2) What are the GLP CMC studies?
- ICH stability 2. ICH impurity analysis 3. Develop prototype clinical formulation (pill, liquid etc)
3) What are the non-GLP animal studies?
Benchmark in vivo models, validate disease models, models in other disease areas. Finalize animal used for GLP/GMP studies.
4) What are the non-GLP ADME studies?
- Optimized analytical method development: - Determine exposure levels in toxicology studies: • For small molecules: use HPLC/MS. Identify molecular structure. • For biologics: use ELISA. It does not show structure and does not demonstrate activity because it uses binding as an endpoint. - Validation of assays: Extraction technique recovery, linearity of standard curve, intra and inter assay precision, bench top and freeze thaw stability, sensitivity (lower limit of quantification), establish QC standards 2. Pk profile 3. Oral bioavailability 4. Determine metabolism of drug
5) What are the GLP ADME studies?
- Comprehensive ADME 2. Bioavailability and Pk: -using optimized bioanalytical method, quantify drug or metabolites usually in plasma -determine bioavailability via Single dose, iv and intended route -determine blood brain barrier bioavailability – measure drug accumulation in brain, brain vs plasma levels 3. GLP Pk profile -rodent and non-rodent, drug availability by intended route, mean residence time, half-life 4. GLP toxicokinetics profile 5. Comprehensive determination of metabolites 6. Multi animal Pk studies: Comparative metabolism: To account for interspecies differences between animals. Done via comparing hepatic microsomes and cytosolic fractions from different species: human, mouse, rat , rabbit, dog, non-human primate, guinea pig etc. Study parameters such as half life and identify metabolites produced. Interspecies scaling improves Pk predictions and allows identification of toxic metabolites that are specific to each species. 7. Metabolic inhibition: to identify certain drugs may inhibit Cyp enzymes or be affected by drugs that inhibit Cyp enzymes
8) What are the requirements in toxicology evaluation?
- Appropriate species: 1 rodent, one second species (dog, pig or monkey generally). These animals should have good exposure and metabolism similar to humans, they must cover all human metabolites. Also, they should have the same pharmacologic effect as humans (same target binding, effect in disease models, pharmacologic effects). The exposures achieved in the test subjects should be sufficient to cover multiples of the intended human dose/exposure in order to establish a safety margin. 2. Higher doses to evaluate possible toxicities: FDA guidance to dose up to 1g/KG 3. Administer compound long enough to support intended clinical study 4. Endpoints: body weight, clinical observations, serum chemistry, hematology, organ weights, histology, drug exposure (toxicokinetics)
9) What are the non-GLP toxicology studies?
(goal: define upper bounds of safe drug administration) 1. Single dose (acute) toxicity: Determination of adverse effects within short time frame of single dose administration. Animals are observed for 14 days after dosing. Not many endpoints with focus on clinical observation and may be non terminal. Identifies single dose MTD. 2. Repeated dose toxicity: Involves a longer schedule of repeated dosing and establishes dosage levels for subsequent toxicity studies. Duration of dosing should ideally match duration of clinical study. Exception (for non-rodent species: if clinical study is >6 months, minimum duration of repeated dose tox study is 6 months in the EU. 3. Preliminary cardiovascular safety
10) What are the GLP toxicology studies?
(full ans in p8) 1. Acute and repeated dose toxicity: • Acute and repeated dose toxicity studies in 2 species (rodent and non-rodent) selected from non-GLP range-finding studies. • More comprehensive: (greater number/gender/species) • More complete toxicology study o Standard toxicology design: o 1) Plasma: drug analysis, 2) tissue: histopathology, 3) blood: clinical pathology, 4) clinical endpoints: survival, body weight, clinical signs, behaviour • Determination of adverse effects resulting from daily dosing to identify MTD and NOAEL (No Observed Adverse Effect Level) 2. Genotoxicity/mutagenicity testing: • in vitro non-mammalian cell system – e.g. Ames Test – Salmonella typhimurium - Determine if cells treated with drug can survive without histidine, indicating mutagenicity • in vitro mammalian cell system – e.g. CHO (Chinese Hamster Ovarian) cells - Determine % chromosomal aberration across a range of drug concentrations • in vivo mammalian system –e.g. mouse micronucleus assay - Immature mice treated with mutagen for period of 2-4 weeks, RBC’s observed under microscope for increased % of micronuclei 3. Carcinogenicity testing: • Long term toxicity testing - ~lifetime exposure • Usually in rats – 24 – 30 months • Mouse or hamster may also be used • 50 / gender / dose level and 100 / gender /control group • Determine potential tumorigenic effects of drug 4. Reproductive toxicology: • Fertility and general reproductive performance – Rats – Dosing of males for 60-80 days prior to mating – Dosing of females for 14 days prior to mating and during gestation and lactation • Potential drug-induced embryotoxicity and teratogenicity -Rat/mouse and Rabbit -Escalating dosages -1 month of treatment in pregnant females during embryonic and fetal development. • Late fetal development, labour, delivery, lactation and newborn viability (Rat or Mouse) – pregnant females using escalating dose levels – Dosing from last gestation day (day 16-17) to end of weaning – If reproductive capacity of offspring is evaluated, study duration is 5-6 months 5. Safety pharmacological core battery: 1. CNS: Irwins test, global nervous system assessment (autonomic, sensorimotor, neuromuscular, behavioural) 2. Respiratory system (in vivo): Whole body plethysmograph chambers (put rodents in chamber before and after drug, as the rats breathe in and out the chamber measures the change in volume). Measures tidal volume and respiratory rate before and after drug. 3. Cardiovascular: measure QT interval prolongation that is mediated by inhibition of hERG ion channel. (hERG channel involved in pumping of K+ out) - In vitro: Recording of K+ current from hERG expressing CHO cell line, generate full concentration effect curve to identify dosage of drugs that changes action potential - In vivo: Telemetry receiver inserted into dogs, measure QT interval change after drug administration and varying dosage. 6. Irritation and sensitisation testing (may or may not be required), dependent on route of administration 1. Rabbits eye test 2. Skin tests – rabbit, guinea pig
11) How are dosages translated from animals to humans?
• Maximum Recommended Starting Dose (MRSD) is converted from NOAEL in animal studies converted to HED divided by a safety factor of at least 10. • Human equivalent dose (HED) can be converted from animal dosage as a ratio of body weight, normalized by body surface area (BSA). (Allometric scaling)
12) Why is allometric scaling better than isometric scaling:
• Isometric scaling (straight conversion based on body weight) may lead to overestimation of human dosage and/or underestimation of toxicity of a given dose. • Allometric scaling takes into account lower metabolism of larger animals (humans) compared to smaller animals (rodents, dogs, etc.). • Max Rubner (1883) demonstrated that while the ratio of blood volume to body weight decreases in larger animals, blood volume is constant to body surface area.
13) How is combination index calculated?
• Km = body weight (kg) divided by BSA (m2) • HED (mg/kg) = animal dose (mg/kg) × (animal Km/human Km)
1) What is the definition of a biomarker?
- A defined characteristic that is measured as an indicator of normal biological processes, pathogenic processes, or responses to an exposure or intervention, including therapeutic interventions. - Molecular, histologic, radiographic, or physiologic characteristics are types of biomarkers.
2) What is the definition of a clinical endpoint?
A characteristic or variable that reflects how a patient feels, functions or how long a patient survives.
3) What is the definition of a surrogate endpoint?
A biomarker intended to substitute a clinical endpoint. A clinical investigator uses epidemiological, therapeutic, pathophysiological or other scientific evidence to select a surrogate endpoint that is expected to predict clinical benefit, harm, or lack of harm.
4) What is a diagnostic biomarker?
- A biomarker used to identify individuals with the disease or condition of interest or to define a subset of the disease. - Examples: • Sweat chloride levels in cystic fibrosis (CF) • Galactomannan in invasive aspergillosis • Blood sugar or HbA1c in DM • Blood pressure in hypertension • Serum creatinine or GFR (glomerular filtration rate) in kidney failure • Ejection fraction in heart failure
5) What is a monitoring biomarker?
- A biomarker measured serially and used to detect a change in the degree or extent of disease. - Monitoring biomarkers may also be used to indicate toxicity or assess safety, or to provide evidence of exposure, including exposures to medical products. - Examples: • HCV-RNA in chronic hepatitis C • INR or PT in warfarin • PSA in prostate cancer • CA-125 in ovarian cancer • BNP or NT-proBNP in pediatric pulmonary hypertension