Presentation is loading. Please wait.

Presentation is loading. Please wait.

Department of Molecular Pharmacology & Neuroscience

Similar presentations


Presentation on theme: "Department of Molecular Pharmacology & Neuroscience"— Presentation transcript:

1 Department of Molecular Pharmacology & Neuroscience
Pharmacogenetics/Pharmacogenomics Neil A. Clipstone, Ph.D. Department of Molecular Pharmacology & Neuroscience

2 Pharmacogenetics and Pharmacogenomics
The study of genetic factors that underlie the variation in drug responses Twin studies Family studies →Some drug responses are genetically determined Despite this: Most patients with same diagnosis Get same prescription At same dose Drug beneficial Drug beneficial but toxic Drug not beneficial No Toxicity Drug not beneficial and Toxic Lower dose or Alternative Therapy Higher dose or Alternative Therapy Alternative Therapy

3 Many major drugs are ineffective for many patients and are harmful to others
- much of this can be attributable to genetic differences

4 Goals of Pharmacogenetics/Pharmacogenomics
Understand how genetic differences influence the DRUG RESPONSE Identify patients that will respond to drug treatment Maximize drug efficacy Optimize drug dosing Minimize drug toxicity Aid in new drug development Right drug Right indication Right patient Right dose

5 Variability in Drug Response:
Genetic factors influencing the drug response Drug Toxic Targets Drug Metabolizing Enzymes Drug Transporters Pharmacodynamics (What drug does to body) Pharmacokinetics (what body does to drug) Variability in Drug Response: Drug Therapeutic DOSING, EFFICACY & ADVERSE EFFECTS

6 Pharmacogenetics and Pharmacokinetics
Most common factor responsible for inter individual differences in drug responses - Differences in drug metabolic enzymes (e.g. CYP450) Many of the enzymes involved in drug metabolism are polymorphic - existence of different alleles of a single gene within the population - different alleles exhibit different degrees of metabolic activity - CYP450 family of drug metabolizing enzymes are highly polymorphic especially CYP2D6, 2C9 and 2C19 Polymorphism may arise from: - single nucleotide changes (SNPs) (coding region, splicing sites, promoter regions) - deletions or insertions - whole gene duplications & amplifications (CNV) Gene polymorphisms can result in: - unchanged enzyme activity - increased enzyme activity - decreased enzyme activity - absent/null enzyme activity

7 Inheritance of polymorphic CYP450 genes
An individual inherits one individual CYP450 gene allele (e.g. 2D6) from each parent Alleles are referred to as wild type or variant Polymorphism occurs when an individual inherits one or more variant alleles - CYP2D6, CYP2C19 & CYP2C9 are most polymorphic (metabolize >40% of all drugs) Individuals are classified phenotypically based upon their degree of metabolism wild type/wildtype - Extensive metabolizer (normal metabolism) variant/ variant - Poor metabolizer (↓↓↓ metabolism) wild type/variant - Intermediate metabolizer (↓ metabolism) wild type/multiple copies - Ultra rapid metabolizer (↑↑↑↑ metabolism) Multiple copy alleles Multiple copies (3-13) wt alleles Increased gene expression Increased metabolic activity Wild type alleles Normal function Most common Variant alleles Reduced or null activity Rare/low frequency

8 Effects of genetic polymorphisms in CYP450 enzymes
on serum drug concentrations AN EXAMPLE Wild type - normal levels of CYP450 enzymatic activity Variant - CYP450 allele displaying reduced enzymatic activity - less efficient conversion of drug to inactive metabolite Less efficient drug metabolism →Increased concentration of active drug →Increased efficacy →Increased risk of toxicity

9 Functional effects of CYP450 polymorphisms on drug action
Extensive Metabolizer (NORMAL) Poor (enzyme variant with decreased activity) Ultra rapid (multiple copies of wild type enzyme) Standard Drugs Prodrugs Increased efficacy due to decreased inactivation of active drug Increased risk toxicity due to accumulation of active drug Typically lower dosing to avoid May need alternative drug Decreased efficacy Decreased serum concn of active drug metabolite due to decreased production of active drug metabolite Likely need alternative drug Decreased efficacy due to rapid inactivated of active drug Typically require higher dosing Increased efficacy due to increased production of active drug metabolite Increased risk of toxicity May require lower dosing to prevent accumulation of active metabolite May require alternative drug Drug (Active) Metabolite (INACTIVE) Prodrug (Inactive) (ACTIVE) CYP450 Phenotype

10 Pharmacogenetics of CYP2D6
CYP2D6 – highly polymorphic > 100 alleles >95% of phenotypes can be accounted for by just 9 alleles Significant polymorphisms and levels of enzyme activity between ethnic groups - potentially clinically significant Alleles *1 *1xN *3 *4 *10 *17 Function Normal Increased None Decreased Mutation NONE Gene duplication Frameshift Splicing defect P34S/S486T T107I/R296C/S486T African Asian European 39% 34% 52% 1.4% 0.03% 0.07% 0.003% 0% 1.3% 3.3% 0.45% 18% 6.7% 42% 2.8% 19% % 0.27% Allelic Frequency 2D6 CYP2D6*1xN - 22% in Oceania

11 Clinical relevance of CYP2D6 polymorphisms
Metabolizes 20-25% of drugs, although represents only 2-4% of hepatic CYP450 enzymes Active Drug Inactive metabolite 2D6 Examples: - Antidepressants (TCA & SSRI) - Antipsychotics Poor metabolizers (nulls) - 2D6*3, *4, *10 & *17 - Decreased metabolism - Higher drug concentrations Increased CNS toxicity with antidepressants & antipsychotics Ultra rapid Metabolizers (duplications) - 2D6*1xN & 2D6*2xN - more efficient metabolism of active drug - lower concentration of active drug decreased efficacy of antidepressants & antipsychotics ProDrug Active Examples: - Codeine (Opioid analgesic) - Tamoxifen (Breast cancer therapy) Poor metabolizers - decreased clinical efficacy (less active metabolite) e.g. codeine- lack of analgesic effect Poor clinical outcome with Tamoxifen in Breast cancer treatment - more conversion of Prodrug to active metabolite - increased efficacy & increased risk of toxicity Risk of codeine overdose with standard dosing i.e. risk of respiratory depression

12 Pharmacogenetics of CYP2C19
> 30 alleles, although 4 alleles account for most of the variability 2C19*1 - normal activity 2C19*2 and *3 – non-functional 2C19* increased expression- increased activity Substrates include: Proton Pump inhibitors (e.g. Omeprazole), antidepressants (e.g.Citalopram), anti-epileptics (e.g. mephenytoin), and anti-platelet drugs (e.g. clopidrogel) Clopidrogel Prodrug (Inactive) CYP2C19*1 CYP2C19*2 or CYP2C19*3 (null activity) Clopidrogel Metabolite (Active) Used to prevent blood clots in HTN & CVD No metabolism No active clopidrogel metabolite No platelet inhibition Clinically ineffective Continued risk of blood clots Platelet Activation

13 Pharmacogenetics of CYP2C9
2C9 – accounts for the metabolism of 15-20% of drugs Major alleles 2C9*2 and *3 - reduced function variants 2C9*15 and *25 – null variants Substrates include: glipizide (anti-diabetic), NSAIDs (analgesic) and warfarin (anti-coagulant) Clinical relevance Warfarin – narrow therapeutic window, population differences in dosing and toxicity Warfarin consists of equal amounts R-warfarin and S-warfarin (most potent) 1A1/2C19 3A4 Inactive Metabolite 2C9 Inactive Metabolite R-warfarin/S-Warfarin (Active) Patients with 2C9*2 or 2C9*3 - decreased metabolic inactivation of S-warfarin - increased concentration of active warfarin with standard dosing - prone to experiencing bleeding events (elevated S-warfarin) - lower warfarin doses required (to account for reduced metabolism)

14 Pharmacogenetics of Phase II enzymes
Phase II enzymes are involved in conjugation reactions that inactivate drugs and promote drug excretion - Genetic variation in phase II enzymes can decrease drug inactivation & elimination, thereby increasing the potential risk of drug toxicities - However, compared to CYP450, there are fewer examples of Phase 2-selective substrates with significant toxicity problems Polymorphic Phase II enzymes Highly Polar conjugate conjugate Functional group Phase I enzymes Decreased activity D D D Lipophilic Drug DRUG METABOLITE (Active/Inactive) DRUG-CONJUGATE Water Soluble Inactive Potential Toxicity EXCRETION (Urine/Bile)

15 (Inactive metabolite)
Example #1 - UDP-glucuronosyl transferase (UGT1A1) - UGT1A1 > 30 alleles - many with reduced activity, although most are quite rare - UGT1A1*28 (reduced expression) is common across most ethnic groups - ~10% of Europeans are homozygous for UGT1A1*28 - UGT1A1*28/UGT1A1*28- cause of Gilbert’s Disease (Indirect Hyperbilirubinemia) - also increased risk for adverse effects from certain drugs that are exclusive UGT1A1 substrates e.g. Irinotecan (SN-38) Irinotecan (Inactive prodrug) SN-38 (Active metabolite) SN-38-Glucuronide (Inactive metabolite) Carboxylesterase Glucuronic acid conjugate UGT1A1 Irinotecan (ProDrug)- UGT1A1 substrate UGT1A1*28 homozygotes - Reduced inactivation of SN-38 - Enhanced SN-38 toxicity Cancer Cell Killing Toxicity

16 Example #2: N-Acetyl Transferases (NATs- NAT1 & NAT2)
One of the first characterized genetic defects in drug metabolism NAT2 is important for the metabolism of ISONIAZID (mainly undergoes Phase 2 metabolism) - Drug used in the treatment of tuberculosis - Narrow therapeutic window- associated hepatotoxicity NAT2- highly polymorphic NAT2*1 – wild type enzyme NAT2*5, *6, *7, *10, 14 & *17- encode defective enzymes NAT2*1/NAT2*1 - “Rapid acetylator Phenotype” - Efficient metabolism of Isoniazid NAT2*5/NAT2*5 - “Slow acetylator Phenotype” - Deficient metabolism of Isoniazid - Significantly increased risk of hepatotoxicity - CNS side effects due to increased production of a Neurotoxin minor side product in rapid acetylators, but significantly increased when Isoniazid acetylation is deficient

17 6-methylmercaptopurine
Example #3 Thiopurine S-Methyltransferase (TPMT)- Involved in the inactivation of thiopurine drugs e.g. 6-mercaptopurine (immunosuppression and cancer) Bone Marrow suppression 6-mercaptopurine (active) TPMT Toxicity 6-methylmercaptopurine (inactive) TPMT*3A- non functional allele present in ~10% Europeans TPMT*3C- non functional allele present in ~10% Africans and Asians ~0.3% inherit two non-functional alleles - ↓↓↓ metabolism & inactivation - ↑↑↑ blood levels of active 6-mercatopurine - ↑↑↑ risk toxicity - fatal BM suppression

18 Pharmacogenetics of Drug Transporters
Drug transporters mediate the influx or efflux of drugs in to or out of cells Play an important role in determining plasma and tissue levels of a drug Genetic polymorphisms in drug transporters can dramatically alter drug disposition and drug responses Decreased activity in influx pump - Reduced cellular uptake - Decreased efficacy - If expressed in liver can affect drug metabolism and elimination - Potential for increased “off target” toxicity Decreased activity in efflux pump - Reduced cell excretion - Potentially increased efficacy - Potentially increased toxicity - Potential for decreased drug excretion - Potential for compromising blood brain barrier Drug Influx/Uptake transporter Efflux Plasma/Tissue drug concentration distribution Efficacy Toxicity Influx

19 Pharmacogenetics of Drug Transporters
Example OATP1B1 (aka SLCO1B1)- hepatic uptake of weakly acidic drugs e.g. statins Hepatic uptake of statin drugs to the liver is important, since that is where they mediate their clinical effects - inefficient hepatic take up of statins results in decreased efficacy and increased systemic bioavailability of the drug with increased potential for toxicity (muscle) >40 SNPs identified: SLCO1B1*15 (Val174Ala) is a common reduced function allele - due to reduced membrane expression Statin-induced Muscle Damage SLCO1B1*1 (wt) Efficient hepatic uptake of drug Statin SLCO1B1*15 Reduced expression Decreased drug uptake Decreased efficacy accumulates in serum Increased statin bioavailability allows for statins to accumulate in muscle and trigger toxicity Requires reduced Statin Dosing blood liver Efficient clinical effect

20 * Pharmacogenetics of Drug Targets/Pharmacodynamic effects
b g a Receptor Gi-protein Drug Downstream Signaling pathway Gene 1 enzyme * Polymorphic residue Genetic variation in pharmacokinetic genes e.g. CYPs influence the concentration of the Drug at the site of its target Genetic variation in the direct drug target or other downstream pharmacodynamic genes can significantly influence the Drug response a) Drug target expression levels b) Affinity for drug c) Efficiency of downstream signaling *- potential sites of polymorphisms

21 - most common form of receptor contains Ser49 and Arg389
Example #1 - Polymorphism in a cell surface receptor (b1 adrenergic receptor) - target for beta blocker drugs used in the treatment of hypertension e.g. metoprolol However, many HTN patients fail to adequately respond to b-blocker therapy - most common form of receptor contains Ser49 and Arg389 Signals very efficiently to b1 adrenergic agonists - two common SNPs result in receptor amino acid substitutions Ser49Gly (~15% Caucasians, ~25% Blacks & ~ 14% Asians) Arg389Gly (~30% Caucasians, ~40% Blacks & ~25% Asians) - Patients with 2 copies of the most common receptor haplotype Ser 49/Arg389 exhibit a much greater reduction in blood pressure in response to metoprolol compared to those expressing variant receptor haplotypes e.g. Gly49/Arg389 & Ser49/Gly389 RELEVANCE: Ability to predict responsiveness based upon genotype would allow beta blockers to be started in those predicted to respond well to the drug b1 adrenergic receptor Ser49 Arg389

22 Example #2: Polymorphism in a signaling molecule downstream from the drug receptor
Selective Serontonin reuptake inhibitors (SSRIs) are used to treat depression They work by blocking serotonin uptake into presynaptic neurons thereby increasing the concentration of serotonin in the synaptic cleft, which is then able to bind to and trigger the serotonin G-protein-coupled receptor expressed on the post-synaptic neuron A common SNP (c.825C>T) is present in the Gi-protein b3 subunit downstream of the serotonin receptor - leads to enhanced serotonin receptor signaling and has been correlated with an improvement in the depressive symptoms of those patients treated with the SSRI class of drugs RELEVANCE: This SNP identifies patients most likely to benefit with drug treatment b g a Serotonin Receptor Gi-protein Downstream signaling Polymorphic gene Presynaptic neuron transporter SSRI *

23 Polygenic effects explain variability of the drug response within populations
Drug response can be affected by multiple independent genes Genetic polymorphisms can affect both Pharmacokinetic and Pharmacodynamic parameters Inheritance of specific genetic differences in multiple individual genes regulating PK and PD parameters can explain the significant variability in drug responses between individuals Drug Transporters Phase I/ Phase II Enzymes Targets Downstream Signaling Pathways Response Potential functional genetic polymorphisms wt/wt var/var wt/var var/wt Patient genotypes

24 Warfarin as an example of the effects of Polygenic effects on drug responses
Warfarin (anticogulant) Used to prevent blood clots (e.g. DVT or stroke) Works by inhibiting VKOR (Vitamin K epoxide reductase) Significant inter patient variability in drug response differences in both EFFICACY & ADVERSE EFFECTS Caused by genetic variants that affect both PK and PD parameters Pharmacokinetics S-warfarin metabolized by CYP2C9 2C9*2 (Arg144Cys)- 12% wild type activity 2C9*3 (Ile358Leu) - 5% of wild type activity - patients with variant alleles exhibit high levels of warfarin and require decreased drug dosing to avoid excessive bleeding Pharmacodynamics Common polymorphism found in the VKORC1 promoter region: -1639G>A G is associated with higher VKORC1 expression and requires a higher warfarin dose for efficacy A is associated with lower VKORC1 expression and requires a lower warfarin dose to avoid excessive bleeding - 37% whites, 14% Africans & 90% Asians carry the A-allele Rare missense mutations in the VKORC1 coding region have reduced affinity for warfarin, but do not affect VKORC1 enzymatic activity- they are all associated with warfarin resistance i.e. patients require very high doses of warfarin to achieve appropriate levels of anticoagulation Reduced vitamin K Vitamin K Epoxide Vitamin K epoxide reductase VKOR (VKORC1) S-Warfarin Vitamin K-dependent g-glutamylcarboxylase Clotting factor precursor Active O2 CO2 Inactive Metabolite 2C9 CLOT FORMATION BLEEDING

25 Pharmacogenetics of adverse drug reactions
Adverse reactions are a huge burden on healthcare ~5th leading cause of death in US - Estimated that 10-20% of adverse effects are due to genetics Pharmacokinetic PK effects often increase systemic drug levels leading to increased risk of toxicity - often polymorphisms in CYP450 metabolic enzymes/Drug transporters etc - most often seen in drugs with narrow therapeutic windows Pharmacodynamic PD effects can influence the interaction of a drug for its target and/or influence the efficiency of drug-dependent downstream signaling pathways leading to an exaggerated drug response that causes an adverse effect Toxic target Genetic polymorphisms in toxic drug targets can lead to drugs being able to bind and influence the activity of unexpected targets, thereby causing an adverse effect e.g. Polymorphisms in the Dopamine D3 receptor allow antipsychotic medications (normally D2 antagonists) to inhibit D3 receptors causing Tardive Dyskinesia

26 Idiosyncratic drug-induced adverse effects/Hypersensitivity
- typically very rare and unrelated to effects on either PK or PD parameters - not predictable from known drug pharmacology (i.e. MOA/affinity etc) - often associated with polymorphisms in immune system genes (e.g. HLA) A. Drug-induced hepatotoxicity e.g. Flucloxacillin used to treat Staphyloccocal infections Drug-induced liver damage most common reason for failed clinical trials and withdrawal of drugs from the market - Flucloxacillin-induced liver damage occurs in 8.5 /100,000 users - Linked to HLA-B*57:01 (other hepatotoxic drugs also linked to HLA alleles) B. Drug-induced hypersensitivity reactions (Severe Allergic-like reactions, often life threatening) e.g. Carbamazepine-induced Steven Johnson Syndrome - linked to HLA-B*1502 in Han Chinese FDA recommends Pharmacogenetic testing to screen Asian patients requiring carbamazepine in order to avoid SJS

27 Pharmacogenetics and the promise of Personalized Medicine
Identify patients that will respond to drug treatment Optimize drug dosing based upon pharmacogenetic information Minimize drug toxicity Patient population all with the same clinical diagnosis Genetic Testing Patient Stratification Identify those that would benefit from drug therapy Determine individualized optimal dose for each patient Identify those that could benefit, but are susceptible to toxicity Determine optimal dosing to treat disease, but avoid toxicity consider alternative therapeutic options for non-responders

28 Pharmacogenomics and Targeted Therapy
Therapy that is targeted towards a specific molecular target based upon unique mutations or genetics Currently, most often used in the treatment of specific cancers Treatment is based upon specific somatic mutations in key oncogenic drivers unique to the patient’s cancer - Drug is only given to those patients with the specific mutation Imatinib (Gleevac®), a tyrosine kinase inhibitor specific for the BCR-ABL Chr breakpoint oncoprotein found in patients with Chronic Myeloid Leukemia Vemurafenib (Zelboraf®)- used in the treatment of malignant melanoma specifically harboring the BRAF V600E oncogenic mutation Gefitnib (Iressa®), an EGFR tyrosine kinase inhibitor used to treat a subset (10-20%) of Non-Small cell lung cancers patients that harbor the L858R activating mutation in their EGFR Maraviroc (Selzentry®), an anti-HIV drug used to block viral entry in patients expressing a specific kind of receptor

29 The Use of Pharmacogenomics in translational research
Drug Discovery & Diagnosis Drug Therapy Trial Control healthy population Disease population (e.g. cancer) Disease population placebo Disease population Drug-treated Genetic Analysis DNA sequencing Clinical data Clinical outcome Efficacy/toxicity Drug disposition Genetic Analysis DNA sequencing RNA/Protein/miRNA expression Statistical analysis Identify genetic mutations specific to disease Identify genetic biomarkers that predict Efficacy/Toxicity/Drug disposition Develop drug specific to mutated gene product Develop biomarker for diagnostics

30 Clinical implementation of pharmacogenetic data
>100 FDA approved medications contain pharmacogenetic information in their labeling However, pharmacogenetic data is currently not widely incorporated into clinical practice Barriers to implementation Physician education and understanding of pharmacogenetics Absence of data demonstrating utility in the clinical setting Lack of evidence for added value to existing prescribing method Availability of alternative medications where genetic variability is not an issue Cost effectiveness Timeliness of turnaround for genetic testing Lack of reimbursement for tests/Insurance coverage Lack of clear clinical guidelines

31

32


Download ppt "Department of Molecular Pharmacology & Neuroscience"

Similar presentations


Ads by Google