Drug Metabolism and Toxicity

58
Drug Metabolism and Toxicity Edna Mao Group Meeting Literature Talk October 13 th , 2021

Transcript of Drug Metabolism and Toxicity

Drug Metabolism and Toxicity

Edna MaoGroup Meeting Literature Talk

October 13th, 2021

Drug discovery and development

Discovery Preclinical development

Clinical trials

FDA review

Approved drug

> 5000 compounds

250 compounds

5 compounds 1 compound

Image credit: Ian B. Perry

Reasons drugs fail in the pipeline

Most common reason for failures in drug

discovery attributed to pharmacokinetic properties

Harmful toxicity another major concern in

drug development

Van de Waterbeemd, H.; Gifford, E. Nat. Rev. Drug. Discov. 2003, 2, 192–204.

Pharmacokinetics

The study of the bodily processes that affect the movement of drugs across the body

Fundamentals of pharmacokinetics

Absorption

Route of administration

Bioavailability

Distribution

Transfer of drug through body by bloodstream

Sites of accumulation

Metabolism

Biotransformation pathways

Mechanism of detoxification/active species formation

Excretion

Method and rate of drug clearance from body

Outline

Introduction to drug metabolism

Common biotransformations

Applications to drug design

Designing around metabolism

Metabolism-induced toxicity

Mechanisms of adverse reactions Reactive intermediates and

structural alerts

Applications to drug design

Designing around drug-induced toxicity Challenges in predicting adverse effects

Outline

Introduction to drug metabolism

Common biotransformations

Applications to drug design

Designing around metabolism

Metabolism-induced toxicity

Mechanisms of adverse reactions Reactive intermediates and

structural alerts

Applications to drug design

Designing around drug-induced toxicity Challenges in predicting adverse effects

Introduction to drug metabolism

Xenobiotic metabolism

The chemical alteration of non-endogenous compounds within the body

The early studies of metabolism

1841-1842

Ure and Keller independently ingest benzoic acid

Observed compound in urine in “copious amounts”

Compound similar to benzoic acid, but contained nitrogen

1845

Compound identified by Dessaignes as hippuric

acid (glycine–benzoic acid conjugate)

Glycine conjugation

first metabolic reaction to be described!

1848-1893

Methylation, aromatic oxidation, and conjugation

reactions characterized

Mechanisms considered “absolutely puzzling”, a “biochemical curiosity”

1945

Lipmann elucidates role of coenzyme A in acetylation reactions

Discovery wins Nobel

Prize in Medicine in 1953

Early 1900’s

Metabolism found to occur primarily in liver

Liver microsome

isolated in 1943 by Claude

1960’s

Discovery and characterization of

cytochrome P450’s and their role in drug

metabolism

1947

Richard Williams publishes “Detoxification mechanisms”, solidifying

drug metabolism as a defined field of study

King, R. S. Drug Metabolism Handbook: Concepts and Applications.; John Wiley & Sons, 2009; pp 3–9.

First pass metabolism

Metabolism by gastrointestinal enzymes

Orally administered drug

Metabolism by gut bacterial enzymes

All orally administered drugs pass through portal vein into liver before distribution to

the rest of body

Fraction of dose enters bloodstream

The concentration of orally administered drugs is greatly

reduced by “first pass metabolism” before reaching systemic circulation

Phase I and II biotransformations

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

Phases of drug metabolism

Phase 1: Modification

Introducing functional handles to drug

Phase 2: Conjugation

Attaching highly hydrophilic groups

Drug molecule Lipophilic,

not excretable

Excretion Bile, urine

Modified compound Polar, hydrophilic

Metabolism

Phases of metabolism

Initial modifications to drug molecules

Adding or uncovering reactive functional groups

Mostly facilitated by CYP enzymes in liver

Phase I metabolism

Conjugation of polar functional groups, usually resulting in deactivation

Facilitated by wide variety of transferase

enzymes

Conjugated molecules are recognized by transporters and excreted

Phase II metabolism

Cytochrome P450s

N

N

N

N

Me

Me

MeMe

Fe

O HO OOH

Heme B

Superfamily of heme-containing monooxygenase enzymes

R R

H

CYPO2 H+

R R

OHH2O

NADPH NADP+

+ + +

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

The Cytochrome P450-dependent mixed-function oxidase system

In human microsomes, P450s found on in mitochondrial and

endoplasmic reticulum membranes

CYP enzymes responsible for 75% of metabolism

Other enzymes that facilitate metabolism

Flavin-containing monooxygenases (FMO) Alcohol dehydrogenase (ALD)

Aldehyde oxidases (AO) Monoamine oxidases (MAO)

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

Phase I biotransformations - Oxidations at carbon centers

Aromatic hydroxylation Epoxidation

Aliphatic hydroxylation

R R

OH

CYPR R

O

CYP

R R

H

R R

OHCYP

R OHADH/ALDH

R OH

O

Alcohol oxidation

Dehalogenation

Halothane

Br

ClCYPF

FF

Br

ClF

FF

OH – HBrCl

F

FF

OH2O

OHF

FF

OH

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

Oxidative dehalogenation

Phase I biotransformations - Oxidations at heteroatoms

N-dealkylation

O-dealkylation

RN

CH3

R

RN

H

RCYP

viaR

NR

OH

RO

CH3 RO

H

CYP

viaR

O OH

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

N-oxidation

Deamination

FMORN

RN

OR

R RR

MAOR

RNH2 H

O

Responsible for the inactivation of endogenous neurotransmitters

Targeted by class of antidepressant and anti-anxiety medications known as “MAO

inhibitors”

Phases of metabolism

Initial modifications to drug molecules

Adding or uncovering reactive functional groups

Mostly facilitated by CYP enzymes in liver

Phase I metabolism

Conjugation of polar functional groups, usually resulting in deactivation

Facilitated by wide variety of transferase

enzymes

Conjugated molecules are recognized by transporters and excreted

Phase II metabolism

Phase II conjugations: glucuronidation

Glucuronidation

ROH

UDP-glucuronosyl transferase (UGT)R

OO

OHO

HOHO

OH

UDP-Glucuronic acid

O

OHO

HOHO

OH O PO

OHO P

O

OHO O N

NH

O

O

HO OH

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

Phase II conjugations

ROH

Sulfotransferase (SULT)R

OS

O

O

OPAPS

(3′-Phosphoadenosine-5′-phosphosulfate)

RNH2

N-acetyltransferaseR

HN

acetyl-CoAMe

O

Glutathione conjugation

O SO

OO P

O

OO O N

O OHPO

OO

N

NN

NH2

Reactive electrophile O

HN

OH

OS

NH

NH2

HO

O OGlutathione transferase (GST)

Glutathione (GSH)

OR

R

ROH

R

Sulfation

Acetylation

Ionescu, C.; Caira, M. R.; Drug Metabolism: Current Concepts.; Springer, 2005.

Phases of drug metabolism

Phase 1: Modification

Introducing functional handles to drug

Phase 2: Conjugation

Attaching highly hydrophilic groups

Drug molecule Lipophilic,

not excretable

Excretion Bile, urine

Modified compound Polar, hydrophilic

Metabolism

Implications of metabolism on drug design

Drugs need to reach their targeted site and remain in the system for a meaningful amount of time in order to elicit the desired pharmacological effects.

Pharmacokinetic properties which are impacted by metabolism:

Clearance Half life

Bioavailability

Prodrugs

Prodrugs: a class of drugs administered in a pharmacologically inactive form, which upon administration is enzymatically converted into its active form in vivo

Prodrug strategy can overcome challenges in :

Absorption Route of administration

Low bioavailability

Case study: Sofosbuvir (Pharmasset/Gilead)

Approved in 2013 for hepatitis C WHO List of Essential Medications

Nucleotide prodrug (ProTide)

O N

HN

O

O

MeO

Sofosbuvir

FHO

PO

OPh

NH

MeO

O

Me

Me

The ProTide approach: Sofosbuvir

Nucleoside analogue drugs tend to suffer from low potency due to slow first phosphorylation

Active form of drug

Nucleosides delivered as the monophosphate suffer from poor absorption and bioavailability due to charged phosphate

O N

HN O

O

MeHO

Nucleoside Nucleoside monophosphate

FHO

O N

HN O

O

MeO

FHO

PO

OO

slow

Nucleosidekinase

Nucleoside triphosphate

O N

HN O

O

MeO

FHO

PO

OOfast

PO

O O

P OOO

Rautio, J.; Meanwell, N. A.; Di, L.; Hageman, M. J. Nat. Rev. Drug. Discov. 2018, 17, 559–587.

Rautio, J.; Meanwell, N. A.; Di, L.; Hageman, M. J. Nat. Rev. Drug. Discov. 2018, 17, 559–587.

Phosphate group masked

Effectively distributed to hepatocyte cytosol (cells expressing high levels of CTSA/CES1)

ProTide hydrolyzed intracellularly (no metabolite detected in plasma)

The ProTide approach: Sofosbuvir

O N

HN

O

O

MeO

Sofosbuvir

FHO

PO

OPh

NH

MeO

O

Me

Me

CTSA/CES1

ester hydrolysis

O N

HN

O

O

MeO

FHO

PO

OPh

NH

MeHO

O

Sofosbuvir triphosphate

Recognized by HCV NS5B polymerase

Incorporated into RNA strand, leading to chain

growth termination

UMP-CMP kinase,NDPK

The ProTide approach: Sofosbuvir

Rautio, J.; Meanwell, N. A.; Di, L.; Hageman, M. J. Nat. Rev. Drug. Discov. 2018, 17, 559–587.

O N

HN

O

O

MeO

FHO

PO

OPh

NH

MeHO

O

–PhOH O N

HN

O

O

MeO

FHO

POH

N

O

O

Me

H2O HINT1 O N

HN O

O

MeO

FHO

P

O

OOO N

HN

O

O

MeO

FHO

PO

NH

MeHO

O O

O N

HN O

O

MeFHO

OPOPOO

OPO

OO O O

Lessons learned from metabolic studies

Information from metabolic studies can be used for rational drug design

Case study:

Ezetimibe (Schering-Plough/Merck) Approved in 2002 for hypercholesterolemia 125th most prescribed medication in 2018

Clader, J. W. J. Med. Chem. 2004, 47 (1), 1–9.

N

O

OMe

OMe

Lead compound: SCH-48461

Clader, J. W. J. Med. Chem. 2004, 47 (1), 1–9.

N

O

OMe

OMe

Lead compound: SCH-48461

Control group showed no decrease in plasma cholesterol until dosed with compound in week 3

Cholesterol-fed hamsters dosed with 1 mg/kg compound displayed a lower total plasma

cholesterol level Dosed group showed increase in plasma

cholesterol when dosing stopped at week 3

Case study: the development of Ezetimbe

Initial results with cholesterol-fed hamsters showed promising efficacy

Case study: the development of Ezetimbe

Rosenblum, S. B., et. al. J. Med. Chem. 1998, 41 (6), 973–980. Clader, J. W. J. Med. Chem. 2004, 47 (1), 1–9.

N

O

OMe

OMe

Lead compound ED50 = 2.2 mg/kg/day

N

O

OH

OMe

Metabolite isolated from bile (19)

Compound is rapidly metabolized in vivo, resulting in poor half life

Relatively high dose needed

Hamsters fed 14C cholesterol

14C cholesterol levels in plasma measured

Demethylated metabolite accounted for majority of

activity

Case study: the development of Ezetimbe

Rosenblum, S. B., et. al. J. Med. Chem. 1998, 41 (6), 973–980. Clader, J. W. J. Med. Chem. 2004, 47 (1), 1–9.

N

O

OMe

OMe

Lead compound ED50 = 2.2 mg/kg/day

N

O

OMe

OMe

OH

(S)-benzylic hydroxylation ED50 = 0.9 mg/kg/day

2x more potent than lead 5x more potent than (R)-hydroxylation

N

O

OMe

OMe

HO

C3 Aryl hydroxylation -16% LCE @ 50 mg/kg/dayLarge decrease in activity

N

O

OMe

OH

N-Aryl hydroxylation -78% LCE @ 50 mg/kg/daySlight decrease in activity

SAR studies

Case study: the development of Ezetimbe

Rosenblum, S. B., et. al. J. Med. Chem. 1998, 41 (6), 973–980.

N

O

OMe

OMe

SCH-48461

Unproductive sites of metabolism

Productive sites of metabolism

N

O

OH

F

OH

F

Ezetimibe

Metabolism blocked

Pre-metabolized

Monkey0.2 0.0005

ED50 (mg/kg)

Hamster2.2 0.04

Dog0.1 0.007

Outline

Introduction to drug metabolism

Common biotransformations

Applications to drug design

Designing around metabolism

Metabolism-induced toxicity

Mechanisms of adverse reactions Reactive intermediates and

structural alerts

Applications to drug design

Designing around drug-induced toxicity Challenges in predicting adverse effects

Adverse drug reactions (ADRs)

Type B (idiosyncratic ADRs)

Not associated with primary pharmacology, mechanism usually not understood

Does not occur at any dose for most people

Poor correlation with animal models

Often not detected until drug has been exposed to a

large population of patients

Attia, S. M. Oxid. Med. Chem. Longev. 2010, 3 (4), 238–258.

Adverse drug reaction:

a response to a drug that is noxious, unintended and occurs at doses normally used in man

Type A

Associated with primary pharmacology of drug

Usually detected in animal models/clinical trials

Can be mitigated through dose adjustments

Bioactivation to reactive metabolites

Drug moleculeDetoxified/deactivated metabolite

Metabolism

Bioactivation to reactive metabolites

Reactive metabolite

Drug moleculeDetoxified/deactivated metabolite

Bioactivation is usually the initial event in most drug-induced toxicities

Metabolism

Mechanisms of adverse reactions

Drug molecule

Metabolism

Reactive intermediate

Covalent binding

DNA Essential cell protein Autologous macromolecule

Mutagenesis, cancer

Cell death, necrosis

Immune response, hypersensitivity

Enzyme bindingEnzyme deactivation

Mechanisms of adverse reactions

Drug molecule

Metabolism

Reactive intermediate

Covalent binding

DNA Essential cell protein Autologous macromolecule

Mutagenesis, cancer

Cell death, necrosis

Immune response, hypersensitivity

Enzyme bindingEnzyme deactivation

Mechanism-based inhibition

General mechanism of C(sp3)–H hydroxylation

In some instances, a substrate can escape the oxygen-rebound step, binding CYP and leading to inactivation

Enzyme deactivated

Substrate escapes oxygen rebound step

SFeIV

O

Cys

R R

H

SFeIV

O

Cys

R R

H

SFeIII

O

Cys

HR

R

R R

OH

Oxygen reboundHAT

R R

Fe

Fe

R R

Reactive intermediate binds enzyme

Mechanism-based inhibition

De Montenallo, O.; Cytochrome P450: Structure, Mechanism, and Biochemistry. 3rd ed.; Springer, 2005.

Mechanism-based inhibition by paroxetine

HN

OO

F

O

Paroxetine

Approved in 1992 for use as antidepressant

74th most prescribed drug in 2018

Known to cause drug-drug interactions

Formed during metabolism

Strong inhibition ofCYP2D6 and CYP2B6

O

O

ROH

Prescribing guidelines warn that paroxetine is not to be taken with certain other drugs

De Montenallo, O.; Cytochrome P450: Structure, Mechanism, and Biochemistry. 3rd ed.; Springer, 2005.

O

O

RH

[Ox]–H+

O

O

Fe Fe

R OO

R

Mechanisms of adverse reactions

Drug molecule

Metabolism

Reactive intermediate

Covalent binding

DNA Essential cell protein Autologous macromolecule

Mutagenesis, cancer

Cell death, necrosis

Immune response, hypersensitivity

Enzyme bindingEnzyme deactivation

Classic example: drug-induced liver damage from acetaminophen

OH

NH

O

MeCYP2E1

O

N

O

MeGSH

OH

NH

O

Me

SNH

NH

O

COOHO

NH2HOOC

Acetaminophen Reactive quinoneintermediate Glutathione conjugate

Endogenous GSH levels depleted

Covalent binding to hepatic proteins Cell death,

liver damage

Most commonly used medication forpain and fever in the US and Europe

On WHO List of Essential Medicines

Leading cause of acute liver failure and drug overdoses in Western countries

Treatment of overdose may involve administratingof N-acetylcysteine (glutathione precursor)

Kalgutkar, A. S. J. Med. Chem. 2020, 63 (12), 6276–6302.

Classic example: Toxicity of benzene

O

O

1e-

O2

O2

Covalent binding

Quinone redox cycling generates reactive oxygen species

Bolton, J. L.; Dunlap, T. Chem. Res. Toxicol. 2017, 30 (1), 13–37.

OH

OH

Nuc

OH

OH

OH OHOH

O

O

OO

Benzene

CYP CYP Reactive quinone intermediates

The structural alert concept

Nelson, S. D. J. Med. Chem. 1982, 25 (7), 753–765.

Structural alerts (toxicophores): functional groups frequently found in drugs associated with adverse reactions

OR

NH

O

Me

R NH

ONH2 N

R

RR

ROR SR

RNO2

XR

Acetanilides Hydrazides Amines

Phenyl rings Heteroaromatics Nitroaromatics

Halogenated compounds

The structural alert concept

Nelson, S. D. J. Med. Chem. 1982, 25 (7), 753–765.

Structural alerts (toxicophores): functional groups frequently found in drugs associated with adverse reactions

Acetanilides Hydrazides Amines

Phenyl rings Heteroaromatics Nitroaromatics

Halogenated compounds

O

N

O

Me

R H

ON2 HR HR

OHN

R

R O X

R RN

O OHR SR

O

OHO

O

R O

Michael acceptors Alkylating/acylating agents Acylating agents, possible fluoride toxicity

Epoxidation leading to reactive intermediates Reactive oxygen species

NR

RR

or

Structural alerts as a method for identifying potential toxicity

Stepan, A. F.; Walker, D. P.; Bauman, J.; Price, D. A.; Baillie, T. A.; Kalgutkar, A. S.; Aleo, M. D. Chem. Res. Toxicol. 2011, 24, 1345– 1410.

Survey of withdrawn drugs found structural alerts in

55 out of 68 drugs (80.8%)

The propensity to form reactive metabolites was

observed in 36 out of the 55 drugs (65%)

Drugs recalled due to idiosyncratic drug-induced liver injury

NH2HO

O

O

Br

Bromfenac Structural alerts: Aniline, bromobenzene

Withdrawn in: 1998

OHN

S

OMe

MeOH

MeMe

OO

Troglitazone Structural alerts: Thiazolidinedione, phenol

Withdrawn in: 2000Linked to 63 cases of liver-failure deaths

Hepatotoxicity is leading cause of idiosyncratic adverse drug reactions

O

N NN ClN

Et

NO

Nefazodone Structural alerts: N-aryl piperazine

Withdrawn in: 2004

S

SO

O

NH

O O

O

Me

O N

Me

Cl

Sitaxentan Structural alerts: 1,3-benzodioxole

Withdrawn in: 2010

Kalgutkar, A. S. J. Med. Chem. 2020, 63 (12), 6276–6302.

Assaying formation of reactive metabolites

Incubation of drug in liver microsome

Addition of appropriate nucleophile

(Glutathione, KCN)

Administration of radio labeled (3H /14C)

compounds

Isolate and quantify radioactive covalently bound proteins and

DNA

Evans, D. C., et. al. Chem. Res. Toxicol. 2004, 17 (1), 3–16.

Reactive metabolite trapping assays

O

R

NR

Reactive metabolites

HO

R

GSH

NR

CN

LC-MS analysis of adducts

Radiolabeling assays

Strategies for mitigating effects of structural alerts

Reports of hepatotoxicity,agranulocytosis

Reduce electron density of aromatic

rings

NH

Cl N

HO

CYP N

Cl N

O

Amodiaquine

Et2N Et2NAnalogues with improved safety profile

O’Neill, P. M., et. al. J. Med. Chem. 2009, 52 (5), 1408–1415.

Steric blocking of metabolically

active sites

NH

F

Ar

Et2N

NH

HO

Ar

HN

tBu

Kalgutkar, A. S., et. al. Bioorg. Med. Chem. Lett. 2009, 19 (6), 1559–1563.

NNH

N

N

O

Me

F

Me

Redesigned compound

Ames negative

Remove structural alert

Block metabolically labile sites

NN

N

N

R

O

Reactive intermediates observed

ClHO

OR

ClO

OR

Strategies for mitigating effects of structural alerts

NNH

N

N

O

Cl

5-HT2C (serotonin) receptor agonist(Pfizer)

Ames positive (mutagenic)

Investigated as bradykinin B1 antagonist

(Merck)

Incubation with human and rat liver microsome

N

N

RO

NR

N

NH

RO

NHRO

N

NH

RO

NHR

O

N

NH

RO

NHR

GS

N

NH

RO

NHRO

GS

N

NH

RO

NHR

GS

GSH

GSH

GSH

– H2O

Irreversible binding to liver microsomal proteins

Tang, C., et. al. Chem. Res. Toxicol. 2005, 18 (6), 934–945.

2,3-diaminopyridine moiety determined to be safety liability

Strategies for mitigating effects of structural alerts

N

NH

OCF3

NH

MeO2C

(Human bradykinin B1 inhibitor constant)

Tang, C., et. al. Chem. Res. Toxicol. 2005, 18 (6), 934–945.

NH

OCF3

NH

MeO2C

Simplify to ethylene diamineAdd conformational

constraint and hydrogen bonding capability

NH

OCF3

NH

MeO2C

O

hBK B1 Ki = 52 μM

Inactive

Bioisosteric replacement of structural alerts

Design principle:Replace 2,3-DAP with

nonaromatic linker

N

NH

OCF3

NH

MeO2C

hBK B1 Ki = 0.012 μM

Lead compound

Structural modification

Wood, M. R., et. al. J. Med. Chem. 2006, 49 (4), 1231–1234.

NH

OCF3

NH

MeO2C

O

Me

MeNH

OCF3

NH

MeO2C

ON

NH

OCF3

NH

MeO2C

120o 109o 116o

Thorpe-Ingolde effect allows for bioisosteric mimicry of 2,3-diaminopyridine bond angle

Large improvement in rat pharmacokinetic profile due to heightened metabolic stability

Bioisosteric replacement of structural alerts

Lead compound Redesigned

Challenges in predicting adverse effects

The ability to predict a priori whether drug bioactivation will lead to toxicity in humans is very limited

Challenges in predicting adverse effects

OH

NH

O

Me

Acetaminophen (APAP)

AMAP

NH

O

Me

OH

Hepatotoxic

Nonhepatotoxic

Dose normalized to give comparable levels of in vivo

covalent binding in mice liver

Adduction of “critical proteins” is required to cause organ toxicity

Not all protein binding is deleterious

Tirmenstein, M. A., Nelson, S. D. J. Biol. Chem. 1989, 264, 9814–9819.

]

Shortcomings of the structural alert concept

Driscoll, J. P.; Sadlowski, C. M.; Shah, N. R.; Feula, A. J. Med. Chem. 2020, 63 (12), 6303–6314.

Not every structural alert results in toxicity

53% of 108 of the most prescribed drugs in 2009 possessed structural alerts

Evidence of reactive metabolite formation in 41% of those cases

OOMe

Cl

N

S

Clopidogrel Thiophene structural alert

OOMe

Cl

N

SOCYP

OOMe

Cl

N

SO

O

OOMe

Cl

NCOOH

SOH

H2O P2Y12 receptor

OOMe

Cl

NCOOH

SS P2Y12

]

]

Shortcomings of the structural alert concept

Driscoll, J. P.; Sadlowski, C. M.; Shah, N. R.; Feula, A. J. Med. Chem. 2020, 63 (12), 6303–6314.

Not every structural alert results in toxicity

53% of 108 of the most prescribed drugs in 2009 possessed structural alerts

Evidence of reactive metabolite formation in 41% of those cases

Irreversible inhibition of

platelet aggregation

OOMe

Cl

N

S

Clopidogrel Thiophene structural alert

OOMe

Cl

N

SOCYP

OOMe

Cl

N

SO

O

OOMe

Cl

NCOOH

SOH

H2O P2Y12 receptor

OOMe

Cl

NCOOH

SS P2Y12

]

Shortcomings of the structural alert concept

Kalgutkar, A. S. J. Med. Chem. 2020, 63 (12), 6276–6302.

Not every IADR associated with a structural alert

“If medicinal chemists abstained from the

synthesis of phenyl ring-containing compounds

because the phenyl ring is considered as a

structural alert, humanity would be deprived of

countless life-saving drugs”Ximelagatran

Rejected for FDA approval in 2009 after clinical trials due to idiosyncratic drug-

induced liver injury

Cannot completely eliminate structural alerts

N

HN

O

H2N NOH

O HN O

OMe

Does not contain any known structural alerts

Lack of thorough

characterization

distinguishing critical

and noncritical proteins

Challenges in predicting adverse effects

Structural alert

analysis often inaccurate

Inter- and intra-species variation in drug metabolism

The ability to predict a priori whether drug bioactivation will lead to organ or immune toxicity is very limited

Drug discovery and development

Discovery Preclinical development

Clinical trials

FDA review

Approved drug

In-vitro studies

In-vivo studies Animal trials Human trials

General population

Higher accuracy of testing results Increasing stakes and cost of test subjects

Image credit: Ian B. Perry

Outline

Introduction to drug metabolism

Common biotransformations

Applications to drug design

Designing around metabolism

Metabolism-induced toxicity

Mechanisms of adverse reactions Reactive intermediates and

structural alerts

Applications to drug design

Designing around drug-induced toxicity Challenges in predicting adverse effects