Download as pdf or txt
Download as pdf or txt
You are on page 1of 125

Dr.

Mahwish Akhtar
Ph.D. (Pharm. Chem.)
Assistant Professor
DCOP, DUHS

MEDICINAL CHEMISTRY I
617
Lecture 16-18
4th year, 7th semester
OBJECTIVES
• At the end of the lecture student will be able to
define
– Biotransformation
– Phases of drug metabolism
– Phase I
– Phase II
DRUG BIOTRANSFORMATION
DRUG BIOTRANSFORMATION
• The metabolism of a drug or toxin in a body is an
example of biotransformation.
• The body typically deals with a foreign compound
by making it more water-soluble, to increase the
rate of its excretion through the urine.
• There are many different process that can occur;
the pathways of drug metabolism can be divided
into:
• phase І
• phase II
DRUG BIOTRANSFORMATION
• Drugs can undergo one of four potential bio-
transformations:
– Active Drug to Inactive Metabolite
• Lidocaine, Nitroglycerin
– Active Drug to Active Metabolite
• Codeine --- morphine
• Tramadol --- O-desmethyltramadol
DRUG BIOTRANSFORMATION
• Drugs can undergo one of four potential bio-
transformations:
– Inactive Drug to Active Metabolite
• Lisdexamphetamine
• Prodrug of Epinephrine: Dipivefrin
– Active Drug to Toxic Metabolite (bio-toxification)
– Conversion of secondary amines in the stomach into
carcinogenic nitrosamines via NO pathway.
– Nicotine into the nitrosated carcinogenic NNK (4-(methylnitrosamino)- 1-(3-
pyridyl)-1-butanone) in the lung.
– Hypoglycin A into the highly toxic MCPA-CoA
Metabolite Examples and notes
activity
Inactive Routes that result in the formation of inactive metabolites are often referred to as
(detoxification) detoxification.

Similar The metabolite may exhibit either a different potency or duration of action or both to the
activity to the original drug.
drug

Different
activity

Toxic
metabolites
DRUG BIOTRANSFORMATION
• Phase I
• Includes oxidative, reductive, and hydrolytic reactions.
• In these type of reactions, a polar group is either
introduced or unmasked, so the drug molecule
becomes more water-soluble and can be excreted.
• Reactions are non-synthetic in nature and in general
produce a more water-soluble and more-active
metabolites.
• The majority of metabolites are generated by a
common hydroxylating enzyme system known
as Cytochrome P450.
DRUG BIOTRANSFORMATION
• Phase II reaction
• These reactions involve covalent attachment
of small polar endogenous molecule such as
glucuronic acid, sulfate, or glycine to form
water-soluble compounds.
• This is also known as a conjugation reaction.
• The final compounds have a larger molecular
weight.
DRUG BIOTRANSFORMATION
Metabolism
Metabolism
DRUG BIOTRANSFORMATION

• Functionalization • Conjugation (Phase 2)


reaction (Phase 1) • Glucuronic acid
• Oxidation Conjugation
• Reduction • Sulphate Conjugation
• Hydrolysis • Amino acid Conjugation
• Glutathione Conjugation
• Water Conjugation
• Acetyl Conjugation
• Methyl Conjugation
Phase I
Phase I

Oxidation Reduction Hydrolysis


• Aromatic moieties • Aldehyde & ketone • Ester & amide
• Olefins • Nitro & azo • Epoxide & arene
• Benzylic, allylic oxide by epoxide
carbon hydrase
• Aliphatic & alicylic
carbon
• Carbon heteroatom
(C-N, C-O, C-S)
• Alcohol & aldehyde
OXIDATION
Aromatic moieties

Olefins

Oxidation Benzylic, allylic


carbon

Aliphatic & alicylic


carbon
Carbon
heteroatom (C-N,
C-O, C-S)

Alcohol & aldehyde


OXIDATION

• Oxidation
– Oxidation of aromatic moieties (aromatic hydroxylation)
• Arenes to arenols
1. Propranolol
2. Phenobarbital
3. Phenytoin
4. Phenylbutazone
5. Atrorvastatin
6. 17α-Eyhinylestradol
7. Warfarin
8. Amphetamine
OXIDATION

■ Mixed function oxidation of arenes to


arenols via an epoxide intermediate
arene oxide
■ Major route of metabolism for drugs
with phenyl ring
■ Occurs primarily at para position
■ Substituents attached to aromatic ring
influence the hydroxylation
OH
■ Activated rings (with electron-rich
substituents) are more susceptible
while deactivated (with electron
withdrawing groups, e.g., Cl, N+R3,
COOH, SO2NHR) are generally slow or OH
resistant to hydroxylation
Oxidation (Aromatic hydroxylation)

Amphetamin
Phenytoi p- e
n hydroxyphenytoin

Warfarin sodium
17-α-Ethinylestradiol Propranolo
l

Phenylbutazon
e
Atorvastati
OXIDATION
OXIDATION (Aromatic hydroxylation)

Antihypertensive drug clonidine undergo little


aromatic hydroxylation and the uricosuric
agent probenecid has not been reported to
undergo any aromatic hydroxylation
Probeneci
Clonidin
d
e

Preferentially the more


electron rich ring is
hydroxylated
Diazepa Chlorpromazin
m e
OXIDATION

• Oxidation of olefins
OXIDATION

• Oxidation of olefins
OXIDATION

• Oxidation of olefins
– 7, 12-dimethylbenz [a] anthracene
– carbamazepine
OXIDATION

• Oxidation of olefins
– 7, 12-dimethylbenz [a] anthracene
– carbamazepine
OXIDATION

• Oxidation of allylic carbon atoms


• Tetrahydrocannabinol
Oxidation at Allylic Carbon
Atoms
Oxidation at Allylic Carbon
Atoms

Pentazocine
BENZYLIC CARBON

• Oxidation of benzylic carbon atoms


– To alcohol
– Aldehyde
– Carboxylic acid
• Tolbutamide
Benzylic Carbon
Hydroxylation
■ Hydroxylate a carbon attached to a phenol group (aromatic ring)
■ R1 and R2 can produce steric hindrance as they get larger and
more branched
■ So a methyl group is most likely to hydroxylate
■ Primary alcohol metabolites are often oxidized further to
aldehyde and carboxylic acids and secondary alcohols are
converted to ketones by soluble alcohol and aldehyde
dehydrogenase

Tolbutamide Metabolism

Dicarboxylic acid is
the major metabolite

Tolmetin sodium
Hydroxylation at C α to C=O and C=N

The benzodiazepines
are classic examples
with both functionalities

The sedative hypnotic


glutethimide possesses
C α to carbonyl function
Aliphatic
hydroxylation
■ Catalyzes hydroxylation of the ω and ω-1
carbons in aliphatic chains
■ Generally need three or more
unbranched carbons

Pentobarbital
Metabolism

Ibuprofen
+
Metabolism
Alicyclic (nonaromatic
ring) Hydroxylation

■ Cyclohexyl group is commonly present in many drug


molecules
■ The mixed function oxydase tend to hydroxylate at the 3 or
4 position of the ring
■ Due to steric factors if position 4 is substituted it is harder
to hydroxylate the molecules

Acetohexamide Metabolism
Oxidation Involving Carbon-
Heteroatom Systems
■ C-N, C-O and occasionally C-S
■ Two basic types of biotransformation processes:
1. Hydroxylation of α-C attached directly to the heteroatom (N,O,S).
The resulting intermediate is often unstable and decomposes
with the cleavage of the C-X bond:

Oxidative N-, O-, and S-dealkylation as well as oxidative deamination


reaction fall under this category
2. Hydroxylation or oxidation of heteroatom (N, S only, e.g., N-
hydroxylation, N-oxide formation, sulfoxide and sulfone formation)
■ Metabolism of some N containing compounds are complicated by the
fact that C or N hydroxylated products may undergo secondary
reactions to form other, more complex metabolic products (e.g.,
oxime, nitrone, nitroso, imino)
C-N systems
■ Aliphatic (1o, 2o, 3o,) and alicyclic (2o and 3o) amines; Aromatic and heterocyclic
nitrogen compounds; Amides
■ Enzymes:
1. CYP mixed-function oxidases: α-C hydroxylation and N-oxidation
2. Amine oxidases or N-oxidases (non-CYP, NADPH dependent flavoprotein
and require O): N-oxidation

■ 3 o Aliphatic and alicyclic


amines are metabolized by
oxidative N-dealkylation
(CYP)
■ Aliphatic 1 o , 2 o amines are
susceptible to oxidative
deamination, N-dealkylation
and N-oxidation reactions
■ Aromatic amines undergoes
similar group of reactions as
aliphatic amines, i.e., both N-
dealkylation and N-oxidation
N-Dealkylation (Deamination)

■ Deamination and N-dealkylation differ only in the point of reference; If the drug is R1 or R2
then it is a deamination reaction and If the drug is R3 or R4 then it is an N-dealkylation
N-Dealkylation (Deamination)

■ In general, least sterically hindered carbon (α) will be hydroxylated first, then the next, etc.
Thus the more substituent on this C, the slower it proceeds; branching on the adjacent
carbon slows it down, i.e. R1 , R 2 = H is fastest.
■ Any group containing an α-H may be removed, e.g., allyl, benzyl. Quaternary carbon
cannot be removed as contain no α-H
■ The more substituents placed on the nitrogen the slower it proceeds (steric hindrance)
■ The larger the substituents are the slower it proceeds (e.g. methyl vs. ethyl). In general,
small alkyl groups like Me, Et and i–Pro are rapidly removed; branching on these
substituents slows it down even more

Imipramine N-
Dealkylation
3 oAmine drugs

Lidocain Disopyramid Tamoxifen


e e

Diphenhydramin Chlorpromazin Benzphetamin Brompheniramin


e e e e
Alicyclic Amines Often Generate Lactams

Alicyclic Amine drugs

Meperidin Morphin Dextromethorpha


e e n
2o & 1o Amines

Generally, dealkylation of secondary amines occurs before deamination. The rate of


deamination is easily influenced by steric factors both on the α-C and on the N; so it is
easier to deaminate a primary amine but much harder for a tertiary amine.
Exceptions: Some 2 o and 3 o amines can undergo deamination directly without
dealkylation.
Alicyclic Amines Often Generate Lactams
N-
Oxidation
Aromatic
amines


amines


amines


amines
■ The attack is on the unbonded electrons so 3o amines can be oxidized
■ Generally, only occurs if nothing else can happen, so it is a rare reaction
■ Performed by both amine oxidases and hepatic MFO’s (mix function
oxidase)
■ Good examples would include amines attached to quaternary carbons
since they cannot be deaminated

Chlorphentermine N-
Hydroxylation Hydroxylamin
e

Nitros
o

Nitr
o
Phentermin
Amantadin
e
e
Amide
s

C-N bond cleavage via α-C hydroxylation (formation of carbinolamide) and N-


hydroxylation reactions
Oxidation involving C-O System (O-Dealkylation)

■ Converts an ether to an alcohol plus a ketone or aldehyde


■ Steric hindrance discussion similar to N-dealkylation

Trimethoprim O-Dealkylation
Codein Phenaceti Indomethaci
e n n

Metoprolo
Prazosi l
n
■ One exception that appears to be a form of O-dealkylation is the
oxidation of ethanol by CYP2E1
■ In this case R3 is hydrogen instead of carbon to form the terminal
alcohol rather than an ether
■ The enzyme involved is CYP2E1 and has been historically referred
to as the Microsomal Ethanol Oxidizing System (MEOS)
Oxidation involving C-S System
■ S-

Dealkylation
Steric hindrance discussion similar to N-
dealkylation

■ Desulfuration

■ S-Oxidation
Oxidative
Dehalogenation

■ Requires two halogens on carbon


■ With three there is no hydrogen available to
replace
■ With one, the reaction generally won’t proceed
■ The intermediate acyl halide is very reactive

Q. What is Gray Baby Syndrome?


REDUCTION & HYDROLYSIS
REDUCTION

• Reduction
• Aldehyde to alcohol
• chlorpheniramine to
aldehyde metabolite
to reduce alcohol and
oxidize to carboxylic
acid
REDUCTION

• Reduction
• Ketone to 2˚ alcohol
(acetophenone)
REDUCTION

• Reduction
• Nitro group (clonazepam) to amine
ce
HYDROLYSIS

• Hydrolysis
HYDROLYSIS

• Hydrolysis (aspirin)

= +
Phase II
Phase II Metabolism
• Phase II is usually the true detoxification of
drugs
• Occurs mostly in cytosol
• Gives products that are generally water
soluble and easily excreted
• Includes sugar conjugation, sulfation,
methylation, acetylation, amino acid
conjugation, glutathione conjugation
Phase II Metabolism
• Transferase: is any one of a class of
enzymes that enact the transfer of specific
functional groups
• Cofactor: a substance (other than the
substrate) whose presence is essential for
the activity of an enzyme.
Phase II Metabolism
Conjugation (Phase 2)
Glucuronic acid Conjugation

Sulphate Conjugation

Amino acid Conjugation


(Glycine, glutamine etc)

Glutathione Conjugation

Acetyl Conjugation (Acetylation)

Methyl Conjugation (Methylation)


Conjugation (Phase 2)

Glucuronic acid Conjugation Sulphate Conjugation

Amino acid Conjugation Amino acid Conjugation


(Glutamine) (Glycine)
Conjugation (Phase 2)

Acetyl Conjugation
(Acetylation)

Glutathione Conjugation

Methyl Conjugation (Methylation)


Conjugation (Phase 2)
Conjugation (Phase 2)
• -OH, -SH, -COOH, -CONH with glucuronic acid to
give glucuronides
• -OH with sulphate to give sulphates derivatives
• -NH2, -CONH2, amino acids, sulpha drugs with
acetyl- to give acetylated derivatives
• -halo, -nitrate, epoxide, sulphate with glutathione to
give glutathione conjugates all tend to be less lipid
soluble and therefore better excreted (less well
reabsorbed)
Phase II Metabolism
Cofactors

S-Adenosyl methionine (SAM)

3'-Phosphoadenosine-5‘-phosphosulfate (PAPS)
-OH, -COOH, -NH2 , -NR2 , -SH, -CH
UDP-glucoronosyl- transferase

GLUCURONIDATION
Glucuronidation

• Most important conjugation reactions


• Cofactor UDP – glucuronic acid is in high
abundance
– Closely related to glycogen synthesis
– Found in all tissues of the body
• Other sugars, glucose, xylose or ribose
may be conjugated
Glucuronidation

Liver has several soluble UDP-Gluc-


Glucuronidation
Glucuronidation

Glucuronic acid conjugation to


phenols, 3°-amines, aromatic amines
Glucuronidation
-OH, -NH2
Sulfotransferase

SULFATION
Sulfation

• Major conjugation pathway for phenols, also


alcohols and amines
• Compounds that can be glucuronidated can
also be sulfated
• Can be competition between the two pathways
• In general, sulfate conjugation predominates at
low substrate concentration and glucuronide
conjugation predominates at high substrate
concentration
Sulfation

(PAPS, 3’-phosphoadenosine-
5’-phosphosulfate)

Examples: ethanol, p-hydroxyacetanilide, 3-


hydroxycoumarin
Sulfation

Sulfation may produce active metabolite


Sulfation
ArX, Arene oxide, Epoxide, Carbocation
Glutathione S- transferase

GLUTATHIONE CONJUGATION
Glutathione Conjugation

• Glutathione is a protective compound (tripeptide,


Gly-Cys-Glu) within the body for removal of
potentially toxic electrophilic compounds
• Many drugs are metabolized in phase I to strong
electrophiles
• React with glutathione to form non-toxic
conjugates
• Glutathione conjugates may be excreted directly in
urine or bile, but are usually metabolized further
Glutathione Conjugation
Glutathione Conjugation
Glutathione Conjugation
-OH, -NH2, -SH
Acetyltransferase

ACETYLATION
Acetylation

Examples: Procainamide, isoniazid, sulfanilimide, histamine

NAT enzyme is found in many tissues, including liver


Acetylation
-OH, -NH2, -SH, heterocyclic N
Methyltransferase

METHYLATION
Methylation
• Addition of methy group
• Cofactor is catechol-O-methyltransferase
(COMT)
Methylation
-COOH
Glycine or Glutamine N- acyltransferase

GLYCINE/GLUTAMINE
GLYCINE/GLUTAMINE
• Glutamine
GLYCINE/GLUTAMINE
Glycine Conjugation
Factors affecting Drug
Metabolism
Factors affecting Drug Metabolism
• Disease Factors
• Liver Disease- cirrhosis, alcoholic liver disease, jaundice, carcinoma
• Major location of drug metabolizing enzymes
• Dysfunction can lead to impaired drug metabolism decreased
enzyme activity
• First pass metabolism effected-may increase 2-4 times
bioavailability
• Result in exaggerated pharmacological responses and adverse
effects
• Cardiac failure causes decreased blood flow to liver
• Hormonal diseases, infections and inflammation can change drug
metabolizing capacity
Factors affecting Drug Metabolism
• Age
• Newborns and infants- metabolize drugs
relatively efficiently but at a rate generally
slower than adults
• Full maturity appears in second decade of life
• Slow decline in function associated with aging
Factors affecting Drug Metabolism
• Genetic Variation
• Wide variability in response to drugs b/w individuals
• Consequences of such variation may be therapeutic failure or an adverse
drug reaction
• Genetic diversity is rule rather than the exception with all proteins,
including drug metabolizing enzymes
• Allelic variants with different catalytic activities from that of the wild-type
form have been identified
• Inheritance leads to subpopulations (genetic polymorphisms) with
different drug metabolizing abilities
– Lack of activity
– Reduction in catalytic ability
– Enhanced activity
• Frequency of polymorphism often varies according to ethnic ancestry of
individual
Genetic factor
Factors affecting Drug Metabolism
• Gender
• Responsiveness to certain drugs is different
for men and women
• Pregnancy-induction of certain drug
metabolizing enzymes occurs in second and
third trimester
• Hormonal changes during development have
a profound effect on drugs metabolism
SAR &QSAR
SAR &QSAR
HANSCH ANALYSIS
• Electronic properties
• Lipophilicity
• Steric effect

FREE-WILSON ANALYSIS
• Substitution at specific position

MIXED APPROACH
• Follow above rules
REFERENCES
• Foye’s the Principles of Medicinal Chemistry, 6th ed,
Lippincott William & Wilkins
• Textbook of medicinal chemistry, Vol 1, by V.
Alagarsamy, Elsevier.

You might also like