0% found this document useful (0 votes)
19 views

Phar MedChem Chemo Unit3 part3

pharma

Uploaded by

Karo Moti
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
19 views

Phar MedChem Chemo Unit3 part3

pharma

Uploaded by

Karo Moti
Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 239

Medicinal Chemistry II

Unit 3 – Chemotherapeutic Agents


Part 3 – Antifungal, Antiviral, Antiprotozoal,
Anthelmintic & Antineoplastic agents
Antifungal agents
Introduction
➢ Fungi are plant-like non-photosynthetic Eukaryotes.
✓ Yeasts, molds, rusts and mushrooms.
✓ Heterotrophic obtain nutrients from environment,
not from endogenous sources.
✓ Saprophytes live in soil or on dead plants &
involved in biodegradation.
➢ Thus, most fungi are beneficial as they take key role
to the mineralization of dead bio matter.
➢ However, few can cause opportunistic infections.
Introduction

➢ Human-fungi-parasitic relationship result in mycotic

illnesses.

➢ Most fungal infections (mycoses) involve superficial

invasion of the skin or mucous membrane of the body

orifices.

➢ These diseases can usually be controlled by local

application of the antifungal agents.


Introduction
➢ Fungi have different shapes and sizes. Some are large while

others are minute, parasitic, and saprophytic cells. They

differ from the following organisms in some important

aspects:

✓ Algae by lack of photosynthetic ability.

✓ Protozoa by the lack of motility, possession of chitinous

cell wall, and ease of culture on simple media.

✓ Bacteria by greater size and having certain intracellular

structure such as mitochondria and nuclear membrane.


Classification Based on the Chemical Structure
1. Antibiotics: Amphotericin B, Nystatin, Griseofulvin.
2. Azoles: (Imidazole, Triazole derivates).
a. Triazoles: Fluconazole, Itraconazole, Terconazole.
b. Imidazoles: Clotrimazole, Ketoconazole,
Miconazole, Bifonazole, Butoconazole, and
Zinoconazole.
3. Fluorinated Pyrimidines: Flucytosine.
4. Chitin Synthetase Inhibitors: Nikomycin Z.
5. Peptides/Proteins: Cispentacin.
6. Miscellaneous: Ciclopirox, Tolnaftate, Naftifine, and
Terbinafine.
1. Antibiotics
Amphotericin B

➢ It is polyene antibiotic obtained from Streptomyces nodosus.


➢ It is an amphoteric compound that consists of seven-
conjugated double bond, an internal ester, a free carbonyl
group, and a glycoside side chain with a primary amino group.
➢ The carbohydrate moiety is D-mycosamine.
1. Antibiotics
Amphotericin B

➢ The conjugated systems are usually of all trans


configurations, so that the ring contains a planner
lipophilic segment and a less rigid hydrophilic
portion.
➢ Amphotericin B is an amphoteric, forming soluble
salts in both basic and acidic environments, and due
to extensive unsaturation, it is unstable, primarily
used as antifungal agents.
1. Antibiotics
Amphotericin B

Mode of action:
➢ The antifungal activity of this drug depends on its binding to a
sterol moiety.
➢ Primarily, ergosterol that is present in the membrane of
sensitive fungi, by virtue of their interaction with the sterols of
cell membranes and polyenes, appear to form pores or
channels.
➢ The result is an increase in the permeability of the membrane,
allowing leakage of a variety of small molecules, such as
intracellular potassium, magnesium, sugars, and metabolites
leading to cellular death.
1. Antibiotics
Nystatin

➢ It is a polyene antibiotic isolated from Streptomyces


noursei. It is structurally similar to amphotericin B and
has the same mechanism of action.
➢ It is used as an antifungal agent.
1. Antibiotics
Griseofulvin

➢ Griseofulvin is used as an antifungal agent.

➢ It is a fungi-static drug that causes disruption of the

mitotic spindle by interacting with polymerized

microtubules.
2. Azole Antifungals (Imidazole & Triazole)

Mode of action:
➢ Azole antifungals inhibit sterol 14-α-demethylase, a
microsomal cytochrome P450-dependent enzyme system,
and thus, impair the biosynthesis of ergosterol for the
cytoplasmic membrane and lead to the accumulation of 14-α-
methyl sterols.
➢ These methyl sterols may disrupt the packing of aryl chains
of phospholipids, the functioning of certain membrane
bound enzyme systems, such as ATPase and enzymes of the
electron transport system, and thus, inhibiting the growth of
fungi.
2. Azole Antifungals (Imidazole & Triazole)
Miconazole and Econazole

➢ Miconazole is a white or almost white powder, very slightly


soluble in water, sparingly soluble in methanol, and slightly
soluble in alcohol.
➢ Econazole is white or almost white crystalline powder, very
slightly soluble in water, soluble in methanol, sparingly
soluble in methylene chloride, and slightly soluble in alcohol.
2. Azole Antifungals (Imidazole & Triazole)
Terconazole and Ketoconazole

Terconazole:
➢ It is metabolized by CYP3A4 on oral administration.
➢ It is a triazole derivative that is used exclusively for the control
of Vulvovaginal moniliasis caused by Candida albicans and
other Candida spp.
2. Azole Antifungals (Imidazole & Triazole)
Terconazole and Ketoconazole

Ketoconazole:
➢ It is extensively metabolized by deacetylase of the microsomal
enzymes and all the metabolites are inactive.
➢ It is a racemic compound, consisting of the cis-2S, 4R, and cis-
2R, 4S isomers.
➢ Ketoconazole is an imidazole antifungal agent, which is a highly
lipophilic compound.
➢ This property leads to high concentrations of ketoconazole in
fatty tissues and purulent exudates.
➢ Ketoconazole is active against Candida spp and Cryptococcus
neoformans.
2. Azole Antifungals (Imidazole & Triazole)
Clotrimazole

➢ Clotrimazole is a white or pale yellow crystalline powder,

practically insoluble in water, soluble in alcohol and in

methylene chloride. It is used as an antifungal agent.


2. Azole Antifungals (Imidazole & Triazole)
Fluconazole

➢ It is a widely used bis-triazole antifungal agent.


➢ It is generally considered to be a fungi-static agent, and it is
principally active against Candida spp and Cryptococcus spp.
➢ Fluconazole has useful activity against Coccidioides immitis,
and is often used to suppress the meningitis produced by the
fungus.
2. Azole Antifungals (Imidazole & Triazole)
Structure - Activity Relationships of Azoles

➢ Basic structural requirement for members of azole class


is a weakly basic imidazole or 1,2,4-triazole ring (pKa of
6.5 to 6.8) bonded by a nitrogen-carbon linkage to rest of
structure.
➢ At molecular level, amidine nitrogen atom (N3 in
imidazoles, N4 in triazoles believed to bind to heme iron of
enzyme-bound cytochrome P450 to inhibit activation of
molecular oxygen & prevent oxidation of steroidal
substrates by enzyme.
2. Azole Antifungals (Imidazole & Triazole)
Structure - Activity Relationships of Azoles

➢ Most potent antifungal azoles possess two or three aromatic

rings, at least one of which is halogen substituted (e.g., 2,4-

dichlorophenyl, 4-chlorophenyl, or 2,4-difluorophenyl and other

nonpolar functional groups. Only 2 and/or 2,4-substitution

yields effective azoles.

➢ Halogen atom that yields most potent compounds is fluorine,

although groups like sulfonic acids have same effects.

➢ Substitution at other positions, yields inactive compounds.


2. Azole Antifungals (Imidazole & Triazole)
Structure - Activity Relationships of Azoles

➢ Presumably, large nonpolar portion of molecules mimics

nonpolar part of substrate for lanosterol l4-α-demethylase, in

shape and size.

➢ Nonpolar functionality confers high lipophilicity to azoles.

➢ Free bases are typically insoluble in water but are soluble in

most organic solvents, like ethanol.

➢ Fluconazole, which possesses two polar triazole moieties, is

exception, it is sufficiently water soluble to be injected

intravenously as a solution of the free base.


2. Azole Antifungals (Imidazole & Triazole)
Structure - Activity Relationships of Azoles

➢ Imidazoles – 1st generation:

─ All contain a 1-substituted imidazole ring.


2. Azole Antifungals (Imidazole & Triazole)
Structure - Activity Relationships of Azoles

➢ Imidazoles – 2nd generation:


─ More hydrophilic & can be administered orally.
─ Imidazole ring still susceptible to metabolic
degradation in vivo, less than 5% excreted
unchanged.
2. Azole Antifungals (Imidazole & Triazole)
Structure - Activity Relationships of Azoles

➢ 1,2,4-Triazoles – 3rd generation:


─ Much more hydrophilic & so administered orally.
─ Triazole ring much less susceptible to degradation
in vivo.
3. Fluorinated Pyrimidines
Flucytosine

Mode of action:
➢ Flucytosine is converted by cytosine deaminase into 5-
flurouracil (5-FU), then, 5-fluoro deoxy uridylic acid is formed.
➢ This false nucleotide inhibits thymidylate synthetase, thus,
depriving the organism of thymidylic acid, an essential DNA
component.
3. Fluorinated Pyrimidines
Flucytosine

➢ It is a potent antimetabolite, which replaces uracil in the


pyrimidine pool and thus, disrupts protein synthesis.
➢ Mammalian cells do not convert flucytosine to fluorouracil. This
fact is crucial for the selective action of this compound.
➢ Flucytosine is the only available antimetabolite drug having
antifungal activity.
4. Chitin Synthetase Inhibitors
Nikomycin

➢ Nikomycin competitively inhibits the chitin synthase, mimicking its


substrate uridine diphosphate-N-acetyl glucosamine of fungi with C.
albicans as the primary target organism.
➢ The enzyme is an integral membrane protein.
➢ Nikomycin is a nucleoside peptide antibiotic that is produced by soil
strains of Steptomyces tendae.
➢ It is found to be more potent than most azoles in inhibiting highly
chitinous, dimorphic fungal pathogens, such as Coccidiodes imitis and
Blastomyces dermatidis.
5. Peptides/Proteins
➢ The inhibitors of glucosamine-6-
phosphate synthase delivered
through peptide transport
systems have been reported as
having anticandidal activity.

➢ It is dipeptide analogue that is


shown in vitro and in vivo
activity against C. albicans.
6. Miscellaneous agents
Naftifine

➢ The drug has fungicidal activity against Tinea cruris and

Corporis spp.

➢ This inhibits squalene 2,3-epoxidase and thus, inhibits fungal

biosynthesis of ergosterol.
6. Miscellaneous agents
Ciclopirox

➢ Ciclopirox It is available as 1% cream on cotton for the

treatment of cutaneous candidiasis and for Tinea corporis, T.

cruris, T. pedis, and Pityriasis versicolour.


6. Miscellaneous agents
Tolnaftate

➢ Tolnaftate is effective for the treatment of most cutaneous


mycoses, such as Trichophyton rubrum and Microsporum canis.
➢ Replacement of the aromatic methyl group by hydroxy or
methoxy or its removal does not affect potency.
➢ Replacement of the complete tolyl group by a α-napthyl or a β-
napthyl substituent does not decrease its potency.
➢ It is nontoxic.
6. Miscellaneous agents
Terbinafine

➢ Terbinafine is active by virtue of its ability to block squalene

epoxidase.

➢ Several CYP450 enzymes, including CYP1A2, CYP2C19,

CYP2C9, CYP2C8, CYP3A4, and CYP2B6, extensively

metabolize it.
Antiviral agents
Introduction
➢ Antiviral agents are substances used in the treatment
and prophylaxis of diseases caused by viruses.
➢ Viral diseases include influenza, rabies, yellow fever,
poliomyelitis, mumps, measles, Ebola, human immuno
deficiency virus (HIV), herpes, warts, and small pox.
➢ Viruses are not proper living things, but consists of a
genome; they are smaller in size with simple chemical
composition, sometimes a few enzymes stored in a
capsule made up of protein and rarely covered with a
lipid layer.
Introduction
➢ The viruses only replicate within the host cell and the viral
replication depends primarily on the metabolic processes of
the invaded cell.
➢ Viruses does not possess cell wall, but they have RNA or DNA
enclosed in a shell of protein known as capsid.
➢ The capsid is composed of several subunits known as
capsomers.
➢ In certain cases, capsid may be surrounded by an outer
protein or lipoprotein envelope.
➢ One group of RNA virus that are known as retro viruses. They
are responsible for acquired immuno deficiency syndrome
(AIDS) and T - Leukemias.
Introduction
➢ Retro viruses contain reverse transcriptase (RT) enzyme
activity that makes a DNA copy from the viral RNA template.
Then, the DNA copy is integrated into the host genome, at
which it is referred to as provirus and is transcribed into both
the genomic RNA and mRNA for translocation into the viral
proteins, giving generation to new virus particles.
➢ Viral life cycle varies according to the species, but they all
share a general pattern that can be sequenced as follows:
Introduction
➢ Adsorption: Attachment of the virus to the host cell.
➢ Penetration: Penetration of virus into the cell.
➢ Uncoating: The genetic material or viral genome (DNA or RNA)
passes into the host cell leaving the capsid covering outside
the host cell.
➢ Transcription: Production of the viral mRNA from the viral
genome.
➢ Translation: The viral genome enters the cytoplasm or the
nucleoplasma and directs or utilizes the host nucleic acid
machinery for the synthesis of the new viral protein and for the
production of more viral genome. The viral protein modifies
the host cell and allows the viral genome to replicate by using
host and viral enzyme.
Introduction
➢ Translation (Cont.): This is often the stage at which the cell is
irreversibly modified and eventually killed.
➢ Assembly of the viral particle: New viral coat protein
assembles into capsid and viral genomes.
➢ Release of the mature virus from the cell and the budding
process or rupture of the cell and repeat of the process, in a
fresh host cell.
Since the host cell machinery is totally utilized for the production
of new virions, the normal cell function is affected. Antiviral
agents have been developed to act at various stages in the viral
replication cycle, such as attachments, replication, and release
of the virus.
Examples of Viruses with Diseases
Classification of Antiviral Agents
I. Nucleoside/tide Reverse Transcriptase Inhibitors
(Ns(t)RTIs).
a. Purine nucleosides and nucleotides.
b. Pyrimidine nucleosides and nucleotides.
c. Miscellaneous NRTIs.
II. Non-nucleoside Reverse Transcriptase Inhibitors
(NNRTIs).
III. Reverse Transcriptase Inhibitors (Anti-HIV agents).
IV. HIV protease Inhibitors (Anti-HIV agents).
V. Miscellaneous.
Classification of Antiviral Agents
I. Nucleoside/tide RT inhibitors (Ns(t)RTIs)
a. Purine nucleosides and nucleotides
Classification of Antiviral Agents
I. Nucleoside/tide RT inhibitors (Ns(t)RTIs)
b. Pyrimidine nucleosides and nucleotides
Classification of Antiviral Agents
I. Nucleoside/tide RT inhibitors (Ns(t)RTIs)
c. Miscellaneous NRTIs
Classification of Antiviral Agents
II. Non-nucleoside RT inhibitors (NNRTIs)
Classification of Antiviral Agents
III. RT inhibitors (Anti-HIV agents)
Classification of Antiviral Agents
IV. HIV protease inhibitors (Anti-HIV agents)
Classification of Antiviral Agents
IV. HIV protease inhibitors (Anti-HIV agents)
Classification of Antiviral Agents
V. Miscellaneous
a. Thiosemicarbazones
Classification of Antiviral Agents
V. Miscellaneous
b. Adamantane amines
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)

➢ Acyclovir has nucleoside-like structure, same nucleic acid


base as deoxy guanosine but, lacks complete sugar ring.
➢ In virally infected cells, it is phosphorylated in three stages to
form a triphosphate which is active agent → acyclovir a
prodrug.
➢ Acyclovir triphosphate prevents DNA replication in two ways.
✓ First, similar to normal deoxy guanosine triphosphate
building block that can bind to DNA polymerase and
inhibit.
✓ Second, DNA polymerase can catalyze attachment of
acyclovir nucleotide to growing DNA chain.
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)

✓ As sugar unit is incomplete & lacks required hydroxyl group


normally present at position 3′, chain cannot be extended
any further, thus acts as a chain terminator.
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)

➢ Oral BA of Acyclovir is quite low (15–30%), to overcome


this, various prodrugs were developed to increase water
solubility.
➢ Valaciclovir is L-valyl ester prodrug absorbed from the
gut, far more effective than Acyclovir (active
transportation) - once absorbed, hydrolyzed to Acyclovir in
liver & gut wall.
➢ Desciclovir is a prodrug of Acyclovir which lacks carbonyl
group at position 6 of purine ring & more water soluble -
once in blood supply, metabolism by cellular xanthine
oxidase oxidizes 6-position to give Acyclovir.
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)

➢ Ganciclovir is Acyclovir analogue, extra hydroxy methylene.


➢ Valganciclovir acts as a prodrug for this compound.
➢ Penciclovir & prodrug Famciclovir analogues of Ganciclovir.
➢ In Famciclovir, the two alcohol groups of Penciclovir masked
as esters making less polar, resulting in better absorption.
➢ Once absorbed, acetyl groups hydrolyzed by esterase &
purine ring oxidized by aldehyde oxidase to generate
Penciclovir.
➢ Phosphorylation reactions take place in virally infected cells.
➢ Some viruses are immune from action of these antiviral agents
- phosphorylation fails to take place because they lack enzyme
thymidine kinase.
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)

➢ Cidofovir, analogue of deoxycytidine 5-monophosphate where

sugar & phosphate groups replaced by acyclic group & a

phosphono methylene group, designed to combat problem.

➢ Phosphono methylene acts as a bio isostere for phosphate

group & used because phosphate group would be susceptible

to enzymatic hydrolysis.

➢ As a phosphate equivalent is now present, the drug does not

require viral thymidine kinase to become activated.

➢ Two more phosphorylation can take place catalyzed by

cellular kinases, to convert Cidofovir to active ‘triphosphate’.


Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Inhibitors of viral RNA dependent DNA polymerase (RT)

➢ In contrast to acyclovir, idoxuridine, trifluridine, vidarabine are


phosphorylated equally well by viral & cellular thymidine
kinases, so there is less selectivity for virally infected cells.
➢ As a result, these drugs have more toxic side effects -
triphosphate inhibits viral DNA polymerase, as well as
thymidylate synthetase.
Nucleoside/tide Reverse Transcriptase Inhibitors (Ns(t)RTIs)
Miscellaneous NRTIs - Ribavirin

➢ It is guanosine (ribonucleic) analog used to stop viral RNA


synthesis and viral mRNA capping, thus it is a nucleoside
inhibitor.
➢ Ribavirin is a prodrug, which when metabolized resembles
purine RNA nucleotides. In this form, it interferes with RNA
metabolism required for viral replication.
➢ It is used in the treatment of influenza type-A and -B, hepatitis,
genital herpes, and Lassa fever.
Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs)

➢ Hydrophobic molecules that bind to allosteric binding site

which is hydrophobic in nature.

➢ Since allosteric binding site is separate from substrate

binding site, NNRTIs are non-competitive, reversible

inhibitors.

➢ Binding of a NNRTI to the allosteric site results in an

induced fit which locks the neighboring substrate-binding

site into an inactive conformation.


Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs)

➢ Nevirapine has rigid butterfly-like conformation that

makes chiral: one ‘wing’ interacts through hydrophobic

and van der Waals interactions with aromatic residues

in the binding site, while the other wing interacts with

aliphatic residues.

➢ Other NNRTI inhibitors bind to the same pocket and

appear to function as π electron donors to aromatic side

chain residues.
Non-nucleoside Reverse Transcriptase Inhibitors (NNRTIs)

Non-nucleoside reverse transcriptase inhibitors in clinical use.


(interactions with amino acids in the binding site are shown in blue).
Reverse Transcriptase Inhibitors (Anti-HIV agents)
➢ Reverse transcription is RNA dependent DNA polymerase.
➢ The drug inhibiting RT interferes with the replication of HIV
and stops the synthesis of further viral particle.

Didanosine

➢ Didanosine is ultimately converted into hypoxanthine, xanthine,


and uric acid through the usual metabolic pathways of purines.
The latter is a nontoxic metabolic product.
➢ It is a nucleoside RT inhibitor recommended for the treatment of
patients with advanced HIV infections.
Reverse Transcriptase Inhibitors (Anti-HIV agents)
Zidovudine (ZDV) [azido-deoxythymidine (AZT)

➢ Most of the administered drug is converted to its inactive glucuronide


metabolite and it is excreted unchanged through urine.
➢ ZDV is a nucleoside RT inhibitor, having activity against HIV, and
hence, it is used for the treatment of AIDS and AIDS-related complex
(ARC). It increases the survival and improves the quality of life of
patients with complications, such as severe weight loss, fever, and
pneumocystis.
➢ As it crosses the blood brain barrier, it has favorable effect on the
neurological symptoms of AIDS.
Reverse Transcriptase Inhibitors (Anti-HIV agents)

Zalcitabine

➢ Zalcitabine exists as white crystals.


➢ It is approved for combination therapy with zidovudine in
advanced HIV infection, who has demonstrated significant
clinical or immunological deterioration, showing intolerance to
zidovudine.
Reverse Transcriptase Inhibitors (Anti-HIV agents)

Lamivudine

➢ It is a nucleoside RT inhibitor, used in combination with ZDV for


the treatment of diseases caused by HIV infection.
HIV protease Inhibitors (Anti-HIV agents)

➢ Unlike reverse transcriptase inhibitors, PIs are not


prodrugs and do not need to be activated.
➢ Therefore, is possible to use in vitro assays
involving virally infected cells in order to test their
antiviral activity.
➢ Protease enzyme can also be isolated, allowing
enzyme assays to be carried out to measure IC50
levels as a measure of how effectively novel drugs
inhibit protease enzyme.
Protease Enzyme

➢ Protease is a type of enzyme that functions mainly

to help us digest different kinds of proteins.

➢ They break down the bonds by a process known as

hydrolysis and convert proteins into smaller chains

called peptides or even smaller units called amino

acids.
Protease in HIV
➢ When viral RNA is translated into a polypeptide
sequence, that sequence is assembled in a long
chain that includes several individual proteins
(reverse transcriptase, protease, integrase).
➢ Before these enzymes become functional, they must
be cut from the longer polypeptide chain.
➢ Viral protease cuts the long chain into its individual
enzyme components which then facilitate the
production of new viruses.
Protease in HIV

➢ HIV-1 protease cleaves poly-protein precursors to


form functional proteins.
Enzyme Inhibition
Protease in HIV
➢ HIV protease enzyme is an example of an enzyme family

called the aspartyl proteases — enzymes which catalyze

the cleavage of peptide bonds and which contain an

aspartic acid in the active site that is crucial to the catalytic

mechanism.

➢ A symmetrical dimer made up of two identical protein

units, each consisting of 99 amino acids.

➢ The active site is at the interface between protein units and

is also symmetrical.
Protease in HIV
➢ The amino acids Asp-25, Thr-26, and Gly-27 from each
monomer are located on the floor of active site, and each
monomer provides a flap to act as the ceiling.
➢ Enzyme has a broad substrate specificity, can cleave a variety
of peptide bonds in viral polypeptides, crucially, cleave bonds
between a proline residue & aromatic residue (phen or tyr).
➢ Cleavage of peptide bond next to proline is unusual, not occur
with mammalian proteases (renin, pepsin, or cathepsin D), so
achieving selectivity on HIV protease over mammalian is
possible.
➢ Symmetrical nature of viral enzyme active site is not present
in mammalian proteases → possibility of drug selectivity.
Protease in HIV
➢ There are eight binding subsites in the enzyme - four on each
protein unit, located on either side of catalytic region.
➢ These subsites accept amino acid side chains of the substrate
& numbered S1–S4 on one side, S1′– S4′ on other side,
relevant side chains on substrate numbered P1–P4 & P1′–P4′.
➢ Peptide bonds in substrate are also involved in hydrogen
bonding interactions with the active site.
➢ A water molecule present in active site w/c acts as a hydrogen
bonding bridge to two isoleucine NH groups on enzyme flaps.
➢ This hydrogen bonding network has the effect of closing the
flaps over the active site once the substrate is bound.
Protease in HIV
Design of HIV Protease Inhibitors (PIs)

➢ Transition-state inhibitors are designed to mimic the

transition state of an enzyme - catalyzed reaction.

➢ Advantage of this approach is transition state is

likely to be bound to active site more strongly than

substrate or product.

➢ In HIV protease-catalyzed reaction, transition state

resembles tetrahedral intermediate.


Design of HIV Protease Inhibitors (PIs)

➢ Structures are inherently unstable, it is necessary to design


an inhibitor which contains a transition-state isostere.
➢ Such isostere have a tetrahedral center to mimic tetrahedral
center of transition state, yet be stable to hydrolysis.
➢ Large number of structures were synthesized incorporating
these isosteres, with hydroxy ethylamine isostere.
Design of HIV Protease Inhibitors (PIs)
Design of HIV Protease Inhibitors (PIs)
➢ Isostere has a hydroxyl group which mimics one of
hydroxyl groups of tetrahedral intermediate & binds to
aspartate residues in active site, stereochemistry of
this group is also important to activity, with R -
configuration generally being preferred.
➢ In theory, it might make sense to design inhibitors
such that all eight subsites are filled to allow stronger
interactions.
➢ However, this leads to structures with a high
molecular weight and, consequently, poor oral
bioavailability.
Design of HIV Protease Inhibitors (PIs)
➢ Therefore, most of PIs designed to have a core unit spanning

S1 to S1′ subsites; further substituents were then added at

either end to fit into the S2/S3 and S2′/S3′ subsites.

➢ Early inhibitors, such as saquinavir have amino acid side

chains that bind to most of the subsites from S3 to S3′.

─ but these compounds have a large molecular weight & a high

peptide character leading to poor pharmacokinetic

properties.

➢ More recent inhibitors designed with increased aqueous

solubility, oral BA using variety of novel P2 & P2′ groups that

reduce molecular weight & peptide character of compounds.


Design of HIV Protease Inhibitors (PIs)
➢ S2 & S2′ subsites of protease enzyme appear to contain
both polar (Asp-29, Asp-30) & hydrophobic (Val-32, Ile-
50, -84) AAs, allowing design of drugs that contain
hydrophobic P2 groups which are also capable of
hydrogen bonding.
➢ Also possible to design a P1 group that span both S1 &
S3 subsites, allowing removal of a P3 moiety, thus
lowering molecular wt.
➢ P2 group is usually attached to P1 by acyl link, because
carbonyl oxygen concerned acts as important hydrogen
bond acceptor to the bridging water molecule.
Design of HIV Protease Inhibitors (PIs)
➢ Variations carried out on carboxyl half of molecule.
✓ Proline fits into S1′ pocket, but was possible to replace with a
bulkier decahydro isoquinoline ring system.
✓ t-butyl ester protecting group occupy S2′ subsite, can be
replaced by t-butyl amide group which proved more stable.
✓ Resulting structure (saquinavir) showed 60-fold ↑activity.
✓ R - stereochemistry of transition-state hydroxyl is essential.
✓ If configuration is S, all activity is lost.
Design of HIV Protease Inhibitors (PIs)
➢ X-ray crystallography of enzyme–saquinavir complex
demonstrated following:
✓ Substituents on drug occupy five subsites S3–S2′.
✓ t-butyl amine nitrogen is positioned in such a way that
further N -substituents incapable of reaching S3′
subsite;
✓ HB interactions between hydroxyl of hydroxy ethylamine
moiety & catalytic aspartates (Asp-25 and Asp-25′);
✓ Carbonyl groups on either side of transition state
isostere act as hydrogen bond acceptors to a bridging
water molecule.
Design of HIV Protease Inhibitors (PIs)
➢ Various efforts made to design simpler analogues of
saquinavir which have lower molecular weight, less
peptide character, and, consequently, better oral
bioavailability.
➢ Symmetrical inhibitors show greater selectivity for viral
protease over mammalian aspartyl proteases, as active
sites of the latter are not symmetrical.
➢ Symmetrical molecules might be less recognizable to
peptidase resulting in improved oral bioavailability.
➢ Development of saquinavir showed that a benzyl residue
was optimum binding group for S1 subsite.
Design of HIV Protease Inhibitors (PIs)
Design of HIV Protease Inhibitors (PIs)
Design of HIV Protease Inhibitors (PIs)
Design of HIV Protease Inhibitors (PIs)
Miscellaneous Antiviral Agents
a. Thiosemicarbazones

➢ Methisazone is an antiviral drug that works by


inhibiting mRNA and protein synthesis, especially in pox
viruses. It has been used in the past to treat smallpox.
➢ It is a thiosemicarbazone, which inhibits the synthesis of
structural viral proteins and interrupts full assembly of mature
virus. Methisazone is a member of indole class.
Miscellaneous Antiviral Agents
b. Adamantane amines
Miscellaneous Antiviral Agents
b. Adamantane amines

➢ The first adamantane derivative used as a drug


was amantadine – first (1967) as an antiviral drug against
various strains of flu and then to treat Parkinson's
disease.
➢ Other drugs among adamantane derivatives have been
patented as antiviral agents against HIV.
➢ Influenza virus strains have developed drug resistance to
amantadine and rimantadine, which are not effective
against prevalent strains as of 2016.
Miscellaneous Antiviral Agents
b. Adamantane amines (SAR)

✓ α –methyl derivative of adamantane produced Rimantadine.


✓ N-Alkyl and N,N- dialkyl derivatives of adamantadine exhibit antiviral
activity similar to that of adamantadine HCl.
✓ Except glycyl derivatives, N–acyl derivatives shows decreased
antiviral action and tromantadine possesses efficacy against clinical
Herpes labialis and H. gentalis.
✓ Replacement of the amino group with OH, SH, CN, or halogen
produced inactive compounds.
✓ Optical isomers and the racemic mixtures of rimantadine are equally
active.
Antiprotozoal agents
Antiamoebic agents & Antimalarials
Antiamoebic agents
Amoebiasis
➢ Amoebiasis affects about 10% of the world’s population,

causing invasive diseases in about 50 million people and

death in about 1,00,000 of these annually.

➢ This infection is, especially, common in lower socio-economic

groups and institutionalized individuals living under crowded

and poor hygienic conditions.

➢ Two morphologically identical, but genetically and

biochemically distinct, species of Entamoeba (E. histolitica

and E. dispar) are the causative organisms.


Amoebiasis
➢ Human beings are the only host for these organisms.

➢ Ingested amoebic cysts from contaminated food or water

survive and form acid gastric contents and transform them

into trophozoites that usually cause colitis which is either

acute or chronic (dysentery).

➢ In some cases, they target the brain and the liver producing

abscesses and systemic diseases.

➢ This parasitic disease is one of the major causes of illness

and death in many countries.


Amoebiasis
World Health Organization (WHO) has classified this disease as
follows:
1. Asymptomatic.
2. Symptomatic.
➢ Intestinal Amoebiasis
✓ Dysentery.
✓ Non-dysenteric colitis.
✓ Ameboma.
✓ Amoebic appendicitis.
➢ Extraintestinal amoebiasis
✓ Hepatic - acute nonsuppurative hepatitis.
✓ Liver abscesses.
3. Cutaneous involvement of other organs.
➢ Lung, brain, and spleen without the obvious liver
involvement are some examples under this category.
Antiamoebic Agents
➢ Antiamoebic agents are drugs used to treat amoebiasis.
➢ The potential drug should be active within the bowel lumen, in
the bowel wall, and particularly in the liver.
➢ The infection is transmitted exclusively by the fecal - oral
route; human beings are the only known hosts.

Classification of Amebicides
➢ Luminal amebicides: Diloxanide furoate. It is active only
against intestinal forms of amoeba.
➢ Systemic amebicides: Chloroquine. These agents have been
employed primarily to treat severe amoebic dysentery or
hepatic abscesses.
➢ Mixed amebicides: Metronidazole, tinidazole, and ornidazole.
These agents are active against both intestinal and systemic
forms of amoeba.
Antiamoebic Agents
Luminal amebicides

Diloxanide Furoate
Metabolism: The diloxanide
furoate is administered orally and
is hydrolyzed in the gut to give
diloxanide.

➢ Diloxanide furoate is dichloro acetamide derivative that is


nontoxic, mainly used in the treatment of chronic amoebiasis. It
is less effective in the treatment of acute intestinal amoebiasis.
Antiamoebic Agents
Mixed amebicides

Metronidazole

➢ It is used in the treatment of intestinal and hepatic


amoebiasis.
➢ The reactive intermediate formed in the parasital reduction of
the 5-nitro group of metronidazole covalently binds to the
DNA of the parasite and triggers the lethal effects.
➢ Potential reactive intermediates include the nitroxide, nitroso,
hydroxylamine, and amine.
Antiamoebic Agents
Mixed amebicides
Metronidazole
Metabolism: In the liver, metabolism of metronidazole leads to
two major metabolites, hydroxylation of the 2-methyl group to 2-
hydroxymethyl metronidazole and oxidation to metronidazole
acetic acid (MAA).
Antiamoebic Agents
Mixed amebicides

Tinidazole

➢ Tinidazole’s mechanism of action is similar to that of


metronidazole. It is used in the treatment of intestinal and
hepatic amoebiasis with a potential greater efficacy than
metronidazole.
➢ Tinidazole is metabolized by hydroxylation at the 2-methyl
group and catalyzed by CYP3A4 to form inactive compound.
➢ Used as an antiprotozoal and antibacterial agent.
Antiamoebic Agents
Mixed amebicides

Ornidazole

➢ It has a longer duration of action than metronidazole and used

as an antiprotozoal.
Antiamoebic Agents
Mixed amebicides

Nitazoxanide

➢ Mode of action: Nitazoxanide is a member of the 5-nitro

heterocycles and is a prodrug forming a short-lived redox

active intermediate. It appears to be more selective than

metronidazole.

➢ Uses: Used as an antiprotozoal agent.


Antiamoebic Agents
Mixed amebicides

Nimorazole

➢ It possesses antiamoebic activity, and hence, used

against intestinal and hepatic amoebiasis.


Antimalarials
Introduction
➢ Antimalarial agents are drugs used for the treatment or
prophylaxis of malaria.
➢ Malaria is caused by four species of Plasmodium, such as
Plasmodium falciparum, P. malariae, P. ovale, and P. vivax.
➢ Three of which produces the mild forms of malaria by
destroying red blood cells in peripheral capillaries and thus,
causing anemia.
➢ The bouts of fever correspond to the reproductive cycle of the
parasite. However, the most dangerous is the P. falciparum.
➢ In this case, the infected red blood cells become sticky and
form lumps in the capillaries of the deep organs of the body and
cause microcirculatory arrest.
Introduction

➢ This disease still affects about 200 millions people

and causes at least 2 million deaths per year.

➢ Two important phases of the parasite life cycle are

the following:

✓ Asexual cycle - occurs in the infected host.

✓ Sexual cycle - occurs in the mosquito.


Life Cycle of Plasmodium
Classification

I. Cinchona alkaloids.
II. 7-Chloro-4-Amino Quinolines.
III. 8-Amino Quinolines.
IV. Acridine derivatives (9-amino acridine derivatives).
V. Antifolates.
a) Biguanides.
b) Diamino pyrimidines.
VI. Sulphonamides and Sulphones.
VII.Miscellaneous drugs.
Cinchona alkaloids
Cinchona alkaloids
➢ Quinine is the chief alkaloid of cinchona bark (known as ‘Fever
Bark’).
➢ It has a colorful history of more than 350 years.
➢ Mechanism of action:
✓ Quinine acts as a blood schizonticide although it also has
gametocytocidal activity against P. vivax and P. malariae.
✓ Because it is a weak base, it is concentrated in the food
vacuoles of P. falciparum.
✓ It is said to act by inhibiting heme polymerase, thereby
allowing accumulation of its cytotoxic substrate, heme.
✓ As a schizonticidal drug, it is less effective and more toxic
than chloroquine.
✓ However, it has a special place in the management of severe
falciparum malaria in areas with known resistance to
chloroquine.
Cinchona alkaloids
➢ The drug is extensively metabolized in the liver and only 10% is

excreted unchanged in the urine. There is no cumulative toxicity

on continued administration.

➢ Quinine is a potentially toxic drug. The typical syndrome of

quinine side effects is called as cinchonism and it can be mild in

usual therapeutic dosage or could be severe in larger doses.

Mild cinchonism consists of ringing in the ears, headache,

nausea and disturbed vision.

➢ Quinidine: The anti-arrhythmic drug related to quinine can also

be used in the treatment of severe P. falciparum malaria.


7-Chloro-4-Amino Quinolines
7-Chloro-4-Amino Quinolines
4-Substituted Quinolines
Mode of Action: Three different mechanism of actions are
suggested for these drugs:
➢ DNA interaction: The mechanism of action for quinine is that the
drug gets intercalated into the DNA of the parasite.
➢ Ferriprotoporphyrin IX: The plasmodium parasite utilizes host
hemoglobin as a source of amino acid. On digestion of the
hemoglobin, the haem is released as ferriprotoporphyrin IX and
it produces hemolysis of the erythrocyte parasites. Therefore,
ferriprotoporphyrin that is released is converted into nontoxic
products and they, in turn, to haemozoites by the polymerase
enzyme. The steps involved in the conversion to haemozoites
are inhibited by the chloroquine.
7-Chloro-4-Amino Quinolines
4-Substituted Quinolines
Mode of Action (Cont.):

➢ Weak base hypothesis: The 4-substituted quinolines have weak

base and because of this pKa they are thought to accumulate in

a location, which is acidic (parasite lysosome pH 4.8–5.2). As

the extracellular fluid of the parasite is at pH 7.4, the weak base

will move towards a more acidic pH of lysosome. Once the acid–

base reaction occurs, elevating the pH in the lysosome, that in

turn reduces the parasite’s ability to digest hemoglobin, thus

reducing the availability of amino acids.


7-Chloro-4-Amino Quinolines
4-Substituted Quinolines

Metabolism of Quinine:

✓ It is metabolized in the liver to 2-hydroxy derivative

followed by additional hydroxylation on the quinoline

ring with the 2,3-dihydroxy derivative, as the major

metabolite.

✓ This metabolite has low activity and is rapidly

excreted in urine.
7-Chloro-4-Amino Quinolines
Chloroquine

➢ Chloroquine is the prototype anti malarial drug, most widely


used to treat all types of malarial infections.
➢ Mechanism of action:
✓ The mechanism of action of chloroquine is unclear.
✓ Being alkaline, the drug reaches high concentration within
the food vacuoles of the parasite and raises its pH.
✓ It is found to induce rapid clumping of the pigment.
7-Chloro-4-Amino Quinolines
Chloroquine
➢ Mechanism of action (Cont.):
✓ Chloroquine inhibits the parasitic enzyme heme polymerase
that converts the toxic heme into non-toxic hemozoin,
thereby resulting in the accumulation of toxic heme within
the parasite.
✓ It may also interfere with the biosynthesis of nucleic acids.
✓ Other mechanisms suggested include formation of drug-
heme complex, intercalation of the drug with the parasitic
DNA etc.
➢ Metabolism:
✓ The drug is metabolized by N-dealkylation through CYP2D6,
and CYP3A4 isoenzymes.
✓ It has been reported that the level of metabolism correlates
closely with degree of resistance.
7-Chloro-4-Amino Quinolines
Hydroxy chloroquine

➢ It is equivalent to the chloroquine, but it is less toxic


and used in the place of chloroquine against normally
sensitive strains. It is mainly used as an antimalarial.
It is also used for the treatment of rheumatoid
arthritis and lupus erythematosus.
7-Chloro-4-Amino Quinolines - SAR

➢ At C-4 position, the dialkylamino alkyl side chain has 2-5


carbon atoms between the nitrogen atoms, particularly the 4-
diethylaminomethyl butyl amino side chain that is optimal for
activity, as in chloroquine and quinacrine.
➢ The substitution of a hydroxyl group on one of the ethyl groups
on the tertiary amine (hydroxy quinoline), reduces toxicity.
➢ Incorporation of an aromatic ring in the side chain (e.g.
amodiaquine) gives a compound with reduced toxicity and
activity.
7-Chloro-4-Amino Quinolines - SAR

➢ The tertiary amine in the side chain is important.


➢ The introduction of an unsaturated bond in the side chain
was not detrimental to activity.
➢ The 7-chloro group in the quinoline nucleus is optimal, the
methyl group in position 3 reduces activity, and an
additional methyl group in position 8 abolishes activity.
➢ The D-isomer of chloroquine is less toxic than its L-
isomer.
8-Amino Quinolines
8-Amino Quinolines

Mode of action:
➢ While the mechanism of action of the 8-amino
quinolines is unknown, it is known that primaquine
can generate reactive oxygen species via an
autoxidation of the 8-amino quinoline group with the
formation of radical anion.
➢ As a result, cell destructive oxidants, such as
hydrogen peroxide, super oxide, and hydroxyl radical
can be formed.
8-Amino Quinolines
Primaquine

➢ In vitro and in vivo studies indicate that the stereochemistry at

the asymmetric carbon is not important for antimalarial activity.

➢ These appears to be less toxicity with the levorotatory isomer.

➢ It is extensively used for the radical cure of relapsing vivax

malaria, but it is not normally employed either for arresting the

severe attacks of the disease or for the suppressive therapy.


8-Amino Quinolines
Primaquine
➢ It invariably kills gametocytes of all the species, or inhibits their
growth and development in the mosquito.
➢ It fails to produce any significant effect on other erythrocytic
stages, and hence, it must not be employed alone for the
treatment of malaria.
➢ Metabolism: Primaquine is totally metabolized by CYP3A4 with
primary metabolites having carboxy primaquine.
➢ Trace amounts of N-acetyl primaquine, aromatic hydroxylated
products, and conjugation metabolites are seen.
Acridine derivatives
(9-amino acridine derivatives)
Acridine derivatives
(9-amino acridine derivatives)
Quinacrine

➢ Quinacrine acts at many sites within the cell, including


intercalation of DNA strands, succinic dehydrogenase,
mitochondrial electron transport, and cholinesterase.
➢ It may be tumorigenic and mutagenic and has been used as a
as a sclerosing agent.
➢ Quinacrine can cause yellow discoloration of the skin and
urine, because of its dye property.
➢ It acts as a schizontocidal and now it is not used as an
antimalarial agent. It is used in the treatment of leishmaniasis
and some tape worm infestations.
Antifolates
a. Biguanides

b. Diamino pyrimidines
Antifolates
a. Biguanides
Mode of action: Biguanides inhibit dihydrofolate reductase
enzyme and interfere in the folic acid metabolism. This leads to
inhibition of the nuclear division in malarial parasites.

➢ Metabolism: Proguanil is a prodrug, which is metabolized in the


liver to diamino triazine (cycloguanil) that acts as a
dihydrofolate reductase inhibitor of Plasmodium species and
inhibits DNA synthesis.
➢ It is used mainly for prophylactic treatment of malaria.
Antifolates
b. Diamino pyrimidines
Mode of Action: It inhibits the reduction of folic acid and
dihydrofolic acid to the active tetrahydrofolate coenzyme form.

Pyrimethamine

➢ It finds its extensive use as a suppressive prophylactic for the


prevention of severe attacks due to P. falciparum and P. vivax.
➢ It is also used in the treatment of toxoplasmosis and as an
immuno suppressive agent.
Antifolates
b. Diamino pyrimidines

Trimethoprim

➢ It is a potent inhibitor of dihydrofolate reductase.


➢ It has been employed in conjugation with sulphamethopyrazine
in the treatment of chloroquine-resistant malaria.
➢ It has also been used in conjugation with sulphonamides in the
treatment of bacterial infections.
➢ Trimethoprim is an antibacterial, effective against malarial
parasite.
Sulphonamides and Sulphones
Sulphonamides and Sulphones
Mode of action:
➢ They only act against the erythrocytic stages of malaria
parasite.
➢ The sulphadoxine interferes with the parasites ability to
synthesize folic acid.
➢ Sulphonamides block the incorporation of p - amino benzoic
acid (PABA) to form dihydropteroic acid. PABA is the central
part of the folate structure.
➢ Sulphonamides exhibit significant toxicity because humans do
not synthesize the vitamin folic acid.
Miscellaneous
Halofantrine

➢ Structurally, Halofantrine differs from all other antimalarial


drugs and belongs to the latest generation of antimalarials.
➢ It is a good example of a drug design that incorporates bio
isosteric principles evidenced by the tri-fluoro methyl moiety. It
is schizonticidal and has no effect on the sporozoite,
gametocyte, or hepatic stages.
➢ It is effective against multi drug resistant (including mefloquine
resistant) P. falciparum malaria.
Miscellaneous
Halofantrine

Metabolism: The drug is metabolized by N-dealkylation to


desbutyl halofantrine by CYP3A4. The metabolites appear to
be several folds more active than the parent drug.
Miscellaneous
Mefloquine

➢ Mefloquine has been found to produce swelling of the P.


falciparum food vacuoles.
➢ It may act by forming toxic complexes with free heme that
damage membranes and interact with other plasmodial
components.
➢ It is effective against the blood forms of falciparum malaria,
including the chloroquine resistant types.
Miscellaneous
Mefloquine
Metabolism of mefloquine: It is metabolized through CYP3A4
oxidation to its major inactive metabolite called carboxy
mefloquine and rest of the amount is excreted unchanged in urine.
Miscellaneous
Artesunate

➢ Artesunate is an ester derivative


of Artemisinin (lactone containing
an unusual peroxide bridge).

➢ The mechanisms of action of artesunate remains unclear and


debatable.
➢ Artesunate is a prodrug that is rapidly converted to its active
form dihydroartemisinin (DHA).
➢ This process involves hydrolysis of the 4-carbon ester group via
plasma esterase enzyme.
➢ It is hypothesized that the cleavage of endoperoxide bridge in
the pharmacophore of DHA generates reactive oxygen
species (ROS), which increases oxidative stress and causes
malarial protein damage via alkylation.
Anthelmintic agents
Introduction
➢ Anthelmintics are drugs used to treat parasitic infections due

to worms. Worms that are pathogenic to human beings,

namely, metazoa are conventionally classified into round

worms (nematodes) and two types of flatworms, that is, flukes

(trematodes) and tapeworms (cestodes).

➢ Anthelmintics act locally either to expel the worms from the

gastrointestinal tract or systemically to eradicate the species

and the developing forms of helminthic that invade the organs

and tissues.
Classification
I. Benzimidazoles.

II. Quinolines and isoquinolines.

III. Piperazine derivatives.

IV. Vinyl pyrimidines.

V. Amides.

VI. Natural products: Avermectins.

VII. Organo phosphorus: Metrifonate.

VIII. Imidazothiazoles: Levamisole.

IX. Nitro derivatives: Niridazole.


I. Benzimidazoles
➢ These are versatile anthelmintic agents particularly effective
against gastrointestinal nematodes.
➢ These are highly effective against ascaris, enterobius,
trichuris, and hookworm infections as single or mixed
infections.
I. Benzimidazoles
Mode of action:
➢ These drugs act by blocking the glucose transportation
in the parasites and lead to the depletion of glycogen
storage of the intracellular microtubules in the cells of
the worms, thereby arresting the nematodes and cell
division in the metaphase.
➢ The major site of action is microtubular protein β tubulin
of the parasite.
➢ These drugs are bound with β tubulin and inhibit the
polymerization.
I. Benzimidazoles

Metabolism:

➢ The parent compound is rapidly and almost completely

metabolized by oxidative and hydrolytic processes.

➢ The phase I oxidative reaction is commonly carried out

by the cytochrome P450 catalyzed reaction, which may

be followed by phase II conjugation.


I. Benzimidazoles

Metabolism:
I. Benzimidazoles

Metabolism:
Mebendazole is metabolized by the reduction of the 5-carbonyl group to
a secondary alcohol, which greatly increases the water solubility of this
compound and thereby potentiates the excretion through urine.
Secondary alcohol and amine are readily conjugated.
I. Benzimidazoles

Metabolism:
Thiabendazole is metabolized through aromatic hydroxylation at the fifth
position catalyzed by CYP1A2. The resulting phenol is conjugated to 5-
hydroxythiabendazole glucuronide.
I. Benzimidazoles

Properties and uses: Flubendazole is a white powder, practically

insoluble in water, in alcohol, and in methylene chloride. It is used

as an anthelmintic agent.
I. Benzimidazoles - SAR
➢ 5-substituents do not necessary increase potency, but when R
group in C5 prevent metabolic inactivation, resulting
compound has greater anthelmintic activity.
➢ 2-substituents may be methyl carbamate (--NHCOCH3),
aromatic or heterocyclic without loss of potency - but those
with aromatic or heterocyclic are more toxic than those with
carbamate e.g., thiabendazole is the most toxic one.
I. Benzimidazoles - SAR
➢ Mebendazole & albendazole are less toxic than
thiabendazole
✓ Do not have heterocyclic ring in 2 position.
✓ Less absorbed from git after oral administration
than in case of thiabendazole that is readily
absorbed from GIT.
II. Quinolines and isoquinolines

➢ Mode of action: Schistosoma mansoni is highly susceptible to


oxamniquine. Adenosine-5′-triphosphate (ATP)-dependent
enzymatic activation of the drug in susceptible schistosomes
forms unstable phosphate esters, which disassociate to yield
a chemically reactive carbocation this intermediate alkylates
the DNA.
➢ Metabolism: It is metabolized by oxidative reaction and it
gives inactive metabolites of acid and alcohol derivatives.
II. Quinolines and isoquinolines

➢ Metabolism: In serum, the major metabolites are 4-


hydroxycyclohexyl carboxylate, but in the urine, 50%–60% of the
initial PZQ exist as dihydroxylated products. Hydroxylation reactions
are catalyzed by CYP2B6 and CYP3A4.
➢ Uses: It is used for the treatment of schistosomiasis and liver fluke
infections. It causes ligamental damage to the worms, which
activates the host defense mechanisms and results in the
destruction of the worms.
III. Piperazine derivatives

➢ The drug is highly effective against both Ascaris lumbricoides


and Enterobius (oxyuris) vermicularis.
➢ These drugs cause the hyperpolarization of the ascaris
muscles by gamma-aminobutyric acid (GABA) agonistic
action, opening Cl– channels, which cause relaxation, depress
responsiveness to the contractile action of acetylcholine, and
produce the suppression of spontaneous spike potentials with
peristalsis.
III. Piperazine derivatives

➢ Mode of action: It selectively acts on the microflora, causes


attraction, and therefore, is readily phagocytosed by the tissue
fixed monocytes. It also has an effect on the muscular activity
of the microflora and causes hyper-polarization to destroy the
worms.
➢ Metabolism: The metabolism of diethyl carbamazepine leads to
the compounds of methyl piperazine and piperazine. Nearly, all
of the metabolites appear in the urine.
➢ It is the drug of choice for treating filariasis infections.
IV. Vinyl pyrimidines

➢ Mode of action: Pyrantel is a depolarizing neuromuscular


blocking agent. It induces marked persistent activation of the
nicotinic receptors, which result in spastic paralysis of the
worm. It is an alternative to mebendazole in the treatment of
ascariasis and enterobiasis.
➢ Properties and uses: It is also as investigational drug for the
treatment of hookworms, moniliformis, and trichostrongylus
infections.
V. Amides

➢ Mode of action: Niclosamide is also a potent molluscicide,


which is effective against Biomphaloria glabrata, the principle
action of the drug may be to inhibit anaerobic phosphorylation
of adenosine diphosphate (ADP) by the mitochondria of the
parasite, an energy producing process.
➢ Properties and uses: Niclosamide exists as yellowish fine
crystals, practically insoluble in water, sparingly soluble in
acetone, and slightly soluble in ethanol. It is used as an
anthelmintic.
VI. Natural products: Avermectins
VI. Natural products: Avermectins

Mode of action: Avermectins specifically open the chloride


channels in the invertebrate system distinct from the GABA-
gated and glutamate-gated chloride channels.
Metabolism: It is metabolized and it gives 3-O-demethyl-22,23-
dihydroavermectin B1α monosaccharide.
Properties and uses:
➢ Avermectins are macrocyclic lactones with broad
antinematicidal activity.
➢ The drug avermectin is now effectively being used to treat
and control onchocera volvulus, the filarial infection
responsible for River blindness.
➢ Ivermectin is a mixture of B1a and B1b (80:20) and is
prepared by catalytic reduction of ivermectin B1 (Abamectin).
VII. Organo phosphorus: Metrifonate

➢ Mode of action: Metrifonate is metabolized and


rearranged in vivo to dichlorvos, which inhibits acetyl
cholinesterase.
➢ Properties and uses: Metrifonate is a white crystalline
powder, soluble in water, in acetone, and in alcohol, and
very soluble in methylene chloride, used as an
anthelmintic.
VIII. Imidazothiazoles: Levamisole

➢ It stimulates the ganglion in the worms, causes tonic paralysis,


which results in the expulsion of live worms.
➢ These also interfere with the carbohydrate metabolism by
inhibiting fumarate reductase.
IX. Nitro derivatives: Niridazole

➢ Niridazole is used as an anthelmintic agent


Antineoplastic agents
Cancer - Introduction
➢ Cancer still remains one of the most feared diseases in
the modern world.
➢ Cancer cells are formed when normal cells lose the
normal regulatory mechanisms that control growth and
multiplication.
➢ They become ‘rogue cells’ and often lose the specialized
characteristics that distinguish one type of cell from
another (for example a liver cell from a blood cell). This is
called a loss of differentiation.
➢ The term neoplasm means new growth and is a more
accurate terminology for the disease.
Cancer - Introduction
➢ The word tumor actually means a local swelling. If the
cancer is localized it is said to be benign.
➢ If the cancer cells invade other parts of the body and set
up secondary tumors - a process known as metastasis -
the cancer is defined as malignant.
➢ It is malignant cancer that is life threatening.
➢ A major problem in treating cancer is the fact that it is not
a single disease. There are more than 200 different
cancers resulting from different cellular defects, and so a
treatment that is effective in controlling one type of
cancer may be ineffective on another.
Causes of Cancer
➢ Possibly as many as 30% of cancers are caused by
smoking, while another 30% are diet related.
➢ Carcinogenic chemicals in smoke, food, and the
environment may cause cancer by inducing gene
mutations or interfering with normal cell differentiation.
➢ The birth of a cancer (carcinogenesis) can be initiated by
a chemical—usually a mutagen—but other triggering
events, such as exposure to further mutagens, are usually
required before a cancer develops.
Causes of Cancer
➢ Viruses have been implicated in at least six human
cancers and are the cause of about 15% of the
world's cancer deaths. For example,
➢ The Epstein–Barr virus is the cause of Burkitt's
lymphoma and nasopharyngeal carcinoma.
➢ Human papillomaviruses are sexually transmitted
and can lead to cancer of the cervix.
➢ Hepatitis B may cause 80% of all liver cancers, and
➢ HIV can cause Kaposi's sarcoma and lymphoma.
Causes of Cancer
➢ The treatments used to combat cancer (radiotherapy
and chemotherapy) can actually induce a different
cancer in surviving patients. For example, 5% of
patients cured of Hodgkin's disease developed acute
leukemia.
➢ Some patients are prone to certain cancers for
genetic reasons. Damaged genes can be passed
from one generation to another, increasing the risk of
cancer in subsequent generations (e.g. certain
breast cancers).
Genetic faults leading to Cancer
Activation of proto-oncogenes:
➢ Proto-oncogenes are genes which normally code for proteins involved
in the control of cell division and differentiation.
➢ If they are mutated, this disrupts the normal function and the cell can
become cancerous. The protooncogene is then defined as an
oncogene. The ras gene is one example. Normally, it codes for a protein
called Ras which is involved in the signaling pathway leading to cell
division.
➢ In normal cells, this protein has a self-regulating ability and can ‘switch
itself off’. If the gene becomes mutated, an abnormal Ras protein is
produced which loses this ability and is continually active, leading to
continuous cell division. It has been shown that mutation of the ras
gene is present in 20–30% of human cancers.
➢ Oncogenes may also be introduced to the cell by viruses.
Genetic faults leading to Cancer
Inactivation of tumor suppression genes (anti-oncogenes):
➢ If DNA is damaged in a normal cell, there are cellular ‘policemen’ that
can detect the damage and block DNA replication.
➢ This gives the cell time to repair the damaged DNA before the next cell
division.
➢ If repair does not prove possible, the cell commits suicide (apoptosis).
Tumor suppression genes are genes which code for proteins that are
involved in these processes of checking, repair, and suicide.
➢ TP53 is an important example of such a gene and codes for the protein
of the same name (p53 protein). If the TP53 gene is damaged, the
repair mechanisms become less efficient, defects are carried forward
from one cell generation to another, and, as the damage increases, the
chances of the cell becoming cancerous increase.
Genetic faults leading to Cancer

Abnormal signaling pathways:

➢ Whether a normal cell grows and divides depends on the various

signals it receives from surrounding cells.

➢ The most important of these signals come from hormones called

growth factors.

➢ These are extracellular chemical messengers which activate protein

kinase receptors in the cell membrane.

➢ The receptors concerned trigger a signal transduction pathway which

eventually reaches the nucleus and instructs the transcription of the

proteins and enzymes required for cell growth and division.


Genetic faults leading to Cancer

Abnormal signaling pathways: (Cont.)


➢ Most, if not all, cancers suffer from some defect in this signaling

process such that the cell is constantly instructed to multiply.

➢ The signaling process is complex, so there are various points at which

it can go wrong.

➢ Many cancer cells are capable of growing and dividing in the absence

of external growth factors.

➢ For example, the Ras protein is a crucial feature in the signal

transduction pathways leading to cell growth and division. Abnormal

Ras protein is locked in the ‘on’ position and is constantly active

despite the lack of an initial signal from a growth factor.


Genetic faults leading to Cancer
Abnormalities in cell cycle regulation:
➢ A cycle of events takes place during cell growth and multiplication
which involves four phases known as G1, S, G2 and M.
➢ As part of this process, decisions have to be made by the cell whether
to move from one stage to another, depending on the balance of those
chemical signals promoting growth and those inhibiting it.
➢ The G1 phase (gap 1) is where a cell is actively growing in size and
preparing to copy its DNA in response to various growth factors or
internal signals.
➢ The next phase is the S phase (synthesis) where replication of DNA
takes place.
➢ Once the cell's chromosomes are copied, there is another interval
called the G2 phase (gap 2) during which the cell readies itself for cell
division. This gap, or interval, is crucial, as it gives the cell time to
check the copied DNA and to repair any damaged copies.
Genetic faults leading to Cancer
Abnormalities in cell cycle regulation: (Cont.)
➢ Finally, there is the M phase (mitosis) where cell division takes place to
produce two daughter cells, each containing a full set of chromosomes.
The daughter cells can then enter the cell cycle again (G1).
➢ Alternatively, they may move into a dormant or resting state (G0).
Genetic faults leading to Cancer
Abnormalities in cell cycle regulation: (Cont.)
➢ Within the cell cycle, there are various decision points which determine
whether the cell should continue to the next phase.
➢ For example, there is a decision point called the restriction point (R)
during the G1 phase which frequently becomes abnormal in tumor
cells.
➢ There are also various surveillance mechanisms known as checkpoints
which assess the integrity of the process.
➢ For example, a delay will take place during the G2 phase if DNA
damage is detected.
➢ This gives sufficient time for damaged DNA to be repaired or for the cell
to commit suicide (apoptosis).
➢ These checkpoints can also be defective in tumor cells.
Genetic faults leading to Cancer

Tissue invasion and metastasis:

➢ Not all cancers are life threatening.

➢ Benign tumors are growths which remain localized in a particular part

of the body and can grow to the size of a football without a fatal result.

➢ Malignant cancers, however, are life threatening because the cells

involved have the ability to break away from the primary tumor, invade a

blood vessel or a lymphatic vessel, travel through the circulation, and

set up tumors elsewhere in the body.

➢ In order to do this, these cells have to overcome a series of controls

that are designed to keep cells in their place.


Treatment of Cancer
➢ There are three traditional approaches to the treatment of

cancer—surgery, radiotherapy, and chemotherapy.

➢ As cancer cells are derived from normal cells, identifying

targets that are unique to cancer cells is not easy - as a

result, most traditional anticancer drugs act against targets

which are present in both types of cell.

➢ Therefore, effectiveness & selectivity of drugs is dependent

on becoming more concentrated in cancer cells than

normal.
Types of Anticancer Chemotherapy
➢ Alkylating Agents.
➢ Plant Alkaloids.
➢ Antitumor Antibiotics.
➢ Antimetabolites.
➢ Miscellaneous Antineoplastic.
Beyond the aforementioned types of chemotherapy,
many other types of chemo treatments exist, such as
targeted therapy, immunotherapy, and hormone
therapy.
Alkylating Agents
Alkylating Agents
Alkylating agents are most active in the resting phase of the
cell. These types of drugs are cell-cycle non-specific. There
are several types of alkylating agents used in chemotherapy
treatments:
➢ Nitrogen Mustards: Mechlorethamine, Cyclophosphamide,
Chlorambucil, Melphalan, and Ifosfamide.
➢ Alkyl sulfonates: Busulfan.
➢ Nitrosoureas: Carmustine, Lomustine and Streptozotocin.
Nitrosoureas are unique because, unlike most types of
chemo treatments, they can cross the blood-brain barrier.
They can be useful in treating brain tumors.
➢ Hydrazines: Dacarbazine, Procarbazine and Temozolomide.
➢ Metal salts: Carboplatin and Cisplatin.
Alkylating Agents
➢ Alkylating agents are highly electrophilic compounds that
react with nucleophiles to form strong covalent bonds.
➢ There are several nucleophilic groups present on the
nucleic acid bases of DNA which can react with
electrophiles.
➢ Drugs with two alkylating groups can react with a nucleic
acid base on each chain of DNA to cross-link the strands
such that replication or transcription is disrupted.
Alkylating Agents
➢ Alternatively, the drug could link two nucleophilic groups on
the same chain such that the drug is attached like a limpet to
the side of the DNA helix.
➢ That portion of DNA then becomes masked from the
enzymes required to catalyze DNA replication and
transcription.
➢ Unfortunately, alkylating agents can alkylate nucleophilic
groups on proteins, as well as DNA, which means they have
poor selectivity and have toxic side effects. They can even
lead to cancer in their own right.
➢ Nevertheless, alkylating drugs are still useful in the
treatment of cancer.
Alkylating Agents - Nitrogen Mustards
➢ The nitrogen mustard compound chlormethine (Mechlorethamine)
was the first alkylating agent to be used medicinally.
➢ The nitrogen atom is able to displace a chloride ion
intramolecularly to form the highly electrophilic aziridinium ion.
Alkylation of DNA can then take place.
➢ As the process can be repeated, cross-linking between chains or
within the one chain will occur, thus these drugs inhibit replication
and act as anticancer agents.
➢ Analogues of chlormethine have been designed to improve
selectivity and to reduce side effects.
➢ Other agents, such as cyclophosphamide, have been designed as
prodrugs and are converted into the alkylating drug once they have
been absorbed into the blood supply.
Alkylating Agents - Nitrogen Mustards
Alkylating Agents - Nitrogen Mustards
Alkylating Agents – Alkyl Sulfonates
➢ Busulfan was synthesized in 1950 as part of a systematic search

for novel alkylating agents.

➢ It is an anticancer agent which causes inter-strand cross-linking

between guanine units.

➢ The sulphonate groups are good leaving groups and play a

similar role to the chlorines in the nitrogen mustards.

➢ However, the mechanism involves a direct SN2 nucleophilic

substitution of the sulphonate groups and does not involve any

intermediates such as the aziridinium ion.


Alkylating Agents – Alkyl Sulfonates
Alkylating Agents – Nitrosoureas
➢ The anticancer agents lomustine and carmustine were
discovered in the 1960s, and are chloroethyl nitrosoureas
which decompose spontaneously in the body to form two
active compounds — an alkylating agent and a
carbamoylating agent.
➢ The organic isocyanate that is formed carbamoylates lysine
residues in proteins and may inactivate DNA repair enzymes.
➢ The alkylating agent reacts initially with a guanine moiety on
one strand of DNA, then with a guanine or cytosine unit on
the other strand to produce interstrand cross-linking.
➢ Streptozotocin is a naturally occurring nitrosourea isolated
from Streptomyces achromogenes.
Alkylating Agents – Nitrosoureas
Alkylating Agents – Hydrazines
➢ Dacarbazine and procarbazine are prodrugs which generate a
methyl diazonium ion as the alkylating agent.
➢ Dacarbazine is activated by N -demethylation in the liver—a
reaction catalyzed by cytochrome P450 enzymes.
➢ Formaldehyde is then lost to form a product which
spontaneously degrades to form 5-aminoimidazole-4-
carboxamide (AIC) and the methyl diazonium ion.
➢ Reaction of this ion with RNA or DNA results in methylation,
mainly at the 7-position of guanine. DNA fragmentation can also
occur.
➢ AIC has no cytotoxic effect and is present naturally as an
intermediate in purine synthesis.
Alkylating Agents – Hydrazines
➢ Temozolomide also acts as a prodrug and is hydrolyzed in the body to
form MTIC (5-(3-methyltriazen-1-yl)imidazole-4-carboxamide), which
decomposes in a similar fashion to form the methyl diazonium ion. The
O6 oxygen atom of guanine groups is particularly methylated by this
agent.
Alkylating Agents – Metal Salts
➢ Cisplatin is one of the most frequently used anticancer
drugs.
➢ The structure is activated within cells and produces
intrastrand cross-linking.
➢ Carboplatin is a derivative of cisplatin with reduced side
effects.
➢ A range of other platinum drugs such as oxaliplatin was
approved in 1999 and is effective against tumors that have
gained resistance to cisplatin and carboplatin.
➢ The diamino cyclohexane ring in oxaliplatin is bad for water
solubility, but this can be counteracted by introducing an
oxalato ligand as the leaving group.
Alkylating Agents – Metal Salts
Plant Alkaloids
Plant Alkaloids
➢ Plant alkaloids are chemotherapy treatments
derived made from certain types of plants.
➢ The vinca alkaloids are made from the periwinkle plant
(catharanthus rosea).
➢ The taxanes are made from the bark of the Pacific Yew
tree (taxus).
➢ The vinca alkaloids and taxanes are also known as
antimicrotubular agents.
➢ The podophyllotoxins are derived from the May apple
plant. Camptothecan analogs are derived from the Asian
"Happy Tree" (Camptotheca acuminata).
Plant Alkaloids
➢ Podophyllotoxins and camptothecan analogs are also
known as topoisomerase inhibitors, which are used in
certain types of chemotherapy.
➢ The plant alkaloids are cell-cycle specific.
➢ This means they attack the cells during various phases of
division.
✓ Vinca alkaloids: Vincristine, Vinblastine,
Tubulin
Vindesine and Vinorelbine.
Inhibitors
✓ Taxanes: Paclitaxel and Docetaxel.
✓ Camptothecan analogs: Camptothecin,
Topoisomerase
Irinotecan and Topotecan. Inhibitors
✓ Podophyllotoxins: Etoposide and Tenisopide.
Tubulin Protein
➢ Tubulin is a structural protein which is crucial to cell division.

➢ The protein acts as a building block for microtubules which

are polymerized and depolymerized during cell division.

➢ Drugs can block this process by either binding to tubulin to

prevent polymerization or binding to the microtubules to

prevent depolymerization.

➢ Agents which prevent polymerization do not prevent

depolymerization.
Topoisomerase Enzymes
➢ Topoisomerases are enzymes that participate in the
overwinding or underwinding of DNA.
➢ The winding problem of DNA arises due to the nature of its
double-helical structure.
➢ In order to prevent and correct these types of topological
problems caused by the double helix, topoisomerases bind to
DNA and cut the phosphate backbone of either one or both the
DNA strands.
➢ This intermediate break allows the DNA to be untangled or
unwound, and, at the end of these processes, the DNA
backbone is resealed again. Thus, topoisomerase enzymes are
required for managing DNA supercoils.
Plant Alkaloids - Vinca alkaloids
➢ Vincristine, Vinblastine, Vindesine and Vinorelbine are

alkaloids derived from the Madagascar periwinkle plant

(Catharanthus roseus , formerly known as Vinca rosea).

➢ The Vinca alkaloids bind to tubulin to prevent

polymerization and are useful anticancer agents.

➢ A range of other natural products have also been found to

prevent the polymerization of microtubules and are

currently being studied as potential anticancer agents.


Plant Alkaloids - Vinca alkaloids
Plant Alkaloids - Taxanes
➢ Paclitaxel (Taxol) and the semi-synthetic analogue docetaxel
are important anticancer agents that inhibit tubulin
depolymerization.
➢ Paclitaxel was isolated from the bark of yew trees (Taxus spp.).
➢ The semisynthetic route for the synthesis of paclitaxel
derivatives involves docetaxel as an intermediate.
➢ The term taxoids is used generally for paclitaxel and its
derivatives.
➢ Tubulin is actually made up of two separate proteins and the
taxoids are found to bind to the β-subunit of tubulin.
➢ The binding of paclitaxel accelerates polymerization and
stabilizes the resultant microtubules, which means that
depolymerization is inhibited. As a result, the cell division cycle
is halted.
Plant Alkaloids - Taxanes
➢ The benzoyl and acetyl substituents, at positions 2 and 4, respectively,
play an important role in this binding interaction, as do the side chain
and the oxetane ring.
➢ These groups dominate the ‘lower’ or ‘southern’ half of the molecule
(as the structure presented below), and so the variations that are
possible in this region are restricted when making analogues.
➢ In contrast, it is possible to carry out more variations in the ‘northern’
half of the molecule. This can affect the in vivo efficacy of the
molecule, allowing modification of aqueous solubility and
pharmacokinetic properties.
Plant Alkaloids - Camptothecan analogs
➢ Camptothecin is a naturally occurring cytotoxic alkaloid
which was extracted from a Chinese bush (Camptotheca
acuminata) in 1966.
➢ It targets the complex between DNA and topoisomerase I.
➢ This leads to DNA cleavage and cell death if DNA synthesis
is in progress, but it has been observed that these agents are
also toxic to cancer cells which are not synthesizing new
DNA.
➢ The camptothecins show selectivity for cancer cells over
normal cells, since Topoisomerase I levels will be high and
more active in cancer cells than normal cells.
Plant Alkaloids - Camptothecan analogs
➢ The lactone group is important for activity, but at blood pH it is in
equilibrium with the less active ring-opened carboxylate structure.
➢ Introducing substituents into the A and B rings can alter the relative
binding affinities of these structures to serum albumin such that the
level of the lactone present is altered favorably.
➢ Unfortunately, camptothecin itself shows poor aqueous solubility and
has unacceptable toxic side effects.
Plant Alkaloids - Camptothecan analogs
➢ Irinotecan and topotecan are clinically useful, semi-
synthetic analogues of camptothecin.
➢ They retain the important lactone group and were
designed to have aqueous solubility by adding
suitable polar functional groups such as alcohols and
amines.
➢ Irinotecan is a urethane prodrug that is converted to
the active phenol by carboxylesterases,
predominantly in the liver.
Plant Alkaloids - Podophyllotoxins
➢ The anticancer agents’ etoposide and teniposide belong to a
group of compounds called the podophyllotoxins, and are semi-
synthetic derivatives of epipodophyllotoxin - an isomer of a
naturally occurring agent called podophyllotoxin.
➢ Both agents act as topoisomerase poisons.
➢ Both are potent anticancer agents which stabilize the covalent
intermediate formed between DNA and topoisomerase II, and
are also thought to produce strand breakage.
➢ The drugs show selectivity for cancer cells, despite the fact that
topoisomerase II is present in both cancer cells and normal
cells. This is thought to be a result of elevated enzyme levels or
enzyme activity in the cancer cells.
Plant Alkaloids - Podophyllotoxins
➢ DNA strand breakage is also thought to occur by a free radical
process involving oxidation of the 4′-phenolic group and the
production of a semiquinone free radical. Evidence supporting
this comes from the fact that the 4′-methoxy structures are
inactive.
➢ The presence of the glucoside sugar moiety also increases the
ability to induce breaks.
Antitumor Antibiotics
Antitumor Antibiotics
➢ Antitumor antibiotics are chemo treatments made from
natural products produced by species of the soil fungus
Streptomyces.
➢ These drugs act during multiple phases of the cell cycle
and are considered cell-cycle specific.
➢ There are several types of antitumor antibiotics:

✓ Anthracyclines: Doxorubicin, Daunorubicin,


Epirubicin, Idarubicin and Mitoxantrone. Intercalating
✓ Chromomycins: Dactinomycin. Drugs on DNA

✓ Miscellaneous: Bleomycin.
Intercalating Drugs on DNA
➢ Intercalating drugs are compounds that contain planar or
heteroaromatic features which slip between the base-pair
layers of the DNA double helix.
➢ Some of these drugs prefer to approach the helix via the major
groove; others prefer access via the minor groove.
➢ Once they are inserted between the nucleic acid base pairs, the
aromatic/heteroaromatic rings are held there by van der Waals
interactions with the base pairs above and below.
➢ Several intercalating drugs also contain ionized groups which
can interact with the charged phosphate groups of the DNA
backbone, thus strengthening the interaction.
➢ Once the structures have become intercalated, a variety of
other processes may take place which prevent replication and
transcription, leading, finally, to cell death.
Antitumor Antibiotics - Anthracyclines
➢ Doxorubicin belongs to a group of naturally occurring antibiotics
called the anthracyclines, and was isolated from Streptomyces
peucetius.
➢ It is very similar in structure to daunorubicin - differing only in one
hydroxyl group.
➢ However, that has an important effect on activity and doxorubicin is
one of the most effective anticancer agents ever discovered.
➢ The drug intercalates into DNA and is an example of a topoisomerase II
poison as it stabilizes the complex formed between DNA and
topoisomerase II.
➢ A second mechanism by which doxorubicin can prove harmful to DNA
involves the hydroxyquinone moiety, which can chelate iron to form a
doxorubicin–DNA–iron complex. Reactive oxygen species are then
generated, leading to single-strand breaks in the DNA chain.
➢ A third proposed mechanism involves intercalated doxorubicin
inhibiting the helicases which unravel DNA into single DNA strands.
Antitumor Antibiotics - Anthracyclines
➢ A variety of other anthracyclines are used in cancer
chemotherapy, mainly daunorubicin, and the second-generation
anthracyclines epirubicin and idarubicin.
➢ Idarubicin lacks the methoxy group at R1, so it is more polar and
has an altered metabolism which prolongs its half-life.
Antitumor Antibiotics - Anthracyclines
➢ Mitoxantrone is a simplified, synthetic analogue of the
anthracyclines where the tetracyclic ring system has been
‘pruned’ back to the planar tricyclic system required for
intercalation.
➢ There are two identical substituent chains present which
make the molecule symmetrical and easier to synthesize.
➢ The sugar ring is lacking because it is thought to be
responsible for cardio toxic side effects.
➢ However, the amino substituent that is normally present
on the sugar is still present within the substituent chains.
Antitumor Antibiotics - Anthracyclines
➢ Structure–activity relationship (SAR) studies on
mitoxantrone identify a pharmacophore involving one of the
phenol groups, a carbonyl group, and the amino group in the
side chain.
➢ It was demonstrated that the amino group linking the side
chain to the tricyclic ring system was important to activity.
Antitumor Antibiotics - Chromomycins
➢ Dactinomycin is a naturally occurring antibiotic that was first isolated
from Streptomyces parvullis in 1953, and was shown to be an effective
anticancer agent in children.
➢ It contains two cyclic pentapeptides, but the important feature is a flat,
tricyclic, heteroaromatic structure which slides into the double helix via
the minor groove.
➢ The molecule is further held in position by
hydrogen bond interactions between the
nucleic acid bases of DNA and the cyclic
pentapeptides positioned on the outside of
the helix.
➢ The 2-amino group of guanine plays a
particularly important role in this
interaction. The resulting bound complex is
very stable and prevents the unwinding of
the double helix.
Antitumor Antibiotics - Miscellaneous
➢ Bleomycins are complex natural products that were isolated from
Streptomyces verticillus in 1962 and are some of the few anticancer
drugs not to cause bone marrow depression.
➢ Their structure includes a bithiazole ring system which intercalates
with DNA.
➢ Once the structure has become intercalated, the nitrogen atoms of the
primary amines, pyrimidine ring, and imidazole ring chelate a ferrous
ion which then interacts with oxygen and is oxidized to a ferric ion,
leading to the generation of superoxide or hydroxyl radicals.
➢ These highly reactive species abstract hydrogen atoms from DNA,
which results in the DNA strands being cut - particularly between
purine and pyrimidine nucleotides.
➢ Bleomycin also appears to prevent the enzyme DNA ligase from
repairing the damage caused.
Antitumor Antibiotics - Miscellaneous
Antimetabolites
Antimetabolites
➢ Antimetabolites are types of chemotherapy

treatments that are very similar to normal

substances within the cell.

➢ When the cells incorporate these substances into the

cellular metabolism, they are unable to divide.

➢ Antimetabolites are cell-cycle specific.

➢ They attack cells at very specific phases in the cycle.


Antimetabolites – Enzyme Inhibitors
➢ Antimetabolites are classified according to the
Enzymes/substances with which they interfere.
✓ Dihydrofolate Reductase Inhibitors: Methotrexate,
Pemetrexed and Pralatrexate.
✓ Inhibitors of Thymidylate Synthase: 5-Fluorouracil,
Capecitabine, and Raltitrexed.
✓ Inhibitors of Ribonucleotide Reductase: Hydroxycarbamide.
✓ Inhibitors of Adenosine Deaminase: Pentostatin.
✓ Inhibitors of DNA Polymerases: Cytarabine, Gemcitabine
and Fludarabine.
✓ Purine Antagonists: 6-Mercaptopurine and 6-Thioguanine.
Antimetabolites - Dihydrofolate Reductase Inhibitors
➢ Dihydrofolate reductase (DHFR) is an enzyme which is crucial in
maintaining levels of the enzyme cofactor tetrahydrofolate (FH4).
➢ Without this cofactor, the synthesis of the DNA building block (dTMP)
would grind to a halt, which, in turn, would slow down DNA synthesis
and cell division.
➢ The enzyme catalyzes the reduction of the vitamin folic acid to FH4 in
two steps via dihydrofolate (FH2).
➢ Once formed, FH4 picks up a single carbon unit to form N5, N10-
methylene FH4, which then acts as a source of one-carbon units for
various biosynthetic pathways, including the methylation of
deoxyuridine monophosphate (dUMP) to form deoxythymidine
monophosphate (dTMP).
➢ N5, N10- methylene FH4 is converted back to FH2 in the process and
dihydrofolate reductase is vital in restoring the N5, N10- methylene FH4
for further reaction.
Antimetabolites - Dihydrofolate Reductase Inhibitors
Antimetabolites - Dihydrofolate Reductase Inhibitors
➢ Methotrexate is one of the most widely used antimetabolites in
cancer chemotherapy.
➢ It is very similar in structure to the natural folates, differing only
in additional amino and methyl groups.
➢ It has a stronger binding affinity for the enzyme owing to an
additional hydrogen bond or ionic bond which is not present
when FH2 binds.
➢ As a result, methotrexate prevents the binding of FH2 and its
conversion to N5, N10- methylene FH4.
➢ Depletion of the cofactor has its greatest effect on the enzyme
thymidylate synthase, resulting in the lowered synthesis of
dTMP.
➢ Pemetrexed and Pralatrexate are related drugs that were
approved in 2004 and 2009 respectively.
Antimetabolites - Dihydrofolate Reductase Inhibitors
Antimetabolites - Inhibitors of Thymidylate Synthase
➢ Methotrexate has an indirect effect on thymidylate synthase by
lowering the amount of N5, N10- methylene FH4 cofactor required.
➢ 5-Fluorouracil is an anticancer drug which inhibits this enzyme
directly. It does so by acting as a prodrug for a suicide substrate.
➢ 5-Fluorouracil is converted in the body to the fluorinated
analogue of 2′-deoxyuridylic acid monophosphate (FdUMP),
which then combines with the enzyme and the cofactor.
➢ The tetrahydrofolate will form a covalent bond to the uracil
skeleton via the methylene unit which is usually transferred to
uracil.
➢ At this stage, a proton is usually lost from position 5 of uracil
(X=H).
Antimetabolites - Inhibitors of Thymidylate Synthase
➢ However, 5-fluorouracil has a fluorine atom at that position instead of
hydrogen (X=F) and the above reaction is impossible, because fluorine
can’t leave as a positive ion.
➢ As a result, the fluorouracil skeleton remains covalently and irreversibly
bound to the active site.
➢ The synthesis of thymidine is now terminated, which, in turn, stops the
synthesis of DNA. Consequently, replication and cell division are
blocked.
Antimetabolites - Inhibitors of Thymidylate Synthase
Antimetabolites - Inhibitors of Thymidylate Synthase
➢ Capecitabine is a prodrug for 5-fluorouracil.
➢ Inhibitors which bind to the cofactor binding region have also been
developed.
➢ Raltitrexed is the first of a new generation of highly specific folate-
based thymidylate synthase inhibitors which leads to DNA
fragmentation and cell death.
➢ Raltitrexed is transported into cells via a reduced folate carrier. Inside
the cell it is extensively polyglutamated, which enhances thymidylate
synthase inhibitory power and duration.
Antimetabolites - Ribonucleotide Reductase Inhibitors
➢ Ribonucleotide reductase is responsible for the conversion of
ribonucleotide diphosphates to deoxyribonucleotide
diphosphates. The enzyme contains an iron cofactor which is
crucial to the reaction mechanism.
➢ Hydroxycarbamide is a clinically useful agent which inhibits the
enzyme by destabilizing the iron center.
Antimetabolites - Adenosine Deaminase Inhibitors
➢ Ribonucleotide reductase is inhibited directly by hydroxycarbamide, but
it can also be inhibited indirectly by increasing the level of natural
allosteric inhibitors such as dATP (allosteric inhibitors).
➢ The enzyme adenosine deaminase catalyzes the deamination of
adenosine to inosine and it is found that inhibition of the enzyme leads
to a build-up of dATP in the cell, which, in turn, inhibits ribonucleotide
reductase.
Antimetabolites - Adenosine Deaminase Inhibitors
➢ The anti-leukemia drug pentostatin is a natural product isolated
from Streptomyces antibioticus, and is a powerful inhibitor of
adenosine deaminase (Ki = 2.5 pM).
➢ It acts as a transition-state inhibitor, mimicking the proposed
tetrahedral nature of the transition state, which is believed to be
similar to the tetrahedral intermediate.
Antimetabolites - Inhibitors of DNA Polymerases
➢ DNA polymerases catalyze the synthesis of DNA using the four
deoxyribonucleotide building blocks dATP, dGTP, dCTP, and
dTTP.
➢ The anticancer drug cytarabine is an analogue of 2′
deoxycytidine and acts as a prodrug.
➢ It is phosphorylated in cells to the corresponding triphosphate
(ara-CTP) which acts as a competitive inhibitor.
➢ In addition, ara-CTP can act as a substrate for DNA
polymerases and become incorporated into the growing DNA
chain.
➢ This can lead to chain termination or prevent replication of the
modified DNA. All of these effects result in the inhibition of
DNA synthesis and repair.
Antimetabolites - Inhibitors of DNA Polymerases
➢ Gemcitabine is an analogue of cytarabine with fewer side
effects.
➢ The purine analogue fludarabine is also metabolized to a
triphosphate and has the same mechanism of action as
cytarabine. It, too, inhibits transcription and can be incorporated
into RNA.
Antimetabolites - Purine Antagonists
➢ The thiopurines 6-mercaptopurine and 6-tioguanine are
prodrugs which are converted to their corresponding
nucleoside monophosphates by cellular enzymes.
➢ The monophosphates then inhibit purine synthesis at a
number of points.
➢ They are also incorporated into RNA and DNA, leading to
complex effects which end in cell death.
➢ Both agents are converted to a common product (thio-
GMP) which is subsequently converted to thio-GTP and
thio-dGTP, before incorporation into RNA and DNA
respectively.
Antimetabolites - Purine Antagonists
Miscellaneous Antineoplastic
Miscellaneous Antineoplastic
➢ The field of anticancer research is a vast one with a
wide diversity of novel structures being investigated.
➢ Many of the useful types of chemotherapy drugs are
unique.
➢ Examples:

✓ Adrenocortical steroid inhibitor: Mitotane.

✓ Enzymes: Asparaginase and Pegaspargase.

✓ Antimicrotubular agent: Estramustine.


Miscellaneous Antineoplastic
Adrenocortical steroid inhibitor
Mitotane:
➢ Mitotane is a steroidogenesis inhibitor and cytostatic antineoplastic
medication which is used in the treatment of adrenocortical
carcinoma and Cushing's syndrome.
➢ It is a derivative of p,p'-DDT and an isomer of p,p'-DDD, and is also
known as 1,1-(dichlorodiphenyl)-2,2-dichloroethane (o,p'-DDD).
➢ Mitotane has direct and selective cytotoxic effects on the adrenal
cortex, via an unknown mechanism, and thereby induces
permanent adrenal atrophy similarly to DDD.
Miscellaneous Antineoplastic
Enzymes
Asparaginase and Pegaspargase:
➢ The rationale behind asparaginase as anticancer agent is that it takes
advantage of the fact that acute lymphoblastic leukemia cells and
some other suspected tumor cells are unable to synthesize the non-
essential amino acid asparagine, whereas normal cells are able to
make their own asparagine; thus, leukemic cells require high amount
of asparagine.
➢ These leukemic cells depend on circulating asparagine.
➢ Asparaginase, however, catalyzes the conversion of L-asparagine to
aspartic acid and ammonia. This deprives the leukemic cell of
circulating asparagine, which leads to cell death.
➢ Pegaspargase, more effective than asparaginase, converts asparagine
to aspartic acid and ammonia.
Miscellaneous Antineoplastic
Antimicrotubular agent
Estramustine:
➢ Estramustine is a derivative of estradiol with a nitrogen mustard moiety.
➢ This gives it alkylating properties.
➢ The nitrogen mustard component is active and can alkylate DNA and
other cellular components such as tubulin components of rapidly
dividing cells.
➢ This causes DNA strand breaks or miscoding events. This leads to
apoptosis and cell death.
➢ Also, due to the drugs estrogen component, it can bind more selectively
to active estrogen receptors.

You might also like