Question 2
Question 2
The evolution of drug discovery for anti-neoplastic agents has come a long way and is
marked by significant breakthroughs that have revolutionized cancer treatment.
The discovery of chemotherapy in the 1940s marked the first significant step. The
first anti-neoplastic agent, Nitrogen Mustard, was discovered during World War II
when soldiers exposed to mustard gas were found to have low white blood cell
counts, suggesting a possible mechanism for cancer treatment. This led to the
development of alkylating agents, a class of drugs that work by attaching an alkyl
group to the DNA of cancer cells, disrupting their growth and reproduction.
In the 1960s, the development of Cytarabine, a pyrimidine analog, marked the
advancement of antimetabolites as anti-cancer drugs. These drugs work by mimicking
the cellular substances needed for DNA synthesis, thereby inhibiting the growth and
replication of cancer cells. Cytarabine has been vastly used for the treatment of acute
myeloid leukemia.
The late 20th century saw the rise of targeted therapies, an advancement that allowed
for more precise treatments with fewer side effects. One remarkable example is the
development of Imatinib (Gleevec) in the late 1990s. As a tyrosine-kinase inhibitor, it
targets specific proteins involved in the growth and development of cancer cells. This
drug marked a new era in cancer treatment, as it effectively transformed chronic
myeloid leukemia from a fatal disease into a manageable condition.
Technological advancements have also played a significant role in the evolution of
anti-neoplastic drug discovery. High throughput screening (HTS) technologies have
enabled rapid testing of thousands of compounds, speeding up the drug discovery
process. Additionally, the advent of computational models and bioinformatics has
facilitated the understanding of cancer at a molecular level, leading to the
development of more targeted therapies.
In recent years, the rise of immunotherapies, such as the checkpoint inhibitor
Pembrolizumab, has opened up a new frontier in cancer treatment. These drugs work
by harnessing the body's immune system to fight cancer, offering a new approach to
anti-neoplastic therapy.
Using the structure of mechlorethamine hydrochloride and guanine, illustrate the mechanism of
DNA alkylation by nitrogen mustards.
bii) Name an analogue of mechlorethamine, and explain how it improves on the prototypical
molecule? [3]
An analogue of mechlorethamine is **Cyclophosphamide**, which is a nitrogen mustard
alkylating agent commonly used in chemotherapy.
1. **Increased Selectivity**:
- Cyclophosphamide is a prodrug that requires metabolic activation in the liver to form its active
alkylating metabolite. This activation step imparts selectivity to the drug, as the conversion
mainly occurs in tumor tissues due to higher levels of specific liver enzymes.
- This selective activation reduces the systemic toxicity associated with mechlorethamine,
allowing for higher doses to be administered to cancer cells while sparing normal tissues.
2. **Enhanced Pharmacokinetics**:
- Cyclophosphamide exhibits improved pharmacokinetic properties compared to
mechlorethamine. It has better oral bioavailability, allowing for convenient administration in pill
form.
- The prodrug nature of cyclophosphamide also provides a longer half-life compared to
mechlorethamine, leading to sustained exposure to the active metabolite.
c. i) What is the function of ribonucleotide reductase, and which vital element is required to
initiate the catalytic cycle? [2]
To initiate the catalytic cycle, ribonucleotide reductase requires the presence of the vital element
**iron** (Fe). Iron plays a crucial role in the active site of ribonucleotide reductase, where it
facilitates the reduction of ribonucleotides to deoxyribonucleotides by donating electrons during
the enzymatic reaction.
ii) Using the chemical structure illustrate the mechanism by which hydroxyurea stops the catalytic
cycle. [2]
- The active form of ribonucleotide reductase includes an iron center (Fe) surrounded by a tyrosyl
radical that participates in the reduction reaction.
- Hydroxyurea (represented as HU) forms a covalent bond with the iron center, preventing the
tyrosyl radical from engaging in the reduction of ribonucleotides.
- This disruption halts the catalytic cycle of ribonucleotide reductase, inhibiting the production of
deoxyribonucleotides and ultimately interfering with DNA synthesis.
d. The structure of imatinib is provided below. Using the chemical structure as well as a narrative
to illustrate structure activity relationships of the molecule with tyrosine kinase enzyme. [10]
Imatinib is a potent tyrosine kinase inhibitor used in the treatment of cancers such as chronic
myeloid leukemia (CML) and gastrointestinal stromal tumors (GIST). Its structure consists of a
**bis-aryl urea** backbone with several key functional groups that contribute to its activity
against tyrosine kinases.
The structure-activity relationship (SAR) of imatinib with tyrosine kinase enzymes can be
elucidated as follows:
2. **3-Phenylaminopyridine Moiety**:
- The 3-phenylaminopyridine moiety in imatinib plays a crucial role in binding to the ATP-
binding pocket of tyrosine kinases. This interaction is vital for blocking the ATP binding site and
inhibiting kinase activity.
3. **Methylpiperazine Ring**:
- The presence of the methylpiperazine ring enhances the solubility and bioavailability of
imatinib. It also contributes to interactions with the enzyme through hydrogen bonding.
5. **4-Methylpiperazine Group**:
- The 4-methylpiperazine group contributes to the overall pharmacokinetic properties of
imatinib, influencing its distribution and metabolism in the body.
3. **Folate Antagonists**:
- These antimetabolites interfere with folate metabolism, crucial for nucleotide synthesis.
- *Examples*:
- Methotrexate: Inhibits dihydrofolate reductase, reducing available folate for DNA synthesis.
- Pemetrexed: Inhibits multiple folate-dependent enzymes, impeding DNA and RNA synthesis.
4. **Antifolate Antimetabolites**:
- These agents block folic acid metabolism, essential for nucleotide synthesis.
- *Examples*:
- Raltitrexed: Inhibits thymidylate synthase, hindering thymidine synthesis.
- Pralatrexate: Blocks dihydrofolate reductase, affecting purine and pyrimidine synthesis.
**IV. Pharmacokinetics**:
- **Absorption**: Varied oral bioavailability. Some require boosting with ritonavir.
- **Distribution**: Widely distributed in tissues.
- **Metabolism**: Extensively metabolized by the liver (CYP3A4).
- **Excretion**: Primarily renal excretion as metabolites.
In conclusion, HIV protease inhibitors play a significant role in the management of HIV/AIDS by
targeting a key enzyme essential for viral replication. Understanding their mechanism of action,
pharmacokinetics, adverse effects, resistance patterns, and clinical applications is crucial for
optimizing treatment outcomes in patients with HIV infection.
ii. Explain how changes in metabolic pathways (e.g., induction, inhibition) can impact the
therapeutic activity, toxicity, and/or drug interactions (9)
**I. Introduction**:
Metabolic pathways play a crucial role in determining the fate of drugs within the body. Changes
in these pathways, such as induction or inhibition, can significantly influence the therapeutic
efficacy, toxicity, and potential drug interactions of medications.
**V. Conclusion**:
Changes in metabolic pathways, whether through induction or inhibition, can have profound
effects on the therapeutic activity, toxicity, and drug interactions of medications. Clinicians must
consider these factors when prescribing drugs to ensure optimal treatment outcomes and
minimize the risk of adverse events associated with altered drug metabolism.
Question 3
a. Explain how genetic differences in drug metabolism result in predictable differences in drug
response (5)
b. Describe the differences in the ways that bacteria and viruses multiply (3)
a. Genetic differences in drug metabolism can lead to variations in drug response due to the role
that enzymes play in metabolizing drugs in the body. Enzymes are proteins encoded by genes,
and variations in these genes can affect the activity of the enzymes involved in drug metabolism.
For instance, some people may have genetic variants that result in lower levels of an enzyme
responsible for metabolizing a specific drug, leading to slower drug clearance from the body. This
can result in higher drug concentrations in the blood, potentially causing toxicity or adverse
effects. On the other hand, individuals with genetic variations that enhance the activity of drug-
metabolizing enzymes may metabolize drugs more rapidly, leading to lower drug levels and
reduced efficacy. Understanding these genetic differences can help predict how individuals will
respond to certain drugs, allowing for personalized medicine approaches that optimize treatment
outcomes and minimize adverse reactions.
b. Bacteria and viruses have distinct mechanisms of multiplication. Bacteria reproduce asexually
through binary fission, where a single bacterial cell divides into two identical daughter cells. This
process results in exponential growth of bacterial populations under favorable conditions. In
contrast, viruses require a host cell to replicate. They invade host cells and hijack the cell's
machinery to produce multiple copies of the virus. This process often leads to the destruction of
the host cell. The replication of viruses involves steps such as attachment to the host cell, entry,
replication of viral genetic material, assembly of new viruses, and release from the host cell.
Unlike bacteria, viruses cannot reproduce independently and rely on host cells to multiply.
c. The effect of some drugs used to treat cancer depends on geometrical isomerism. One
successful anti-cancer drug is cisplatin, whose formula is PtCl2 (NH3)2. Describe the structure of
cisplatin by referring to the following:
i. The meaning of the term geometrical isomerism as applied to cisplatin
ii. Diagrams to show the structure of cisplatin and its geometrical isomer
iii. The types of bonding in cisplatin. (6)
ii. **Diagrams Showing the Structure of Cisplatin and Its Geometrical Isomer**:
Cl NH3
\ /
Pt
/ \
NH3 Cl
- **Coordinate Covalent Bonds**: The bonding in cisplatin involves coordinate covalent bonds.
The ligands (chloride ions and ammonia molecules) donate electron pairs to the platinum atom,
forming these coordinate covalent bonds. The platinum atom acts as the Lewis acid, accepting
electron pairs from the Lewis bases (ligands) to form stable complexes.
In summary, cisplatin exhibits geometrical isomerism due to the arrangement of its ligands
around the central platinum atom. The two possible geometric isomers, cisplatin and transplatin,
have distinct spatial arrangements of the ligands. The bonding in cisplatin involves coordinate
covalent bonds, where the ligands provide electron pairs to the central platinum atom, forming
stable coordination complexes.
The catalytic mechanism of the Cytochrome P450 (CYP450) enzyme involves several steps that
are crucial for its function as a monooxygenase enzyme. Here is a detailed explanation of the
catalytic mechanism of the CYP450 enzyme:
1. **Substrate Binding**:
- The substrate, typically a hydrophobic molecule like a drug or xenobiotic, enters the active site
of the CYP450 enzyme.
- The active site of CYP450 contains a heme cofactor with an iron atom at its center. This iron
atom can form complexes with oxygen molecules.
2. **Reduction of CYP450**:
- NADPH-cytochrome P450 reductase transfers two electrons from NADPH to the CYP450
enzyme.
- This reduction step converts the iron atom in the heme group from Fe3+ to Fe2+, allowing it to
bind oxygen molecules more effectively.
3. **Oxygen Activation**:
- Molecular oxygen (O2) binds to the reduced form of the iron atom (Fe2+), forming a reactive
oxygen species known as an iron-oxo intermediate.
- This iron-oxo species is responsible for oxidizing the substrate in the subsequent steps.
4. **Substrate Oxidation**:
- The substrate interacts with the iron-oxo species, leading to the transfer of an oxygen atom
from the iron-oxo intermediate to the substrate.
- This results in the oxidation of the substrate and the formation of a metabolite.
5. **Product Release**:
- The oxidized product (metabolite) is released from the active site of the CYP450 enzyme.
- The enzyme returns to its original state, ready to catalyze another round of reactions.
6. **Regeneration**:
- NADPH-cytochrome P450 reductase regenerates the reduced form of the CYP450 enzyme by
transferring electrons from NADPH to the enzyme.
- This regeneration step completes the catalytic cycle of the CYP450 enzyme, allowing it to
continue metabolizing substrates.
Question 4
a. Describe how the following factors affect the action local anaesthetics
i. Lipid solubility [5]
ii. pH [4]
iii. Vasodilation [5]
**Factors Affecting the Action of Local Anesthetics**:
**ii. pH**:
- **Effect**: pH affects the ionization state of local anesthetics.
- **Explanation**: Local anesthetics exist in either ionized (charged) or non-ionized (uncharged)
forms. The ionized form (at high pH) is less able to penetrate the cell membrane, while the non-
ionized form (at low pH) is more lipid-soluble and can penetrate membranes efficiently.
- **Clinical Impact**: In areas with lower pH (e.g., inflamed tissues), local anesthetics exist
mostly in their non-ionized form, enhancing their membrane penetration and efficacy. Adjusting
the pH of the local anesthetic solution can optimize its action in different clinical scenarios.
**iii. Vasodilation**:
- **Effect**: Vasodilation influences the absorption and systemic distribution of local anesthetics.
- **Explanation**: Vasodilation at the injection site can increase the systemic absorption of local
anesthetics, leading to higher plasma levels and potentially systemic side effects. Conversely,
vasoconstriction can reduce systemic uptake and prolong the duration of action at the injection
site.
- **Clinical Impact**: Using vasoconstrictors like epinephrine with local anesthetics can reduce
systemic absorption, enhance local anesthesia duration, and decrease the risk of systemic
toxicity. However, caution is necessary in areas with compromised blood flow or where
vasoconstrictors are contraindicated.
Nitrogen mustards, such as Mechlorethamine Hydrochloride, are alkylating agents that form
covalent bonds with DNA, leading to DNA damage and disruption of cell division. Here is an
illustration of the mechanism of DNA alkylation by nitrogen mustards using Mechlorethamine
Hydrochloride and Guanine:
2. **Structure of Guanine**:
- Guanine is one of the four nucleobases found in DNA. It contains sites susceptible to
alkylation, particularly the N7 position on the imidazole ring.
3. **Mechanism**:
- **Step 1 - Activation**: Mechlorethamine Hydrochloride is activated in an enzymatic or non-
enzymatic process to form an electrophilic species capable of alkylating DNA.
- **Step 3 - DNA Damage**: Covalent bonding between Mechlorethamine and Guanine alters
the structure of the DNA molecule. This modification inhibits proper base pairing, disrupts DNA
replication and transcription, and triggers cellular responses leading to cell death.
- **Step 4 - Repair or Apoptosis**: Cells may attempt to repair the damaged DNA using repair
mechanisms. If repair is unsuccessful, the cell may undergo apoptosis, programmed cell death, to
prevent the propagation of damaged DNA.
d. Anthracyclines generally have the structure presented below. Which R group(s) differ for
doxorubicin and daunorubicin? How do the functional groups differ? [3]
Anthracyclines, like doxorubicin and daunorubicin, share a common basic structure. The major
structural differences between these two drugs lie in their R groups. Here are the specific R group
differences and the functional groups that are altered between doxorubicin and daunorubicin:
1. **Doxorubicin**:
- **R Group Differences**: In doxorubicin, the R group consists of a **sugar moiety**,
particularly a **daunosamine** sugar.
- **Functional Group Differences**: The sugar moiety in doxorubicin contributes to its solubility
and stability. It affects the pharmacokinetics and pharmacodynamics of the drug.
2. **Daunorubicin**:
- **R Group Differences**: In daunorubicin, the R group comprises a **different sugar
moiety** known as **rhodomycin**.
- **Functional Group Differences**: The presence of the rhodomycin sugar in daunorubicin
alters its biological activity and some pharmacological properties compared to doxorubicin.
ei) What is the mechanism by which antimetabolites such as gemcitabine inhibit ribonucleotide
reductase, and which structural modification is required [4]
Antimetabolites such as gemcitabine are chemotherapy drugs that interfere with nucleotide
metabolism in cancer cells. One of the key targets of gemcitabine is ribonucleotide reductase
(RNR), an enzyme responsible for converting ribonucleotides to deoxyribonucleotides, essential
for DNA synthesis. The mechanism by which gemcitabine inhibits RNR involves:
1. **Mechanism**:
- **Inhibition of Ribonucleotide Reduction**: Gemcitabine acts as a nucleoside analog that
undergoes intracellular phosphorylation to its active form, gemcitabine diphosphate.
- **Competitive Inhibition**: Gemcitabine diphosphate competes with the natural substrate,
deoxycytidine triphosphate (dCTP), for binding to the active site of ribonucleotide reductase.
Question 6
a. State the structure activity relationship (S.A.R) of the compound below. [8]
b. Classify antiviral agents. Explain the mechanism of action and synthesis of one drug from 2
different classes [9]
2. **Protease Inhibitors**: These drugs target viral proteases that are essential for the cleavage
of viral polyproteins into functional proteins, thereby inhibiting viral replication. Examples include
ritonavir (HIV) and boceprevir (Hepatitis C).
3. **Entry Inhibitors**: These agents block viral entry into host cells by targeting viral attachment
and fusion processes. Examples include enfuvirtide (HIV) and maraviroc (HIV).
4. **Neuraminidase Inhibitors**: These drugs target the viral enzyme neuraminidase, preventing
the release of new virions from infected cells and limiting viral spread. Oseltamivir (Tamiflu) and
zanamivir fall into this class, used for influenza.
**Mechanism of Action and Synthesis of Acyclovir (Nucleoside Analog) and Ritonavir (Protease
Inhibitor)**:
1. **Acyclovir**:
- **Mechanism of Action**: Acyclovir is a nucleoside analog that inhibits viral DNA synthesis by
acting as a chain terminator. Once phosphorylated by viral thymidine kinase, acyclovir
triphosphate competes with deoxyguanosine triphosphate for incorporation into viral DNA,
leading to chain termination and inhibition of viral replication.
- **Synthesis**: Acyclovir synthesis involves several steps. Initially, guanosine is converted to
acycloguanosine through a chemical modification process. Acycloguanosine is then further
phosphorylated to acyclovir monophosphate, which is subsequently converted to its active form,
acyclovir triphosphate, within the infected cells, enabling it to exert its antiviral effects.
2. **Ritonavir**:
- **Mechanism of Action**: Ritonavir is a protease inhibitor that binds to the active site of the
HIV protease enzyme, blocking its function. By inhibiting the cleavage of viral polyproteins into
functional proteins essential for viral maturation, ritonavir prevents the production of infectious
viral particles.
- **Synthesis**: The synthesis of ritonavir involves multiple chemical steps, including the
formation of key intermediates and the final assembly of the compound. The intricate process
culminates in the creation of ritonavir, which exhibits potent inhibitory activity against the HIV
protease enzyme.
Antilipidemic drugs are medications used to lower lipid levels in the blood, particularly
cholesterol and triglycerides. These drugs target various receptors and molecules involved in lipid
metabolism to reduce the risk of cardiovascular diseases. The types of drug-receptor interactions
of antilipidemic drugs include:
5. **PCSK9 Inhibitors**:
- **Type of Interaction**: Neutralization
- **Mechanism**: PCSK9 inhibitors (such as evolocumab) neutralize proprotein convertase
subtilisin/kexin type 9 (PCSK9), a protein that degrades the LDL receptor. By blocking PCSK9,
these drugs increase the number of LDL receptors on the liver cells, promoting LDL clearance
from the bloodstream.
Thyroxine (T4) is a critical thyroid hormone that regulates various metabolic processes in the
body. The metabolism and excretion pathways of T4 are crucial for maintaining thyroid hormone
levels within the physiological range. Here is a detailed and precise description of the metabolism
and excretion pathways of thyroxine (T4):
1. **Aromatic Ring**:
- Many local anesthetics, including Lidocaine, feature an aromatic ring in their structure. This
aromatic ring is often crucial for binding to specific sites on sodium channels in nerve cells,
blocking the influx of sodium ions and thereby inhibiting nerve conduction. Variations in the size
and substitutions on the aromatic ring can affect the potency and duration of action of the local
anesthetic.
2. **Amino Group**:
- Local anesthetics typically contain an amino group, which is essential for their interaction with
the sodium channels. The amino group may be tertiary, as in the case of Lidocaine, contributing
to the compound's potency by increasing membrane solubility and facilitating its penetration into
nerve cells.
4. **Hydrophobic Tail**:
- The presence of a hydrophobic tail in local anesthetics, often attached to the aromatic ring,
impacts their potency and lipophilicity. This tail region helps the compound penetrate nerve cell
membranes and reach the site of action. Modifying the length or structure of the hydrophobic
tail can influence the local anesthetic's efficacy and duration of action.
5. **Stereochemistry**:
- The spatial arrangement of atoms in a local anesthetic molecule can significantly affect its
activity. Stereoisomers of a local anesthetic may exhibit varying affinities for sodium channels,
leading to differences in potency, duration of action, and side effect profiles.
6. **Ionization State**:
- The pKa of a local anesthetic determines its ionization state and influences its ability to cross
biological membranes. Local anesthetics need to be in their non-ionized form to pass through cell
membranes and reach their target sites within nerve cells effectively.
Question 8
a) Draw an illustrated diagram of insulin and describe how it work. [15]
Insulin is a highly sophisticated peptide hormone that plays a crucial role in regulating
metabolism, especially glucose metabolism, in the body. The advanced and precise mechanisms
of how insulin works can be outlined as follows:
1. **Mechanism of Insulin Secretion**:
- Insulin is synthesized and stored in pancreatic beta cells as preproinsulin and proinsulin. In
response to elevated blood glucose levels post-meal, glucose enters beta cells via GLUT2
transporters.
- Increased intracellular glucose levels lead to the closure of ATP-sensitive potassium channels,
causing depolarization of the cell membrane.
- Depolarization triggers the opening of voltage-gated calcium channels, allowing calcium influx
into the cell.
- Elevated calcium levels stimulate insulin granule exocytosis, releasing insulin into the
bloodstream.
6. **Metabolic Homeostasis**:
- Overall, insulin plays a central role in maintaining metabolic homeostasis by regulating
glucose, protein, and lipid metabolism to ensure a balance of energy production, storage, and
utilization within the body.
Discuss three mechanisms for drugs that are used to treat Diabetes Mellitus (DM). In each case,
name the drug or class involved. [10]
Question 10
a. Differentiate phase I and phase II Metabolic reactions, giving examples [8]
Phase I and Phase II metabolic reactions are essential processes in the body's metabolism,
dealing primarily with the modification and transformation of drugs and xenobiotics. Here are
more detailed differences and examples of each phase:
Phase I Metabolic Reactions:
- Definition: Phase I metabolism involves reduction, oxidation, or hydrolysis reactions that
convert lipophilic compounds into more water-soluble molecules for easier excretion.
- Characteristics:
- Phase I reactions add or expose a polar functional group (e.g., -NH₂ or -OH) to the substrate,
making it more hydrophilic.
- These reactions can activate, deactivate, or render a compound toxic.
- Predominantly occur in the liver, with the hepatic cytochrome P450 system being the primary
oxidation system.
- Examples:
- Oxidation: Involves the addition of oxygen to the molecule.
- Reduction: Involves the gain of electrons or a decrease in oxidation state.
- Hydrolysis: The cleavage of chemical bonds by water.
Optimizing Potency:
* Understanding Quantal and Graded Dose-Response Curves: Potency refers to the amount of
drug needed to produce a given effect. It is often expressed as the quantal dose response ED50
or the graded dose response EC50. A highly potent drug evokes a larger response at lower
concentrations.
* Enhancing Receptor Affinity: Increasing the affinity of a drug for its target receptor can enhance
potency, as it allows the drug to produce a quantal effect in a larger proportion of the population
or achieve 50% of its maximal effect at lower doses.
* Chemical Optimization: Chemical modifications to the drug molecule can be explored to
improve its potency. This may involve altering the drug's structure to enhance its binding affinity
to the target receptor or to increase its bioavailability and tissue distribution.
Optimizing Efficacy:
* Maximizing Functional Response: Efficacy refers to the maximal effect that a drug produces
irrespective of concentration. Maximizing the drug's ability to produce a maximum functional
response is essential for optimizing efficacy.
* Receptor Activation: Enhancing the drug's ability to activate the target receptor and elicit a
maximal response while occupying a relatively low proportion of the receptors can optimize
efficacy.
* Therapeutic Effectiveness: While efficacy is important, it's also crucial to consider how well a
drug works in real-world use. Assessing a drug's effectiveness in clinical settings and patient
populations is essential for optimizing its therapeutic impact.
c. Discuss what causes attrition of drugs during preclinical and clinical trials [6]
The attrition of drugs during preclinical and clinical trials can be attributed to several factors,
impacting the success and progression of drug development. Here are the key causes identified
from the provided search results:
1. Safety Concerns and Cardiovascular Toxicity:
* Safety issues, including cardiovascular toxicity, represent a major cause of drug attrition
during preclinical and clinical development. Cardiovascular toxicity has been highlighted as a
significant concern, contributing to the withdrawal of medicines and impacting the duration of
drug therapies, particularly in the case of anticancer drugs .
2. Lack of Efficacy:
* Lack of efficacy in the intended indication has emerged as a major reason for late-stage drug
development failure. This is often established in randomized controlled clinical trials and can lead
to the discontinuation of drug candidates. The failure to demonstrate efficacy effectively indicates
that the target plays no role in the disease, leading to high rates of late-stage failure .
3. Preclinical Safety and Efficacy Testing:
* Preclinical and clinical safety concerns remain significant causes of drug attrition. Non-clinical
and clinical safety issues have been identified as major concerns during preclinical or clinical drug
development, influencing the progression of drug candidates into late clinical development and
marketing approval phases .
4. Inadequate Predictive Models:
* The need for more accurate prediction of the risk of drug-induced cardiotoxicity in preclinical
and early clinical stages has been emphasized. Inadequate predictive models for assessing the
risk of cardiovascular toxicity contribute to the progression of high-risk drug candidates into later
stages of development, leading to potential safety concerns and attrition .
5. Ethical and Regulatory Challenges:
* Ethical concerns related to animal testing in preclinical stages pose challenges, as some
individuals oppose such practices. Additionally, the design of clinical trials is based on the data
required by regulators, influencing the progression of drug development and the decision-making
process.
d. Prontosil, a sulfonamide derivative with the structure shown below, is inactive against
microorganisms in vitro, but shows activity in vivo. Explain why this is so and draw the structure
of the active compound derived from Prontosil [6]
H
|
H--N--C6H5
|
H--S--O
|
H
Prontosil, a sulfonamide derivative, is initially inactive against microorganisms in vitro because it
is a prodrug. When ingested, it undergoes metabolic processes in the body that convert it into
the active form. The primary reason for its inactivity in vitro is that Prontosil itself lacks the
necessary chemical properties to inhibit bacterial growth directly. However, when it is
metabolized, it generates the active compound responsible for antimicrobial activity.
Question 9
a)Discuss any three (3) conditions that promote drugs entry into the Central Nervous System
(CNS). [10]
b) Benzylpenicillin is a narrow spectrum antibiotic. Discuss, illustrating and naming any three
changes to transform its activity to a broad spectrum antibiotic class. [15]