Lesson 3 Flashcards

1
Q

What is the primary way that drugs are eliminated?
Which mechanism allows for reabsorption of drugs in the intestine ?
What kind of molecules must can be secreted into the urine?(ionized or non ionized? Choose one)
What is the effect of lipophilic drugs on renal excretion and reabsorption?

A

Elimination of drugs occurs primarily through renal mechanism
Secretion into bile also possible, but allows for re-absorption in the intestine

Secretion into the urine requires ionized or hydrophilic molecules, but:
1.Most drugs are not small molecules that are highly ionized at body pH
2.Most drugs are poorly ionized and lipophilic
=> This decreases renal excretion and facilitates renal tubular reabsorption
3.Many drugs are highly protein bound, and therefore not efficiently filtered in the kidney
4.Most drugs would have a long duration of action if termination of their effects depended only on renal excretion

Inactivation versus elimination of the active drug

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

What is the primary way that drugs are eliminated?
Which mechanism allows for reabsorption of drugs in the intestine ?
What kind of molecules must can be secreted into the urine?(ionized or non ionized? Choose one)
What is the effect of lipophilic drugs on renal excretion and reabsorption?

A

Elimination of drugs occurs primarily through renal mechanism
Secretion into bile also possible, but allows for re-absorption in the intestine

Secretion into the urine requires ionized or hydrophilic molecules, but the problem with most drugs is that :
1.Most drugs are not small molecules that are highly ionized at body pH
2.Most drugs are poorly ionized and lipophilic
=> This decreases renal excretion (decreases excretion because drug is hydrophobic or lipophilic but drug has to be hydrophilic before it can be excreted cuz urine is made up primarily of water molecules) and facilitates renal tubular reabsorption
3.Many drugs are highly protein bound, and therefore not efficiently filtered in the kidney
4.Most drugs would have a long duration of action if termination of their effects depended only on renal excretion
Inactivation versus elimination of the active drug
The solution to all the above problems is drug metabolism

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Why are hydrophilic molecules ionized?

A

Charge and Water Interaction**:
- Ionized = Charged: Ionized drugs have gained or lost an electron, resulting in a charge.
- Hydrophilic = Water-loving: Hydrophilic substances are attracted to water molecules due to their polarity and ability to form hydrogen bonds.

Mnemonic: “Ions love water”. This simple phrase reminds you that ionized drugs, being charged, have an affinity for water.

  1. Contrasting Properties:
    • Hydrophobic = Water-repelling: Hydrophobic substances are repelled by water and tend to be non-polar.
    • Ionized drugs are not hydrophobic: Since they interact favorably with water, ionized drugs are hydrophilic.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Why is drug metabolism important?
Why are most metabolic products less pharmacologically active?

Which drugs are exceptions to this statement that most metabolic products less pharmacologically active?

A

Why is drug metabolism so important? Elimination of drugs and chemicals by the kidney is often compromised because the drug/chemical is too nonpolar, lipophilic and readily “reabsorbed” from tubular fluid. Metabolism can convert the drug to a more hydrophilic compound reducing reabsorption.

Most metabolic products ar else pharmacologically active because through metabolism, the products become more hydrophilic for renal excretion but for a drug to be very pharmacologically active, it must be hydrophobic or lipophilic
Most metabolic products are less pharmacologically active

Important exceptions:
Where the metabolite is more active - 3 examples
1.(Prodrugs, e.g. Erythromycin-succinate (less irritation of GI) –> Erythromycin, enaliprilat -> enalapril, codeine) : Erythromycin succinate is a pro drug and these pro drugs are only active when metabolized that’s why they are exceptions.
2.Where the metabolite is toxic (acetaminophen): cuz we’re saying metabolism makes the metabolite less pharmacologically active than the parent drug but in cases where metabolites are toxic, the metabolites are more pharmacologically active than the parent drugs. Just not in a good way
3.Where the metabolite is carcinogenic: cuz we’re saying metabolism makes the metabolite less pharmacologically active than the parent drug but in cases where metabolites are carcinogenic, the metabolites are more pharmacologically active than the parent drugs. Just not in a good way

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What is the relationship between the bio transformation of drugs and normal biochemical processes occurring in the body

A

Close relationship between the biotransformation of drugs and normal biochemical processes occurring in the body:
-Metabolism of drugs involves many pathways associated with the synthesis of endogenous substrates such as steroid hormones, cholesterol and bile acids
-Many of the enzymes involved in drug metabolism are principally designed for the metabolism of endogenous compounds
-These enzymes metabolize drugs only because the drugs resemble the natural compound

Certainly! Let’s break it down:

  1. Enzymes Shared Between Drugs and Body: The same enzymes in your body that help break down drugs also help make and regulate natural substances like hormones (like estrogen or testosterone), cholesterol (which is important for cell function), and bile acids (which help digest fats).
  2. Competition for Enzymes: When you take a drug, it can compete with these natural substances for the enzymes that break them down. This competition can affect how well the drug works and how your body handles these natural substances.
  3. Effects on Natural Processes: Some drugs can change how these natural substances are made or broken down in your body. For example, medications like statins can lower cholesterol levels by affecting how your body produces cholesterol.
  4. Why It Matters: Understanding how drugs interact with these natural processes helps doctors predict how drugs might affect you. It also helps them manage any side effects or interactions between different drugs you might be taking.

In essence, drug metabolism isn’t just about how drugs break down in your body—it’s also about how they interact with and affect the natural processes that keep your body running smoothly.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

State three example of drugs with more active metabolites

A

Erythromycin succinate – gram+ antibiotic; pH sensitive (so it needs enteric coating), nonpolar, esterified (succinic acid, proprionic acid); converted by cell esterases

Enaliprilat - ACE-Inhibitor; prodrug; esterase converts to Enalapril (active)

Codeine – O-demethylation (or oxygen demethylation) to morphine – more active analgesic than codeine; CYP2D6 metabolic enzyme; deficient in 10% caucasians, 2% in asians; reduced analgesia for same dosage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Difference between ionotropic and chronotropic drugs
In the body, what drug is metabolized to acetaminophen?
What enzymes are involved in this process?
What process does acetaminophen go through to be metabolized ?
What toxic metabolite is produced from this reaction?
How do these metabolites lead to liver damage?

A

Focus**: Ionotropic drugs focus on changing the force or strength of heart contractions, affecting myocardial contractility.
- Focus: Chronotropic drugs focus on altering the rate of heart contractions, affecting heart rate.

heart cells.
- Examples:
- Positive Ionotropic Agents: Digoxin (increases contraction force by affecting calcium levels), Dopamine (increases force at moderate doses).
- Negative Ionotropic Agents: Beta-blockers like Metoprolol (decrease contraction force by blocking beta-adrenergic receptors), Verapamil (reduces force by blocking calcium channels).

  • Definition: Chronotropic drugs influence the rate or speed of heart contractions by affecting electrical signals that regulate heart rhythm.
  • Examples:
    • Positive Chronotropic Agents: Epinephrine (increases heart rate and contraction strength via beta-adrenergic stimulation), Atropine (increases heart rate by blocking muscarinic receptors).
    • Negative Chronotropic Agents: Beta-blockers like Atenolol (decrease heart rate by blocking beta-adrenergic receptors), Diltiazem (slows heart rate by blocking calcium channels).
  • Ionotropic Drugs: Used to optimize cardiac output in conditions like heart failure or shock, where enhancing or reducing contraction strength is beneficial.
  • Chronotropic Drugs: Employed to manage heart rate abnormalities such as bradycardia (slow heart rate) or tachycardia (fast heart rate), adjusting heart rhythm as needed

Phenacetin: 1887, analgesic, antipyretic, negative inotropic.
Present in APC headache mix: (aspirin+phenacetin+caffeine)
Use today: “cutting” cocaine due to its analgesic properties, adulterant; chronic use leads to renal papillary necrosis due to toxic metabolites

Ethyl ester  de-ethylation to acetaminophen (CYP2A13, CYP1A2)
Acetaminophen more potent than phenacetin
Phenacetin & acetaminophen conjugated with glucuronic acid or sulfate for elimination
Phenacetic metabolized by monooxygenase hydroxylation to toxic metabolites –
NAPQI = N-acetyl-(1,4) benzoquinone imine and epoxides
Acetaminophen also conjugated and hydroxylated (CYP2E1, CYP2A6, CYP1A2)
Hydroxylation leads to toxic metabolite imine and epoxide
Detoxification: conjugation with Hepatic cell glutathione (GSH)
If Overdose – deplete hepatic GSH; metabolites  mitochondrial dysfunction, oxidative damage to proteins, liver cell necrosis and hepatic failure
Therapy: N-acetyl cysteine or methionine

Phenacetin, historically used as an analgesic and antipyretic, is notable for its negative inotropic effects on the heart. Originally a component of the APC headache mixture (Aspirin, Phenacetin, Caffeine), it’s now primarily associated with illicit practices such as “cutting” cocaine due to its analgesic properties. Chronic use of phenacetin can lead to renal papillary necrosis due to toxic metabolites.

In the body, phenacetin is metabolized into acetaminophen (paracetamol) through de-ethylation processes primarily involving CYP2A13 and CYP1A2 enzymes. Acetaminophen is more potent than phenacetin and is further metabolized through hydroxylation by enzymes like CYP2E1, CYP2A6, and CYP1A2. This metabolic pathway can produce toxic metabolites such as N-acetyl-(1,4) benzoquinone imine (NAPQI) and epoxides.

Toxic metabolites like NAPQI and epoxides can lead to liver damage by depleting hepatic cell glutathione (GSH), causing mitochondrial dysfunction, oxidative stress, and liver cell necrosis. Treatment for acetaminophen overdose typically involves administering N-acetylcysteine or methionine to replenish GSH levels and mitigate liver damage.

Understanding the metabolic pathways and toxicity of phenacetin and acetaminophen underscores the importance of safe usage and monitoring to prevent severe adverse effects like hepatic failure.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What are the two main phases of drug metabolism

Which of the two phases combines with endogenous substrates to make drugs more soluble ?

A

Phase I Reactions:
-Convert parent compound into a more polar (=hydrophilic) metabolite by adding or unmasking(exposing. Any reaction that removes a functional group or breaks bonds in a drug molecule, exposes rhat drug molecule and makes it more polar) functional groups (-OH, -SH, -NH2, -COOH, etc.) on a drug molecule
-Often these metabolites are rendered inactive by the conversion
-The reaction product may be sufficiently polar to be excreted readily

Phase II Reactions:
-Conjugation with endogenous substrate to further increase aqueous solubility.
-Conjugation with glucoronide, sulfate, acetate, amino acid
Phase I usually precedes phase II reactions

some drugs only need to undergo phase1 reaction and that’s enough for them. Others require both reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What organ is the principal site for drug metabolism?
What is the first pass effect

A

Liver is principal site of drug metabolism:
Other sites include the gut(midozalam,cyclosporine,L dopa), lungs(metabolizes inhaled drugs example is propanolol and albuterol and inhaled corticosteroids ) , skin(metabolizes topical drugs, Topical corticosteroids, nitroglycerin (transdermal), local anesthetics.) and kidneys(vitamin D and penicillin or beta lactam antibiotics or cephalothin are metabolized by the kidneys)
For orally administered compounds, there is the
“First Pass Effect”
Intestinal metabolism
Liver metabolism
Enterohepatic recycling
Gut microorganisms - glucuronidases

The first-pass effect, also known as first-pass metabolism, refers to the phenomenon where a drug undergoes metabolism in the liver or intestines before reaching systemic circulation. Here’s a concise explanation:

  1. Definition: The first-pass effect describes the metabolism of a drug that occurs when it is absorbed from the gastrointestinal tract (via oral administration) or enters the liver (via portal circulation) before reaching the systemic circulation.so the liver metabolizes the drugs before they get into systemic circulation. This makes the amount of the drug available, smaller than it was when it was taken in. So it passed through mouth and through intestines to the liver then to systemic circulation.
  2. Mechanism: When a drug is absorbed orally, it travels through the portal vein to the liver first. In the liver, enzymes metabolize some of the drug before it can enter the general bloodstream and reach other organs or tissues.
  3. Significance: The extent of first-pass metabolism can significantly affect the bioavailability of a drug, which is the fraction of the administered dose that reaches systemic circulation unchanged and is available for therapeutic action.
  4. Examples: Drugs that undergo extensive first-pass metabolism include propranolol (a beta-blocker), lidocaine (a local anesthetic), and nitroglycerin (used for angina). For example, nitroglycerin undergoes rapid metabolism in the liver, which limits its oral bioavailability, so it is often administered sublingually or transdermally.
  5. Clinical Considerations: Understanding the first-pass effect is crucial for dosing and route of administration decisions in pharmacotherapy. Drugs that undergo extensive first-pass metabolism may require higher oral doses or alternative routes (such as sublingual, transdermal, or intravenous) to achieve therapeutic concentrations.

In summary, the first-pass effect highlights how drugs are metabolized in the liver or intestines before entering systemic circulation, impacting their bioavailability and therapeutic effectiveness.

Liver metabolism
Enterohepatic recycling
Gut microorganisms - glucuronidases

So from mouth to stomach to intestines where it is primarily absorbed (more absorbed in the duodenum and jejunum of the small intestine cuz of the plenty villi, increased blood supply) then it moves through the enterocytes(intestinal epithelial cells) into portal circulation and passes through the portal vein and into the liver for metabolism before it gets into the systemic circulation and loves to target organs.

Phase I and II - Summary:
Products are generally more water soluble
These reactions products are ready for (renal) excretion
There are many complementary, sequential and competing pathways
Phase I and Phase II metabolism are a coupled interactive system interfacing with endogenous metabolic pathways(endogenous because the body already has its own metabolic pathways and the drugs just take advantage of these pathways to act)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is the end result of hydrophilic drugs in the body?
What is the end result of lipophilic drugs in the body?

A

Hydrophilic drugs (drugs that like water) absorbed in GI tract, pass through liver, get excreted by kidney.

B. Lipophilic (hydrophobic) drug without metabolism, absorbed into blood, may get excreted into tubular fluid in kidney but reabsorbed – no elimination.

Lipophilic drugs are absorbed through the gastrointestinal tract and enter the bloodstream.
• Due to their affinity for fats, these drugs tend to distribute well into fatty tissues and other lipid-rich areas of the body.

C. Lipophilic drug is slowly metabolized by liver enzymes. The liver tries to
Metabolize it. In this case, the hydrophilic metabolite formed from the liver enzymes trying to do some metabolism on the lipophilic drug is the only one eliminated, then the rest of the unchanged drug recirculates.

D. Lipophilic drug rapidly metabolized by liver enzymes, nearly complete elimination by kidney.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

State the three main phase 1 metabolism reactions
Which of the reactions often converts prodrugs to their active forms or deactivating drugs

  1. Which enzyme is most commonly involved in the hydroxylation of drugs during Phase I metabolism?
    • A) Esterase
    • B) Alcohol dehydrogenase
    • C) Cytochrome P450
    • D) UDP-glucuronosyltransferase
      **
  2. In the context of Phase I metabolism, what is the primary outcome of N-oxidation reactions?
    • A) Addition of a hydroxyl group to a drug molecule
    • B) Conversion of a drug molecule into a more polar form by adding oxygen to nitrogen
    • C) Hydrolysis of ester or amide bonds
    • D) Reduction of nitro groups to amino groups

**

  1. Which of the following drugs is primarily metabolized through Phase I hydrolysis reactions rather than oxidation or reduction?
    • A) Acetaminophen
    • B) Diazepam
    • C) Propranolol
    • D) Aspirin

*

  1. Which Phase I metabolic reaction is responsible for the conversion of codeine to morphine?
    • A) Hydrolysis
    • B) Oxidation
    • C) Reduction
    • D) Dealkylation
  2. Which of the following statements about dealkylation reactions in Phase I metabolism is TRUE?
    • A) Dealkylation reactions typically result in the addition of an oxygen atom to the drug molecule.
    • B) Dealkylation involves the removal of alkyl groups from the drug molecule, which can result in the formation of a more active or toxic metabolite.
    • C) Dealkylation is a form of hydrolysis that breaks ester bonds in the drug molecule.
    • D) Dealkylation reactions are primarily mediated by UDP-glucuronosyltransferases.
A

Phase I Reactions:
Oxidation(OILRIG. For charged species,Oxidation is loss and reduction is gain)
Reduction
Hydrolytic cleavage(breaking bonds using water molecules. Breaks chemical bonds through the addition of water, often converting prodrugs to their active forms or deactivating drugs)

The above are the three main phase 1 reactions

Phase one reactions include oxidation, reduction, and hydrolysis, which introduce or unmask a functional group on the drug. The other options— acetylation, methylation, and sulphate conjuration-are phase two reactions,
Alkylation (Methylation)(adding an alkyl or methyl group)
Dealkylation(removing an alkyl group)
Ring cyclization
N-carboxylation
Dimerization
Transamidation
Isomerization
Decarboxylation

More interested in top 5

Here’s a clearer breakdown:

Phase I reactions generally involve modifications to the drug molecule itself to introduce or expose functional groups. These include:
1. Oxidation
2. Reduction
3. Hydrolysis

Main ones are the above.
8. Hydroxylation

Phase II reactions typically involve conjugation reactions where an endogenous substrate is added to the drug molecule to increase its solubility. These include:
1. Glucuronidation
2. Sulfation (Sulphate Conjugation)
3. Acetylation
4. Methylation
5. Amino Acid Conjugation
6. Glutathione Conjugation

  • Methylation and Acetylation: These are phase II reactions as they involve the addition of a methyl group (methylation) or an acetyl group (acetylation) to the drug or its metabolites, increasing solubility for excretion.
  • Reduction and Sulphate Conjugation: Reduction is a phase I reaction, while sulphate conjugation is a phase II reaction.

If you have specific sources that describe alkylation (including methylation) as phase I, they might be discussing specific biochemical contexts or exceptions. In general pharmacology, methylation and acetylation are recognized as phase II reactions.

Alcohol dehydrogenase (ADH) is an enzyme involved in Phase I reactions of drug metabolism. It primarily catalyzes the oxidation of alcohols into aldehydes. For example, ADH converts ethanol (an alcohol) into acetaldehyde (an aldehyde). This reaction is an oxidation process, which is one of the main types of Phase I metabolic reactions

Certainly! Here’s a summary:

  • Hydroxylation:
    • What: Addition of a hydroxyl group (-OH) to a molecule.
    • Enzyme: Cytochrome P450 (CYP) enzymes.
    • Type: Oxidation reaction.
    • Purpose: Increases polarity, making the molecule more water-soluble and preparing it for further metabolism.
  • Hydrolysis:
    • What: Breakdown of a molecule by adding water, splitting it into smaller molecules.
    • Enzyme: Hydrolases (e.g., esterases, amidases).
    • Type: Decomposition reaction.
    • Purpose: Cleaves bonds in complex molecules, facilitating their excretion or further metabolism.

Here are five challenging MCQs related to Phase I metabolism reactions:

  1. Which enzyme is most commonly involved in the hydroxylation of drugs during Phase I metabolism?
    • A) Esterase
    • B) Alcohol dehydrogenase
    • C) Cytochrome P450
    • D) UDP-glucuronosyltransferase
    Answer: C
  2. In the context of Phase I metabolism, what is the primary outcome of N-oxidation reactions?
    • A) Addition of a hydroxyl group to a drug molecule
    • B) Conversion of a drug molecule into a more polar form by adding oxygen to nitrogen
    • C) Hydrolysis of ester or amide bonds
    • D) Reduction of nitro groups to amino groups
    Answer: B
  3. Which of the following drugs is primarily metabolized through Phase I hydrolysis reactions rather than oxidation or reduction?
    • A) Acetaminophen
    • B) Diazepam
    • C) Propranolol
    • D) Aspirin
    Answer: D
  4. Which Phase I metabolic reaction is responsible for the conversion of codeine to morphine?
    • A) Hydrolysis
    • B) Oxidation
    • C) Reduction
    • D) Dealkylation
    Answer: B
  5. Which of the following statements about dealkylation reactions in Phase I metabolism is TRUE?
    • A) Dealkylation reactions typically result in the addition of an oxygen atom to the drug molecule.
    • B) Dealkylation involves the removal of alkyl groups from the drug molecule, which can result in the formation of a more active or toxic metabolite.
    • C) Dealkylation is a form of hydrolysis that breaks ester bonds in the drug molecule.
    • D) Dealkylation reactions are primarily mediated by UDP-glucuronosyltransferases.
    Answer: B

These questions are designed to test a deeper understanding of the specific mechanisms and outcomes of Phase I drug metabolism reactions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

State the two types of oxidation reactions
State the difference between deimination and deamination

A

Two types of oxidation reactions:
Oxygen is incorporated into the drug molecule (e.g. hydroxylation, epoxidations, sulfoxidations)
Oxidation causes the loss of part of the drug molecule
(e.g. oxidative deimination, dealkylation)

Microsomal Mixed Function Oxidases (MFOs)
-“Microsomes”
form in vitro after cell homogenization and fractionation of ER
Rough ER microsomes are primarily associated with protein synthesis
Smooth ER (are usually associated with steroid hormones) microsomes contain a class of oxidative enzymes called cytochrome P450 enzymes

-“Mixed Function Oxidases” or “Monooxygenases”
These enzymes require a reducing agent (NADPH) and molecular oxygen
(one oxygen atom appearing in the product and the other in the form of water)

Deimination and deamination are related terms in biochemistry, but they refer to different processes:

Deimination: Also known as citrullination, it’s the conversion of an arginine amino acid residue in a protein to a citrulline residue. This process is catalyzed by enzymes called peptidylarginine deiminases (PADs) and plays a role in various cellular processes, including protein function regulation and immune responses.

Deamination: Refers to the removal of an amino group (-NH2) from a molecule, resulting in the formation of a keto or aldehyde group. In biochemistry, deamination reactions are crucial for the breakdown of amino acids, nucleotides, and other biomolecules. Enzymes like deaminases and amidases catalyze these reactions.

Let me know if you’d like more details or have specific questions about these processes!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What kind of reactions do MFOs catalyze?
Mixed function oxidases consists of which 2 enzymes?
Why are MFOs called mixed function?
What by products are formed in the CYP450 catalytic cycle?

A

Yes, Cytochrome P450 enzymes are a type of mixed-function oxidase (MFO).

Mixed-function oxidases are enzymes that catalyze oxidation reactions, using one substrate as the electron donor (usually NADPH or NADH which give two electrons and one of these electron reduces the ferric (Fe+++) in the CYP450 to ferrous (Fe++) ) and another substrate as the electron acceptor(CYP450) while the other electron enables the activation of molecular oxygen). This means they oxidize one molecule while reducing another.

In the context of mixed-function oxidases (also known as monooxygenases), such as the cytochrome P450 (CYP450) enzyme system, NADPH indeed undergoes oxidation as it loses electrons, and the electron acceptor (typically cytochrome P450) is reduced. Here is how it works:

Oxidation and Reduction in the Cytochrome P450 System:

1.	Role of NADPH:
•	NADPH (Nicotinamide Adenine Dinucleotide Phosphate) serves as an electron donor.
•	When NADPH donates electrons, it is oxidized to NADP⁺. 

Cytochrome P450 enzymes specifically catalyze monooxygenase reactions, which involve the insertion of one oxygen atom into the substrate, resulting in a hydroxylated product. The general reaction is:

Substrate + NADPH + H+ + O2 → Hydroxylated product + NADP+ + H2O

The “mixed-function” designation refers to the ability of these enzymes to perform multiple functions, including:
1. Oxidation
2. Reduction
3. Electron transfer

MFO consists of two enzymes:
1.A Flavoprotein, NADPH-cytochrome c reductase
One mole of this enzyme contains one mole each of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD)
Enzyme is also called NADPH-cytochrome P450 reductase

2.Cytochrome P450
named based on its light absorption at 450 nm when complexed with carbon monoxide
is a hemoprotein containing an iron atom which can alternate between the ferrous (Fe++) and ferric (Fe+++) states; “has REDOX properties”
Electron acceptor
Serves as terminal oxidase
Its relative abundance compared to NADPH-cytochrome P450 reductase makes it the rate-limiting step in the oxidation reactions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

What is the CP450 enzyme and why is it called 450?
Which organelle contains CP450

A

These enzymes contain a heme group (iron-porphyrin complex) and are responsible for catalyzing oxidation reactions, often involving the transfer of electrons. The name “P450” comes from the characteristic absorbance peak at 450 nm in the presence of carbon monoxide. Cytochrome P450 gets its name from its unique spectral properties. The “P450” designation refers to the enzyme’s ability to absorb light at a specific wavelength:

  • “P” stands for “pigment” (referring to the heme group, a colored component of the enzyme)
  • “450” represents the wavelength (in nanometers) at which the enzyme absorbs light, specifically the Soret band (a characteristic peak in the absorption spectrum of heme proteins)

In the presence of carbon monoxide (CO), the enzyme’s absorption peak shifts to 450 nm, which is why it’s called P450. This spectral property allows researchers to detect and quantify the enzyme’s activity.

The “Cytochrome” part of the name refers to the enzyme’s classification as a hemoprotein, containing a heme group (a porphyrin ring with an iron atom at its center). Cytochromes are a family of proteins that play crucial roles in various cellular processes, including electron transport and oxidation reactions.

Microsomes, which are vesicles derived from the smooth ER, contain Cytochrome P450 enzymes and are often used in vitro to study drug metabolism and toxicity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

How many families and sub families of cytochrome P450 genes do humans have ?
Which cytochrome is responsible for both drug and steroid metabolism

A

Humans have 18 families of cytochrome P450 genes and 43 subfamilies:

—CYP1 drug metabolism (3 subfamilies, 3 genes, 1 pseudogene)
-CYP2 drug and steroid metabolism (13 subfamilies, 16 genes, 16 pseudogenes)
-CYP3 drug metabolism (1 subfamily, 4 genes, 2 pseudogenes)
-CYP4 arachidonic acid or fatty acid metabolism (5 subfamilies, 11 genes, 10 pseudogenes)
-CYP5 Thromboxane A2 synthase (1 subfamily, 1 gene)
—CYP7A bile acid biosynthesis 7-alpha hydroxylase of steroid nucleus (1 subfamily member)
—CYP7B brain specific form of 7-alpha hydroxylase (1 subfamily member)
—CYP8A prostacyclin synthase (1 subfamily member)
—CYP8B bile acid biosynthesis (1 subfamily member)
—CYP11 steroid biosynthesis (2 subfamilies, 3 genes)
CYP17 steroid biosynthesis (1 subfamily, 1 gene) 17-alpha hydroxylase
—CYP19 steroid biosynthesis (1 subfamily, 1 gene) aromatase forms estrogen
—CYP20 Unknown function (1 subfamily, 1 gene)
—CYP21 steroid biosynthesis (1 subfamily, 1 gene, 1 pseudogene)
—CYP24 vitamin D degradation (1 subfamily, 1 gene)
—CYP26A retinoic acid hydroxylase important in development (1 subfamily member)
—CYP26B probable retinoic acid hydroxylase (1 subfamily member)
—CYP26C probabvle retinoic acid hydroxylase (1 subfamily member)
—CYP27A bile acid biosynthesis (1 subfamily member)
—CYP27B Vitamin D3 1-alpha hydroxylase activates vitamin D3 (1 subfamily member)
—CYP27C Unknown function (1 subfamily member)
—CYP39 7 alpha hydroxylation of 24 hydroxy cholesterol (1 subfamily member)
—CYP46 cholesterol 24-hydroxylase (1 subfamily member)
—CYP51 cholesterol biosynthesis (1 subfamily, 1 gene, 3 pseudogenes) lanosterol 14-alpha demethylase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

State five inhibitors and five inducers of Cytochrome P450 enzymes

Here are 15 challenging MCQs based on Cytochrome P450 (CYP450) inhibitors and inducers:

  1. Which of the following drugs is a known CYP450 inhibitor that can increase the plasma concentration of drugs metabolized by CYP450 enzymes?
    • A) Carbamazepine
    • B) Rifampin
    • C) Ketoconazole
    • D) Phenobarbital

**

  1. Which CYP450 inducer is most likely to decrease the effectiveness of oral contraceptives by increasing their clearance?
    • A) St. John’s Wort
    • B) Cimetidine
    • C) Erythromycin
    • D) Fluconazole

**

  1. Which of the following drugs would be least affected by the CYP450 inhibitor, grapefruit juice?
    • A) Diazepam
    • B) Lovastatin
    • C) Warfarin
    • D) Carbamazepine

**

  1. Which CYP450 inhibitor is also known to affect the metabolism of drugs metabolized by CYP3A4?
    • A) Isoniazid
    • B) Omeprazole
    • C) Metronidazole
    • D) Sodium valproate

**

  1. Which of the following drugs is both a CYP450 inducer and can lead to decreased levels of drugs metabolized by CYP3A4?
    • A) Rifampin
    • B) Amiodarone
    • C) Ciprofloxacin
    • D) Ketoconazole

**

  1. Which drug is a CYP450 inducer and can reduce the effectiveness of certain anticoagulants, such as warfarin?
    • A) Phenobarbital
    • B) Fluconazole
    • C) Erythromycin
    • D) Sulfonamides

**

  1. Which CYP450 inhibitor is known to interact with warfarin and increase its anticoagulant effect?
    • A) Fluconazole
    • B) St. John’s Wort
    • C) Carbamazepine
    • D) Rifampin
      *
  2. Chronic alcohol consumption has what effect on CYP450 enzymes compared to acute alcohol consumption?
    • A) Chronic alcohol increases CYP450 activity, while acute alcohol decreases it.
    • B) Acute alcohol increases CYP450 activity, while chronic alcohol decreases it.
    • C) Both chronic and acute alcohol decrease CYP450 activity.
    • D) Both chronic and acute alcohol increase CYP450 activity.

*

  1. Which CYP450 inhibitor is also used to treat fungal infections and has significant drug-drug interactions due to its inhibition of CYP3A4?
    • A) Metronidazole
    • B) Erythromycin
    • C) Fluconazole
    • D) Omeprazole

**

  1. Which of the following substances is a CYP450 inducer that could potentially increase the metabolism of drugs such as cyclosporine A?
    • A) Griseofulvin
    • B) Fluconazole
    • C) Cimetidine
    • D) Erythromycin
  2. Which of the following drugs, when used in combination with CYP450 inhibitors, could potentially lead to drug toxicity due to decreased metabolism?
    • A) Carbamazepine
    • B) Phenobarbital
    • C) Rifampin
    • D) Warfarin

**

  1. Which CYP450 inducer is also known for causing reduced therapeutic effects of antiepileptic drugs due to increased clearance?
    • A) Phenobarbital
    • B) Cimetidine
    • C) Omeprazole
    • D) Metronidazole

**

  1. Which CYP450 inhibitor is frequently used in the treatment of peptic ulcers and can interact with drugs metabolized by CYP450 enzymes?
    • A) St. John’s Wort
    • B) Omeprazole
    • C) Rifampin
    • D) Carbamazepine

**

  1. Which of the following drugs is a potent CYP450 inducer that can lead to increased clearance of medications like oral contraceptives?
    • A) Ketoconazole
    • B) Griseofulvin
    • C) Erythromycin
    • D) Fluconazole
  2. Which CYP450 inhibitor could result in a significant increase in the plasma concentration of drugs that are substrates for CYP3A4 when consumed concurrently?
    • A) Rifampin
    • B) Phenobarbital
    • C) Grapefruit juice
    • D) St. John’s Wort
A

СУР450 INHIBITORS
* DECREASE CYP450 ACTIVITY. This makes DRUG CLEARANCE SLOWER +
DRUGS HAVE GREATER EFFECT *
S Sodium valproate
T TICLODIPINE
I ISONIAZID
C CIMETIDINE
K KETOCONAZOLE
F FLUCONAZOLE
A ALCOHOL (ACUTE / BINGE),AMIODARONE
C—CIPROFLOXACIN,CHLORAMPHENICOL
E ERYTHROMYCIN
S SULFONAMIDES-antibiotic such as sulfadiazine

GROUP GRAPEFRUIT JUICE

C CRANBERRY juice
O OMEPRAZOLE
M METRONIDAZOLE

Mnemonic is inhibit STICK FACES GROUP. COM

СУР450 INDUCERS
* INCREASE CYP 45O ACTIVITY
DRUG CLEARANCE FASTER +
DRUGS HAVE LESSER EFFECT *
B BARBITUATES
R RIFAMPIN
A ALCOHOL (CHRONIC)
C CARBAMAZEPINES
S St JOHN’S WORT

G GRISEOFULVIN
P PHENYTOIN
P PHENOBARBITAL
S SULFONYLUREAS

mnemonic is: induce the BRACS GPPS.
BS CRAP GPS

Here are 15 challenging MCQs based on Cytochrome P450 (CYP450) inhibitors and inducers:

  1. Which of the following drugs is a known CYP450 inhibitor that can increase the plasma concentration of drugs metabolized by CYP450 enzymes?
    • A) Carbamazepine
    • B) Rifampin
    • C) Ketoconazole
    • D) Phenobarbital
    Answer: C
  2. Which CYP450 inducer is most likely to decrease the effectiveness of oral contraceptives by increasing their clearance?
    • A) St. John’s Wort
    • B) Cimetidine
    • C) Erythromycin
    • D) Fluconazole
    Answer: A
  3. Which of the following drugs would be least affected by the CYP450 inhibitor, grapefruit juice?
    • A) Diazepam
    • B) Lovastatin
    • C) Warfarin
    • D) Carbamazepine
    Answer: D

Carbamazepine: Primarily metabolized by CYP3A4, but carbamazepine is also an inducer of CYP3A4. This means it can actually increase the activity of CYP3A4, potentially reducing the impact of grapefruit juice on its metabolism.

  1. Which CYP450 inhibitor is also known to affect the metabolism of drugs metabolized by CYP3A4?
    • A) Isoniazid
    • B) Omeprazole
    • C) Metronidazole
    • D) Sodium valproate
    Answer: B
  2. Which of the following drugs is both a CYP450 inducer and can lead to decreased levels of drugs metabolized by CYP3A4?
    • A) Rifampin
    • B) Amiodarone
    • C) Ciprofloxacin
    • D) Ketoconazole
    Answer: A
  3. Which drug is a CYP450 inducer and can reduce the effectiveness of certain anticoagulants, such as warfarin?
    • A) Phenobarbital
    • B) Fluconazole
    • C) Erythromycin
    • D) Sulfonamides
    Answer: A
  4. Which CYP450 inhibitor is known to interact with warfarin and increase its anticoagulant effect?
    • A) Fluconazole
    • B) St. John’s Wort
    • C) Carbamazepine
    • D) Rifampin
    Answer: A
  5. Chronic alcohol consumption has what effect on CYP450 enzymes compared to acute alcohol consumption?
    • A) Chronic alcohol increases CYP450 activity, while acute alcohol decreases it.
    • B) Acute alcohol increases CYP450 activity, while chronic alcohol decreases it.
    • C) Both chronic and acute alcohol decrease CYP450 activity.
    • D) Both chronic and acute alcohol increase CYP450 activity.
    Answer: A
  6. Which CYP450 inhibitor is also used to treat fungal infections and has significant drug-drug interactions due to its inhibition of CYP3A4?
    • A) Metronidazole
    • B) Erythromycin
    • C) Fluconazole
    • D) Omeprazole
    Answer: C
  7. Which of the following substances is a CYP450 inducer that could potentially increase the metabolism of drugs such as cyclosporine A?
    • A) Griseofulvin
    • B) Fluconazole
    • C) Cimetidine
    • D) Erythromycin
    Answer: A
  8. Which of the following drugs, when used in combination with CYP450 inhibitors, could potentially lead to drug toxicity due to decreased metabolism?
    • A) Carbamazepine
    • B) Phenobarbital
    • C) Rifampin
    • D) Warfarin
    Answer: D
  9. Which CYP450 inducer is also known for causing reduced therapeutic effects of antiepileptic drugs due to increased clearance?
    • A) Phenobarbital
    • B) Cimetidine
    • C) Omeprazole
    • D) Metronidazole
    Answer: A
  10. Which CYP450 inhibitor is frequently used in the treatment of peptic ulcers and can interact with drugs metabolized by CYP450 enzymes?
    • A) St. John’s Wort
    • B) Omeprazole
    • C) Rifampin
    • D) Carbamazepine
    Answer: B
  11. Which of the following drugs is a potent CYP450 inducer that can lead to increased clearance of medications like oral contraceptives?
    • A) Ketoconazole
    • B) Griseofulvin
    • C) Erythromycin
    • D) Fluconazole
    Answer: B
  12. Which CYP450 inhibitor could result in a significant increase in the plasma concentration of drugs that are substrates for CYP3A4 when consumed concurrently?
    • A) Rifampin
    • B) Phenobarbital
    • C) Grapefruit juice
    • D) St. John’s Wort
    Answer: C

These questions are designed to assess a deeper understanding of how various substances influence CYP450 enzyme activity and the implications for drug interactions and metabolism.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Ethanol induces which cytochrome enzyme?
What about barbiturate? What cytochrome enzyme does it induce?
What cytochrome enzyme does cigarette smoke and charred food induce?

A

Induction of P450 enzymes:
PPAR (peroxisome proliferator activated receptor) ligands (e.g.clofibrate) induce P450 enzymes

CYP1 family are induced by aromatic hydrocarbons
(cigarette smoke; charred food)
CYP2E enzymes induced by ethanol
CYP2B enzymes induced 40-50 fold by barbiturates

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

How can polymorphism cause differences in drug metabolism

Which of the following is an example of a cytochrome P450 enzyme that shows significant genetic polymorphism?
• A) CYP3A4
• B) CYP2E1
• C) CYP2D6
• D) CYP1A2

A

Polymorphisms cause differences in drug metabolism:
CYP2C19 has a polymorphism that changes the enzyme’s ability to metabolize mephenytoin (a marker drug). In Caucasians, the polymorphism for the poor metabolizer phenotype is only seen in 3% of the population. However, it is seen in 20% of the asian population.
=> It is important to be aware of a person’s race when drugs are given that are metabolized differently by different populations

Which of the following is an example of a cytochrome P450 enzyme that shows significant genetic polymorphism?
• A) CYP3A4
• B) CYP2E1
• C) CYP2D6
• D) CYP1A2
Answer: C
Why: Got it! Let’s use the specific example of CYP2C19 and mephenytoin to illustrate how polymorphisms can change drug metabolism:

  1. CYP2C19 Enzyme: This enzyme is responsible for metabolizing various drugs, including mephenytoin, an anticonvulsant.
  2. Polymorphisms: Genetic variations in the CYP2C19 gene can lead to different versions of the enzyme that work at different speeds.
  1. Poor Metabolizers (PM):
    • Genetic Variant: Some people have a version of the CYP2C19 gene that produces an enzyme with little to no activity.
    • Effect on Mephenytoin: These individuals cannot effectively metabolize mephenytoin. The drug stays in their body longer, increasing the risk of side effects.
  2. Extensive Metabolizers (EM):
    • Genetic Variant: These people have the “normal” version of the CYP2C19 gene, producing an enzyme with standard activity.
    • Effect on Mephenytoin: They metabolize mephenytoin at a normal rate, achieving the desired therapeutic effect without excessive side effects.
  3. Ultra-Rapid Metabolizers (UM):
    • Genetic Variant: Some individuals have a version of the CYP2C19 gene that produces an enzyme with higher-than-normal activity.
    • Effect on Mephenytoin: They metabolize mephenytoin very quickly, which may lead to lower drug levels in the body and reduced effectiveness.
  • Dose Adjustment: Knowing a patient’s CYP2C19 genotype helps doctors adjust the dose of mephenytoin. For example, a poor metabolizer might need a lower dose to avoid side effects, while an ultra-rapid metabolizer might need a higher dose for the drug to be effective.

Polymorphisms in the CYP2C19 gene create different versions of the enzyme with varying abilities to metabolize drugs like mephenytoin. This affects how much of the drug is needed and how effective or safe it is for the patient. Understanding these differences helps tailor drug therapy to individual needs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Which is a chief CP450 inhibitor ?

Which enzyme is NOT primarily involved in the metabolism of acetaminophen?

•	A) CYP2E1
•	B) UGTs (UDP-glucuronosyltransferases)
•	C) SULTs (sulfotransferases)
•	D) CYP3A4

Here are 10 MCQs based on the information provided about Cytochrome P450 enzymes and drug interactions:

  1. Which of the following drugs induces CYP2B and leads to increased metabolism of other drugs?
    • A) Ketoconazole
    • B) Grapefruit juice
    • C) Barbiturates
    • D) Erythromycin
  2. Which enzyme is inhibited by ketoconazole, leading to reduced metabolism of other drugs?
    • A) CYP2B
    • B) CYP3A4
    • C) CYP1A2
    • D) CYP2D6
  3. Grapefruit juice is known to inhibit which of the following Cytochrome P450 enzymes?
    • A) CYP2B
    • B) CYP3A4
    • C) CYP2D6
    • D) CYP2E1
  4. Which drug listed is a substrate for CYP3A4?
    • A) Acetaminophen
    • B) Codeine
    • C) Diazepam
    • D) Warfarin
  5. Which of the following drugs would have its metabolism reduced by grapefruit juice?
    • A) Lovastatin
    • B) Lidocaine
    • C) Taxol
    • D) All of the above
  6. What effect does barbiturate use have on CYP2B and related drug metabolism?
    • A) Inhibition of CYP2B, leading to decreased metabolism of other drugs
    • B) Induction of CYP2B, leading to increased metabolism of other drugs
    • C) No effect on CYP2B
    • D) Induction of CYP3A4, leading to increased metabolism of other drugs
  7. Which of the following drugs is NOT a substrate for CYP3A4?
    • A) Cyclosporin A
    • B) Diazepam
    • C) Acetaminophen
    • D) Warfarin
  8. How does grapefruit juice affect the concentration of drugs metabolized by CYP3A4?
    • A) Decreases the concentration by enhancing drug metabolism
    • B) Increases the concentration by inhibiting CYP3A4
    • C) Has no effect on drug concentrations
    • D) Decreases the concentration by inducing CYP3A4
  9. Which drug listed is used as an immunosuppressant and is a substrate for CYP3A4?
    • A) Codeine
    • B) Erythromycin
    • C) Cyclosporin A
    • D) Taxol
  10. If a patient consumes grapefruit juice while taking a CYP3A4 substrate, what is the likely outcome?
    • A) Decreased drug efficacy due to increased metabolism
    • B) Increased drug toxicity due to reduced metabolism
    • C) No change in drug levels
    • D) Enhanced drug metabolism due to CYP3A4 induction

Which of the following drugs is most likely to have a 12-fold increase in concentration due to grapefruit juice consumption?
• A) Lidocaine
• B) Warfarin
• C) Codeine
• D) Taxol

Which of the following drug interactions would be least affected by grapefruit juice consumption?
• A) Lovastatin
• B) Cyclosporin A
• C) Erythromycin
• D) Diazepam

2.	A patient on warfarin begins taking a medication that induces CYP2B. What is the expected effect on warfarin therapy?
•	A) Increased anticoagulant effect due to enhanced metabolism
•	B) Decreased anticoagulant effect due to increased metabolism
•	C) No effect on warfarin metabolism
•	D) Increased bleeding risk due to decreased metabolism


3.	Which enzyme’s activity would be most impacted by ketoconazole, leading to increased plasma levels of drugs metabolized by this enzyme?
•	A) CYP2E1
•	B) CYP2D6
•	C) CYP3A4
•	D) CYP1A2

4.	In the context of drug metabolism, which of the following is a key mechanism by which barbiturates alter drug metabolism?
•	A) Inhibition of CYP2B leading to decreased metabolism
•	B) Induction of CYP2B leading to increased metabolism of other drugs
•	C) Inhibition of CYP3A4 leading to decreased drug metabolism
•	D) Induction of CYP3A4 leading to decreased drug metabolism

5.	Which of the following statements about CYP3A4 substrates is TRUE?
•	A) All CYP3A4 substrates are affected similarly by grapefruit juice.
•	B) CYP3A4 substrates generally exhibit increased metabolism when CYP3A4 is inhibited.
•	C) CYP3A4 substrates can have significantly increased plasma concentrations when CYP3A4 activity is inhibited by substances such as grapefruit juice.
•	D) Grapefruit juice exclusively affects CYP3A4 substrates by decreasing their metabolism and does not affect their plasma concentrations.
A

P450s and drug interactions:
Barbiturates induce CYP2B => increased metabolism of other drugs
Antifungals (e.g. ketoconazole) inhibit fungal CYP51 and unintentionally also human CYP3A4
=> reduced metabolism of other drugs
Grapefruit juice contains a CYP3A4 inhibitor =>12 fold increase in some drug concentrations. Grapefruit juice is a Chief CP450 inhibitor
CYP3A4 Substrates: • Acetominophen (Tylenol) • Codeine (narcotic) • Cyclosporin A (immunosuppressant), • Diazepam (Valium) • Erythromycin (Antibiotic) • Lidocaine (local anaesthetic), • Lovastatin (HMGCoA reductase inhibitor), • Taxol (cancer drug), • Warfarin (anticoagulant).

Which enzyme is NOT primarily involved in the metabolism of acetaminophen?

•	A) CYP2E1
•	B) UGTs (UDP-glucuronosyltransferases)
•	C) SULTs (sulfotransferases)
•	D) CYP3A4

Answer: D

Here are 10 MCQs based on the information provided about Cytochrome P450 enzymes and drug interactions:

  1. Which of the following drugs induces CYP2B and leads to increased metabolism of other drugs?
    • A) Ketoconazole
    • B) Grapefruit juice
    • C) Barbiturates
    • D) Erythromycin
    Answer: C
  2. Which enzyme is inhibited by ketoconazole, leading to reduced metabolism of other drugs?
    • A) CYP2B
    • B) CYP3A4
    • C) CYP1A2
    • D) CYP2D6
    Answer: B
  3. Grapefruit juice is known to inhibit which of the following Cytochrome P450 enzymes?
    • A) CYP2B
    • B) CYP3A4
    • C) CYP2D6
    • D) CYP2E1
    Answer: B
  4. Which drug listed is a substrate for CYP3A4?
    • A) Acetaminophen
    • B) Codeine
    • C) Diazepam
    • D) Warfarin
    Answer: C
  5. Which of the following drugs would have its metabolism reduced by grapefruit juice?
    • A) Lovastatin
    • B) Lidocaine
    • C) Taxol
    • D) All of the above
    Answer: D
  6. What effect does barbiturate use have on CYP2B and related drug metabolism?
    • A) Inhibition of CYP2B, leading to decreased metabolism of other drugs
    • B) Induction of CYP2B, leading to increased metabolism of other drugs
    • C) No effect on CYP2B
    • D) Induction of CYP3A4, leading to increased metabolism of other drugs
    Answer: B
  7. Which of the following drugs is NOT a substrate for CYP3A4?
    • A) Cyclosporin A
    • B) Diazepam
    • C) Acetaminophen
    • D) Warfarin
    Answer: C
  8. How does grapefruit juice affect the concentration of drugs metabolized by CYP3A4?
    • A) Decreases the concentration by enhancing drug metabolism
    • B) Increases the concentration by inhibiting CYP3A4
    • C) Has no effect on drug concentrations
    • D) Decreases the concentration by inducing CYP3A4
    Answer: B
  9. Which drug listed is used as an immunosuppressant and is a substrate for CYP3A4?
    • A) Codeine
    • B) Erythromycin
    • C) Cyclosporin A
    • D) Taxol
    Answer: C
  10. If a patient consumes grapefruit juice while taking a CYP3A4 substrate, what is the likely outcome?
    • A) Decreased drug efficacy due to increased metabolism
    • B) Increased drug toxicity due to reduced metabolism
    • C) No change in drug levels
    • D) Enhanced drug metabolism due to CYP3A4 induction
    Answer: B

Nn Which of the following drugs is most likely to have a 12-fold increase in concentration due to grapefruit juice consumption?
• A) Lidocaine
• B) Warfarin
• C) Codeine
• D) Taxol
Answer: B

Which of the following drug interactions would be least affected by grapefruit juice consumption?
• A) Lovastatin
• B) Cyclosporin A
• C) Erythromycin
• D) Diazepam
Answer: C
2. A patient on warfarin begins taking a medication that induces CYP2B. What is the expected effect on warfarin therapy?
• A) Increased anticoagulant effect due to enhanced metabolism
• B) Decreased anticoagulant effect due to increased metabolism
• C) No effect on warfarin metabolism
• D) Increased bleeding risk due to decreased metabolism
Answer: B
3. Which enzyme’s activity would be most impacted by ketoconazole, leading to increased plasma levels of drugs metabolized by this enzyme?
• A) CYP2E1
• B) CYP2D6
• C) CYP3A4
• D) CYP1A2
Answer: C
4. In the context of drug metabolism, which of the following is a key mechanism by which barbiturates alter drug metabolism?
• A) Inhibition of CYP2B leading to decreased metabolism
• B) Induction of CYP2B leading to increased metabolism of other drugs
• C) Inhibition of CYP3A4 leading to decreased drug metabolism
• D) Induction of CYP3A4 leading to decreased drug metabolism
Answer: B
5. Which of the following statements about CYP3A4 substrates is TRUE?
• A) All CYP3A4 substrates are affected similarly by grapefruit juice.
• B) CYP3A4 substrates generally exhibit increased metabolism when CYP3A4 is inhibited.
• C) CYP3A4 substrates can have significantly increased plasma concentrations when CYP3A4 activity is inhibited by substances such as grapefruit juice.
• D) Grapefruit juice exclusively affects CYP3A4 substrates by decreasing their metabolism and does not affect their plasma concentrations.
Answer: C

These questions cover the basics of Cytochrome P450 interactions, including induction, inhibition, and the impact of substances like grapefruit juice on drug metabolism.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

State four things needed for drug oxidation to occur

A

Drug oxidation requires:
1.Cytochrome P450(this enzyme usually or mainly catalyzes oxidation reactions even though it can catalyze other reactions. In this, a substrate either gains oxygen atoms or loses hydrogen atoms)
2.Cytochrome P450 reductase: Cytochrome P450 Reductase:
• Function: This enzyme transfers electrons from NADPH to the CYP450 enzyme. It is essential for the catalytic cycle of CYP450, enabling the reduction of the heme iron.
• Mechanism: Cytochrome P450 reductase receives electrons from NADPH and sequentially transfers them to the CYP450 enzyme.
3.NADPH(NADPH (nicotinamide adenine dinucleotide phosphate)**: This serves as the electron donor for the reduction of cytochrome P450 reductase, which in turn supplies electrons to CYP450 enzymes.)
4.Molecular oxygen: Molecular oxygen is required for the oxidation reactions catalyzed by CYP450 enzymes. It acts as the oxidizing agent, with one oxygen atom incorporated into the substrate and the other reduced to water.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Explain how the oxidation cycle occurs
Which form of oxidized CP450 enzyme combines with the drug to form an iron cytochrome P450 complex?

Fe2+ ferrous iron or Fe3+ ferric iron

1. Which step in the cytochrome P450 catalytic cycle directly involves the reduction of molecular oxygen?

A) The combination of oxidized cytochrome P450 with a drug substrate
B) The donation of an electron from NADPH to cytochrome P450 reductase
C) The formation of the “activated oxygen”-cytochrome P450-substrate complex
D) The transfer of “activated” oxygen to the drug substrate
E) The dissociation of the oxidized product from cytochrome P450

**

2. What role does NADPH play in the cytochrome P450 enzyme system?

A) It donates electrons to reduce molecular oxygen
B) It binds directly to the substrate to facilitate oxidation
C) It combines with cytochrome P450 to form a binary complex
D) It is oxidized by cytochrome P450 reductase during the reaction
E) It transfers “activated” oxygen to the drug substrate

**

3. In the cytochrome P450 catalytic cycle, what is the oxidation state of iron after the initial combination of oxidized cytochrome P450 with a drug substrate?

A) Fe2+
B) Fe3+
C) Fe4+
D) Fe5+
E) The oxidation state remains unchanged

4. Which of the following statements best explains the significance of the “activated oxygen” in the cytochrome P450 cycle?

A) It allows the enzyme to form a binary complex with the drug substrate
B) It acts as a reducing agent to regenerate cytochrome P450
C) It stabilizes the cytochrome P450-drug complex during the reaction
D) It provides the oxidative power necessary for drug substrate oxidation
E) It initiates the formation of NADPH

5. If the cytochrome P450 reductase is defective, which step in the P450 cycle would be directly impaired, leading to decreased drug metabolism?

A) Binding of the drug substrate to cytochrome P450
B) Initial electron donation to cytochrome P450
C) Formation of the binary complex
D) Transfer of “activated” oxygen to the drug substrate
E) Dissociation of the oxidized product

A

The cycle involves four steps:
NB : Fe2+ is ferrous iron and Fe3+ is ferric iron
1.Oxidized (Fe3+ or ferric iron) cytochrome P-450 combines with a drug substrate to form a binary complex.(Iron cytochrome P450 complex)
2.NADPH donates an electron to the cytochrome P-450 reductase, which in turn reduces the oxidized cytochrome P-450-drug complex (so the complex gains an electron since reduction has occurred)
3.A second electron is introduced from NADPH via the same cytochrome P-450 reductase, which serves to reduce molecular oxygen and form an “activated oxygen”-cytochrome P-450-substrate complex.
4.This complex in turn transfers “activated” oxygen to the drug substrate to form the oxidized product. The potent oxidizing properties of this activated oxygen permit oxidation of a large number of substrates.
so this continues like a positive feedback loop and keeps going on and on and on

  1. Formation of the Binary Complex:
    • Oxidized (Fe³⁺ or ferric iron) cytochrome P450 binds to a drug substrate, forming a binary complex.
    • At this stage, cytochrome P450 is in its oxidized state (Fe³⁺).
    1. First Electron Donation:
      • NADPH donates an electron to cytochrome P450 reductase, which then reduces the cytochrome P450-drug complex from Fe³⁺ to Fe²⁺ (ferrous iron). This is the reduction of the complex.
    2. Second Electron Donation and Formation of Activated Oxygen:
      • A second electron is donated by NADPH via cytochrome P450 reductase. This electron is used to reduce molecular oxygen (O₂) bound to the cytochrome P450.
      • This results in the formation of an “activated oxygen”-cytochrome P450-substrate complex. The oxygen is partially reduced and now in a highly reactive state, making this part of the cycle more complicated as it involves both reduction (to activate oxygen) and oxidation (due to the subsequent steps).
    3. Oxidation of the Substrate:
      • The activated oxygen is transferred to the drug substrate, leading to its oxidation and the formation of the oxidized drug product.

Clarification on the Confusion:

•	The term “oxidized cytochrome P450” refers to the Fe³⁺ state of the enzyme at the beginning of the cycle.
•	The “activated oxygen” complex forms after the reduction of the enzyme and molecular oxygen. This complex is what enables the oxidation of the drug substrate.

Corrected Understanding:

•	Oxidation in this context refers to the final step where the substrate is oxidized.
•	Reduction occurs earlier when the cytochrome P450 is reduced from Fe³⁺ to Fe²⁺ and when oxygen is reduced to form the reactive “activated oxygen” species.

Given this explanation, the formation of the “activated oxygen” complex involves reduction of oxygen, not oxidation. The oxidation happens when this “activated oxygen” is transferred to the substrate.

No, CYP450 reductase does not transfer the activated oxygen itself. Instead, the transfer of activated oxygen is performed by the cytochrome P450 enzyme (CYP450) after it is activated. However, cytochrome P450 reductase plays a crucial supporting role in the process.

  1. Cytochrome P450 System:
    • The cytochrome P450 enzyme system is composed of two key proteins:
      • Cytochrome P450 enzyme (CYP450)
      • NADPH-cytochrome P450 reductase (CPR)
  2. Function of CYP450 Reductase:
    • CYP450 reductase is an electron transfer protein that shuttles electrons from NADPH (nicotinamide adenine dinucleotide phosphate) to the CYP450 enzyme.
    • This transfer of electrons is essential for the activation of molecular oxygen (O₂) bound to the heme iron (Fe) of the CYP450 enzyme.
  3. Steps in Oxygen Activation and Transfer:
    • Substrate Binding: A drug (substrate) binds to the CYP450 enzyme.
    • Electron Transfer by CYP450 Reductase:
      • CYP450 reductase transfers electrons from NADPH to the heme iron of the CYP450 enzyme in two sequential steps. This process reduces the heme iron from Fe³⁺ to Fe²⁺.
    • Oxygen Binding and Activation:
      • Molecular oxygen (O₂) binds to the reduced Fe²⁺ form of CYP450.
      • A second electron is transferred, leading to the formation of a highly reactive iron-oxo species (FeO³⁺), often referred to as Compound I.
    • Oxygen Transfer to the Substrate:
      • The activated oxygen (Compound I) in the CYP450 enzyme transfers to the substrate, oxidizing it (e.g., hydroxylation, dealkylation).

While CYP450 reductase does not directly transfer activated oxygen, it is critical in providing the necessary electrons that enable the CYP450 enzyme to activate oxygen and subsequently transfer it to the substrate during the oxidation process. The actual transfer of activated oxygen to the drug substrate is carried out by the CYP450 enzyme, not by CYP450 reductase.

Here are five challenging MCQs based on the steps of the cytochrome P450 cycle you described:

1. Which step in the cytochrome P450 catalytic cycle directly involves the reduction of molecular oxygen?

A) The combination of oxidized cytochrome P450 with a drug substrate
B) The donation of an electron from NADPH to cytochrome P450 reductase
C) The formation of the “activated oxygen”-cytochrome P450-substrate complex
D) The transfer of “activated” oxygen to the drug substrate
E) The dissociation of the oxidized product from cytochrome P450

Answer: C) The formation of the “activated oxygen”-cytochrome P450-substrate complex

2. What role does NADPH play in the cytochrome P450 enzyme system?

A) It donates electrons to reduce molecular oxygen
B) It binds directly to the substrate to facilitate oxidation
C) It combines with cytochrome P450 to form a binary complex
D) It is oxidized by cytochrome P450 reductase during the reaction
E) It transfers “activated” oxygen to the drug substrate

Answer: A) It donates electrons to reduce molecular oxygen

3. In the cytochrome P450 catalytic cycle, what is the oxidation state of iron after the initial combination of oxidized cytochrome P450 with a drug substrate?

A) Fe2+
B) Fe3+
C) Fe4+
D) Fe5+
E) The oxidation state remains unchanged

Answer: B) Fe3+

4. Which of the following statements best explains the significance of the “activated oxygen” in the cytochrome P450 cycle?

A) It allows the enzyme to form a binary complex with the drug substrate
B) It acts as a reducing agent to regenerate cytochrome P450
C) It stabilizes the cytochrome P450-drug complex during the reaction
D) It provides the oxidative power necessary for drug substrate oxidation
E) It initiates the formation of NADPH

Answer: D) It provides the oxidative power necessary for drug substrate oxidation

5. If the cytochrome P450 reductase is defective, which step in the P450 cycle would be directly impaired, leading to decreased drug metabolism?

A) Binding of the drug substrate to cytochrome P450
B) Initial electron donation to cytochrome P450
C) Formation of the binary complex
D) Transfer of “activated” oxygen to the drug substrate
E) Dissociation of the oxidized product

Answer: B) Initial electron donation to cytochrome P450

These questions are designed to test a deep understanding of the cytochrome P450 catalytic cycle and its components.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

What kind of phase 1 reaction is Aromatic hydroxylation?
What is aromatic hydroxylation

A

It is an Oxidation reaction:

Aromatic hydroxylation is a cytochrome P450-mediated oxidation process(the process is mediated by some P450enzymes) that adds a hydroxyl group to the ring structure of aromatic compounds like benzene derivatives. This aids in the metabolism of such drugs.
This metabolic reaction enhances water solubility, aiding in the elimination of drugs and toxins from the body.

23
Q

What kind of phase 1 reaction is Aliphatic hydroxylation,Epoxidation?
What is epoxidation?
What is Aliphatic hydroxylation ?

A

They are oxidation reactions in phase 1 metabolism

  1. Epoxidation: Involves the formation of an epoxide ring structure through the addition of an oxygen atom to a double bond. Cytochrome P450 enzymes can catalyze epoxidation reactions, which are important in detoxification processes.
  2. Aliphatic hydroxylation: Refers to the addition of a hydroxyl group (-OH) to aliphatic (non-aromatic) compounds. Cytochrome P450 enzymes also catalyze aliphatic hydroxylation reactions, aiding in the metabolism of various drugs and chemicals.
24
Q

Dealkylation is not an oxidation reaction under phase 1 reaction true or false?

A

It’s true wai. It’s under phase 2 saf. Check chat gpt. It’s mainly phase 2
False. Dealkylation can be oxidative or not oxidative depending on the specific substrate or enzyme involved

Dealkylation in phase 1 metabolism can involve oxidative or non-oxidative mechanisms, depending on the specific substrate and enzyme involved:

  1. Oxidative Dealkylation: In this process, cytochrome P450 enzymes catalyze the removal of alkyl groups from a substrate molecule through oxidation. This typically involves introducing oxygen atoms into the substrate molecule, resulting in the removal of the alkyl group as part of the metabolite.
  2. Non-Oxidative Dealkylation: Some dealkylation reactions may occur through non-oxidative mechanisms, such as hydrolysis or other types of enzymatic cleavage that do not involve the introduction of oxygen atoms into the substrate.

Therefore, while dealkylation can be an oxidative reaction when catalyzed by certain cytochrome P450 enzymes, it’s important to note that other enzymes and pathways may facilitate dealkylation through different mechanisms in phase 1 metabolism.

25
Q

What is the difference between alkylation and methylation?

A

Alkylation**: Typically refers to the addition of an alkyl group to a substrate molecule, which can modify its activity or properties. This process is not as common in phase 1 metabolism compared to phase 2 metabolism.

  • Methylation: Specifically refers to the addition of a methyl group, which can occur as part of phase 1 or phase 2 metabolism. In phase 1 metabolism, methylation reactions can occur as oxidative demethylation processes catalyzed by enzymes such as cytochrome P450 enzymes.
26
Q

State four other oxidative reactions catalyzed by CP450 enzyme apart from aromatic hydroxylation,aliphatic hydroxylation,EPO ideation and Dealkylation

A

O-demethylation:example is O demethylation of codeine. Then example for Dealkylation is N Demethylation of diazepam
O demethylation is the removal of a methyl group (CH3) from an oxygen atom in a molecule and N demethylation is the removal of a methyl group (CH3) from an nitrogen atom in a molecule
S-demethylation:removal of a methyl group from a sulfur atom in a molecule
N-oxidation: N-oxidation is a phase 1 metabolic reaction where an oxygen atom is added to a nitrogen atom (N) within a molecule, resulting in the formation of an N-oxide group (N=O)
N-hydroxylation: N-hydroxylation is a phase 1 metabolic reaction where a hydroxyl group (-OH) is added to a nitrogen atom (N) within a molecule

So de- attached to any process or reaction is removal of a functional group

27
Q

State four other oxidative reactions catalyzed by CP450 enzyme apart from aromatic hydroxylation,aliphatic hydroxylation,EPO ideation and Dealkylation

A

O-demethylation:example is O demethylation of codeine. Then example for Dealkylation is N Demethylation of diazepam
O demethylation is the removal of a methyl group (CH3) from an oxygen atom in a molecule and N demethylation is the removal of a methyl group (CH3) from an nitrogen atom in a molecule
S-demethylation:removal of a methyl group from a sulfur atom in a molecule
N-oxidation: N-oxidation is a phase 1 metabolic reaction where an oxygen atom is added to a nitrogen atom (N) within a molecule, resulting in the formation of an N-oxide group (N=O)
N-hydroxylation: N-hydroxylation is a phase 1 metabolic reaction where a hydroxyl group (-OH) is added to a nitrogen atom (N) within a molecule

So de- attached to any process or reaction is removal of a functional group

28
Q

State five other enzymes (apart from CP450 )that catalyze oxidation reactions and explain the reactions they catalyze
Which of these enzymes are mainly present in the liver and are located in the smooth ER
Which of these enzymes are in the cytosol?

A

-Flavin containing monoxygenase system:
Present mainly in liver but some is expressed in gut and lung
Located in smooth endoplasmic reticulum
Oxidizes compounds containing sulfur and nitrogen Using NADH and NADPH as cofactors

-Alcohol dehydrogenase (cytosol): Alcohol dehydrogenase**: Located in the cytosol, it catalyzes the oxidation of alcohols to aldehydes or ketones, using NAD+ as a cofactor.

-Aldehyde oxidation (cytosol): Also in the cytosol, this enzyme oxidizes aldehydes to carboxylic acids, using NAD+ as a cofactor.
-Xanthine oxidase: Found in various tissues including the liver, it catalyzes the oxidation of hypoxanthine to xanthine and xanthine to uric acid, utilizing molecular oxygen and producing hydrogen peroxide.

-Amine oxidases:
Under amine oxidases,We have 1.Monoamine oxidase (nerve terminals, mitochondria. Found in nerve terminals and mitochondria, it catalyzes the oxidation of monoamines (e.g., neurotransmitters) using molecular oxygen and producing ammonia and hydrogen peroxide.
2.Diamine oxidase found in liver microsomes(Primarily endogenous metabolism. liver microsomes, it primarily metabolizes endogenous compounds like histamine.
)

29
Q

Which enzyme catalyzes oxidative reactions that occurs at the neuromuscular junctions and catalyzes oxidative deamination of endogenous catecholamines in housing epinephrine ?
What inhibits this enzyme?

A

Monoamine Oxidases (MAO) – isoforms MAO-A; MAO-B:

Catalyze oxidative deamination of endogenous catecholamines (epinephrine)

Yes, monoamine oxidases (MAOs) are enzymes that break down catecholamines, which include neurotransmitters like dopamine, norepinephrine, and epinephrine. MAOs do this by deaminating the catecholamines, which means they remove an amine group from the molecule, leading to their inactivation and breakdown. There are two main types of MAOs: MAO-A and MAO-B, each with slightly different substrate specificities, but both are involved in the metabolism of catecholamines.

Located in nerve terminals and peripheral tissues
Substrates for catecholamine metabolism found in foods (tyramine) can cause a drug/food interaction

Inhibited by class of antidepressants called MAO inhibitors
(Inhibition of MAO isoforms in the CNS also effects levels of serotonin – Tranylcypromine – irreversible inhibitor)
These drugs can cause severe or fatal drug/drug interactions with drugs that increase release of catecholamines or inhibit their reuptake in nerve terminals (Meperidine, pentazocine, dextromethorphan, SSRI antidepressants)

30
Q

state three foods that contain tyramine
Why will tyramine cause tyramine induced crisis
What is the relationship between tyramine and Monoamine oxidase?
State two mono amine oxidase inhibitors and their effect on tyramine

A

Yes, substrates for catecholamine metabolism found in foods, such as tyramine, can indeed cause significant drug-food interactions, particularly with medications that inhibit monoamine oxidase (MAO) enzymes.

Tyramine can cause hypertension due to its ability to induce the release of norepinephrine (noradrenaline) from nerve endings in the sympathetic nervous system. Here’s how this works:

  1. Role of Tyramine:• Tyramine is a naturally occurring monoamine compound found in various foods like aged cheeses, cured meats, fermented products, and some wines.
    • Normally, tyramine is rapidly metabolized by an enzyme called monoamine oxidase (MAO), particularly by the MAO-A subtype, which breaks down excess tyramine in the gut and liver.

Tyramine is a substance found in various foods and beverages, including aged cheeses, cured meats, and certain alcoholic beverages. Normally, MAO enzymes in the gut and liver break down tyramine and prevent its accumulation in the bloodstream. However, if someone takes medications that inhibit MAO (MAO inhibitors or MAOIs), such as certain antidepressants (e.g., phenelzine, tranylcypromine), the ability to metabolize tyramine is reduced.

When tyramine-containing foods are consumed while on MAOIs, the body cannot efficiently metabolize tyramine. This can lead to a sudden increase in tyramine levels in the bloodstream, causing a condition known as tyramine-induced hypertensive crisis. Symptoms of this crisis include severe headache, rapid heart rate, nausea, and potentially life-threatening increases in blood pressure.

Therefore, individuals taking MAOIs are usually advised to follow a strict dietary regimen that avoids high-tyramine foods to prevent such interactions and associated health risks.

31
Q

State the drugs that :
1.increase catecholamine levels
2. Inhibit reuptake of catecholamines
Why will monoamine oxidase inhibitors cause severe drug drug interactions with drugs that increase release of catecholamines or inhibit Their reuptake in nerve terminals ?

A

. MAO inhibitors (MAOIs), such as tranylcypromine, are a class of antidepressants that irreversibly inhibit monoamine oxidase enzymes, including MAO-A and MAO-B. This inhibition not only affects the breakdown of neurotransmitters like serotonin, dopamine, and norepinephrine (making their levels increase) but also leads to interactions with other drugs and certain foods.

  1. Increased Catecholamine Release: Drugs such as meperidine (a painkiller), pentazocine (a painkiller), and dextromethorphan (a cough suppressant) can increase the release of catecholamines (e.g., dopamine, norepinephrine) from nerve terminals.
  2. Inhibition of Reuptake: Selective serotonin reuptake inhibitors (SSRIs), which are another class of antidepressants that increase serotonin levels by inhibiting its reuptake, can also interact with MAOIs.

When MAOIs are combined with drugs that increase catecholamine release or inhibit reuptake, the concurrent inhibition of MAO which will destroy the catecholamines, can lead to excessive accumulation of these neurotransmitters. This can cause a condition known as serotonin syndrome or hypertensive crisis, characterized by symptoms such as agitation, confusion, high blood pressure, rapid heart rate, and potentially life-threatening complications.

32
Q

Give one example of monoamine oxidase inhibitors?
Between phase I and phase II reactions, which is the true detoxification step in the metabolism process?
What is the main function of phase I reactions?
Drugs can only be subject to one phase I pathway,true or false

A

Tranylcypromine

Phase II is the true “detoxification” step in the metabolism process. Phase I of drug metabolism primarily involves the modification of drugs through processes like oxidation, reduction, or hydrolysis. These reactions, often catalyzed by enzymes like cytochrome P450, introduce or expose functional groups on the drug molecules. However, the products of Phase I reactions are typically more reactive and less water-soluble than the original compounds.

Here’s why Phase I isn’t considered the “true detoxification” step:

  1. Intermediate Metabolites: Phase I often generates intermediate metabolites that may still be biologically active and potentially toxic. These metabolites may require further processing in Phase II to make them more water-soluble and less harmful.
  2. Increased Reactivity: The modifications in Phase I can sometimes make the drug molecules more reactive, which can potentially lead to the formation of toxic byproducts if not further processed.example is prodrugs
  3. Conjugation for Excretion: The primary role of Phase I is to prepare the drug for further processing, rather than to complete the detoxification. Phase II conjugation reactions are crucial for rendering the drug and its metabolites more excretable and less toxic.

In summary, while Phase I is essential for modifying drugs and making them more amenable to further processing, Phase II is considered the “true detoxification” step because it involves the conjugation of these intermediates into more water-soluble, less toxic compounds ready for excretion.

main function:
These reactions
Main function of phase I reactions is to prepare chemicals for phase II metabolism (by creating functional groups that place the drugs in a correct chemical state to be acted upon by Phase II conjugative mechanisms ) and subsequent excretion

False:
Drugs can be subject to several Phase I pathways
Almost any drug can undergo modifications by drug-metabolizing enzyme systems

33
Q

State three different reduction reactions for drug metabolism
Reduction is associated with gain of what and loss of what?
Oxidation is associated with gain of what and loss of what?

A

Azo reduction:
1. Azo Reduction: Azo compounds contain a diazenyl functional group (-N=N-). Reduction of azo bonds involves the enzymatic cleavage of the azo group(cleavage occurs by the addition of electrons facilitated by azoreductase), often mediated by various enzymes in the liver or gut microbiota. This reduction can split the compound into two amines.

  1. Nitrogen Reduction: Reduction of nitro groups (-NO2) involves the enzymatic addition of electrons and protons to convert the nitro group to an amine (-NH2). This reduction is often catalyzed by nitroreductase enzymes.
  2. Dehalogenation: Involves the removal of halogen atoms (e.g., chlorine, bromine,fluorine,iodine) from organic compounds. Dehalogenation reactions are mediated by various enzymes and can occur in phase 1 metabolism to make the compound more reactive for subsequent conjugation or excretion

To remember the three main types of reduction reactions in phase I drug metabolism, you can use a mnemonic or associative method. The three types of reduction reactions are:

  1. Reduction of Nitro Groups
  2. Reduction of Carbonyl Groups
  3. Reduction of Azides

Here’s a way to remember them:

  1. Nitro Groups:
    • “Nitro”: Nitro groups (–NO₂) are reduced to amines (–NH₂).
    • Example: Nitrobenzene is reduced to aniline.
  2. Carbonyl Groups:
    • “Carbs”: Carbonyl groups (–C=O) can be reduced to alcohols (–CH₂OH) or other less oxidized forms.
    • Example: Ketones are reduced to secondary alcohols.
  3. Azides:
    • “Aren’t”: Azides (–N₃) are reduced to amines (–NH₂).
    • Example: Organic azides can be reduced to amines.
  • Reduction of Nitro Groups: Remember that nitro groups are often reduced to amines, which are more “amine-ly” functional.
  • Reduction of Carbonyl Groups: Carbonyl groups (like ketones and aldehydes) become alcohols, which are generally “carb-ony” (more hydrated) forms.
  • Reduction of Azides: Azides are reduced to amines, which can be remembered as a type of “azide” becoming a more basic “amine.”

By associating the functional groups with their reduced forms and using a mnemonic, you can more easily recall the types of reduction reactions involved in phase I drug metabolism.

Hydrolysis reactions under reduction:
Ester hydrolysis
Amide hydrolysis

Reduction: Gain of electrons (often associated with the gain of hydrogen or loss of oxygen).
• Oxidation: Loss of electrons (often associated with the gain of oxygen or loss of hydrogen).

34
Q

What is hydrolysis (or hydrolytic cleavage)under phase 1 reactions?
State two hydrolysis reactions and how they occur

A

Hydrolysis is a chemical reaction where a molecule is cleaved into two or more parts by the addition of a molecule of water (H2O). This process involves breaking a chemical bond in a compound with the insertion of a water molecule, resulting in the formation of two or more new compounds.

  1. Ester Hydrolysis: Involves the cleavage of an ester bond (-COO-) by water, resulting in the formation of an alcohol and a carboxylic acid (or its conjugate base).Example: ( R-COOR’ + H2O \rightarrow R-COOH + R’OH )
  2. Amide Hydrolysis: Involves the cleavage of an amide bond (-CONH-) by water, resulting in the formation of a carboxylic acid and an amine.Example: ( R-CONH2 + H2O \rightarrow R-COOH + NH3 )

Hydrolysis reactions are important in biological systems, including drug metabolism, where enzymes catalyze these reactions to break down complex molecules into simpler components that can be readily eliminated from the body. In drug metabolism, hydrolysis often occurs as part of phase 1 reactions, preparing drugs for further modification (phase 2 conjugation) or direct excretion.

35
Q

State seven conjugation reactions under phase II reactions

A

Conjugation reactions:
-Glucuronidation by UDP-Glucuronosyltransferase:
(on -OH, -COOH, -NH2, -SH groups)
-Sulfation by Sulfotransferase:
(on -NH2, -SO2NH2, -OH groups)
-Acetylation by acetyltransferase:
(on -NH2, -SO2NH2, -OH groups)
-Amino acid conjugation
(on -COOH groups)
-Glutathione conjugation by Glutathione-S-transferase:
(to epoxides or organic halides)
-Fatty acid conjugation
(on -OH groups)
-Condensation reactions

36
Q

Quantitatively,which reaction is the most important phase II pathway for drugs and endogenous compounds?
Which enzyme primarily carries out this process?
This enzyme has how many isoforms in man?
Why will enterohepatic recycling occur and what is the importance of this recycling ?
Where are the products from this reaction usually excreted in ?

A

Glucuronidation ( = conjugation drugs and endogenous compounds with alpha-D-glucuronic acid to form glucuronides)
Quantitatively the most important phase II pathway for drugs and endogenous compounds
Products are often excreted via (in) the bile into the intestines.
Enterohepatic recycling may occur due to gut glucuronidases: Enterohepatic Recycling**: After excretion into bile, glucuronides can be hydrolyzed back into their parent compounds by gut bacteria β-glucuronidases, potentially reabsorbing them into circulation, thus prolonging their presence in the body.

Requires enzyme UDP-glucuronosyltransferase (UGT) which primarily carries out the glucuronidation process:
Genetic family of enzymes
Metabolizes a broad range of structurally diverse endogenous and exogenous compounds
Structurally related family with approximately 16 isoforms in man

37
Q

Glucuronidation requires a high energy intermediate. What is the name of this intermediate and how is it formed? What is its role in glucuronidation

A

UDP-Glucuronic Acid.

UDP-glucuronic acid is the activated form of glucuronic acid used in glucuronidation reactions. It serves as a high-energy intermediate that is essential for the conjugation of glucuronic acid to various substrates during glucuronidation.

How is it formed?
1. Formation of UDP-Glucuronic Acid:
- Glucuronic acid is derived from glucose metabolism in cells.
- To become active and ready for conjugation, glucuronic acid binds with uridine triphosphate (UTP), a molecule that provides energy for biochemical reactions.
- This binding forms UDP-glucuronic acid, which is a high-energy intermediate necessary for glucuronidation.

OR

UDP-glucuronic acid is synthesized from UDP-glucose, which is a nucleotide sugar formed from glucose and uridine triphosphate (UTP). The process involves several enzymatic steps:

  1. Formation of UDP-Glucose: Glucose undergoes phosphorylation by ATP (adenosine triphosphate) to form glucose-1-phosphate. Then, UDP-glucose pyrophosphorylase catalyzes the transfer of UDP (uridine diphosphate) from UTP to glucose-1-phosphate, forming UDP-glucose.
  2. Conversion to UDP-Glucuronic Acid: UDP-glucose is further converted to UDP-glucuronic acid through the action of UDP-glucose dehydrogenase. This enzyme catalyzes the oxidation of UDP-glucose to UDP-glucuronic acid.
  3. Importance of UDP-Glucuronic Acid:
    • UDP-glucuronic acid acts as a donor molecule during glucuronidation reactions.
    • Enzymes called UDP-glucuronosyltransferases (UGTs) use UDP-glucuronic acid to attach glucuronic acid to drugs, toxins, or other substances that need to be eliminated from the body.
    • This attachment (conjugation) modifies the substance, making it more water-soluble.
  4. Function in Detoxification and Elimination:
    • Glucuronidation converts lipid-soluble substances into water-soluble glucuronides.
    • These glucuronides can then be easily excreted from the body via urine or bile.
    • This process is crucial for detoxifying drugs, toxins, and endogenous compounds, reducing their activity and facilitating their safe removal from the body.

In essence, UDP-glucuronic acid is a crucial molecule that provides the glucuronic acid needed for attaching to substances during glucuronidation. This conjugation process enhances the body’s ability to eliminate various compounds efficiently and protect against their potential harmful effects.

38
Q

Glucuronidation requires a high energy intermediate. What is the name of this intermediate and how is it formed? What is its role in glucuronidation
How does it make drugs more soluble and what’s the name of the enzyme that aids it

A

UDP-Glucuronic Acid.

UDP-glucuronic acid is the activated form of glucuronic acid used in glucuronidation reactions. It serves as a high-energy intermediate that is essential for the conjugation of glucuronic acid to various substrates during glucuronidation.

How is it formed?
1. Formation of UDP-Glucuronic Acid:
- Glucuronic acid is derived from glucose metabolism in cells.
- To become active and ready for conjugation, glucuronic acid binds with uridine triphosphate (UTP), a molecule that provides energy for biochemical reactions.
- This binding forms UDP-glucuronic acid, which is a high-energy intermediate necessary for glucuronidation.

OR

UDP-glucuronic acid is synthesized from UDP-glucose, which is a nucleotide sugar formed from glucose and uridine triphosphate (UTP). The process involves several enzymatic steps:

  1. Formation of UDP-Glucose: Glucose undergoes phosphorylation by ATP (adenosine triphosphate) to form glucose-1-phosphate. Then, UDP-glucose pyrophosphorylase catalyzes the transfer of UDP (uridine diphosphate) from UTP to glucose-1-phosphate, forming UDP-glucose.
  2. Conversion to UDP-Glucuronic Acid: UDP-glucose is further converted to UDP-glucuronic acid through the action of UDP-glucose dehydrogenase. This enzyme catalyzes the oxidation of UDP-glucose to UDP-glucuronic acid.
  3. Importance of UDP-Glucuronic Acid:
    • UDP-glucuronic acid acts as a donor molecule during glucuronidation reactions.
    • Enzymes called UDP-glucuronosyltransferases (UGTs) use UDP-glucuronic acid to attach glucuronic acid to drugs, toxins, or other substances that need to be eliminated from the body.
    • This attachment (conjugation) modifies the substance, making it more water-soluble.
  4. Function in Detoxification and Elimination:
    • Glucuronidation converts lipid-soluble substances into water-soluble glucuronides.
    • These glucuronides can then be easily excreted from the body via urine or bile.
    • This process is crucial for detoxifying drugs, toxins, and endogenous compounds, reducing their activity and facilitating their safe removal from the body.

In essence, UDP-glucuronic acid is a crucial molecule that provides the glucuronic acid needed for attaching to substances during glucuronidation. This conjugation process enhances the body’s ability to eliminate various compounds efficiently and protect against their potential harmful effects.

39
Q

Explain Glucuronidation Pathway and Enterohepatic Recirculation

A

Glucuronidation is a crucial metabolic pathway involved in the detoxification and elimination of drugs and endogenous substances. Here’s how it relates to enterohepatic recirculation and the excretion routes:

  1. Conjugation Process: Glucuronidation involves the enzymatic attachment of glucuronic acid (derived from UDP-glucuronic acid) to a substrate molecule (e.g., drug or toxin) by UDP-glucuronosyltransferase enzymes (UGTs). This forms a glucuronide conjugate.
  2. Increased Water Solubility: The glucuronide conjugate is more water-soluble than the parent compound, which facilitates its excretion from the body.
  1. Bile Excretion: Glucuronides are often excreted into bile by the liver. From there, they enter the intestines.
  2. Intestinal Fate: In the intestines, glucuronides may encounter bacterial β-glucuronidases, which can hydrolyze the glucuronide bond, releasing the parent compound.
  3. Reabsorption: The released parent compound may be reabsorbed from the intestines back into the bloodstream (enterohepatic recirculation), thus prolonging its presence in the body.
  1. Urine: Glucuronides and their metabolites are primarily excreted in urine after being filtered by the kidneys. This route is significant for water-soluble metabolites.
  2. Feces: Some glucuronides that undergo hydrolysis in the intestines may be excreted in feces, particularly if they are not reabsorbed.

Glucuronidation enhances the water solubility of substances, aiding in their elimination through urine and bile. However, enterohepatic recirculation can complicate this process by potentially reabsorbing hydrolyzed metabolites from the intestines. This pathway is crucial for maintaining drug levels and metabolic balance in the body, impacting both drug efficacy and toxicity.

40
Q

What is N-glucuronidation?
Which compounds does it usually work with?

A

N-glucuronidation:
Occurs with amines (mainly aromatic )
Occurs with amides and sulfonamides

N-glucuronidation is a specific type of glucuronidation where the glucuronic acid moiety is conjugated directly to nitrogen atoms within molecules. This process occurs primarily with:

  1. Amines: Particularly aromatic amines, where the glucuronic acid attaches to the amino group (-NH2) on the aromatic ring structure.
  2. Amides and Sulfonamides: In these compounds, the glucuronic acid can attach to the nitrogen atom within the amide (-CONH-) or sulfonamide (-SO2NH-) functional groups.
  • Enzymatic Action: UDP-glucuronosyltransferase (UGT) enzymes catalyze the transfer of glucuronic acid from UDP-glucuronic acid to the nitrogen atom of the substrate molecule.
  • Detoxification: N-glucuronidation plays a role in detoxifying aromatic amines and other nitrogen-containing compounds, making them more water-soluble for excretion.
  • Metabolism: This pathway contributes to the metabolism and elimination of various drugs and endogenous compounds that contain amine, amide, or sulfonamide groups.
41
Q

What is O-glucuronidation
State two ways that it occurs

A

O-glucuronidation:
Occurs by ester linkages with carboxylic acids
Occurs by ether linkages with phenols and alcohols

O-glucuronidation is another specific type of glucuronidation where the glucuronic acid moiety is conjugated to oxygen atoms within molecules. This process occurs through two main types of linkages:

  1. Ester Linkages with Carboxylic Acids: In this type of O-glucuronidation, the glucuronic acid attaches to the hydroxyl group (-OH) of carboxylic acids, forming an ester linkage (-COO-).
  2. Ether Linkages with Phenols and Alcohols: Here, the glucuronic acid attaches to the hydroxyl group (-OH) of phenols (aromatic compounds) or alcohols (aliphatic compounds), forming an ether linkage (-O-).
  • Enzymatic Action: UDP-glucuronosyltransferase (UGT) enzymes catalyze the transfer of glucuronic acid from UDP-glucuronic acid to the oxygen atom of the substrate molecule.
  • Enhanced Water Solubility: O-glucuronidation increases the water solubility of compounds by introducing a hydrophilic glucuronic acid moiety, aiding in their excretion from the body.
  • Metabolism: This pathway is crucial for the metabolism and elimination of various drugs and endogenous compounds that contain hydroxyl groups susceptible to glucuronidation.

O-glucuronidation, along with N-glucuronidation and other conjugation pathways, plays a vital role in phase II metabolism, facilitating the detoxification and elimination of lipophilic (fat-soluble) substances that are otherwise difficult to excrete through urine or bile.

42
Q

Under conjugation reacrions or pathways, what are the two reactions that compete against each other and why?

A

Sulfation and glucuronidation are competing pathways:
Sulfation predominates at low substrate concentrations
Glucuronidation predominates at higher concentrations
There is relatively less PAPS (PAPS (3’-Phosphoadenosine-5’-phosphosulfate)) in cell cytosol compared to UDPGA(UDP-Glucuronic Acid:
)

Yes, sulfation occurs in the cytosol. Sulfotransferases, the enzymes responsible for sulfation, are located in the cytosol of cells. Glucuronidation primarily occurs in the endoplasmic reticulum of cells, not the cytosol. The enzymes responsible for this process, known as UDP-glucuronosyltransferases (UGTs), are located in the endoplasmic reticulum.

43
Q

What is sulfation
What is the name of the beefy rich donor it requires?
Sulfation is the major pathway for the metabolism of which compounds?

A

Sulfation:
Major pathway for phenols but also occurs for alcohols, amines and thiols
Energy rich donor required:(PAPS (3’-Phosphoadenosine-5’-phosphosulfate)

Sulfation, also known as sulfonation or sulfurylation, is a phase II metabolic pathway where a sulfate group (-OSO3H) is conjugated to a substrate molecule. This process involves the transfer of a sulfonate group from 3’-phosphoadenosine-5’-phosphosulfate (PAPS) to the substrate molecule, catalyzed by enzymes known as sulfotransferases (SULTs). Here are key aspects of sulfation:

  1. Substrate Specificity: Sulfation primarily targets hydroxyl (-OH) and amine (-NH2) groups on substrates, although other functional groups can also be sulfated.
    It is a conjugation reaction because it involves the covalent attachment of a sulfate group (-OSO3H) to substrate molecule.
  2. Endogenous and Exogenous Substances: Sulfation is involved in the metabolism of a wide range of compounds, including hormones (e.g., thyroid hormones), neurotransmitters (e.g., dopamine), drugs, and environmental toxins.
  3. Enhanced Water Solubility: Similar to glucuronidation, sulfation increases the water solubility of substrates, facilitating their excretion via urine.
  4. Regulation of Biological Activity: Sulfation can modify the biological activity of substrates. For example, sulfation of neurotransmitters like dopamine converts them into inactive forms, regulating their signaling in the nervous system.
44
Q

What is the enzyme that catalyzes sulfation ?
What types of this enzyme are non specific and which type is speciifc

A

Sulfotransferases (=SULTs) catalyze transfer of sulfate to substrates:
Phenol, alcohol and arylamine sulfotransferases are fairly non-specific
Steroid sulfotransferases are very specific

45
Q

What is acetylation under conjugation pathways
This pathway is a common reaction for which compounds?
What co factor does this pathway require?
What enzyme is responsible to catalyze this pathway?
Why is this reaction important in sulfonamide metabolism?
Which part of the body does this reaction mainly take place?

A

Acetylation:
Common reaction for aromatic amines and sulfonamides
Requires co-factor acetyl-CoA
Responsible enzyme is N-acetyltransferase
Takes place mainly in the liver
Important in sulfonamide metabolism because acetyl-sulfonamides are less soluble than the parent compound and may cause renal toxicity due to precipitation in the kidney

46
Q

What is fat acid conjugation and which part of the body does it occur in
Which drugs are metabolized by fat acid conjugation?
Do they have a long or short half life

A

Fatty Acid Conjugation:
Stearic and palmitic acids are conjugated to drug by esterification reaction.
Stearic acid is 18 carbons
Palmitic is 16 carbons.
So remember that Stearic acid is 16 plus 2.
Then palmitic acid is the actual 16.
• Stearic Acid: Stearic acid is the fatty acid in this reaction. It is a long-chain saturated fatty acid.
• Drug as the Alcohol: The drug (or its relevant functional group) acts as the alcohol in the esterification reaction. In this case, the drug would typically have a hydroxyl group (-OH) that reacts with the carboxyl group (-COOH) of stearic acid.

Esterification Reaction Overview:

•	Reaction: The esterification reaction involves the reaction between an alcohol (the drug in this case) and a fatty acid (stearic acid). The hydroxyl group of the alcohol reacts with the carboxyl group of the fatty acid to form an ester linkage and water.
•	Product: The product of this reaction is an ester, where the drug is esterified with stearic acid. This process can affect the drug’s solubility, distribution, and pharmacokinetics.

Summary:

•	Stearic Acid is the fatty acid.
•	The Drug (containing a hydroxyl group) is the alcohol.

This conjugation can influence drug properties, such as its release rate and absorption, by altering its chemical structure and solubility characteristics.

fat acid conjugation Occurs in liver microsomal fraction
(Cannabinols are metabolized in this fashion => long half-life)
Fatty acid conjugation refers to the process where fatty acids, such as stearic acid and palmitic acid, are conjugated to drugs through esterification reactions. This pathway typically occurs in the liver microsomal fraction, involving enzymes that facilitate the attachment of fatty acids to drug molecules. Here are key points about fatty acid conjugation:

  1. Mechanism:
    • Esterification: Fatty acids are attached to drugs via ester bonds. This modification can alter the physicochemical properties of the drug, affecting its solubility and metabolism.
  2. Liver Microsomes:
    • Liver microsomes contain enzymes capable of catalyzing esterification reactions, allowing for the conjugation of fatty acids to drugs.
  3. Metabolic Impact:
    • Cannabinoids, such as THC (tetrahydrocannabinol), can undergo fatty acid conjugation. This process is notable because it contributes to their lipophilicity, which can prolong their half-life in the body. Lipophilic compounds tend to be absorbed and distributed into fatty tissues, leading to a longer retention time.
  4. Biological Role:
    • Fatty acid conjugation plays a role in the metabolism and clearance of various drugs and xenobiotics. By modifying the chemical structure of drugs through fatty acid esterification, this pathway can influence their pharmacokinetics and pharmacodynamics.

In summary, fatty acid conjugation is a phase II metabolic pathway involving the esterification of fatty acids, such as stearic and palmitic acids, to drug molecules. This process, occurring predominantly in liver microsomes, affects the lipophilicity and metabolic fate of drugs, including cannabinoids, and can contribute to their extended presence in the body.

47
Q

How does acetylation occur?

A

Acetylation is a phase II conjugation pathway where an acetyl group (-COCH3) is transferred to a substrate molecule. This process is catalyzed by enzymes known as N-acetyltransferases (NATs). Here’s an overview of acetylation in drug metabolism and other biological processes:

  1. Enzymatic Action: N-acetyltransferases (NATs) catalyze the transfer of an acetyl group from acetyl-CoA (acetyl coenzyme A) to a substrate molecule. Acetyl-CoA serves as the acetyl group donor in this reaction.
  2. Substrate Specificity: Acetylation typically targets substrates containing amino (-NH2) groups, such as aromatic amines and hydrazines. It can also occur on hydroxyl (-OH) groups and occasionally on other functional groups.
  3. Formation of Acetylated Products: The addition of an acetyl group modifies the chemical structure of the substrate, often increasing its water solubility and facilitating its excretion from the body.
  • Detoxification: Acetylation plays a role in the detoxification of certain drugs, environmental toxins, and endogenous compounds by making them more polar and easier to eliminate.
  • Metabolism of Aromatic Amines: Acetylation is particularly important in the metabolism of aromatic amines, where it converts these potentially carcinogenic compounds into less toxic and more readily excreted forms.
  • Genetic Variation: The activity of NAT enzymes, particularly NAT2 (one of the NAT isoforms), varies among individuals due to genetic polymorphisms. This can affect the rate at which certain drugs and chemicals are acetylated and cleared from the body.
  • Isoniazid: A drug used to treat tuberculosis, which undergoes acetylation in the liver. The acetylated form is inactive and readily excreted in urine.
  • Sulfonamides: Some sulfonamide antibiotics are acetylated, altering their pharmacokinetics and enhancing their elimination.

In summary, acetylation is a phase II conjugation pathway that involves the enzymatic addition of an acetyl group to substrates, primarily affecting their solubility and metabolism in the body.

48
Q

Under conjugation pathways, what is amino acid conjugation?
What are the usual amino acids involved

A

Amino Acid Conjugation:
ATP-dependent acid:CoA ligase forms active CoA-amino acid conjugates which then react with drugs by N-Acetylation:
Usual amino acids involved are:
Glycine. Glutamine, Ornithine, Arginine

  1. Activation of Amino Acids:
    • Amino acid conjugation begins with the activation of amino acids (such as glycine, glutamine, ornithine, or arginine) by enzymes called ATP-dependent acid:CoA ligases.
    • These enzymes attach the amino acids to a molecule called coenzyme A (CoA), forming CoA-amino acid conjugates. This step requires ATP (adenosine triphosphate) as an energy source.
  2. Reaction with Drugs:
    • Once activated, these CoA-amino acid conjugates can then react with drugs or toxins in a process known as N-acetylation.
    • N-acetylation involves transferring the amino acid (now attached to CoA) to the drug molecule, typically at specific sites like amino (-NH2) or hydroxyl (-OH) groups on the drug.
  3. Purpose and Outcome:
    • The addition of amino acids to drugs makes them more water-soluble and less toxic.
    • These conjugated forms are easier for the body to eliminate through urine or bile, compared to their original, potentially more harmful forms.
49
Q

Under conjugation pathways, what is glutathione conjugation?
What is the importance of glutathione
Glutathione is a tripeptide made up of what three amino acids?

A

Glutathione Conjugation:
Tripeptide Gly-Cys-Glu; is conjugated by glutathione-S-transferase (GST)
Glutathione is a protective factor for removal of potentially toxic compounds
Conjugated compounds can subsequently be attacked by
g-glutamyltranspeptidase and a peptidase to yield the cysteine conjugate => product can be further acetylated to N-acetylcysteine conjugate

Glutathione Conjugation:

  1. Tripeptide: Glutathione is a small molecule composed of three amino acids: glycine, cysteine, and glutamate (Gly-Cys-Glu).
  2. Enzyme Involvement: Glutathione-S-transferase (GST) catalyzes the attachment of glutathione to drugs or toxins, forming glutathione conjugates.
  3. Protective Role: Glutathione conjugation is crucial for detoxifying and eliminating potentially harmful compounds. It protects cells by neutralizing toxins and aiding in their safe removal from the body.
  4. Further Processing: Once conjugated with glutathione, the compounds may undergo additional enzymatic reactions. For example, gamma-glutamyltransferase and peptidases can convert glutathione conjugates into cysteine conjugates, which may then be further metabolized, often ending in the formation of N-acetylcysteine conjugates.
50
Q

What are drug agonists?
State the two types

A

Agonists:
Can be drugs or endogenous ligands for the receptor
Increasing concentrations of the agonist will produce an increase in the biological response:
Full Agonist: Evokes 100% of the maximum possible effect
Partial Agonist: Produces the same type of biological response, but cannot achieve 100% even at very high doses

What are Agonists?

Agonists are substances (like drugs) that bind to a receptor and activate it to produce a biological response. There are two main types of agonists: full agonists and partial agonists.

Full Agonist

•	A full agonist binds to a receptor and fully activates it, producing the maximum possible effect that the receptor can achieve.
•	Think of a full agonist as a key that fits perfectly into a lock (the receptor) and opens it completely.
•	Example: Morphine is a full agonist at the opioid receptors. When morphine binds to these receptors, it fully activates them, resulting in strong pain relief.

Partial Agonist

•	A partial agonist also binds to the same receptor as a full agonist, but even when it binds, it cannot activate the receptor fully.
•	This means that a partial agonist produces a weaker response compared to a full agonist, even when you increase the dose.
•	Think of a partial agonist as a key that only partially opens the lock. It works, but not as well as a full agonist.
•	Example: Buprenorphine is a partial agonist at opioid receptors. It activates the receptors, but not as strongly as morphine does, which means it produces a limited effect (less pain relief compared to morphine).

Key Differences:

•	Full Agonist: Produces 100% of the receptor’s maximum response.
•	Partial Agonist: Produces a response, but never reaches 100%, no matter how much you give

Partial agonists are often used to provide safer, more controlled treatments, especially in situations where the full activation of a receptor could lead to serious side effects, overdose, or addiction

Partial agonists provide a ceiling effect for receptor activation, meaning they do not activate the receptor to its full capacity. This limits the maximum effect, reducing the risk of side effects or toxicity.
• For example, buprenorphine is a partial agonist at the opioid receptor. It provides pain relief and helps manage opioid dependence but is less likely to cause respiratory depression (a life-threatening side effect) compared to full agonists like morphine or fentanyl.

51
Q

What are drug antagonists
State the two types and explain them
Explain inverse agonists

A

Antagonists:
Block or reverse the effects of agonists. They have no effects on their own

Competitive Antagonists: Compete with agonist for receptor binding => Agonist appears less potent, but can still achieve 100% effect (but at higher concentrations). So on the graph, the two lines showed that an agonist at the 100% effect, the agonist plus the competitive antagonist will get the same 100% effect if the agonist concentration is increased cuz it will be able to overcome to competitive antagonist.
- Effect on Potency: Competitive antagonists reduce the potency of agonists, meaning higher concentrations of agonists are required to achieve the same effect.
- Effect on Efficacy: Despite reducing potency, competitive antagonists do not affect the maximal response that an agonist can achieve. At sufficiently high concentrations of agonist, competitive antagonists can be overcome, allowing the agonist to reach its full effect

Non-competitive Antagonists: Bind to receptor at different site and either prevent agonist binding or the agonist effect => maximal achievable response reduced
For this one,agonist plus non competitive antagonist reduces the effect as compared to the agonist alone. This is because the non competitive antagonist has taken over the receptors the agonist could’ve bound to. So no matter how much you increase the agonist concentration, it still won’t overcome the effect of the non competitive antagonist. Until the body produces new receptors. Binding Site and Mechanism:**
- Non-competitive antagonists bind to a site on the receptor molecule that is distinct from the agonist binding site. This alternate binding site may overlap with the orthosteric site (where agonists bind) or be allosteric (different from the original or primary binding site where the agonist binds)
- Binding of non-competitive antagonists induces a conformational change in the receptor that prevents agonists from binding effectively or inhibits the receptor’s ability to activate downstream signaling pathways.

  1. Effect on Agonist Response:
    • Efficacy Reduction: Non-competitive antagonists reduce the maximal response that an agonist can produce, regardless of the concentration of the agonist. This reduction occurs because the antagonist interferes with the receptor’s ability to activate signaling pathways, even when agonists are present.
    • No Effect on Agonist Potency: Unlike competitive antagonists, non-competitive antagonists do not affect the potency (EC50) of agonists. Agonists still bind to their receptors with the same affinity, but the resulting response is dampened or blocked by the antagonist’s action.

Mechanism of Action:**
- Non-competitive antagonists may act through several mechanisms:
- Direct Blockade: By physically hindering the conformational changes necessary for receptor activation.
- Indirect Modulation: By stabilizing a receptor conformation that cannot activate downstream signaling pathways.
- Allosteric Modulation: By binding to allosteric sites and altering receptor function indirectly.

The non competitive antagonist can be reversible or irreversible Some non-competitive antagonists bind transiently to receptors, allowing for recovery of receptor function once the antagonist dissociates.
- Irreversible Binding: Irreversible non-competitive antagonists form covalent bonds or otherwise stable interactions with receptors, resulting in prolonged or permanent receptor blockade until new receptors are synthesized.

Inverse Agonists: Not the same as antagonists! Inverse agonists trigger a negative response (= reduce baseline) (e.g. diazepam = full agonist = anticonvulsant BUT inverse agonists of benzodiazepin receptor are convulsants)

Certainly! Let’s delve into the concept of inverse agonists and how they differ from agonists and antagonists, using diazepam as an example:

Inverse agonists are ligands that bind to the same receptor site as agonists but induce an opposite pharmacological response. Unlike antagonists, which simply block the activity of agonists without inducing any effect of their own, inverse agonists actively reduce the constitutive (baseline) activity of receptors.

  1. Diazepam as an Agonist:
    • Diazepam is a benzodiazepine that acts as a full agonist at GABA-A receptors in the brain.
    • As an agonist, diazepam enhances the effect of the neurotransmitter gamma-aminobutyric acid (GABA) at these receptors.
    • This leads to increased chloride ion influx into neurons, resulting in neuronal hyperpolarization and suppression of neuronal excitability.
    • Clinically, diazepam is used as an anxiolytic (anti-anxiety), muscle relaxant, and anticonvulsant due to its ability to enhance GABAergic inhibition.
  2. Inverse Agonists of Benzodiazepine Receptors:
    • Despite diazepam being a full agonist at benzodiazepine receptors, some ligands act as inverse agonists at these same receptors.
    • Inverse agonists at benzodiazepine receptors induce a pharmacological response opposite to that of diazepam.
    • Instead of enhancing GABAergic inhibition, these inverse agonists reduce the baseline activity of the receptor. This can lead to a net increase in neuronal excitability, potentially manifesting as convulsive activity or exacerbation of anxiety symptoms.
  • Mechanism: Diazepam as an agonist enhances receptor activity and promotes its intended therapeutic effects by enhancing GABAergic neurotransmission.
  • Inverse Agonists: These compounds reduce the constitutive activity of the receptor, leading to effects opposite to those seen with agonists like diazepam.
  • Understanding the distinction between agonists, antagonists, and inverse agonists is crucial in pharmacology and drug development.
  • Diazepam illustrates how a compound can act as a beneficial agonist in one context (e.g., anticonvulsant) while inverse agonists at the same receptor can lead to unintended effects (e.g., convulsant activity).

In summary, while diazepam functions as a GABA-A receptor agonist to exert its therapeutic effects, inverse agonists at the benzodiazepine receptor site can induce effects contrary to those of diazepam, highlighting the importance of receptor modulation specificity in pharmacological treatments.

Sure, here’s another example that might make it clearer:

  • Agonist Effect: Histamine is a chemical in your body that causes itchiness and swelling when it attaches to its receptors. Imagine it’s like a key that makes your skin itchy when it fits into its lock (the receptor).
  • Inverse Agonist Effect: Now, imagine there’s a different key that fits into the same lock as histamine but instead of making your skin itchy, it actually makes the itchiness go away or reduces swelling. This key would be an inverse agonist because it does the opposite of what histamine normally does.

So, while histamine causes itchiness and swelling (acting like a normal key), an inverse agonist at the histamine receptor would do the opposite and relieve itchiness or reduce swelling. It’s like having keys that either cause a problem, solve it, or do nothing.

52
Q

Phase I and II - Summary:

Products are generally more water soluble
These reactions products are ready for (renal) excretion
There are many complementary, sequential and competing pathways: Phase I and Phase II reactions encompass multiple pathways that can be complementary (enhancing metabolism), sequential (one reaction preparing for the next), or competing (different pathways competing for the same substrate).
Phase I and Phase II metabolism are a coupled interactive system interfacing with endogenous metabolic pathways
True or false

A
  1. Interactive System: Phase I and Phase II metabolism interact with each other and with endogenous metabolic pathways. Phase I reactions often prepare substrates for Phase II conjugation, and Phase II reactions convert Phase I metabolites into more readily excretable forms.
53
Q

Difference between UDP AND ALPHA D GLUCURONIC ACID

A

Yes, I understand the confusion. Here’s a clearer explanation of how these components relate to each other in the glucuronidation process:

  1. UDP-Glucuronic Acid (UDPGA): This is the active form of glucuronic acid in the body. It serves as the donor of the glucuronic acid group during the conjugation reaction.
  2. Alpha-D-Glucuronic Acid: This is the form of glucuronic acid that gets attached to the drug or substrate during the glucuronidation process. It is not directly added to the drug; instead, it is part of UDPGA, which donates the glucuronic acid group.
  3. Glucuronidation Reaction:
    • In this process, UDP-glucuronic acid (UDPGA) provides the glucuronic acid group.
    • The enzyme UDP-glucuronosyltransferase (UGT) facilitates the transfer of the glucuronic acid from UDPGA to the drug or substrate.
    • The result is the formation of a glucuronide conjugate, which is a compound where the glucuronic acid is attached to the drug or substrate.

Summary: UDP-glucuronic acid (UDPGA) contains alpha-D-glucuronic acid, which is transferred to the drug during the glucuronidation process. The drug then forms a glucuronide conjugate, making it more water-soluble and easier for the body to excrete.