week 21 p2 Flashcards

1
Q

Why do drugs
(or any other chemical)
cause toxicity? Reason 1: ADME-driven toxicity

A
  • Chemical can disrupt ADME processes
    • (e.g. disruption of phase I and II enzymes).
    • I this disruption alters the pharmacokinetic profile of the chemical
    • (e.g. leading to a higher-than usual concentration in the body, and therefore increasing the risk of toxicity).
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Effect of drugs on CYP450s (a example ofADME-driven

A
  • CYP450s can be inhibited or induced by chemicals. Inhibition of CYPs can lead to unwanted drug–drug interactions due to the resulting large variations of drug concentrations between patients.
    • The poor metabolization of the drug can result in higher-then-usual concentrations, increasing the risk trigger toxicological effects.
    • CYP450-mediated effects are particularly important to determine the risk of drug combinations (and cocktails of non-pharma chemicals
    • CYP enzymes can increase ROS and alter the redox balance, creating oxidative stress, through their catalytic cycle and contribute to disease development.
    • However, some substrates, modified by CYP enzymes to create reactive intermediates or products, can also contribute to disease development. Such as hyperopic lung injury, ROS mediated hepatotxity( veith, et al, 2017)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Effects of drugs that causes ADME-driven (Example: Statins + CYP3A4 Inhibitors)

A

• Statins- lower the level of low-density lipoprotein (LDL) cholesterol in the blood.
High-dose statin monotherapy can lead to rhabdomyolysis (risk:1/10,000 people)

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

What can toxicity of CYP3A54 inhibitor and statins cause

A

• Rhabdomyolysis is a serious syndrome due to a direct or indirect muscle injury.
• It results from the death of muscle fibres and release of their contents into the bloodstream.
This can lead to serious complications such as renal failure.

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

Issues with Example: Statins + CYP3A4 Inhibitors

A

• Some statins are particularly susceptible to drug-drug interaction dynamics (e.g. simvastatin and lovastatin).
• The risk of rhabdomyolysis is increased with concomitant use of certain drugs
Potent inhibitors of CYP34A can significantly increase the serum concentrations of the active forms of simvastatin, lovastatin, and atorvastatin.

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

What are the drugs most likely to interact with statins include:

A

• fibrates (especially gemfibrozil)
• azole antifungal agents,
• Amiodarone
• macrolides (especially erythromycin and clarithromycin, but not azithromycin)
• protease inhibitors
calcium-channel blockers (especially verapamil and diltiazem)

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

Effects of drugs that causes ADME-drive Example: Omeprazol + Clopidogrel

A

• Omeprazole – Proton pump inhibitor (e.g. used in the treatment of gastroesophageal reflux disease, peptic ulcer disease, to prevent upper gastrointestinal bleeding in people who are at high risk)
• Omeprazole, a CYP2C19 inhibitor, decreases the antiplatelet activity of clopidogrel by inhibiting the biotransformation of the clopidogrel pro drug into its active metabolite.
• Clopidogrel - is an antiplatelet medication used to reduce the risk of heart disease and stroke in those at high risk.
CYP1A2 and CYP2B6, then by CYP2C19, CYP2C9, CYP2B6 and CYP3A. Clopidogrel is a prodrug, which is activated in two steps, first by CYP2C19

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

Another example of ADME-mediated toxicity: is disruption of drug distribution

A

• Two drugs may compete for the same albumin binding site.
• According to their affinity to the binding site, one drug may displace the other causing an increase in the free concentration of the displaced drug.
This increase in free concentration may increase the risk of developing adverse effects, as it results in an increase of drug concentration at the target site.

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

example of ADME-mediated toxicity: is disruption of drug distribution

A

• the administration of aspirin to a patient treated chronically with warfarin results in displacement of the latter from its binding site.
the increase in plasma concentration of free warfarin causes the development of serious haemorrhagic reactions.

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

Another reason that causes toxity is Reason 2: Bioactivation

A

• The metabolic transformation of the chemical leads to a novel chemical species that is toxic.- bioactivation.
most important metabolic systems involved in bioactivation reactions are CYP450 and peroxidase enzymes, producing either electrophilic or radical metabolites.

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

The process of Bioactivation

A

• Xenobiotic , nontoxic metabolite, reactive metabolites
• Bids to cellular binding
Results in toxity and cellular repair

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

What is reactive metabolites: Electrophiles

A

Reactive metabolites are usually electron deficient molecules and are referred to as electrophiles (molecules containing positive centres).
They are typically either positively charged or have a partial positive charge (δ+).

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

What is the process of reactive metabolites : Electrophiles

A

• If not detoxified properly, electrophiles can react with electron rich species, i.e. nucleophiles (molecules containing negative centres),
• through covalent bond formation.
• The nucleophiles usually contain atoms such as S, N, or O that have a lone pair of electrons,
• which can form a new bond to the electrophile.
Such nucleophiles are present on macromolecules such as proteins, nucleic acids and lipids.

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

How does electrophiles work

A

• Electrophiles act as acceptors of electrons in a reaction.
• By accepting some electrons from nucleophiles, electrophiles will “quench” their positive or partial positive charge
making a more overall stable species.

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

How Effects of reactive metabolites: Electrophiles

A
  • Chemically reactive metabolites can directly react with proteins
  • causing changes in protein structure or protein folding.
  • These modified proteins are processed by antigen presenting cells
  • and can look “foreign” to the immune system leading to an immune response.
  • Chemically reactive electrophiles can also covalently react with nucleic acids on the DNA
  • thereby causing changes in DNA structure or gene expression.
  • Changes in DNA can lead to mutagenicity, teratogenicity or carcinogenicity.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What is Types of reactive metabolites : Free radicals

A

Free radical refers to compounds having an unpaired electron

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

How does free radicals happen

A

• Since electrons need to be paired to form a chemical bond, a free radical cannot react covalently with nucleophiles. Rather they:
• react with another free radical to form a covalent bond
• abstract a hydrogen atom from a neutral molecule to generate a new radical
abstract an electron to form an anion and generate a radical cation

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

What are the biological effects of reactive metabolites

A

• Toxicity may accrue through formation of a chemically reactive metabolites, which, if not detoxified,
• can cause the modification of biological macromolecules.
As a result of their high reactivity, reactive metabolites are often considered to be short-lived. This is not always true, however, because reactive intermediates can be transported from one tissue to another, where they may exert their deleterious effects.(Attia 2010)

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

What is Effects of reactive metabolites: quinones

A

• Cause different toxicological effects in vivo including acute cytotoxicity, immunotoxicity, genotoxicity and carcinogenesis.
• Can cause cellular damage through alkylation of crucial cellular proteins and/or DNA.
• Quinones are also highly redox active molecules, which can redox cycle with their semiquinone radicals leading to formation of ROS including superoxide, hydrogen peroxide, and ultimately the hydroxyl radical.
Production of ROS can cause severe oxidative stress within cells through the formation of oxidized cellular macromolecules, including lipids, proteins, and DNA .

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

What is the examples of toxicity involving quinones

A

• Benzene-induced leukemia

Remoxipride-induced aplastic anemia

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

What Is the consideration of paracentamol

A

• can trigger severe liver toxicity in human patients at high doses
• The major portion of paracetamol is conjugated with either sulfate or glucuronic acid to form water-soluble metabolites
• and only small amounts of the reactive intermediate, believed to be N-acetyl-p-benzoquinonimine (NAPQI), are formed by the cytochrome P450 enzymes.
• When therapeutic doses of paracetamol are ingested, the small amount of reactive intermediate forms is efficiently deactivated by conjugation with the antioxidant glutathione (GSH).
What happens where there is large amount of paracetamol ingested
• the sulphate and glucuronide cofactors (PAPS and UDPGA) become depleted, resulting in more of the paracetamol being metabolized to the reactive intermediate.
• As long as GSH is available, most of the reactive intermediate can be detoxified. When the concentration of GSH in the liver also becomes depleted,
• NAPQI covalently bind to sulfhydryl (-SH) groups of various cellular proteins increases, resulting in hepatic necrosis.
• If sufficiently large amounts of paracetamol are ingested, as in drug overdoses and suicide attempts, extensive liver damage and death may result.

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

How does effects toxicity Reason 3: non-specific toxic responses

A
  • Chemicals can induce non-specific toxic responses (i.e. non-mediated by specific drug-target interactions)
  • Examples: Oxidative stress, Activation of cellular stress responses , DNA damage
  • Apoptosis, Necrosis ,Immune or allergic response
23
Q

How does Oxidative stress happen

A

Chemically reactive molecules containing oxygen are termed reactive oxygen species (ROS).
Reactivity may be due to the presence of unpaired electrons, but there are also reactive nonradical species such as hydrogen peroxide (H2O2).
ROS are generated during the normal metabolism of oxygen via diverse enzymatic pathways.

24
Q

Why can drug induce oxidative stress?

A

• During the course of the CYP reaction mechanism, there are various steps that can generate ROS.
• Substrate-p450 reaction cycle- ROS-protein modifications, lip peroxisation, oxidation DNA dmage
Toxic effect

25
Q

How does oxidative stress occur vy drugs

A

• When mitochondrial DNA is the target of oxidation, it can lead to mutations, rearrangements, and transcriptional errors that impair important mitochondrial components, leading to more oxidative stress and eventual cell death.
Molecular modifications in surviving cells can cause alterations in gene expression, and, depending on the severity and duration of ROS exposure, prosurvival or proapoptotic response pathways may be activated.

26
Q

What can ROS Lead to DNA damage

A

Oxidation of DNA leads to the formation of lesions including:
• oxidized bases (purines and pyrimidines)
• abasi sites (also called apurinic/apyrimidinic (AP) sites)
DNA single- and/or double-strand breaks

27
Q

What effecrs occurs with DNA damage caused by ROS

A

• Guanine is the most susceptible DNA base because of its low oxidation potential, and there are multiple oxidized guanine products .
• Two of the most common modifications are 8-oxo-7,8-dihydroguanine (8-oxoGua) and 2,6-diamino-4-hydroxy-5-formamidopyrimidine (FapyGua).
• Of all the DNA oxidation products, 8-oxoGua is the most abundant, stable, and well studied and therefore often used as a biomarker of oxidative stress.
It is a strongly promutagenic lesion, since it promotes the mismatched incorporation of dATP instead of dCTP opposite the lesion during replication, inducing a GC to TA transversion.

28
Q

What does Mutagenesis/Carcinogenesis/Teratogenesis mean

A

• Mutagen: an agent that can induce or increase the frequency of mutation in an organism.
• Teratogen: any agent that interferes with normal embryonic development.
Carcinogen: any substance or agent that tends to produce a cancer.

29
Q

What does Drug hypersensitivity

A

• Drug hypersensitivity reactions (DHR) include allergic, exaggerated pharmacologic, and pseudo-allergic reactions
to medications that result from an enhanced immunologic or inflammatory response.

30
Q

What is Reason 4: target-mediated toxicity (drug that causes toxicity)

A

• Chemicals can interact with specific biological targets (e.g. receptors, enzymes, ion channels).
Such interaction causes the alteration of normal biological pathways leading to the disruption of the system. This can occur at any exposure level.

31
Q

What is the example of drug development for Target-mediated toxicity

A

• Pharmaceuticals companies use a range of computational and in vitro assays to predict
whether a new drug can interact with targets that are known to be associated with serious adverse effects.
Eg GCPRs, ion channels, enzymes

32
Q

Examples of safety targets - hERG

A

• hERG (the human Ether-à-go-go-Related Gene) is a gene (KCNH2) that codes for a protein known as Kv11.1,
• the alpha subunit of a potassium ion channel.
• This ion channel is essential to regulate the electrical activity of the heart
Has good aspects and bad too

33
Q

How does hERG work and show does it cause toxic

A

• hERG channel mediates the repolarizing IKr current in the cardiac action potential, which helps coordinate the heart’s beating
• hERG inhibition can result in a potentially fatal disorder called long QT syndrome
A number of drugs have the tendency to inhibit hERG, lengthening the QT and potentially leading to a fatal irregularity of the heartbeat

34
Q

What is hERG safety margin

A
  • > 30 fold between efficacious free Cmax in patients and hERG in vitro IC50
  • > 20-fold between efficacious free Cmax in patients and in vivo EC10 (e.g. for QT prolongation)
35
Q

Linking chemical targets to toxic effects

A

• Adverse Outcome Pathway
• concept proposed by the OECD to harmonise the description of toxic responses is the Adverse Outcome Pathway (AOP).
An AOP is an analytical construct that describes a sequential chain of causally linked events at different levels of biological organisation that lead to an adverse health effect.

36
Q

Example of AOP is Cardiotoxicity mediated by the blockade of L-type calcium channel

A

LTCC-blockade leading to heart failure via the disruption of cardiac contractility
• When this blockage occurs, calcium current , binding to tropin C, contractile response and ejection fraction decrease servery causing heart failure

37
Q

What is Adverse drug reaction

A

a response to a drug that is noxious, unintended and occurs at doses normally used in man for the prophylaxis, diagnosis, or therapy of disease or for modification of the physiological function

38
Q

what are the types of Adverse drug reaction

A

• Type A reaction-
be detected in animal models of pharmacology and/or toxicology; they exhibit simple dose-response relationships and are usually avoided in the clinic via dose adjustments.
• Type B-are unrelated to known drug pharmacology, and although they are dose dependent in susceptible individuals, they do not occur at any dose in most patients.

39
Q

What can Type B reaction effects

A

• can affect any organ, but the most common organs involved are liver, skin, and bone marrow.
The life-threatening type B adverse drug reactions include:Hepatotoxicity
• ,severe cutaneous reactions, aplastic anaemia ,blood dyscrasias
• Amongst these, hepatotoxicity is the most frequent reason for drug withdrawal and is also the major cause of attrition in drug discovery/development.
(Attia, 201)

40
Q

What is Primary pharmacodynamics

A

identifies the desired properties of a molecule

41
Q

Whast is Secondary pharmacodynamics

A

identifies other properties of a molecule which are of no particular importance for safety

42
Q

What does safety pharmcology involve a combination of (usually) sequential tests

A

in silico - performed on computer or via computer simulation (eg. Prediction of drug-enzyme interaction; PK-PD modelling)
in vitro – performed or taking place in a test tube, culture dish, or elsewhere outside a living organism (eg. micronucleus test)
in vivo - performed or taking place in a living organism (eg mouse)

43
Q

How do we assess potential toxicity during drug development?

A
  • To identify undesirable pharmacodynamic properties
  • To evaluate adverse pharmacodynamic and/or pathophysiological effectso
  • To investigate the mechanism of the adverse pharmacodynamic effects observed and/or suspected
44
Q

What are the Three main categories of studies

A

Core battery studies- Investigate the effects of the test substance on vital functions (i.e. cardiovascular

  1. Follow-up studies-Provide greater depth of understanding than, or additional knowledge to, that provided by the core battery on vital functions
  2. Supplemental studies- Evaluate potential adverse pharmacodynamic effects on organ system function not addressed by the core battery or repeated dose toxicity studies
45
Q

What is typically measured as the Core Battery?

A
  • -CNS-motor activity, behavioural changes, coordination, sensory/motor reflex responses, body temperature
  • Cardiovascular system -blood pressure, heart rate, ECG; In vivo, in vitro and/or ex vivo methods incl. methods for repolarization and conductance abnormalities should also be considered.
  • RS-respiratory rate and tidal volume or haemoglobin oxygen saturation.
46
Q

What is typically done as the Core Battery?

A

• Example of in vivo test Irwin test -CNS
• Vitro hERG and vivo -CS
Vivo using plethyography- RS

47
Q

Example of in vivo test: Repeated Dose 90-day Oral Toxicity Study in Rodents (in vivo)

A
  • Orally administrated in gradual doses in several groups for 90 days
  • Animal are observed using OECD(principles regulations)
  • The animals doses are for 7 days every week for 90 days
  • Administrated by a gavage cannula
  • Maximum volume of liquid is on times
48
Q

What is the procedures for vitro studies

A
  • A chemical series has been identified and activity at the primary therapeutic target has been confirmed, secondary profiling can be performed to assess its promiscuity
  • Second, these data can then be used to develop a lead optimization plan, which can include analysis of the SAR at off-targets in order to influence chemical design.
  • Third, at the end of lead optimization, the data can be used to select the candidate drug from a shortlist.
  • the value of the data is enhanced when interpreted in the context of the predicted therapeutic free plasma concentration, (Gintant et al 2016)qsar
49
Q

Wht is QSAR models

A
  • Structure-activity relationship (SAR) and quantitative structure-activity relationship (QSAR) models -
  • are mathematical models that can be used to predict the physicochemical, biological and environmental fate properties of compounds from the knowledge of their chemical structure.
  • These models are available for free or as commercial software’s.
  • use of (Q)SARs has deliver reliable information that is comparable to and sufficient to fulfil the information requirements.
50
Q

What is Example of in vitro test - In vitro hERG assay

A

• Many drugs have been shown to block hERG channel.
• This blockade can delay repolarization of the cardiac action potential and lead to prolongation of the QT interval on the electrocardiogram.
• This can potentially initiate the arrhythmia known as Torsades de Pointes (TdP) with fatal consequences.
• In vitro hERG have become standard components in cardiac safety evaluation during nonclinical drug development.

51
Q

what is the Integrated risk assessment

A

• In vitro IC50 of your safety target (e.g. hERG)
• In vivo EC50 (or any other reference value, e.g. EC10) of you safety endpoin (e.g. QT prolongation)
Human Cmax

52
Q

What is ToxCast: Toxicity Forecaster

A

• Mechanistic screenings
• Using a high-throughput robotic screening system,
• testing 10,000 environmental chemicals for their potential to disrupt biological pathways that may result in toxicit

53
Q

What is ChEMBL

A

• a manually curated database of bioactive molecules with drug-like properties.
• It brings together chemical, bioactivity and genomic data
to aid the translation of genomic information into effective new drugs